Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Publication year range
1.
Nat Rev Mol Cell Biol ; 23(6): 383, 2022 06.
Article in English | MEDLINE | ID: mdl-35352008

Subject(s)
Career Choice
2.
Mol Cell ; 76(3): 412-422.e5, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31522988

ABSTRACT

The function of the CCCTC-binding factor (CTCF) in the organization of the genome has become an important area of investigation, but the mechanisms by which CTCF dynamically contributes to genome organization are not clear. We previously discovered that CTCF binds to large numbers of endogenous RNAs, promoting its self-association. In this regard, we now report two independent features that disrupt CTCF association with chromatin: inhibition of transcription and disruption of CTCF-RNA interactions through mutations of 2 of its 11 zinc fingers that are not required for CTCF binding to its cognate DNA site: zinc finger 1 (ZF1) or zinc finger 10 (ZF10). These mutations alter gene expression profiles as CTCF mutants lose their ability to form chromatin loops and thus the ability to insulate chromatin domains and to mediate CTCF long-range genomic interactions. Our results point to the importance of CTCF-mediated RNA interactions as a structural component of genome organization.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Mouse Embryonic Stem Cells/metabolism , RNA/metabolism , Animals , Binding Sites , CCCTC-Binding Factor/chemistry , CCCTC-Binding Factor/genetics , Cell Line , Chromatin/chemistry , Chromatin/genetics , Mice , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Interaction Domains and Motifs , RNA/chemistry , RNA/genetics , Structure-Activity Relationship , Transcription, Genetic , Zinc Fingers
3.
Mol Cell ; 70(6): 1149-1162.e5, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29932905

ABSTRACT

Polycomb repressive complex 2 (PRC2) maintains gene silencing by catalyzing methylation of histone H3 at lysine 27 (H3K27me2/3) within chromatin. By designing a system whereby PRC2-mediated repressive domains were collapsed and then reconstructed in an inducible fashion in vivo, a two-step mechanism of H3K27me2/3 domain formation became evident. First, PRC2 is stably recruited by the actions of JARID2 and MTF2 to a limited number of spatially interacting "nucleation sites," creating H3K27me3-forming Polycomb foci within the nucleus. Second, PRC2 is allosterically activated via its binding to H3K27me3 and rapidly spreads H3K27me2/3 both in cis and in far-cis via long-range contacts. As PRC2 proceeds further from the nucleation sites, its stability on chromatin decreases such that domains of H3K27me3 remain proximal, and those of H3K27me2 distal, to the nucleation sites. This study demonstrates the principles of de novo establishment of PRC2-mediated repressive domains across the genome.


Subject(s)
Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins/metabolism , Animals , Chromatin/metabolism , Gene Silencing , Histone Code , Histones/metabolism , Lysine/metabolism , Methylation , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells , Protein Binding , Protein Processing, Post-Translational
4.
Proc Natl Acad Sci U S A ; 117(50): 31914-31922, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33257571

ABSTRACT

Inhibiting membrane association of RAS has long been considered a rational approach to anticancer therapy, which led to the development of farnesyltransferase inhibitors (FTIs). However, FTIs proved ineffective against KRAS-driven tumors. To reveal alternative therapeutic strategies, we carried out a genome-wide CRISPR-Cas9 screen designed to identify genes required for KRAS4B membrane association. We identified five enzymes in the prenylation pathway and SAFB, a nuclear protein with both DNA and RNA binding domains. Silencing SAFB led to marked mislocalization of all RAS isoforms as well as RAP1A but not RAB7A, a pattern that phenocopied silencing FNTA, the prenyltransferase α subunit shared by farnesyltransferase and geranylgeranyltransferase type I. We found that SAFB promoted RAS membrane association by controlling FNTA expression. SAFB knockdown decreased GTP loading of RAS, abrogated alternative prenylation, and sensitized RAS-mutant cells to growth inhibition by FTI. Our work establishes the prenylation pathway as paramount in KRAS membrane association, reveals a regulator of prenyltransferase expression, and suggests that reduction in FNTA expression may enhance the efficacy of FTIs.


Subject(s)
Cell Membrane/metabolism , Dimethylallyltranstransferase/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Neoplasms/pathology , Nuclear Matrix-Associated Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Estrogen/metabolism , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , CRISPR-Cas Systems/genetics , Computational Biology , Datasets as Topic , Gene Knockdown Techniques , Humans , Matrix Attachment Region Binding Proteins/genetics , Neoplasms/genetics , Nuclear Matrix-Associated Proteins/genetics , Protein Prenylation , Protein Subunits/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Estrogen/genetics
5.
Proc Natl Acad Sci U S A ; 117(49): 31343-31352, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229554

ABSTRACT

Development of progenitor B cells (ProB cells) into precursor B cells (PreB cells) is dictated by immunoglobulin heavy chain checkpoint (IgHCC), where the IgHC encoded by a productively rearranged Igh allele assembles into a PreB cell receptor complex (PreBCR) to generate signals to initiate this transition and suppressing antigen receptor gene recombination, ensuring that only one productive Igh allele is expressed, a phenomenon known as Igh allelic exclusion. In contrast to a productively rearranged Igh allele, the Igh messenger RNA (mRNA) (IgHR) from a nonproductively rearranged Igh allele is degraded by nonsense-mediated decay (NMD). This fact prohibited firm conclusions regarding the contribution of stable IgHR to the molecular and developmental changes associated with the IgHCC. This point was addressed by generating the IghTer5H∆TM mouse model from IghTer5H mice having a premature termination codon at position +5 in leader exon of IghTer5H allele. This prohibited NMD, and the lack of a transmembrane region (∆TM) prevented the formation of any signaling-competent PreBCR complexes that may arise as a result of read-through translation across premature Ter5 stop codon. A highly sensitive sandwich Western blot revealed read-through translation of IghTer5H message, indicating that previous conclusions regarding a role of IgHR in establishing allelic exclusion requires further exploration. As determined by RNA sequencing (RNA-Seq), this low amount of IgHC sufficed to initiate PreB cell markers normally associated with PreBCR signaling. In contrast, the IghTer5H∆TM knock-in allele, which generated stable IgHR but no detectable IgHC, failed to induce PreB development. Our data indicate that the IgHCC is controlled at the level of IgHC and not IgHR expression.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Alleles , Animals , Biomarkers/metabolism , Genetic Loci , Mice, Inbred C57BL , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
6.
Nat Immunol ; 10(6): 655-64, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19448632

ABSTRACT

Coordinated recombination of homologous antigen receptor loci is thought to be important for allelic exclusion. Here we show that homologous immunoglobulin alleles pair in a stage-specific way that mirrors the recombination patterns of these loci. The frequency of homologous immunoglobulin pairing was much lower in the absence of the RAG-1-RAG-2 recombinase and was restored in Rag1-/- developing B cells with a transgene expressing a RAG-1 active-site mutant that supported DNA binding but not cleavage. The introduction of DNA breaks on one immunoglobulin allele induced ATM-dependent repositioning of the other allele to pericentromeric heterochromatin. ATM activated by the cleaved allele acts in trans on the uncleaved allele to prevent biallelic recombination and chromosome breaks or translocations.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Immunoglobulins/genetics , Protein Serine-Threonine Kinases/genetics , Recombination, Genetic , Tumor Suppressor Proteins/genetics , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/metabolism , Cells, Cultured , DNA Breaks , Gene Rearrangement , Mice , Mice, Inbred C57BL , Mice, Knockout , VDJ Recombinases/metabolism
7.
Nat Immunol ; 9(4): 396-404, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18297074

ABSTRACT

Variable-(diversity)-joining (V(D)J) recombination at loci encoding the immunoglobulin heavy chain (Igh) and immunoglobulin light chain (Igk) takes place sequentially during successive stages in B cell development. Using three-dimensional DNA fluorescence in situ hybridization, here we identify a lineage-specific and stage-specific interchromosomal association between these two loci that marks the transition between Igh and Igk recombination. Colocalization occurred between pericentromerically located alleles in pre-B cells and was mediated by the 3' Igk enhancer. Deletion of this regulatory element prevented association of the Igh and Igk loci, inhibited pericentromeric recruitment and locus 'decontraction' of an Igh allele, and resulted in greater distal rearrangement of the gene encoding the variable heavy-chain region. Our data indicate involvement of the Igk locus and its 3' enhancer in directing the Igh locus to a repressive nuclear subcompartment and inducing the Igh locus to decontract.


Subject(s)
Enhancer Elements, Genetic/immunology , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Heavy Chains/genetics , Immunoglobulins/genetics , Precursor Cells, B-Lymphoid/immunology , 3' Flanking Region/genetics , Animals , Chromosomes/genetics , Chromosomes/metabolism , Genes, Immunoglobulin Heavy Chain/physiology , Immunoglobulins/physiology , Mice , Mice, Knockout , Precursor Cells, B-Lymphoid/metabolism , Recombination, Genetic
8.
Immunity ; 34(3): 303-14, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21435585

ABSTRACT

T cell fate is associated with mutually exclusive expression of CD4 or CD8 in helper and cytotoxic T cells, respectively. How expression of one locus is temporally coordinated with repression of the other has been a long-standing enigma, though we know RUNX transcription factors activate the Cd8 locus, silence the Cd4 locus, and repress the Zbtb7b locus (encoding the transcription factor ThPOK), which is required for CD4 expression. Here we found that nuclear organization was altered by interplay among members of this transcription factor circuitry: RUNX binding mediated association of Cd4 and Cd8 whereas ThPOK binding kept the loci apart. Moreover, targeted deletions within Cd4 modulated CD8 expression and pericentromeric repositioning of Cd8. Communication between Cd4 and Cd8 thus appears to enable long-range epigenetic regulation to ensure that expression of one excludes the other in mature CD4 or CD8 single-positive (SP) cells.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Core Binding Factor alpha Subunits/immunology , Gene Expression Regulation/immunology , Animals , Epigenomics , Flow Cytometry , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C57BL
9.
Mol Cell ; 47(6): 873-85, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22864115

ABSTRACT

Class switch recombination (CSR) has the potential to generate genomic instability in B cells as activation-induced cytidine deaminase (AID), which mediates this process, is known to target many sites outside Igh. Nonetheless we do not fully understand what factors influence AID targeting genome-wide. Given that errors in CSR can lead to dangerous, oncogenic chromosomal translocations it is important to identify the elements that determine which genes are at risk of being "hit" and could be involved in aberrant rearrangements. Here we have investigated the influence of nuclear organization in determining "off-target" activity and the choice of fusion partners. Our studies indicate that the vast majority of known AID-mediated Igh translocation partners are found in chromosomal domains that contact this locus during class switching. Further, these interaction domains can be used to identify other genes that are hit by AID.


Subject(s)
B-Lymphocytes/cytology , Cytidine Deaminase/metabolism , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Class Switching , Translocation, Genetic , Animals , B-Lymphocytes/metabolism , Cytidine Deaminase/genetics , Genomic Instability , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Somatic Hypermutation, Immunoglobulin
10.
Nucleic Acids Res ; 44(18): 8714-8725, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27439714

ABSTRACT

Use of low resolution single cell DNA FISH and population based high resolution chromosome conformation capture techniques have highlighted the importance of pairwise chromatin interactions in gene regulation. However, it is unlikely that associations involving regulatory elements act in isolation of other interacting partners that also influence their impact. Indeed, the influence of multi-loci interactions remains something of an enigma as beyond low-resolution DNA FISH we do not have the appropriate tools to analyze these. Here we present a method that uses standard 4C-seq data to identify multi-loci interactions from the same cell. We demonstrate the feasibility of our method using 4C-seq data sets that identify known pairwise and novel tri-loci interactions involving the Tcrb and Igk antigen receptor enhancers. We further show that the three Igk enhancers, MiEκ, 3'Eκ and Edκ, interact simultaneously in this super-enhancer cluster, which add to our previous findings showing that loss of one element decreases interactions between all three elements as well as reducing their transcriptional output. These findings underscore the functional importance of simultaneous interactions and provide new insight into the relationship between enhancer elements. Our method opens the door for studying multi-loci interactions and their impact on gene regulation in other biological settings.


Subject(s)
Chromosomes/metabolism , Genetic Loci , Nucleic Acid Conformation , Sequence Analysis, DNA/methods , Chromatin/metabolism , Enhancer Elements, Genetic , Estrogen Receptor beta/metabolism , Genome , Receptors, Antigen, T-Cell, alpha-beta
11.
Proc Natl Acad Sci U S A ; 112(5): E458-66, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25609670

ABSTRACT

The genes encoding the variable (V) region of the B-cell antigen receptor (BCR) are assembled from V, D (diversity), and J (joining) elements through a RAG-mediated recombination process that relies on the recognition of recombination signal sequences (RSSs) flanking the individual elements. Secondary V(D)J rearrangement modifies the original Ig rearrangement if a nonproductive original joint is formed, as a response to inappropriate signaling from a self-reactive BCR, or as part of a stochastic mechanism to further diversify the Ig repertoire. VH replacement represents a RAG-mediated secondary rearrangement in which an upstream VH element recombines with a rearranged VHDHJH joint to generate a new BCR specificity. The rearrangement occurs between the cryptic RSS of the original VH element and the conventional RSS of the invading VH gene, leaving behind a footprint of up to five base pairs (bps) of the original VH gene that is often further obscured by exonuclease activity and N-nucleotide addition. We have previously demonstrated that VH replacement can efficiently rescue the development of B cells that have acquired two nonproductive heavy chain (IgH) rearrangements. Here we describe a novel knock-in mouse model in which the prerearranged IgH locus resembles an endogenously rearranged productive VHDHJH allele. Using this mouse model, we characterized the role of VH replacement in the diversification of the primary Ig repertoire through the modification of productive VHDHJH rearrangements. Our results indicate that VH replacement occurs before Ig light chain rearrangement and thus is not involved in the editing of self-reactive antibodies.


Subject(s)
Immunoglobulin Variable Region/genetics , Animals , B-Lymphocytes/immunology , Cell Compartmentation , Mice , Mice, Transgenic , Stochastic Processes
12.
Immunity ; 28(3): 335-45, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18280186

ABSTRACT

Productive rearrangement of the immunoglobulin heavy-chain locus triggers a major developmental checkpoint that promotes limited clonal expansion of pre-B cells, thereby culminating in cell-cycle arrest and rearrangement of light-chain loci. By using Irf4-/-Irf8-/- pre-B cells, we demonstrated that two pathways converge to synergistically drive light-chain rearrangement, but not simply as a consequence of cell-cycle exit. One pathway was directly dependent on transcription factor IRF-4, whose expression was elevated by pre-B cell receptor signaling. IRF-4 targeted the immunoglobulin 3'Ekappa and Elambda enhancers and positioned a kappa allele away from pericentromeric heterochromatin. The other pathway was triggered by attenuation of IL-7 signaling and activated the iEkappa enhancer via binding of the transcription factor E2A. IRF-4 also regulated expression of chemokine receptor Cxcr4 and promoted migration of pre-B cells in response to the chemokine ligand CXCL12. We propose that IRF-4 coordinates the two pathways regulating light-chain recombination by positioning pre-B cells away from IL-7-expressing stromal cells.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Gene Rearrangement, B-Lymphocyte, Light Chain/immunology , Interferon Regulatory Factors/immunology , Interleukin-7/immunology , Signal Transduction/immunology , Animals , B-Lymphocytes/cytology , Blotting, Western , Cell Movement/immunology , Electrophoretic Mobility Shift Assay , Flow Cytometry , Immunoprecipitation , In Situ Hybridization, Fluorescence , Interferon Regulatory Factors/metabolism , Interleukin-7/metabolism , Mice , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/immunology
13.
PLoS Comput Biol ; 12(3): e1004780, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26938081

ABSTRACT

4C-Seq has proven to be a powerful technique to identify genome-wide interactions with a single locus of interest (or "bait") that can be important for gene regulation. However, analysis of 4C-Seq data is complicated by the many biases inherent to the technique. An important consideration when dealing with 4C-Seq data is the differences in resolution of signal across the genome that result from differences in 3D distance separation from the bait. This leads to the highest signal in the region immediately surrounding the bait and increasingly lower signals in far-cis and trans. Another important aspect of 4C-Seq experiments is the resolution, which is greatly influenced by the choice of restriction enzyme and the frequency at which it can cut the genome. Thus, it is important that a 4C-Seq analysis method is flexible enough to analyze data generated using different enzymes and to identify interactions across the entire genome. Current methods for 4C-Seq analysis only identify interactions in regions near the bait or in regions located in far-cis and trans, but no method comprehensively analyzes 4C signals of different length scales. In addition, some methods also fail in experiments where chromatin fragments are generated using frequent cutter restriction enzymes. Here, we describe 4C-ker, a Hidden-Markov Model based pipeline that identifies regions throughout the genome that interact with the 4C bait locus. In addition, we incorporate methods for the identification of differential interactions in multiple 4C-seq datasets collected from different genotypes or experimental conditions. Adaptive window sizes are used to correct for differences in signal coverage in near-bait regions, far-cis and trans chromosomes. Using several datasets, we demonstrate that 4C-ker outperforms all existing 4C-Seq pipelines in its ability to reproducibly identify interaction domains at all genomic ranges with different resolution enzymes.


Subject(s)
DNA, Catalytic/chemistry , DNA, Catalytic/genetics , Genome/physiology , Restriction Mapping/methods , Sequence Analysis, DNA/methods , Software , Algorithms , Base Sequence , Binding Sites , Molecular Sequence Data , Protein Binding
14.
Nature ; 471(7336): 119-23, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368836

ABSTRACT

Misrepair of DNA double-strand breaks produced by the V(D)J recombinase (the RAG1/RAG2 proteins) at immunoglobulin (Ig) and T cell receptor (Tcr) loci has been implicated in pathogenesis of lymphoid malignancies in humans and in mice. Defects in DNA damage response factors such as ataxia telangiectasia mutated (ATM) protein and combined deficiencies in classical non-homologous end joining and p53 predispose to RAG-initiated genomic rearrangements and lymphomagenesis. Although we showed previously that RAG1/RAG2 shepherd the broken DNA ends to classical non-homologous end joining for proper repair, roles for the RAG proteins in preserving genomic stability remain poorly defined. Here we show that the RAG2 carboxy (C) terminus, although dispensable for recombination, is critical for maintaining genomic stability. Thymocytes from 'core' Rag2 homozygotes (Rag2(c/c) mice) show dramatic disruption of Tcrα/δ locus integrity. Furthermore, all Rag2(c/c) p53(-/-) mice, unlike Rag1(c/c) p53(-/-) and p53(-/-) animals, rapidly develop thymic lymphomas bearing complex chromosomal translocations, amplifications and deletions involving the Tcrα/δ and Igh loci. We also find these features in lymphomas from Atm(-/-) mice. We show that, like ATM-deficiency, core RAG2 severely destabilizes the RAG post-cleavage complex. These results reveal a novel genome guardian role for RAG2 and suggest that similar 'end release/end persistence' mechanisms underlie genomic instability and lymphomagenesis in Rag2(c/c) p53(-/-) and Atm(-/-) mice.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Disease Progression , Genomic Instability , Lymphoma/genetics , Lymphoma/pathology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Chromosome Deletion , Chromosomes, Mammalian/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Rearrangement, T-Lymphocyte/genetics , Genes, Immunoglobulin Heavy Chain/genetics , Genes, p53/genetics , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Mice , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, Antigen, T-Cell/genetics , Recombination, Genetic/genetics , Thymus Gland/cytology , Translocation, Genetic/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
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
17.
J Immunol ; 188(12): 6084-92, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22581861

ABSTRACT

Ag receptor diversity involves the introduction of DNA double-stranded breaks during lymphocyte development. To ensure fidelity, cleavage is confined to the G(0)-G(1) phase of the cell cycle. One established mechanism of regulation is through periodic degradation of the RAG2 recombinase protein. However, there are additional levels of protection. In this paper, we show that cyclical changes in the IL-7R signaling pathway functionally segregate pro-B cells according to cell cycle status. In consequence, the level of a downstream effector of IL-7 signaling, phospho-STAT5, is inversely correlated with cell cycle expression of Rag, a key gene involved in recombination. Higher levels of phopho-STAT5 in S-G(2) correlate with decreased Rag expression and Rag relocalization to pericentromeric heterochromatin. These cyclical changes in transcription and locus repositioning are ablated upon transformation with v-Abl, which renders STAT5 constitutively active across the cell cycle. We propose that this activity of the IL-7R/STAT5 pathway plays a critical protective role in development, complementing regulation of RAG2 at the protein level, to ensure that recombination does not occur during replication. Our data, suggesting that pro-B cells are not a single homogeneous population, explain inconsistencies in the role of IL-7 signaling in regulating Igh recombination.


Subject(s)
B-Lymphocyte Subsets/immunology , Cell Cycle/immunology , Interleukin-7/immunology , Precursor Cells, B-Lymphoid/immunology , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Flow Cytometry , Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain/immunology , Genes, RAG-1 , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , In Situ Hybridization, Fluorescence , Interleukin-7/metabolism , Mice , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Signal Transduction/immunology , Transcription, Genetic
18.
Immunol Rev ; 237(1): 43-54, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20727028

ABSTRACT

Perhaps no process has provided more insight into the fine manipulation of locus accessibility than antigen receptor rearrangement. V(D)J recombination is carried out by the lymphoid-specific recombination-activating (RAG 1 and 2) proteins and the non-homologous end joining machinery; yet, it occurs only at specific loci (or portions of loci) during specific developmental stages. This spatiotemporal restriction of recombination is achieved through precise alterations in locus accessibility. In this article, we discuss the work of our laboratory in elucidating how nuclear sublocalization, chromosome conformation, and locus interactions contribute to regulating this complex process. We also discuss what is known about how key factors in B-cell development (such as the ubiquitously expressed helix loop helix protein E2A, the B-cell specific transcription factors EBF1 and Pax5, and the interleukin-7 cytokine signaling pathway) exert their effects through changes in nuclear dynamics.


Subject(s)
B-Lymphocytes/immunology , Chromosomes/genetics , Gene Rearrangement , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/genetics , T-Lymphocytes/immunology , Animals , Chromosomes/metabolism , Humans , VDJ Recombinases/physiology
19.
Nat Commun ; 15(1): 6027, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39025865

ABSTRACT

Aberrations in the capacity of DNA/chromatin modifiers and transcription factors to bind non-coding regions can lead to changes in gene regulation and impact disease phenotypes. However, identifying distal regulatory elements and connecting them with their target genes remains challenging. Here, we present MethNet, a pipeline that integrates large-scale DNA methylation and gene expression data across multiple cancers, to uncover cis regulatory elements (CREs) in a 1 Mb region around every promoter in the genome. MethNet identifies clusters of highly ranked CREs, referred to as 'hubs', which contribute to the regulation of multiple genes and significantly affect patient survival. Promoter-capture Hi-C confirmed that highly ranked associations involve physical interactions between CREs and their gene targets, and CRISPR interference based single-cell RNA Perturb-seq validated the functional impact of CREs. Thus, MethNet-identified CREs represent a valuable resource for unraveling complex mechanisms underlying gene expression, and for prioritizing the verification of predicted non-coding disease hotspots.


Subject(s)
DNA Methylation , Gene Expression Regulation, Neoplastic , Neoplasms , Promoter Regions, Genetic , Humans , Neoplasms/genetics , DNA Methylation/genetics , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Cell Rep ; 43(8): 114547, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39083377

ABSTRACT

During chronic infection, virus-specific CD8+ cytotoxic T lymphocytes (CTLs) progressively lose their ability to mount effective antiviral responses. This "exhaustion" is coupled to persistent upregulation of inhibitory receptor programmed death-1 (PD-1) (Pdcd1)-key in suppressing antiviral CTL responses. Here, we investigate allelic Pdcd1 subnuclear localization and transcription during acute and chronic lymphocytic choriomeningitis virus (LCMV) infection in mice. Pdcd1 alleles dissociate from transcriptionally repressive chromatin domains (lamin B) in virus-specific exhausted CTLs but not in naive or effector CTLs. Relative to naive CTLs, nuclear positioning and Pdcd1-lamina dissociation in exhausted CTLs reflect loss of Pdcd1 promoter methylation and greater PD-1 upregulation, although a direct correlation is not observed in effector cells, 8 days post-infection. Genetic deletion of B lymphocyte-induced maturation protein 1 (Blimp-1) enhances Pdcd1-lamina dissociation in effector CTLs, suggesting that Blimp-1 contributes to maintaining Pdcd1 localization to repressive lamina. Our results identify mechanisms governing Pdcd1 subnuclear localization and the broader role of chromatin dynamics in T cell exhaustion.

SELECTION OF CITATIONS
SEARCH DETAIL