Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell ; 173(5): 1165-1178.e20, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29706548

ABSTRACT

Cohesin extrusion is thought to play a central role in establishing the architecture of mammalian genomes. However, extrusion has not been visualized in vivo, and thus, its functional impact and energetics are unknown. Using ultra-deep Hi-C, we show that loop domains form by a process that requires cohesin ATPases. Once formed, however, loops and compartments are maintained for hours without energy input. Strikingly, without ATP, we observe the emergence of hundreds of CTCF-independent loops that link regulatory DNA. We also identify architectural "stripes," where a loop anchor interacts with entire domains at high frequency. Stripes often tether super-enhancers to cognate promoters, and in B cells, they facilitate Igh transcription and recombination. Stripe anchors represent major hotspots for topoisomerase-mediated lesions, which promote chromosomal translocations and cancer. In plasmacytomas, stripes can deregulate Igh-translocated oncogenes. We propose that higher organisms have coopted cohesin extrusion to enhance transcription and recombination, with implications for tumor development.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Genome , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomes/metabolism , DNA-Binding Proteins , Humans , Mice , Mutagenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Cohesins
2.
Nat Immunol ; 19(12): 1366-1378, 2018 12.
Article in English | MEDLINE | ID: mdl-30420627

ABSTRACT

Thymocyte development requires a complex orchestration of multiple transcription factors. Ablating either TCF-1 or HEB in CD4+CD8+ thymocytes elicits similar developmental outcomes including increased proliferation, decreased survival, and fewer late Tcra rearrangements. Here, we provide a mechanistic explanation for these similarities by showing that TCF-1 and HEB share ~7,000 DNA-binding sites genome wide and promote chromatin accessibility. The binding of both TCF-1 and HEB was required at these shared sites for epigenetic and transcriptional gene regulation. Binding of TCF-1 and HEB to their conserved motifs in the enhancer regions of genes associated with T cell differentiation promoted their expression. Binding to sites lacking conserved motifs in the promoter regions of cell-cycle-associated genes limited proliferation. TCF-1 displaced nucleosomes, allowing for chromatin accessibility. Importantly, TCF-1 inhibited Notch signaling and consequently protected HEB from Notch-mediated proteasomal degradation. Thus, TCF-1 shifts nucleosomes and safeguards HEB, thereby enabling their cooperation in establishing the epigenetic and transcription profiles of CD4+CD8+ thymocytes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Gene Expression Regulation/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Lymphopoiesis/immunology , Thymocytes/immunology , Animals , CD4 Antigens/immunology , CD8 Antigens/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Cell ; 159(7): 1524-37, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25483777

ABSTRACT

The antibody gene mutator activation-induced cytidine deaminase (AID) promiscuously damages oncogenes, leading to chromosomal translocations and tumorigenesis. Why nonimmunoglobulin loci are susceptible to AID activity is unknown. Here, we study AID-mediated lesions in the context of nuclear architecture and the B cell regulome. We show that AID targets are not randomly distributed across the genome but are predominantly grouped within super-enhancers and regulatory clusters. Unexpectedly, in these domains, AID deaminates active promoters and eRNA(+) enhancers interconnected in some instances over megabases of linear chromatin. Using genome editing, we demonstrate that 3D-linked targets cooperate to recruit AID-mediated breaks. Furthermore, a comparison of hypermutation in mouse B cells, AID-induced kataegis in human lymphomas, and translocations in MEFs reveals that AID damages different genes in different cell types. Yet, in all cases, the targets are predominantly associated with topological complex, highly transcribed super-enhancers, demonstrating that these compartments are key mediators of AID recruitment.


Subject(s)
B-Lymphocytes/metabolism , Carcinogenesis , Cytidine Deaminase/genetics , Enhancer Elements, Genetic , Animals , DNA Damage , Humans , Lymphoma/metabolism , Mice
5.
Mol Cell ; 72(2): 222-238.e11, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30293786

ABSTRACT

DNA polymerase stalling activates the ATR checkpoint kinase, which in turn suppresses fork collapse and breakage. Herein, we describe use of ATR inhibition (ATRi) as a means to identify genomic sites of problematic DNA replication in murine and human cells. Over 500 high-resolution ATR-dependent sites were ascertained using two distinct methods: replication protein A (RPA)-chromatin immunoprecipitation (ChIP) and breaks identified by TdT labeling (BrITL). The genomic feature most strongly associated with ATR dependence was repetitive DNA that exhibited high structure-forming potential. Repeats most reliant on ATR for stability included structure-forming microsatellites, inverted retroelement repeats, and quasi-palindromic AT-rich repeats. Notably, these distinct categories of repeats differed in the structures they formed and their ability to stimulate RPA accumulation and breakage, implying that the causes and character of replication fork collapse under ATR inhibition can vary in a DNA-structure-specific manner. Collectively, these studies identify key sources of endogenous replication stress that rely on ATR for stability.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , DNA Replication/genetics , Microsatellite Repeats/genetics , Animals , Cell Cycle Proteins/genetics , Chromatin/genetics , Chromatin Immunoprecipitation/methods , DNA Breaks, Double-Stranded , DNA Damage/genetics , Female , Genomic Instability/genetics , Humans , Mice , Replication Protein A/genetics
6.
Mol Cell ; 67(4): 566-578.e10, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28803781

ABSTRACT

50 years ago, Vincent Allfrey and colleagues discovered that lymphocyte activation triggers massive acetylation of chromatin. However, the molecular mechanisms driving epigenetic accessibility are still unknown. We here show that stimulated lymphocytes decondense chromatin by three differentially regulated steps. First, chromatin is repositioned away from the nuclear periphery in response to global acetylation. Second, histone nanodomain clusters decompact into mononucleosome fibers through a mechanism that requires Myc and continual energy input. Single-molecule imaging shows that this step lowers transcription factor residence time and non-specific collisions during sampling for DNA targets. Third, chromatin interactions shift from long range to predominantly short range, and CTCF-mediated loops and contact domains double in numbers. This architectural change facilitates cognate promoter-enhancer contacts and also requires Myc and continual ATP production. Our results thus define the nature and transcriptional impact of chromatin decondensation and reveal an unexpected role for Myc in the establishment of nuclear topology in mammalian cells.


Subject(s)
B-Lymphocytes/metabolism , Cell Cycle , Cell Nucleus/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , Histones/metabolism , Lymphocyte Activation , Proto-Oncogene Proteins c-myc/metabolism , Acetyl Coenzyme A/metabolism , Acetylation , Adenosine Triphosphate/metabolism , Animals , B-Lymphocytes/immunology , Cell Line , Chromatin/chemistry , Chromatin/genetics , DNA Methylation , Epigenesis, Genetic , Genotype , Histones/chemistry , Immunity, Humoral , Methylation , Mice, Inbred C57BL , Mice, Knockout , Nucleic Acid Conformation , Phenotype , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/genetics , Single Molecule Imaging , Structure-Activity Relationship , Time Factors , Transcription, Genetic
7.
Proc Natl Acad Sci U S A ; 119(32): e2201493119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35921443

ABSTRACT

Understanding the mechanisms promoting chromosomal translocations of the rearranging receptor loci in leukemia and lymphoma remains incomplete. Here we show that leukemias induced by aberrant activation of ß-catenin in thymocytes, which bear recurrent Tcra/Myc-Pvt1 translocations, depend on Tcf-1. The DNA double strand breaks (DSBs) in the Tcra site of the translocation are Rag-generated, whereas the Myc-Pvt1 DSBs are not. Aberrantly activated ß-catenin redirects Tcf-1 binding to novel DNA sites to alter chromatin accessibility and down-regulate genome-stability pathways. Impaired homologous recombination (HR) DNA repair and replication checkpoints lead to retention of DSBs that promote translocations and transformation of double-positive (DP) thymocytes. The resulting lymphomas, which resemble human T cell acute lymphoblastic leukemia (T-ALL), are sensitive to PARP inhibitors (PARPis). Our findings indicate that aberrant ß-catenin signaling contributes to translocations in thymocytes by guiding Tcf-1 to promote the generation and retention of replication-induced DSBs allowing their coexistence with Rag-generated DSBs. Thus, PARPis could offer therapeutic options in hematologic malignancies with active Wnt/ß-catenin signaling.


Subject(s)
Cell Transformation, Neoplastic , Genomic Instability , Hepatocyte Nuclear Factor 1-alpha , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Thymocytes , Translocation, Genetic , beta Catenin , Animals , Cell Transformation, Neoplastic/genetics , DNA Breaks, Double-Stranded , Genomic Instability/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , Thymocytes/pathology , Translocation, Genetic/genetics , beta Catenin/genetics , beta Catenin/metabolism
8.
Nat Immunol ; 13(1): 86-94, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22080921

ABSTRACT

Cell fate depends on the interplay between chromatin regulators and transcription factors. Here we show that activity of the Mi-2ß nucleosome-remodeling and histone-deacetylase (NuRD) complex was controlled by the Ikaros family of lymphoid lineage-determining proteins. Ikaros, an integral component of the NuRD complex in lymphocytes, tethered this complex to active genes encoding molecules involved in lymphoid differentiation. Loss of Ikaros DNA-binding activity caused a local increase in chromatin remodeling and histone deacetylation and suppression of lymphoid cell-specific gene expression. Without Ikaros, the NuRD complex also redistributed to transcriptionally poised genes that were not targets of Ikaros (encoding molecules involved in proliferation and metabolism), which induced their reactivation. Thus, release of NuRD from Ikaros regulation blocks lymphocyte maturation and mediates progression to a leukemic state by engaging functionally opposing epigenetic and genetic networks.


Subject(s)
Lymphocytes/enzymology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Animals , Base Sequence , Cell Differentiation/genetics , Chromatin Assembly and Disassembly , Gene Expression Profiling , Gene Regulatory Networks , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Leukemia/genetics , Lymphocytes/immunology , Mice , Nucleotide Motifs , Protein Binding , Thymocytes/metabolism
9.
Proc Natl Acad Sci U S A ; 111(1): 391-6, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24371308

ABSTRACT

Deregulated activation of ß-catenin in cancer has been correlated with genomic instability. During thymocyte development, ß-catenin activates transcription in partnership with T-cell-specific transcription factor 1 (Tcf-1). We previously reported that targeted activation of ß-catenin in thymocytes (CAT mice) induces lymphomas that depend on recombination activating gene (RAG) and myelocytomatosis oncogene (Myc) activities. Here we show that these lymphomas have recurring Tcra/Myc translocations that resulted from illegitimate RAG recombination events and resembled oncogenic translocations previously described in human T-ALL. We therefore used the CAT animal model to obtain mechanistic insights into the transformation process. ChIP-seq analysis uncovered a link between Tcf-1 and RAG2 showing that the two proteins shared binding sites marked by trimethylated histone-3 lysine-4 (H3K4me3) throughout the genome, including near the translocation sites. Pretransformed CAT thymocytes had increased DNA damage at the translocating loci and showed altered repair of RAG-induced DNA double strand breaks. These cells were able to survive despite DNA damage because activated ß-catenin promoted an antiapoptosis gene expression profile. Thus, activated ß-catenin promotes genomic instability that leads to T-cell lymphomas as a consequence of altered double strand break repair and increased survival of thymocytes with damaged DNA.


Subject(s)
Genomic Instability , Lymphocyte Activation , Lymphoma/genetics , T-Lymphocytes/cytology , beta Catenin/metabolism , Animals , Apoptosis , Base Sequence , Cell Survival , DNA Breaks, Double-Stranded , DNA Methylation , DNA Repair , Disease Models, Animal , Genes, RAG-1/genetics , Hepatocyte Nuclear Factor 1-alpha , Histones/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Recombination, Genetic , T Cell Transcription Factor 1/metabolism , Thymocytes/cytology , Translocation, Genetic , beta Catenin/genetics
10.
Blood ; 122(6): 902-11, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23741008

ABSTRACT

Bcl11b is a T-cell specific gene in hematopoiesis that begins expression during T-lineage commitment and is required for this process. Aberrant expression of BCL11B or proto-oncogene translocation to the vicinity of BCL11B can be a contributing factor in human T-ALL. To identify the mechanism that controls its distinctive T-lineage expression, we corrected the identified Bcl11b transcription start site and mapped a cell-type-specific differentially methylated region bracketing the Bcl11b promoter. We identified a 1.9-kb region 850 kb downstream of Bcl11b, "Major Peak," distinguished by its dynamic histone marking pattern in development that mirrors the pattern at the Bcl11b promoter. Looping interactions between promoter-proximal elements including the differentially methylated region and downstream elements in the Major Peak are required to recapitulate the T-cell specific expression of Bcl11b in stable reporter assays. Functional dissection of the Major Peak sequence showed distinct subregions, in which TCF-1 sites and a conserved element were required for T-lineage-specific activation and silencing in non-T cells. A bacterial artificial chromosome encompassing the full Bcl11b gene still required the addition of the Major Peak to exhibit T-cell specific expression. Thus, promoter-proximal and Major Peak sequences are cis-regulatory elements that interact over 850 kb to control expression of Bcl11b in hematopoietic cells.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Repressor Proteins/genetics , T-Lymphocytes/cytology , Tumor Suppressor Proteins/genetics , Animals , Cell Lineage , CpG Islands , DNA Methylation , Gene Silencing , Genes, Reporter , Hematopoietic Stem Cells , Histones/metabolism , Mice , Promoter Regions, Genetic , Proto-Oncogene Mas , Repressor Proteins/metabolism , T-Lymphocytes/immunology , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism
11.
Blood ; 122(18): 3149-59, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24002445

ABSTRACT

Ikaros is a critical regulator of lymphocyte development and homeostasis; thus, understanding its transcriptional regulation is important from both developmental and clinical perspectives. Using a mouse transgenic reporter approach, we functionally characterized a network of highly conserved cis-acting elements at the Ikzf1 locus. We attribute B-cell and myeloid but not T-cell specificity to the main Ikzf1 promoter. Although this promoter was unable to counter local chromatin silencing effects, each of the 6 highly conserved Ikzf1 intronic enhancers alleviated silencing. Working together, the Ikzf1 enhancers provided locus control region activity, allowing reporter expression in a position and copy-independent manner. Only 1 of the Ikzf1 enhancers was responsible for the progressive upregulation of Ikaros expression from hematopoietic stem cells to lymphoid-primed multipotent progenitors to T-cell precursors, which are stages of differentiation dependent on Ikaros for normal outcome. Thus, Ikzf1 is regulated by both epigenetic and transcriptional factors that target its enhancers in both redundant and specific fashions to provide an expression profile supportive of normal lymphoid lineage progression and homeostasis. Mutations in the Ikzf1 regulatory elements and their interacting factors are likely to have adverse effects on lymphopoiesis and contribute to leukemogenesis.


Subject(s)
Enhancer Elements, Genetic/genetics , Ikaros Transcription Factor/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcriptional Activation , Animals , B-Lymphocytes/metabolism , Base Sequence , Binding Sites/genetics , Brain/metabolism , Epigenesis, Genetic , Flow Cytometry , Gene Regulatory Networks , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ikaros Transcription Factor/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Myeloid Cells/metabolism , Sequence Homology, Amino Acid , T-Lymphocytes/metabolism , Transcription Factors/metabolism
12.
Proc Natl Acad Sci U S A ; 108(50): 20060-5, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22109558

ABSTRACT

Although transcriptional programs associated with T-cell specification and commitment have been described, the functional hierarchy and the roles of key regulators in structuring/orchestrating these programs remain unclear. Activation of Notch signaling in uncommitted precursors by the thymic stroma initiates the T-cell differentiation program. One regulator first induced in these precursors is the DNA-binding protein T-cell factor 1 (Tcf-1), a T-cell-specific mediator of Wnt signaling. However, the specific contribution of Tcf-1 to early T-cell development and the signals inducing it in these cells remain unclear. Here we assign functional significance to Tcf-1 as a gatekeeper of T-cell fate and show that Tcf-1 is directly activated by Notch signals. Tcf-1 is required at the earliest phase of T-cell determination for progression beyond the early thymic progenitor stage. The global expression profile of Tcf-1-deficient progenitors indicates that basic processes of DNA metabolism are down-regulated in its absence, and the blocked T-cell progenitors become abortive and die by apoptosis. Our data thus add an important functional relationship to the roadmap of T-cell development.


Subject(s)
Cell Lineage/immunology , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Bone Marrow Cells/cytology , Cell Movement , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Notch , Signal Transduction , Stem Cells/cytology , T Cell Transcription Factor 1/deficiency , Thymocytes/cytology , Thymocytes/immunology
13.
Blood ; 117(23): 6120-31, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21508411

ABSTRACT

Cyclin-dependent kinase-6 (CDK6) is required for early thymocyte development and tumorigenesis. To mechanistically dissect the role of CDK6 in thymocyte development, we generated and analyzed mutant knock-in mice and found that mice expressing a kinase-dead Cdk6 allele (Cdk6(K43M)) had a pronounced reduction in thymocytes and hematopoietic stem cells and progenitor cells (Lin⁻Sca-1⁺c-Kit⁺ [LSK]). In contrast, mice expressing the INK4-insensitive, hyperactive Cdk6(R31C) allele displayed excess proliferation in LSK and thymocytes. However, this is countered at least in part by increased apoptosis, which may limit progenitor and thymocyte expansion in the absence of other genetic events. Our mechanistic studies demonstrate that CDK6 kinase activity contributes to Notch signaling because inactive CDK6 kinase disrupts Notch-dependent survival, proliferation, and differentiation of LSK, with concomitant alteration of Notch target gene expression, such as massive up-regulation of CD25. Further, knockout of CD25 in Cdk6(K43M) mice rescued most defects observed in young mice. These results illustrate an important role for CDK6 kinase activity in thymocyte development that operates partially through modulating Notch target gene expression. This role of CDK6 as a downstream mediator of Notch identifies CDK6 kinase activity as a potential therapeutic target in human lymphoid malignancies.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Cyclin-Dependent Kinase 6/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Signal Transduction/physiology , Thymus Gland/enzymology , Alleles , Animals , Cell Survival/physiology , Cyclin-Dependent Kinase 6/genetics , Gene Knock-In Techniques , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/therapy , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Mice , Mice, Knockout , Receptors, Notch/genetics , Receptors, Notch/metabolism
14.
Proc Natl Acad Sci U S A ; 106(21): 8641-6, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19423665

ABSTRACT

The molecular requirements for invariant Valpha14-bearing natural killer T cells (iNKT) in the thymus are poorly understood. A minute population of approximately 500 newly selected CD69(+)CD24(+) stage 0 (ST0) iNKT cells gives rise to approximately 100 times more CD44(neg/lo)CD24(-) stage 1 (ST1) cells, which then generate similar frequencies of CD44(hi)CD24(-) stage 2 (ST2) and mature iNKT cells. Although the increased number of ST1 compared with ST0 cells indicates the initiation of a proliferation wave in the very early stages of iNKT cell development, details about the controlling mechanism are currently lacking. Here, we show that the transcription factor c-Myc is required for iNKT cell development. Conditional ablation of c-Myc in double-positive thymocytes specifically impacted iNKT but not conventional T cell development. Within the iNKT population, a progressive reduction of iNKT cells was observed starting at ST1 (approximately 50-fold) and ST2 (approximately 350-fold), with a complete lack of mature cells in thymus, spleen, and liver. ST0/ST1 c-Myc-deficient iNKT cells showed reduced proliferation. In contrast, annexin V staining did not reveal increased apoptosis, and transgenic overexpression of BCL-2 did not rescue iNKT cell development in c-Myc-deficient mice. Moreover, expression of known iNKT differentiation factors such as Plzf and Gata3 was not dramatically altered. These, findings provide compelling evidence that c-Myc mediates an intrathymic proliferation wave immediately after agonist selection of iNKT cells and illustrate the importance of this expansion for the generation of mature iNKT cells in vivo.


Subject(s)
Cell Differentiation/immunology , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Cell Lineage/immunology , Cell Proliferation , DNA, Complementary/genetics , Gene Expression Regulation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics
15.
J Immunol ; 183(6): 3873-84, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717519

ABSTRACT

Thymic maturation of T cells depends on the intracellular interpretation of alphabetaTCR signals by processes that are poorly understood. In this study, we report that beta-catenin/Tcf signaling was activated in double-positive thymocytes in response to alphabetaTCR engagement and impacted thymocyte selection. TCR engagement combined with activation of beta-catenin signaled thymocyte deletion, whereas Tcf-1 deficiency rescued from negative selection. Survival/apoptotis mediators including Bim, Bcl-2, and Bcl-x(L) were alternatively influenced by stabilization of beta-catenin or ablation of Tcf-1, and Bim-mediated beta-catenin induced thymocyte deletion. TCR activation in double-positive cells with stabilized beta-catenin triggered signaling associated with negative selection, including sustained overactivation of Lat and Jnk and a transient activation of Erk. These observations are consistent with beta-catenin/Tcf signaling acting as a switch that determines the outcome of thymic selection downstream the alphabetaTCR cascade.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction/immunology , T Cell Transcription Factor 1/physiology , Thymus Gland/cytology , beta Catenin/physiology , Animals , Apoptosis Regulatory Proteins/immunology , Cell Survival/immunology , Hepatocyte Nuclear Factor 1-alpha , Mice , Mice, Knockout , T Cell Transcription Factor 1/deficiency , Thymus Gland/physiology
17.
J Exp Med ; 214(1): 49-58, 2017 01.
Article in English | MEDLINE | ID: mdl-27998928

ABSTRACT

Activation-induced cytidine deaminase (AID) converts cytosine into uracil to initiate somatic hypermutation (SHM) and class switch recombination (CSR) of antibody genes. In addition, this enzyme produces DNA lesions at off-target sites that lead to mutations and chromosome translocations. However, AID is mostly cytoplasmic, and how and exactly when it accesses nuclear DNA remains enigmatic. Here, we show that AID is transiently in spatial contact with genomic DNA from the time the nuclear membrane breaks down in prometaphase until early G1, when it is actively exported into the cytoplasm. Consistent with this observation, the immunoglobulin (Igh) gene deamination as measured by uracil accumulation occurs primarily in early G1 after chromosomes decondense. Altering the timing of cell cycle-regulated AID nuclear residence increases DNA damage at off-target sites. Thus, the cell cycle-controlled breakdown and reassembly of the nuclear membrane and the restoration of transcription after mitosis constitute an essential time window for AID-induced deamination, and provide a novel DNA damage mechanism restricted to early G1.


Subject(s)
Cytidine Deaminase/metabolism , G1 Phase , Animals , Cell Cycle , Cell Nucleus/enzymology , Cells, Cultured , DNA Damage , Deamination , Genes, Immunoglobulin , Mice , Mice, Inbred C57BL , Transcription, Genetic
18.
Blood ; 112(3): 457-8, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18650457
19.
Sci Transl Med ; 6(225): 225ra28, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24574339

ABSTRACT

The density and type of lymphocytes that infiltrate colon tumors are predictive of the clinical outcome of colon cancer. High densities of T helper 17 (T(H)17) cells and inflammation predict poor outcome, whereas infiltration by T regulatory cells (Tregs) that naturally suppress inflammation is associated with longer patient survival. However, the role of Tregs in cancer remains controversial. We recently reported that Tregs in colon cancer patients can become proinflammatory and tumor-promoting. These properties were directly linked with their expression of RORγt (retinoic acid-related orphan receptor-γt), the signature transcription factor of T(H)17 cells. We report that Wnt/ß-catenin signaling in T cells promotes expression of RORγt. Expression of ß-catenin was elevated in T cells, including Tregs, of patients with colon cancer. Genetically engineered activation of ß-catenin in mouse T cells resulted in enhanced chromatin accessibility in the proximity of T cell factor-1 (Tcf-1) binding sites genome-wide, induced expression of T(H)17 signature genes including RORγt, and promoted T(H)17-mediated inflammation. Strikingly, the mice had inflammation of small intestine and colon and developed lesions indistinguishable from colitis-induced cancer. Activation of ß-catenin only in Tregs was sufficient to produce inflammation and initiate cancer. On the basis of these findings, we conclude that activation of Wnt/ß-catenin signaling in effector T cells and/or Tregs is causatively linked with the imprinting of proinflammatory properties and the promotion of colon cancer.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Colitis/metabolism , Colon/metabolism , Colonic Neoplasms/metabolism , Inflammation Mediators/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , beta Catenin/metabolism , Animals , Binding Sites , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Chromatin Assembly and Disassembly , Colitis/genetics , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Genes, APC , Hepatocyte Nuclear Factor 1-alpha , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Wnt Signaling Pathway , beta Catenin/genetics
20.
Cell Rep ; 3(5): 1678-1689, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23707059

ABSTRACT

The "CTCF code" hypothesis posits that CTCF pleiotropic functions are driven by recognition of diverse sequences through combinatorial use of its 11 zinc fingers (ZFs). This model, however, is supported by in vitro binding studies of a limited number of sequences. To study CTCF multivalency in vivo, we define ZF binding requirements at ∼50,000 genomic sites in primary lymphocytes. We find that CTCF reads sequence diversity through ZF clustering. ZFs 4-7 anchor CTCF to ∼80% of targets containing the core motif. Nonconserved flanking sequences are recognized by ZFs 1-2 and ZFs 8-11 clusters, which also stabilize CTCF broadly. Alternatively, ZFs 9-11 associate with a second phylogenetically conserved upstream motif at ∼15% of its sites. Individually, ZFs increase overall binding and chromatin residence time. Unexpectedly, we also uncovered a conserved downstream DNA motif that destabilizes CTCF occupancy. Thus, CTCF associates with a wide array of DNA modules via combinatorial clustering of its 11 ZFs.


Subject(s)
Genome , Repressor Proteins/metabolism , Animals , B-Lymphocytes/metabolism , Binding Sites , CCCTC-Binding Factor , Chromosome Mapping , Male , Mice , Mice, Inbred C57BL , Nucleotide Motifs , Photobleaching , Repressor Proteins/chemistry , Repressor Proteins/genetics , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL