Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 169(4): 693-707.e14, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475897

ABSTRACT

The spatial organization of chromosomes influences many nuclear processes including gene expression. The cohesin complex shapes the 3D genome by looping together CTCF sites along chromosomes. We show here that chromatin loop size can be increased and that the duration with which cohesin embraces DNA determines the degree to which loops are enlarged. Cohesin's DNA release factor WAPL restricts this loop extension and also prevents looping between incorrectly oriented CTCF sites. We reveal that the SCC2/SCC4 complex promotes the extension of chromatin loops and the formation of topologically associated domains (TADs). Our data support the model that cohesin structures chromosomes through the processive enlargement of loops and that TADs reflect polyclonal collections of loops in the making. Finally, we find that whereas cohesin promotes chromosomal looping, it rather limits nuclear compartmentalization. We conclude that the balanced activity of SCC2/SCC4 and WAPL enables cohesin to correctly structure chromosomes.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , Chromatin/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Acetyltransferases/metabolism , CCCTC-Binding Factor , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins , Fatty Acid Elongases , Gene Editing , Humans , Multiprotein Complexes/metabolism , Repressor Proteins/metabolism , Cohesins
2.
Mol Cell ; 81(15): 3082-3095.e6, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34197738

ABSTRACT

To understand how chromatin domains coordinate gene expression, we dissected select genetic elements organizing topology and transcription around the Prdm14 super enhancer in mouse embryonic stem cells. Taking advantage of allelic polymorphisms, we developed methods to sensitively analyze changes in chromatin topology, gene expression, and protein recruitment. We show that enhancer insulation does not rely strictly on loop formation between its flanking boundaries, that the enhancer activates the Slco5a1 gene beyond its prominent domain boundary, and that it recruits cohesin for loop extrusion. Upon boundary inversion, we find that oppositely oriented CTCF terminates extrusion trajectories but does not stall cohesin, while deleted or mutated CTCF sites allow cohesin to extend its trajectory. Enhancer-mediated gene activation occurs independent of paused loop extrusion near the gene promoter. We expand upon the loop extrusion model to propose that cohesin loading and extrusion trajectories originating at an enhancer contribute to gene activation.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin/genetics , Enhancer Elements, Genetic , Animals , CCCTC-Binding Factor/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression , Mice , Mouse Embryonic Stem Cells , Nuclear Receptor Coactivator 2/genetics , Promoter Regions, Genetic , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Cohesins
3.
Nucleic Acids Res ; 51(8): 3770-3792, 2023 05 08.
Article in English | MEDLINE | ID: mdl-36942484

ABSTRACT

During every cell cycle, both the genome and the associated chromatin must be accurately replicated. Chromatin Assembly Factor-1 (CAF-1) is a key regulator of chromatin replication, but how CAF-1 functions in relation to the DNA replication machinery is unknown. Here, we reveal that this crosstalk differs between the leading and lagging strand at replication forks. Using biochemical reconstitutions, we show that DNA and histones promote CAF-1 recruitment to its binding partner PCNA and reveal that two CAF-1 complexes are required for efficient nucleosome assembly under these conditions. Remarkably, in the context of the replisome, CAF-1 competes with the leading strand DNA polymerase epsilon (Polϵ) for PCNA binding. However, CAF-1 does not affect the activity of the lagging strand DNA polymerase Delta (Polδ). Yet, in cells, CAF-1 deposits newly synthesized histones equally on both daughter strands. Thus, on the leading strand, chromatin assembly by CAF-1 cannot occur simultaneously to DNA synthesis, while on the lagging strand these processes may be coupled. We propose that these differences may facilitate distinct parental histone recycling mechanisms and accommodate the inherent asymmetry of DNA replication.


Subject(s)
Chromatin , Histones , Histones/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , Chromatin/genetics , DNA Replication , DNA/genetics
4.
Mol Cell ; 61(3): 461-473, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26833089

ABSTRACT

Detailed genomic contact maps have revealed that chromosomes are structurally organized in megabase-sized topologically associated domains (TADs) that encompass smaller subTADs. These domains segregate in the nuclear space to form active and inactive nuclear compartments, but cause and consequence of compartmentalization are largely unknown. Here, we combined lacO/lacR binding platforms with allele-specific 4C technologies to track their precise position in the three-dimensional genome upon recruitment of NANOG, SUV39H1, or EZH2. We observed locked genomic loci resistant to spatial repositioning and unlocked loci that could be repositioned to different nuclear subcompartments with distinct chromatin signatures. Focal protein recruitment caused the entire subTAD, but not surrounding regions, to engage in new genomic contacts. Compartment switching was found uncoupled from transcription changes, and the enzymatic modification of histones per se was insufficient for repositioning. Collectively, this suggests that trans-associated factors influence three-dimensional compartmentalization independent of their cis effect on local chromatin composition and activity.


Subject(s)
Cell Nucleus/metabolism , Chromosome Segregation , Embryonic Stem Cells/metabolism , Genetic Loci , Lac Operon , Lac Repressors/metabolism , Animals , Cells, Cultured , Chromatin/metabolism , Chromatin Assembly and Disassembly , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lac Repressors/genetics , Methyltransferases/genetics , Methyltransferases/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Nanog Homeobox Protein , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transfection
5.
Nucleic Acids Res ; 50(6): 3190-3202, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35234910

ABSTRACT

Bovine leukemia virus (BLV)-induced tumoral development is a multifactorial phenomenon that remains incompletely understood. Here, we highlight the critical role of the cellular CCCTC-binding factor (CTCF) both in the regulation of BLV transcriptional activities and in the deregulation of the three-dimensional (3D) chromatin architecture surrounding the BLV integration site. We demonstrated the in vivo recruitment of CTCF to three conserved CTCF binding motifs along the provirus. Next, we showed that CTCF localized to regions of transitions in the histone modifications profile along the BLV genome and that it is implicated in the repression of the 5'Long Terminal Repeat (LTR) promoter activity, thereby contributing to viral latency, while favoring the 3'LTR promoter activity. Finally, we demonstrated that BLV integration deregulated the host cellular 3D chromatin organization through the formation of viral/host chromatin loops. Altogether, our results highlight CTCF as a new critical effector of BLV transcriptional regulation and BLV-induced physiopathology.


Subject(s)
Leukemia Virus, Bovine , Virus Latency , CCCTC-Binding Factor/metabolism , Chromatin , Leukemia Virus, Bovine/genetics , Leukemia Virus, Bovine/metabolism , Promoter Regions, Genetic , Terminal Repeat Sequences/genetics
6.
Circ Res ; 128(1): 115-129, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33107387

ABSTRACT

RATIONALE: ANP (atrial natriuretic peptide) and BNP (B-type natriuretic peptide), encoded by the clustered genes Nppa and Nppb, are important prognostic, diagnostic, and therapeutic proteins in cardiac disease. The spatiotemporal expression pattern and stress-induction of the Nppa and Nppb are tightly regulated, possibly involving their coregulation by an evolutionary conserved enhancer cluster. OBJECTIVE: To explore the physiological functions of the enhancer cluster and elucidate the genomic mechanism underlying Nppa-Nppb coregulation in vivo. METHODS AND RESULTS: By analyzing epigenetic data we uncovered an enhancer cluster with super enhancer characteristics upstream of Nppb. Using CRISPR/Cas9 genome editing, the enhancer cluster or parts thereof, Nppb and flanking regions or the entire genomic block spanning Nppa-Nppb, respectively, were deleted from the mouse genome. The impact on gene regulation and phenotype of the respective mouse lines was investigated by transcriptomic, epigenomic, and phenotypic analyses. The enhancer cluster was essential for prenatal and postnatal ventricular expression of Nppa and Nppb but not of any other gene. Enhancer cluster-deficient mice showed enlarged hearts before and after birth, similar to Nppa-Nppb compound knockout mice we generated. Analysis of the other deletion alleles indicated the enhancer cluster engages the promoters of Nppa and Nppb in a competitive rather than a cooperative mode, resulting in increased Nppa expression when Nppb and flanking sequences were deleted. The enhancer cluster maintained its active epigenetic state and selectivity when its target genes are absent. In enhancer cluster-deficient animals, Nppa was induced but remained low in the postmyocardial infarction border zone and in the hypertrophic ventricle, involving regulatory sequences proximal to Nppa. CONCLUSIONS: Coordinated ventricular expression of Nppa and Nppb is controlled in a competitive manner by a shared super enhancer, which is also required to augment stress-induced expression and to prevent premature hypertrophy.


Subject(s)
Atrial Natriuretic Factor/genetics , Enhancer Elements, Genetic , Hypertrophy, Left Ventricular/genetics , Multigene Family , Myocardial Infarction/genetics , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/genetics , Animals , Atrial Natriuretic Factor/metabolism , Binding Sites , Binding, Competitive , CRISPR-Cas Systems , Cell Line , Disease Models, Animal , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Humans , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/metabolism , Promoter Regions, Genetic
7.
Mol Cell ; 60(4): 676-84, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26527277

ABSTRACT

CCCTC-binding factor (CTCF) is an architectural protein involved in the three-dimensional (3D) organization of chromatin. In this study, we assayed the 3D genomic contact profiles of a large number of CTCF binding sites with high-resolution 4C-seq. As recently reported, our data also suggest that chromatin loops preferentially form between CTCF binding sites oriented in a convergent manner. To directly test this, we used CRISPR/Cas9 genome editing to delete core CTCF binding sites in three loci, including the CTCF site in the Sox2 super-enhancer. In all instances, CTCF and cohesin recruitment were lost, and chromatin loops with distal, convergent CTCF sites were disrupted or destabilized. Re-insertion of oppositely oriented CTCF recognition sequences restored CTCF and cohesin recruitment, but did not re-establish chromatin loops. We conclude that CTCF binding polarity plays a functional role in the formation of higher-order chromatin structure.


Subject(s)
Chromatin/chemistry , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Animals , Binding Sites , CCCTC-Binding Factor , CRISPR-Cas Systems , Cell Cycle Proteins/metabolism , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/cytology , Mice , Protein Binding , Cohesins
8.
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
9.
Methods ; 170: 17-32, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31351925

ABSTRACT

Chromosome conformation capture (3C) methods measure DNA contact frequencies based on nuclear proximity ligation, to uncover in vivo genomic folding patterns. 4C-seq is a derivative 3C method, designed to search the genome for sequences contacting a selected genomic site of interest. 4C-seq employs inverse PCR and next generation sequencing to amplify, identify and quantify its proximity ligated DNA fragments. It generates high-resolution contact profiles for selected genomic sites based on limited amounts of sequencing reads. 4C-seq can be used to study multiple aspects of genome organization. It primarily serves to identify specific long-range DNA contacts between individual regulatory DNA modules, forming for example regulatory chromatin loops between enhancers and promoters, or architectural chromatin loops between cohesin- and CTCF- associated domain boundaries. Additionally, 4C-seq contact profiles can reveal the contours of contact domains and can identify the structural domains that co-occupy the same nuclear compartment. Here, we present an improved step-by-step protocol for sample preparation and the generation of 4C-seq sequencing libraries, including an optimized PCR and 4C template purification strategy. In addition, a data processing pipeline is provided which processes multiplexed 4C-seq reads directly from FASTQ files and generates files compatible with standard genome browsers for visualization and further statistical analysis of the data such as peak calling using peakC. The protocols and the pipeline presented should readily allow anyone to generate, visualize and interpret their own high resolution 4C contact datasets.


Subject(s)
Chromatin/genetics , Data Analysis , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Chromatin/chemistry , Datasets as Topic , Gene Library , Nucleic Acid Conformation , Regulatory Sequences, Nucleic Acid , Sequence Analysis, DNA/methods , Software
10.
Nature ; 501(7466): 227-31, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23883933

ABSTRACT

It is becoming increasingly clear that the shape of the genome importantly influences transcription regulation. Pluripotent stem cells such as embryonic stem cells were recently shown to organize their chromosomes into topological domains that are largely invariant between cell types. Here we combine chromatin conformation capture technologies with chromatin factor binding data to demonstrate that inactive chromatin is unusually disorganized in pluripotent stem-cell nuclei. We show that gene promoters engage in contacts between topological domains in a largely tissue-independent manner, whereas enhancers have a more tissue-restricted interaction profile. Notably, genomic clusters of pluripotency factor binding sites find each other very efficiently, in a manner that is strictly pluripotent-stem-cell-specific, dependent on the presence of Oct4 and Nanog protein and inducible after artificial recruitment of Nanog to a selected chromosomal site. We conclude that pluripotent stem cells have a unique higher-order genome structure shaped by pluripotency factors. We speculate that this interactome enhances the robustness of the pluripotent state.


Subject(s)
Chromatin/chemistry , Chromatin/metabolism , Chromosome Positioning , Genome/genetics , Imaging, Three-Dimensional , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Binding Sites , Cell Line , Chromatin/genetics , Chromatin Immunoprecipitation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Molecular Imaging , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Organ Specificity , Promoter Regions, Genetic , SOXB1 Transcription Factors/metabolism
11.
PLoS Genet ; 7(9): e1002262, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21931560

ABSTRACT

Translesion DNA synthesis (TLS) is a DNA damage tolerance mechanism in which specialized low-fidelity DNA polymerases bypass replication-blocking lesions, and it is usually associated with mutagenesis. In Saccharomyces cerevisiae a key event in TLS is the monoubiquitination of PCNA, which enables recruitment of the specialized polymerases to the damaged site through their ubiquitin-binding domain. In mammals, however, there is a debate on the requirement for ubiquitinated PCNA (PCNA-Ub) in TLS. We show that UV-induced Rpa foci, indicative of single-stranded DNA (ssDNA) regions caused by UV, accumulate faster and disappear more slowly in Pcna(K164R/K164R) cells, which are resistant to PCNA ubiquitination, compared to Pcna(+/+) cells, consistent with a TLS defect. Direct analysis of TLS in these cells, using gapped plasmids with site-specific lesions, showed that TLS is strongly reduced across UV lesions and the cisplatin-induced intrastrand GG crosslink. A similar effect was obtained in cells lacking Rad18, the E3 ubiquitin ligase which monoubiquitinates PCNA. Consistently, cells lacking Usp1, the enzyme that de-ubiquitinates PCNA exhibited increased TLS across a UV lesion and the cisplatin adduct. In contrast, cells lacking the Rad5-homologs Shprh and Hltf, which polyubiquitinate PCNA, exhibited normal TLS. Knocking down the expression of the TLS genes Rev3L, PolH, or Rev1 in Pcna(K164R/K164R) mouse embryo fibroblasts caused each an increased sensitivity to UV radiation, indicating the existence of TLS pathways that are independent of PCNA-Ub. Taken together these results indicate that PCNA-Ub is required for maximal TLS. However, TLS polymerases can be recruited to damaged DNA also in the absence of PCNA-Ub, and perform TLS, albeit at a significantly lower efficiency and altered mutagenic specificity.


Subject(s)
DNA Repair , DNA Replication , DNA/biosynthesis , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination , Animals , Cisplatin/pharmacology , DNA/drug effects , DNA/genetics , DNA Damage , DNA, Single-Stranded/biosynthesis , DNA, Single-Stranded/genetics , Mice , Mutagenesis , Proliferating Cell Nuclear Antigen/genetics , Ubiquitin/genetics , Ubiquitin/metabolism , Ultraviolet Rays
12.
Cell Genom ; 4(1): 100460, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38190099

ABSTRACT

Single-nucleotide polymorphisms (SNPs) near the ERAP2 gene are associated with various autoimmune conditions, as well as protection against lethal infections. Due to high linkage disequilibrium, numerous trait-associated SNPs are correlated with ERAP2 expression; however, their functional mechanisms remain unidentified. We show by reciprocal allelic replacement that ERAP2 expression is directly controlled by the splice region variant rs2248374. However, disease-associated variants in the downstream LNPEP gene promoter are independently associated with ERAP2 expression. Allele-specific conformation capture assays revealed long-range chromatin contacts between the gene promoters of LNPEP and ERAP2 and showed that interactions were stronger in patients carrying the alleles that increase susceptibility to autoimmune diseases. Replacing the SNPs in the LNPEP promoter by reference sequences lowered ERAP2 expression. These findings show that multiple SNPs act in concert to regulate ERAP2 expression and that disease-associated variants can convert a gene promoter region into a potent enhancer of a distal gene.


Subject(s)
Autoimmune Diseases , Polymorphism, Single Nucleotide , Humans , Polymorphism, Single Nucleotide/genetics , Genetic Predisposition to Disease/genetics , Autoimmune Diseases/genetics , Promoter Regions, Genetic/genetics , Aminopeptidases/genetics
13.
J Exp Med ; 204(8): 1989-98, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17664295

ABSTRACT

B cells use translesion DNA synthesis (TLS) to introduce somatic mutations around genetic lesions caused by activation-induced cytidine deaminase. Monoubiquitination at lysine(164) of proliferating cell nuclear antigen (PCNA(K164)) stimulates TLS. To determine the role of PCNA(K164) modifications in somatic hypermutation, PCNA(K164R) knock-in mice were generated. PCNA(K164R/K164R) mutants are born at a sub-Mendelian frequency. Although PCNA(K164R/K164R) B cells proliferate and class switch normally, the mutation spectrum of hypermutated immunoglobulin (Ig) genes alters dramatically. A strong reduction of mutations at template A/T is associated with a compensatory increase at G/C, which is a phenotype similar to polymerase eta (Poleta) and mismatch repair-deficient B cells. Mismatch recognition, monoubiquitinated PCNA, and Poleta likely cooperate in establishing mutations at template A/T during replication of Ig genes.


Subject(s)
Adenine/chemistry , Cytosine/chemistry , Gene Expression Regulation , Immunoglobulins/genetics , Mutagenesis , Mutation , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/physiology , Animals , B-Lymphocytes/metabolism , Cell Proliferation , DNA-Directed DNA Polymerase/metabolism , Homozygote , Mice , Models, Biological , Phenotype , Ubiquitin/chemistry , Ubiquitin/metabolism
14.
Nat Struct Mol Biol ; 29(6): 563-574, 2022 06.
Article in English | MEDLINE | ID: mdl-35710842

ABSTRACT

Developmental gene expression is often controlled by distal regulatory DNA elements called enhancers. Distant enhancer action is restricted to structural chromosomal domains that are flanked by CTCF-associated boundaries and formed through cohesin chromatin loop extrusion. To better understand how enhancers, genes and CTCF boundaries together form structural domains and control expression, we used a bottom-up approach, building series of active regulatory landscapes in inactive chromatin. We demonstrate here that gene transcription levels and activity over time reduce with increased enhancer distance. The enhancer recruits cohesin to stimulate domain formation and engage flanking CTCF sites in loop formation. It requires cohesin exclusively for the activation of distant genes, not of proximal genes, with nearby CTCF boundaries supporting efficient long-range enhancer action. Our work supports a dual activity model for enhancers: its classic role of stimulating transcription initiation and elongation from target gene promoters and a role of recruiting cohesin for the creation of chromosomal domains, the engagement of CTCF sites in chromatin looping and the activation of distal target genes.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Binding Sites , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Enhancer Elements, Genetic/genetics , Cohesins
15.
Blood ; 114(11): 2280-9, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19608748

ABSTRACT

In the germinal center (GC), B cells proliferate dramatically and diversify their immunoglobulin genes, which increases the risk of malignant transformation. The GC B-cell reaction relies on crosstalk with follicular dendritic cells (FDCs), to which the costimulatory receptor CD137 on FDCs and its ligand on GC B cells potentially contribute. We report that mice deficient for CD137 ligand (CD137L) are predisposed to develop B-cell lymphoma, with an incidence of approximately 60% at 12 months of age. Lymphoma membrane markers were characteristic of GC B cells. Longitudinal histologic analysis identified the GC as site of oncogenic transformation and classified 85% of the malignancies found in approximately 200 mice as GC-derived B-cell lymphoma. To delineate the mechanism underlying lymphomagenesis, gene expression profiles of wild-type and CD137L-deficient GC B cells were compared. CD137L deficiency was associated with enhanced expression of a limited gene set that included Bcl-10 and the GC response regulators Bcl-6, Spi-B, Elf-1, Bach2, and activation-induced cytidine deaminase. Among these are proto-oncogenes that mediate GC B-cell lymphoma development in humans. We conclude that CD137L ordinarily regulates the GC B-cell response and thereby acts as a tumor suppressor.


Subject(s)
4-1BB Ligand , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Germinal Center/metabolism , Lymphoma, B-Cell/metabolism , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Cell CLL-Lymphoma 10 Protein , Basic-Leucine Zipper Transcription Factors/biosynthesis , Basic-Leucine Zipper Transcription Factors/genetics , Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Cytidine Deaminase/biosynthesis , Cytidine Deaminase/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Ephrin-A2/biosynthesis , Ephrin-A2/genetics , Genetic Predisposition to Disease , Humans , Lymphoma, B-Cell/genetics , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6 , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
16.
Blood ; 113(16): 3706-15, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19023113

ABSTRACT

Subjects with X-linked hyper-IgM syndrome (X-HIgM) have a markedly reduced frequency of CD27(+) memory B cells, and their Ig genes have a low level of somatic hypermutation (SHM). To analyze the nature of SHM in X-HIgM, we sequenced 209 nonproductive and 926 productive Ig heavy chain genes. In nonproductive rearrangements that were not subjected to selection, as well as productive rearrangements, most of the mutations were within targeted RGYW, WRCY, WA, or TW motifs (R = purine, Y = pyrimidine, and W = A or T). However, there was significantly decreased targeting of the hypermutable G in RGYW motifs. Moreover, the ratio of transitions to transversions was markedly increased compared with normal. Microarray analysis documented that specific genes involved in SHM, including activation-induced cytidine deaminase (AICDA) and uracil-DNA glycosylase (UNG2), were up-regulated in normal germinal center (GC) B cells, but not induced by CD40 ligation. Similar results were obtained from light chain rearrangements. These results indicate that in the absence of CD40-CD154 interactions, there is a marked reduction in SHM and, specifically, mutations of AICDA-targeted G residues in RGYW motifs along with a decrease in transversions normally related to UNG2 activity.


Subject(s)
B-Lymphocytes/enzymology , Cytidine Deaminase/biosynthesis , DNA Glycosylases/biosynthesis , Gene Expression Regulation, Enzymologic/genetics , Hyper-IgM Immunodeficiency Syndrome, Type 1/genetics , Immunoglobulin Heavy Chains/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Adolescent , Adult , B-Lymphocytes/immunology , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/genetics , CD40 Ligand/immunology , CD40 Ligand/metabolism , Child , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , DNA Glycosylases/genetics , DNA Glycosylases/immunology , DNA Mutational Analysis , Gene Expression Regulation, Enzymologic/immunology , Germinal Center/enzymology , Germinal Center/immunology , Humans , Hyper-IgM Immunodeficiency Syndrome, Type 1/enzymology , Hyper-IgM Immunodeficiency Syndrome, Type 1/immunology , Immunoglobulin Heavy Chains/immunology , Immunologic Capping/genetics , Immunologic Capping/immunology , Immunologic Memory/genetics , Male , Mutation , Somatic Hypermutation, Immunoglobulin/immunology , Up-Regulation/genetics , Up-Regulation/immunology
17.
Nat Commun ; 12(1): 3361, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099699

ABSTRACT

In routine diagnostic pathology, cancer biopsies are preserved by formalin-fixed, paraffin-embedding (FFPE) procedures for examination of (intra-) cellular morphology. Such procedures inadvertently induce DNA fragmentation, which compromises sequencing-based analyses of chromosomal rearrangements. Yet, rearrangements drive many types of hematolymphoid malignancies and solid tumors, and their manifestation is instructive for diagnosis, prognosis, and treatment. Here, we present FFPE-targeted locus capture (FFPE-TLC) for targeted sequencing of proximity-ligation products formed in FFPE tissue blocks, and PLIER, a computational framework that allows automated identification and characterization of rearrangements involving selected, clinically relevant, loci. FFPE-TLC, blindly applied to 149 lymphoma and control FFPE samples, identifies the known and previously uncharacterized rearrangement partners. It outperforms fluorescence in situ hybridization (FISH) in sensitivity and specificity, and shows clear advantages over standard capture-NGS methods, finding rearrangements involving repetitive sequences which they typically miss. FFPE-TLC is therefore a powerful clinical diagnostics tool for accurate targeted rearrangement detection in FFPE specimens.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Lymphoma, B-Cell/genetics , Lymphoma, Non-Hodgkin/genetics , Paraffin Embedding/methods , Tissue Fixation/methods , Translocation, Genetic , Computational Biology/methods , Gene Rearrangement , Genes, bcl-2/genetics , Genes, myc/genetics , Humans , In Situ Hybridization, Fluorescence/methods , Lymphoma, B-Cell/diagnosis , Lymphoma, Non-Hodgkin/diagnosis , Proto-Oncogene Proteins c-bcl-6/genetics , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity
18.
Nat Protoc ; 15(2): 364-397, 2020 02.
Article in English | MEDLINE | ID: mdl-31932773

ABSTRACT

We present the experimental protocol and data analysis toolbox for multi-contact 4C (MC-4C), a new proximity ligation method tailored to study the higher-order chromatin contact patterns of selected genomic sites. Conventional chromatin conformation capture (3C) methods fragment proximity ligation products for efficient analysis of pairwise DNA contacts. By contrast, MC-4C is designed to preserve and collect large concatemers of proximity ligated fragments for long-molecule sequencing on an Oxford Nanopore or Pacific Biosciences platform. Each concatemer of proximity ligation products represents a snapshot topology of a different individual allele, revealing its multi-way chromatin interactions. By inverse PCR with primers specific for a fragment of interest (the viewpoint) and DNA size selection, sequencing is selectively targeted to thousands of different complex interactions containing this viewpoint. A tailored statistical analysis toolbox is able to generate background models and three-way interaction profiles from the same dataset. These profiles can be used to distinguish whether contacts between more than two regulatory sequences are mutually exclusive or, conversely, simultaneously occurring at chromatin hubs. The entire procedure can be completed in 2 w, and requires standard molecular biology and data analysis skills and equipment, plus access to a third-generation sequencing platform.


Subject(s)
Chromatin/chemistry , Chromatin/genetics , Sequence Analysis, DNA/methods , Humans , K562 Cells , Molecular Conformation
19.
Dev Cell ; 48(6): 765-779.e7, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30773489

ABSTRACT

Specialized adult somatic cells, such as cardiomyocytes (CMs), are highly differentiated with poor renewal capacity, an integral reason underlying organ failure in disease and aging. Among the least renewable cells in the human body, CMs renew approximately 1% annually. Consistent with poor CM turnover, heart failure is the leading cause of death. Here, we show that an active version of the Hippo pathway effector YAP, termed YAP5SA, partially reprograms adult mouse CMs to a more fetal and proliferative state. One week after induction, 19% of CMs that enter S-phase do so twice, CM number increases by 40%, and YAP5SA lineage CMs couple to pre-existing CMs. Genomic studies showed that YAP5SA increases chromatin accessibility and expression of fetal genes, partially reprogramming long-lived somatic cells in vivo to a primitive, fetal-like, and proliferative state.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aging/physiology , Chromatin/metabolism , Heart/growth & development , Organogenesis , Phosphoproteins/metabolism , Action Potentials , Animals , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Cycle , Cell Cycle Proteins , Cell Lineage , Cell Proliferation , Diploidy , Enhancer Elements, Genetic/genetics , Gain of Function Mutation/genetics , Gene Expression Regulation, Developmental , Heart Ventricles/anatomy & histology , Mice, Transgenic , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Promoter Regions, Genetic/genetics , Transcription Factor AP-1/metabolism , Transgenes , YAP-Signaling Proteins
20.
Nat Genet ; 50(8): 1151-1160, 2018 08.
Article in English | MEDLINE | ID: mdl-29988121

ABSTRACT

Chromatin folding contributes to the regulation of genomic processes such as gene activity. Existing conformation capture methods characterize genome topology through analysis of pairwise chromatin contacts in populations of cells but cannot discern whether individual interactions occur simultaneously or competitively. Here we present multi-contact 4C (MC-4C), which applies Nanopore sequencing to study multi-way DNA conformations of individual alleles. MC-4C distinguishes cooperative from random and competing interactions and identifies previously missed structures in subpopulations of cells. We show that individual elements of the ß-globin superenhancer can aggregate into an enhancer hub that can simultaneously accommodate two genes. Neighboring chromatin domain loops can form rosette-like structures through collision of their CTCF-bound anchors, as seen most prominently in cells lacking the cohesin-unloading factor WAPL. Here, massive collision of CTCF-anchored chromatin loops is believed to reflect 'cohesin traffic jams'. Single-allele topology studies thus help us understand the mechanisms underlying genome folding and functioning.


Subject(s)
Chromatin/genetics , Enhancer Elements, Genetic/genetics , Alleles , Animals , CCCTC-Binding Factor/genetics , Mice , Nucleic Acid Conformation , Regulatory Sequences, Nucleic Acid/genetics , beta-Globins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL