Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Curr Opin Genet Dev ; 88: 102244, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39146885

ABSTRACT

A number of factors contribute to cell type-specific CTCF chromatin binding, but how they act in concert to determine binding stability and functionality has not been fully elucidated. In this review, we tie together different layers of regulation to provide a holistic view of what is known. What emerges from these studies is a multifaceted system in which DNA sequence, DNA and chromatin accessibility, and cell type-specific transcription factors together contribute to CTCF binding profile and function. We discuss these findings in the light of disease settings in which changes in the chromatin landscape and transcriptional programming can disrupt CTCF's binding profile and involvement in looping.


Subject(s)
CCCTC-Binding Factor , Chromatin , Repressor Proteins , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Humans , Chromatin/genetics , Chromatin/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Protein Binding , Animals , Transcription Factors/metabolism , Transcription Factors/genetics , Gene Expression Regulation , Binding Sites
2.
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
3.
Cell Rep ; 43(8): 114547, 2024 Aug 27.
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.


Subject(s)
Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , T-Lymphocytes, Cytotoxic , Animals , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Cell Nucleus/metabolism , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chronic Disease , Promoter Regions, Genetic/genetics , Genetic Loci
4.
Res Sq ; 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39070636

ABSTRACT

Although only a fraction of CTCF motifs are bound in any cell type, and approximately half of the occupied sites overlap cohesin, the mechanisms underlying cell-type specific attachment and ability to function as a chromatin organizer remain unknown. To investigate the relationship between CTCF and chromatin we applied a combination of imaging, structural and molecular approaches, using a series of brain and cancer associated CTCF mutations that act as CTCF perturbations. We demonstrate that binding and the functional impact of WT and mutant CTCF depend not only on the unique properties of each protein, but also on the genomic context of bound sites. Our studies also highlight the reciprocal relationship between CTCF and chromatin, demonstrating that the unique binding properties of WT and mutant proteins have a distinct impact on accessibility, TF binding, cohesin overlap, chromatin interactivity and gene expression programs, providing insight into their cancer and brain related effects.

5.
bioRxiv ; 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-38370764

ABSTRACT

Here we used a series of CTCF mutations to explore CTCF's relationship with chromatin and its contribution to gene regulation. CTCF's impact depends on the genomic context of bound sites and the unique binding properties of WT and mutant CTCF proteins. Specifically, CTCF's signal strength is linked to changes in accessibility, and the ability to block cohesin is linked to its binding stability. Multivariate modelling reveals that both CTCF and accessibility contribute independently to cohesin binding and insulation, however CTCF signal strength has a stronger effect. CTCF and chromatin have a bidirectional relationship such that at CTCF sites, accessibility is reduced in a cohesin-dependent, mutant specific fashion. In addition, each mutant alters TF binding and accessibility in an indirect manner, changes which impart the most influence on rewiring transcriptional networks and the cell's ability to be reprogrammed. Collectively, the mutant perturbations provide a rich resource for determining CTCF's site-specific effects.

6.
Res Sq ; 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37577603

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 novel cis regulatory elements (CREs) in a 1Mb 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 CRISPRi based scRNA 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.

7.
Nat Rev Mol Cell Biol ; 23(6): 383, 2022 06.
Article in English | MEDLINE | ID: mdl-35352008

Subject(s)
Career Choice
8.
Nat Genet ; 54(2): 202-212, 2022 02.
Article in English | MEDLINE | ID: mdl-35145304

ABSTRACT

CCCTC-binding factor (CTCF) is critical to three-dimensional genome organization. Upon differentiation, CTCF insulates active and repressed genes within Hox gene clusters. We conducted a genome-wide CRISPR knockout (KO) screen to identify genes required for CTCF-boundary activity at the HoxA cluster, complemented by biochemical approaches. Among the candidates, we identified Myc-associated zinc-finger protein (MAZ) as a cofactor in CTCF insulation. MAZ colocalizes with CTCF at chromatin borders and, similar to CTCF, interacts with the cohesin subunit RAD21. MAZ KO disrupts gene expression and local contacts within topologically associating domains. Similar to CTCF motif deletions, MAZ motif deletions lead to derepression of posterior Hox genes immediately after CTCF boundaries upon differentiation, giving rise to homeotic transformations in mouse. Thus, MAZ is a factor contributing to appropriate insulation, gene expression and genomic architecture during development.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Genes, Homeobox , Homeodomain Proteins/genetics , Transcription Factors/metabolism , Animals , CCCTC-Binding Factor/chemistry , CCCTC-Binding Factor/genetics , CRISPR-Cas Systems , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Chromatin/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Gene Editing , Gene Expression , Gene Expression Regulation, Developmental , Mice , Transcription Factors/chemistry , Transcription Factors/genetics
9.
Epigenomics ; 14(6): 327-330, 2022 03.
Article in English | MEDLINE | ID: mdl-35195040

ABSTRACT

In this interview, Professor Jane Skok speaks with Storm Johnson, commissioning editor for Epigenomics, on her work to date in the field of chromosome architecture and regulatory elements. Jane Skok's lab uses sophisticated microscopic techniques to visualize recombination in individual cells, tracing the dynamic changes in chromosome architecture and nuclear location at different stages of this complex process. This line of research unites two lifelong passions: science and art. After completing her PhD in immunology and genetics at the Imperial Cancer Research Fund in Lincoln's Inn Fields, Dr Skok took 12 years off and pursued training in art while caring for her young children. She then returned to science, joining David Gray's lab at Imperial College London as a postdoctoral fellow to study B cell biology and acquired expertise in Mandy Fisher's lab to understand how nuclear organization of the antigen receptor genes regulate V(D)J recombination and allelic exclusion. Dr Skok continued to pursue these questions in her own lab at University College London and elucidated the roles of Pax5, locus contraction and nuclear subcompartmentalization in maintaining allelic exclusion. In 2006, Dr Skok was recruited to New York University School of Medicine, where her lab has revealed the activities of several signaling factors in guiding B cell development and they made the surprising discovery that the RAG proteins and the DNA damage response factor ATM help ensure allelic exclusion at the immunoglobulin gene loci. More recently, those at the Skok lab have turned their attention to understanding how localized and long-range chromatin contacts impact gene regulation in health and disease settings.


Subject(s)
B-Lymphocytes , V(D)J Recombination , Child , Child, Preschool , Chromatin , Epigenomics , Female , Humans
10.
Cancer Discov ; 12(4): 1022-1045, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34911733

ABSTRACT

Resistance to targeted therapies is an important clinical problem in HER2-positive (HER2+) breast cancer. "Drug-tolerant persisters" (DTP), a subpopulation of cancer cells that survive via reversible, nongenetic mechanisms, are implicated in resistance to tyrosine kinase inhibitors (TKI) in other malignancies, but DTPs following HER2 TKI exposure have not been well characterized. We found that HER2 TKIs evoke DTPs with a luminal-like or a mesenchymal-like transcriptome. Lentiviral barcoding/single-cell RNA sequencing reveals that HER2+ breast cancer cells cycle stochastically through a "pre-DTP" state, characterized by a G0-like expression signature and enriched for diapause and/or senescence genes. Trajectory analysis/cell sorting shows that pre-DTPs preferentially yield DTPs upon HER2 TKI exposure. Cells with similar transcriptomes are present in HER2+ breast tumors and are associated with poor TKI response. Finally, biochemical experiments indicate that luminal-like DTPs survive via estrogen receptor-dependent induction of SGK3, leading to rewiring of the PI3K/AKT/mTORC1 pathway to enable AKT-independent mTORC1 activation. SIGNIFICANCE: DTPs are implicated in resistance to anticancer therapies, but their ontogeny and vulnerabilities remain unclear. We find that HER2 TKI-DTPs emerge from stochastically arising primed cells ("pre-DTPs") that engage either of two distinct transcriptional programs upon TKI exposure. Our results provide new insights into DTP ontogeny and potential therapeutic vulnerabilities. This article is highlighted in the In This Issue feature, p. 873.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Phosphatidylinositol 3-Kinases/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction
11.
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
12.
Stem Cell Reports ; 15(6): 1233-1245, 2020 12 08.
Article in English | MEDLINE | ID: mdl-32976761

ABSTRACT

Methylation of histone 3 at lysine 9 (H3K9) constitutes a roadblock for cellular reprogramming. Interference with methyltransferases or activation of demethylases by the cofactor ascorbic acid (AA) facilitates the derivation of induced pluripotent stem cells (iPSCs), but possible interactions between specific methyltransferases and AA treatment remain insufficiently explored. We show that chemical inhibition of the methyltransferases EHMT1 and EHMT2 counteracts iPSC formation in an enhanced reprogramming system in the presence of AA, an effect that is dependent on EHMT1. EHMT inhibition during enhanced reprogramming is associated with rapid loss of H3K9 dimethylation, inefficient downregulation of somatic genes, and failed mesenchymal-to-epithelial transition. Furthermore, transient EHMT inhibition during reprogramming yields iPSCs that fail to efficiently give rise to viable mice upon blastocyst injection. Our observations establish novel functions of H3K9 methyltransferases and suggest that a functional balance between AA-stimulated enzymes and EHMTs supports efficient and less error-prone iPSC reprogramming to pluripotency.


Subject(s)
Cellular Reprogramming , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/enzymology , Animals , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Induced Pluripotent Stem Cells/cytology , Methylation , Mice
14.
Genome Biol ; 21(1): 108, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393311

ABSTRACT

BACKGROUND: Ubiquitously expressed CTCF is involved in numerous cellular functions, such as organizing chromatin into TAD structures. In contrast, its paralog, CTCFL, is normally only present in the testis. However, it is also aberrantly expressed in many cancers. While it is known that shared and unique zinc finger sequences in CTCF and CTCFL enable CTCFL to bind competitively to a subset of CTCF binding sites as well as its own unique locations, the impact of CTCFL on chromosome organization and gene expression has not been comprehensively analyzed in the context of CTCF function. Using an inducible complementation system, we analyze the impact of expressing CTCFL and CTCF-CTCFL chimeric proteins in the presence or absence of endogenous CTCF to clarify the relative and combined contribution of CTCF and CTCFL to chromosome organization and transcription. RESULTS: We demonstrate that the N terminus of CTCF interacts with cohesin which explains the requirement for convergent CTCF binding sites in loop formation. By analyzing CTCF and CTCFL binding in tandem, we identify phenotypically distinct sites with respect to motifs, targeting to promoter/intronic intergenic regions and chromatin folding. Finally, we reveal that the N, C, and zinc finger terminal domains play unique roles in targeting each paralog to distinct binding sites to regulate transcription, chromatin looping, and insulation. CONCLUSION: This study clarifies the unique and combined contribution of CTCF and CTCFL to chromosome organization and transcription, with direct implications for understanding how their co-expression deregulates transcription in cancer.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Animals , Embryonic Stem Cells , Female , Humans , Male , Mice
15.
Curr Opin Genet Dev ; 61: 44-52, 2020 04.
Article in English | MEDLINE | ID: mdl-32334335

ABSTRACT

CTCF plays a key role in organizing chromatin into TAD structures but it can also function as a transcription factor. CTCFL (CTCF-like), the paralog of CTCF, is normally transiently expressed in pre-meiotic male germ cells together with ubiquitously expressed CTCF. It plays a unique role in spermatogenesis by regulating expression of testis-specific genes. Genetic alterations in CTCF and its paralog CTCFL have both been found in numerous cancers, but it remains unknown to what extent CTCFL deregulates transcription on its own or by opposing CTCF. Here, we discuss some of the potential mechanisms by which these two proteins could alter gene regulation and contribute to oncogenic transcriptional programs.


Subject(s)
CCCTC-Binding Factor/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , Transcription Factors/genetics , Chromatin/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasms/pathology , Promoter Regions, Genetic
16.
Nat Genet ; 52(4): 378-387, 2020 04.
Article in English | MEDLINE | ID: mdl-32203468

ABSTRACT

Mutations in genes involved in DNA methylation (DNAme; for example, TET2 and DNMT3A) are frequently observed in hematological malignancies1-3 and clonal hematopoiesis4,5. Applying single-cell sequencing to murine hematopoietic stem and progenitor cells, we observed that these mutations disrupt hematopoietic differentiation, causing opposite shifts in the frequencies of erythroid versus myelomonocytic progenitors following Tet2 or Dnmt3a loss. Notably, these shifts trace back to transcriptional priming skews in uncommitted hematopoietic stem cells. To reconcile genome-wide DNAme changes with specific erythroid versus myelomonocytic skews, we provide evidence in support of differential sensitivity of transcription factors due to biases in CpG enrichment in their binding motif. Single-cell transcriptomes with targeted genotyping showed similar skews in transcriptional priming of DNMT3A-mutated human clonal hematopoiesis bone marrow progenitors. These data show that DNAme shapes the topography of hematopoietic differentiation, and support a model in which genome-wide methylation changes are transduced to differentiation skews through biases in CpG enrichment of the transcription factor binding motif.


Subject(s)
Cell Differentiation/genetics , DNA Methylation/genetics , Hematopoiesis/genetics , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/physiology , Humans , Male , Mice , Mice, Transgenic , Mutation/genetics , Transcription, Genetic/genetics , Transcriptome/genetics
17.
Nat Commun ; 10(1): 4843, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31649247

ABSTRACT

CTCF and cohesin play a key role in organizing chromatin into topologically associating domain (TAD) structures. Disruption of a single CTCF binding site is sufficient to change chromosomal interactions leading to alterations in chromatin modifications and gene regulation. However, the extent to which alterations in chromatin modifications can disrupt 3D chromosome organization leading to transcriptional changes is unknown. In multiple myeloma, a 4;14 translocation induces overexpression of the histone methyltransferase, NSD2, resulting in expansion of H3K36me2 and shrinkage of antagonistic H3K27me3 domains. Using isogenic cell lines producing high and low levels of NSD2, here we find oncogene activation is linked to alterations in H3K27ac and CTCF within H3K36me2 enriched chromatin. A logistic regression model reveals that differentially expressed genes are significantly enriched within the same insulated domain as altered H3K27ac and CTCF peaks. These results identify a bidirectional relationship between 2D chromatin and 3D genome organization in gene regulation.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/genetics , Multiple Myeloma/genetics , Repressor Proteins/genetics , Binding Sites , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression/genetics , Humans , Logistic Models , Cohesins
18.
Nat Commun ; 10(1): 4768, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31628339

ABSTRACT

B-1a cells are long-lived, self-renewing innate-like B cells that predominantly inhabit the peritoneal and pleural cavities. In contrast to conventional B-2 cells, B-1a cells have a receptor repertoire that is biased towards bacterial and self-antigens, promoting a rapid response to infection and clearing of apoptotic cells. Although B-1a cells are known to primarily originate from fetal tissues, the mechanisms by which they arise has been a topic of debate for many years. Here we show that in the fetal liver versus bone marrow environment, reduced IL-7R/STAT5 levels promote immunoglobulin kappa gene recombination at the early pro-B cell stage. As a result, differentiating B cells can directly generate a mature B cell receptor (BCR) and bypass the requirement for a pre-BCR and pairing with surrogate light chain. This 'alternate pathway' of development enables the production of B cells with self-reactive, skewed specificity receptors that are peculiar to the B-1a compartment. Together our findings connect seemingly opposing lineage and selection models of B-1a cell development and explain how these cells acquire their unique properties.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Pre-B Cell Receptors/immunology , Receptors, Antigen, B-Cell/immunology , Animals , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Differentiation/genetics , Immunoglobulin Light Chains, Surrogate/genetics , Immunoglobulin Light Chains, Surrogate/immunology , Immunoglobulin Light Chains, Surrogate/metabolism , Liver/embryology , Liver/immunology , Liver/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Pre-B Cell Receptors/genetics , Pre-B Cell Receptors/metabolism , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism
19.
Cell Rep ; 26(1): 108-118.e4, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30605667

ABSTRACT

B cell development is a highly regulated process that requires stepwise rearrangement of immunoglobulin genes to generate a functional B cell receptor (BCR). The polycomb group protein BMI1 is required for B cell development, but its function in developing B cells remains poorly defined. We demonstrate that BMI1 functions in a cell-autonomous manner at two stages during early B cell development. First, loss of BMI1 results in a differentiation block at the pro-B cell to pre-B cell transition due to the inability of BMI1-deficient cells to transcribe newly rearranged Igh genes. Accordingly, introduction of a pre-rearranged Igh allele partially restored B cell development in Bmi1-/- mice. In addition, BMI1 is required to prevent premature p53 signaling, and as a consequence, Bmi1-/- large pre-B cells fail to properly proliferate. Altogether, our results clarify the role of BMI1 in early B cell development and uncover an unexpected function of BMI1 during VDJ recombination.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Gene Rearrangement, B-Lymphocyte , Genes, Immunoglobulin , Polycomb Repressive Complex 1/immunology , Proto-Oncogene Proteins/immunology , Tumor Suppressor Protein p53/immunology , Animals , Cell Differentiation/physiology , Female , Gene Expression , Male , Mice , Mice, Knockout , Polycomb Repressive Complex 1/deficiency , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics
20.
Genome Biol ; 19(1): 216, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30541598

ABSTRACT

BACKGROUND: The organization of chromatin in the nucleus plays an essential role in gene regulation. About half of the mammalian genome comprises transposable elements. Given their repetitive nature, reads associated with these elements are generally discarded or randomly distributed among elements of the same type in genome-wide analyses. Thus, it is challenging to identify the activities and properties of individual transposons. As a result, we only have a partial understanding of how transposons contribute to chromatin folding and how they impact gene regulation. RESULTS: Using PCR and Capture-based chromosome conformation capture (3C) approaches, collectively called 4Tran, we take advantage of the repetitive nature of transposons to capture interactions from multiple copies of endogenous retrovirus (ERVs) in the human and mouse genomes. With 4Tran-PCR, reads are selectively mapped to unique regions in the genome. This enables the identification of transposable element interaction profiles for individual ERV families and integration events specific to particular genomes. With this approach, we demonstrate that transposons engage in long-range intra-chromosomal interactions guided by the separation of chromosomes into A and B compartments as well as topologically associated domains (TADs). In contrast to 4Tran-PCR, Capture-4Tran can uniquely identify both ends of an interaction that involve retroviral repeat sequences, providing a powerful tool for uncovering the individual transposable element insertions that interact with and potentially regulate target genes. CONCLUSIONS: 4Tran provides new insight into the manner in which transposons contribute to chromosome architecture and identifies target genes that transposable elements can potentially control.


Subject(s)
DNA Transposable Elements , Gene Expression Regulation , Genomics/methods , Animals , Endogenous Retroviruses , Humans , Mice , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL