Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nucleic Acids Res ; 51(20): 11024-11039, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37823593

ABSTRACT

The promyelocytic leukemia (PML) protein organizes nuclear aggregates known as PML nuclear bodies (PML-NBs), where many transcription factors localize to be regulated. In addition, associations of PML and PML-NBs with chromatin are described in various cell types, further implicating PML in transcriptional regulation. However, a complete understanding of the functional consequences of PML association to DNA in cellular contexts where it promotes relevant phenotypes is still lacking. We examined PML chromatin association in triple-negative breast cancer (TNBC) cell lines, where it exerts important oncogenic functions. We find that PML associates discontinuously with large heterochromatic PML-associated domains (PADs) that contain discrete gene-rich euchromatic sub-domains locally depleted of PML. PML promotes heterochromatic organization in PADs and expression of pro-metastatic genes embedded in these sub-domains. Importantly, this occurs outside PML-NBs, suggesting that nucleoplasmic PML exerts a relevant gene regulatory function. We also find that PML plays indirect regulatory roles in TNBC cells by promoting the expression of pro-metastatic genes outside PADs. Our findings suggest that PML is an important transcriptional regulator of pro-oncogenic metagenes in TNBC cells, via transcriptional regulation and epigenetic organization of heterochromatin domains that embed regions of local transcriptional activity.


Subject(s)
Chromatin , Triple Negative Breast Neoplasms , Humans , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , Epigenesis, Genetic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor
2.
Nucleic Acids Res ; 50(8): 4500-4514, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35451487

ABSTRACT

Histone H3.3 is an H3 variant which differs from the canonical H3.1/2 at four residues, including a serine residue at position 31 which is evolutionarily conserved. The H3.3 S31 residue is phosphorylated (H3.3 S31Ph) at heterochromatin regions including telomeres and pericentric repeats. However, the role of H3.3 S31Ph in these regions remains unknown. In this study, we find that H3.3 S31Ph regulates heterochromatin accessibility at telomeres during replication through regulation of H3K9/K36 histone demethylase KDM4B. In mouse embryonic stem (ES) cells, substitution of S31 with an alanine residue (H3.3 A31 -phosphorylation null mutant) results in increased KDM4B activity that removes H3K9me3 from telomeres. In contrast, substitution with a glutamic acid (H3.3 E31, mimics S31 phosphorylation) inhibits KDM4B, leading to increased H3K9me3 and DNA damage at telomeres. H3.3 E31 expression also increases damage at other heterochromatin regions including the pericentric heterochromatin and Y chromosome-specific satellite DNA repeats. We propose that H3.3 S31Ph regulation of KDM4B is required to control heterochromatin accessibility of repetitive DNA and preserve chromatin integrity.


Subject(s)
Heterochromatin , Histones , Animals , Mice , Histones/genetics , Histones/metabolism , Heterochromatin/genetics , Histone Demethylases/metabolism , Phosphorylation , Chromatin Assembly and Disassembly
3.
BMC Genomics ; 23(1): 658, 2022 Sep 17.
Article in English | MEDLINE | ID: mdl-36115964

ABSTRACT

BACKGROUND: LncRNAs are tissue-specific and emerge as important regulators of various biological processes and as disease biomarkers. HOTAIR is a well-established pro-oncogenic lncRNA which has been attributed a variety of functions in cancer and native contexts. However, a lack of an exhaustive, cell type-specific annotation questions whether HOTAIR functions are supported by the expression of multiple isoforms. RESULTS: Using a capture long-read sequencing approach, we characterize HOTAIR isoforms expressed in human primary adipose stem cells. We find HOTAIR isoforms population displays varied splicing patterns, frequently leading to the exclusion or truncation of canonical LSD1 and PRC2 binding domains. We identify a highly cell type-specific HOTAIR isoform pool regulated by distinct promoter usage, and uncover a shift in the HOTAIR TSS usage that modulates the balance of HOTAIR isoforms at differentiation onset. CONCLUSION: Our results highlight the complexity and cell type-specificity of HOTAIR isoforms and open perspectives on functional implications of these variants and their balance to key cellular processes.


Subject(s)
Cell Differentiation , Protein Isoforms , RNA, Long Noncoding , Biomarkers , Cell Differentiation/genetics , Histone Demethylases , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
4.
BMC Genomics ; 22(1): 499, 2021 Jul 03.
Article in English | MEDLINE | ID: mdl-34217222

ABSTRACT

BACKGROUND: Mechanisms underlying genome 3D organization and domain formation in the mammalian nucleus are not completely understood. Multiple processes such as transcriptional compartmentalization, DNA loop extrusion and interactions with the nuclear lamina dynamically act on chromatin at multiple levels. Here, we explore long-range interaction patterns between topologically associated domains (TADs) in several cell types. RESULTS: We find that TAD long-range interactions are connected to many key features of chromatin organization, including open and closed compartments, compaction and loop extrusion processes. Domains that form large TAD cliques tend to be repressive across cell types, when comparing gene expression, LINE/SINE repeat content and chromatin subcompartments. Further, TADs in large cliques are larger in genomic size, less dense and depleted of convergent CTCF motifs, in contrast to smaller and denser TADs formed by a loop extrusion process. CONCLUSIONS: Our results shed light on the organizational principles that govern repressive and active domains in the human genome.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin , Animals , Chromosomes , Gene Expression , Genome, Human , Humans
5.
EMBO J ; 36(19): 2829-2843, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28814448

ABSTRACT

The development of many sporadic cancers is directly initiated by carcinogen exposure. Carcinogens induce malignancies by creating DNA lesions (i.e., adducts) that can result in mutations if left unrepaired. Despite this knowledge, there has been remarkably little investigation into the regulation of susceptibility to acquire DNA lesions. In this study, we present the first quantitative human genome-wide map of DNA lesions induced by ultraviolet (UV) radiation, the ubiquitous carcinogen in sunlight that causes skin cancer. Remarkably, the pattern of carcinogen susceptibility across the genome of primary cells significantly reflects mutation frequency in malignant melanoma. Surprisingly, DNase-accessible euchromatin is protected from UV, while lamina-associated heterochromatin at the nuclear periphery is vulnerable. Many cancer driver genes have an intrinsic increase in carcinogen susceptibility, including the BRAF oncogene that has the highest mutation frequency in melanoma. These findings provide a genome-wide snapshot of DNA injuries at the earliest stage of carcinogenesis. Furthermore, they identify carcinogen susceptibility as an origin of genome instability that is regulated by nuclear architecture and mirrors mutagenesis in cancer.


Subject(s)
Carcinogens/toxicity , Cell Transformation, Neoplastic , Drug Resistance/genetics , Genomic Instability/drug effects , Genomic Instability/genetics , Mutagenesis , Base Sequence/physiology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cells, Cultured , DNA Damage , Drug Resistance/drug effects , Epigenesis, Genetic/drug effects , Humans , Melanoma/etiology , Melanoma/genetics , Mutagenesis/drug effects , Mutagenesis/genetics , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Ultraviolet Rays , Melanoma, Cutaneous Malignant
6.
Development ; 145(1)2018 01 03.
Article in English | MEDLINE | ID: mdl-29158440

ABSTRACT

At zygotic genome activation (ZGA), changes in chromatin structure are associated with new transcription immediately following the maternal-to-zygotic transition (MZT). The nuclear architectural proteins cohesin and CCCTC-binding factor (CTCF) contribute to chromatin structure and gene regulation. We show here that normal cohesin function is important for ZGA in zebrafish. Depletion of the cohesin subunit Rad21 delays ZGA without affecting cell cycle progression. In contrast, CTCF depletion has little effect on ZGA, whereas complete abrogation is lethal. Genome-wide analysis of Rad21 binding reveals a change in distribution from pericentromeric satellite DNA and other locations, including the miR-430 locus (the products of which are responsible for maternal transcript degradation), to genes, as embryos progress through the MZT. After MZT, a subset of Rad21 binding overlaps the pioneer factor Pou5f3, which activates early expressed genes. Rad21 depletion disrupts the formation of nucleoli and RNA polymerase II foci, suggestive of global defects in chromosome architecture. We propose that Rad21/cohesin redistribution to active areas of the genome is key to the establishment of chromosome organization and the embryonic developmental program.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , MicroRNAs/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zygote/metabolism , Animals , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Genome/physiology , Genome-Wide Association Study , MicroRNAs/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , Zygote/cytology , Cohesins
7.
Stem Cells ; 38(1): 146-159, 2020 01.
Article in English | MEDLINE | ID: mdl-31502731

ABSTRACT

Clinical-grade mesenchymal stromal cells (MSCs) can be expanded from bone marrow and adipose tissue to treat inflammatory diseases and degenerative disorders. However, the influence of their tissue of origin on their functional properties, including their immunosuppressive activity, remains unsolved. In this study, we produced paired bone marrow-derived mesenchymal stromal cell (BM-MSC) and adipose-derived stromal cell (ASC) batches from 14 healthy donors. We then compared them using transcriptomic, phenotypic, and functional analyses and validated our results on purified native MSCs to infer which differences were really endowed by tissue of origin. Cultured MSCs segregated together owing to their tissue of origin based on their gene expression profile analyzed using differential expression and weighted gene coexpression network analysis. This translated into distinct immune-related gene signatures, phenotypes, and functional cell interactions. Importantly, sorted native BM-MSCs and ASCs essentially displayed the same distinctive patterns than their in vitro-expanded counterparts. As a whole, ASCs exhibited an immune profile consistent with a stronger inhibition of immune response and a lower immunogenicity, supporting the use of adipose tissue as a valuable source for clinical applications.


Subject(s)
Mesenchymal Stem Cells/metabolism , Transcriptome/genetics , Adult , Cell Differentiation , Cells, Cultured , Female , Humans , Male , Middle Aged , Phenotype , Young Adult
8.
Mol Reprod Dev ; 88(3): 187-200, 2021 03.
Article in English | MEDLINE | ID: mdl-33634579

ABSTRACT

In this study, the complexity of chromatin integrity was investigated in frozen-thawed semen samples from 37 sires with contrasting fertility, expressed as 56-day non-return rates (NR56). Protamine deficiency, thiols, and disulfide bonds were assessed and compared with previously published data for DNA fragmentation index (DFI) and high DNA stainability (HDS). In addition, in vitro embryo development and sperm DNA methylation were assessed using semen samples from 16 of these bulls. The percentages of DFI and HDS were negatively associated with NR56 and cleavage rate and positively associated with sperm protamine deficiency (p < 0.05). Significant differences in cleavage and blastocyst rates were observed between bulls of high and low NR56. However, once fertilization occurred, further development into blastocysts was not associated with NR56. The differential methylation analysis showed that spermatozoa from bulls of low NR56 were hypermethylated compared to bulls of high NR56. Pathway analysis showed that genes annotated to differentially methylated cytosines could participate in different biological pathways and have important biological roles related to bull fertility. In conclusion, sperm cells from Norwegian Red bulls of inferior fertility have less compact chromatin structure, higher levels of DNA damage, and are hypermethylated compared with bulls of superior fertility.


Subject(s)
Chromatin/metabolism , DNA Methylation , Fertility/physiology , Spermatozoa/metabolism , Animals , Cattle , DNA Fragmentation , Embryonic Development/physiology , Male , Semen Analysis , Semen Preservation
9.
Proc Natl Acad Sci U S A ; 115(18): 4737-4742, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29669917

ABSTRACT

ATRX (alpha thalassemia/mental retardation X-linked) complexes with DAXX to deposit histone variant H3.3 into repetitive heterochromatin. Recent genome sequencing studies in cancers have revealed mutations in ATRX and their association with ALT (alternative lengthening of telomeres) activation. Here we report depletion of ATRX in mouse ES cells leads to selective loss in ribosomal RNA gene (rDNA) copy number. Supporting this, ATRX-mutated human ALT-positive tumors also show a substantially lower rDNA copy than ALT-negative tumors. Further investigation shows that the rDNA copy loss and repeat instability are caused by a disruption in H3.3 deposition and thus a failure in heterochromatin formation at rDNA repeats in the absence of ATRX. We also find that ATRX-depleted cells are reduced in ribosomal RNA transcription output and show increased sensitivity to RNA polymerase I (Pol I) transcription inhibitor CX5461. In addition, human ALT-positive cancer cell lines are also more sensitive to CX5461 treatment. Our study provides insights into the contribution of ATRX loss of function to tumorigenesis through the loss of rDNA stability and suggests the therapeutic potential of targeting Pol I transcription in ALT cancers.


Subject(s)
DNA, Neoplasm/metabolism , DNA, Ribosomal/metabolism , Gene Dosage , Mutation , Neoplasm Proteins/metabolism , Neoplasms/metabolism , X-linked Nuclear Protein/metabolism , Benzothiazoles/pharmacology , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA, Ribosomal/genetics , Genomic Instability , Humans , Naphthyridines/pharmacology , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , RNA Polymerase I/antagonists & inhibitors , RNA Polymerase I/genetics , RNA Polymerase I/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , X-linked Nuclear Protein/genetics
10.
Hum Mol Genet ; 27(8): 1447-1459, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29438482

ABSTRACT

The p.R482W hotspot mutation in A-type nuclear lamins causes familial partial lipodystrophy of Dunnigan-type (FPLD2), a lipodystrophic syndrome complicated by early onset atherosclerosis. Molecular mechanisms underlying endothelial cell dysfunction conferred by the lamin A mutation remain elusive. However, lamin A regulates epigenetic developmental pathways and mutations could perturb these functions. Here, we demonstrate that lamin A R482W elicits endothelial differentiation defects in a developmental model of FPLD2. Genome modeling in fibroblasts from patients with FPLD2 caused by the lamin A R482W mutation reveals repositioning of the mesodermal regulator T/Brachyury locus towards the nuclear center relative to normal fibroblasts, suggesting enhanced activation propensity of the locus in a developmental model of FPLD2. Addressing this issue, we report phenotypic and transcriptional alterations in mesodermal and endothelial differentiation of induced pluripotent stem cells we generated from a patient with R482W-associated FPLD2. Correction of the LMNA mutation ameliorates R482W-associated phenotypes and gene expression. Transcriptomics links endothelial differentiation defects to decreased Polycomb-mediated repression of the T/Brachyury locus and over-activation of T target genes. Binding of the Polycomb repressor complex 2 to T/Brachyury is impaired by the mutated lamin A network, which is unable to properly associate with the locus. This leads to a deregulation of vascular gene expression over time. By connecting a lipodystrophic hotspot lamin A mutation to a disruption of early mesodermal gene expression and defective endothelial differentiation, we propose that the mutation rewires the fate of several lineages, resulting in multi-tissue pathogenic phenotypes.


Subject(s)
Endothelial Cells/metabolism , Fetal Proteins/genetics , Gene Expression Regulation, Developmental , Lamin Type A/genetics , Lipodystrophy, Familial Partial/genetics , Polycomb-Group Proteins/genetics , T-Box Domain Proteins/genetics , Adolescent , Adult , Case-Control Studies , Cell Differentiation/genetics , Cell Lineage/genetics , Endothelial Cells/pathology , Female , Fetal Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Regulatory Networks , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Lamin Type A/metabolism , Lipodystrophy, Familial Partial/metabolism , Lipodystrophy, Familial Partial/pathology , Male , Mesoderm/metabolism , Mesoderm/pathology , Middle Aged , Mutation , Polycomb-Group Proteins/metabolism , Primary Cell Culture , Protein Binding , Signal Transduction , T-Box Domain Proteins/metabolism
11.
Genome Res ; 27(6): 913-921, 2017 06.
Article in English | MEDLINE | ID: mdl-28341773

ABSTRACT

Maintenance of chromatin homeostasis involves proper delivery of histone variants to the genome. The interplay between different chaperones regulating the supply of histone variants to distinct chromatin domains remains largely undeciphered. We report a role of promyelocytic leukemia (PML) protein in the routing of histone variant H3.3 to chromatin and in the organization of megabase-size heterochromatic PML-associated domains that we call PADs. Loss of PML alters the heterochromatic state of PADs by shifting the histone H3 methylation balance from K9me3 to K27me3. Loss of PML impairs deposition of H3.3 by ATRX and DAXX in PADs but preserves the H3.3 loading function of HIRA in these regions. Our results unveil an unappreciated role of PML in the large-scale organization of chromatin and demonstrate a PML-dependent role of ATRX/DAXX in the deposition of H3.3 in PADs. Our data suggest that H3.3 loading by HIRA and ATRX-dependent H3K27 trimethylation constitute mechanisms ensuring maintenance of heterochromatin when the integrity of these domains is compromised.


Subject(s)
Carrier Proteins/genetics , Heterochromatin/metabolism , Histones/genetics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Nucleosomes/metabolism , Promyelocytic Leukemia Protein/genetics , X-linked Nuclear Protein/genetics , Animals , Carrier Proteins/metabolism , Chromatin Assembly and Disassembly , Co-Repressor Proteins , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescence Recovery After Photobleaching , Gene Expression Regulation , Heterochromatin/ultrastructure , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones , Nuclear Proteins/metabolism , Nucleosomes/ultrastructure , Promyelocytic Leukemia Protein/metabolism , Signal Transduction , X-linked Nuclear Protein/metabolism
12.
J Pathol ; 249(4): 509-522, 2019 12.
Article in English | MEDLINE | ID: mdl-31372995

ABSTRACT

The mechanisms by which lamin A/C in CD4+ T-cells control intestinal homeostasis and can cause inflammatory bowel disease (IBD) are unknown. Here, we explore lamin A/C in a mouse model of IBD. Adoptive transfer to Rag1-/- mice of Lmna-/- CD4+ T-cells, which have enhanced regulatory T-cells (Treg) differentiation and function, induced less severe IBD than wild-type T-cells. Lamin A/C deficiency in CD4+ T-cells enhanced transcription of the Treg master regulator FOXP3, thus promoting Treg differentiation, and reduced Th1 polarization, due to epigenetic changes in the Th1 master regulator T-bet. In mesenteric lymph nodes, retinoic acid (RA) released by CD103+ dendritic cells downregulated lamin A/C in CD4+ T-cells, enhancing Treg differentiation. However, non-RA-producing CD103- dendritic cells predominated in peripheral lymph nodes, facilitating lamin A/C expression in CD4+ T-cells and therefore Th1 differentiation. Our findings establish lamin A/C as a key regulator of Th differentiation in physiological conditions and show it as a potential immune-regulatory target in IBD. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cell Differentiation , Colitis/prevention & control , Colon/metabolism , Lamin Type A/deficiency , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Adoptive Transfer , Animals , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lamin Type A/genetics , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice, Knockout , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Th1 Cells/immunology , Tretinoin/metabolism
13.
Biochem Biophys Res Commun ; 511(4): 882-888, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30850162

ABSTRACT

Targeted deposition of histone variant H3.3 into chromatin is paramount for proper regulation of chromatin integrity, particularly in heterochromatic regions including repeats. We have recently shown that the promyelocytic leukemia (PML) protein prevents H3.3 from being deposited in large heterochromatic PML-associated domains (PADs). However, to what extent PML modulates H3.3 loading on chromatin in other areas of the genome remains unexplored. Here, we examined the impact of PML on targeting of H3.3 to genes and repeat regions that reside outside PADs. We show that loss of PML increases H3.3 deposition in subtelomeric, telomeric, pericentric and centromeric repeats in mouse embryonic fibroblasts, while other repeat classes are not affected. Expression of major satellite, minor satellite and telomeric non-coding transcripts is altered in Pml-null cells. In particular, telomeric Terra transcripts are strongly upregulated, in concordance with a marked reduction in H4K20me3 at these sites. Lastly, for most genes H3.3 enrichment or gene expression outcomes are independent of PML. Our data argue towards the importance of a PML-H3.3 axis in preserving a heterochromatin state at centromeres and telomeres.


Subject(s)
Centromere/metabolism , Fibroblasts/metabolism , Histones/metabolism , Promyelocytic Leukemia Protein/metabolism , Telomere/metabolism , Animals , Cells, Cultured , Heterochromatin/metabolism , Mice
14.
Arterioscler Thromb Vasc Biol ; 38(4): 854-869, 2018 04.
Article in English | MEDLINE | ID: mdl-29449332

ABSTRACT

OBJECTIVE: Endothelial upregulation of adhesion molecules serves to recruit leukocytes to inflammatory sites and appears to be promoted by NOTCH1; however, current models based on interactions between active NOTCH1 and NF-κB components cannot explain the transcriptional selectivity exerted by NOTCH1 in this context. APPROACH AND RESULTS: Observing that Cre/Lox-induced conditional mutations of endothelial Notch modulated inflammation in murine contact hypersensitivity, we found that IL (interleukin)-1ß stimulation induced rapid recruitment of RELA (v-rel avian reticuloendotheliosis viral oncogene homolog A) to genomic sites occupied by NOTCH1-RBPJ (recombination signal-binding protein for immunoglobulin kappa J region) and that NOTCH1 knockdown reduced histone H3K27 acetylation at a subset of NF-κB-directed inflammatory enhancers. CONCLUSIONS: Our findings reveal that NOTCH1 signaling supports the expression of a subset of inflammatory genes at the enhancer level and demonstrate how key signaling pathways converge on chromatin to coordinate the transition to an infla mmatory endothelial phenotype.


Subject(s)
Endothelial Cells/drug effects , Histones/metabolism , Inflammation/prevention & control , Interleukin-1beta/pharmacology , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Acetylation , Animals , Appendicitis/metabolism , Appendicitis/pathology , Cells, Cultured , Dermatitis, Contact/genetics , Dermatitis, Contact/metabolism , Dermatitis, Contact/pathology , Dipeptides/pharmacology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptor, Notch1/genetics , Signal Transduction/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
15.
Nucleic Acids Res ; 45(10): 5691-5706, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28334823

ABSTRACT

Histone chaperones prevent promiscuous histone interactions before chromatin assembly. They guarantee faithful deposition of canonical histones and functionally specialized histone variants into chromatin in a spatial- and temporally-restricted manner. Here, we identify the binding partners of the primate-specific and H3.3-related histone variant H3.Y using several quantitative mass spectrometry approaches, and biochemical and cell biological assays. We find the HIRA, but not the DAXX/ATRX, complex to recognize H3.Y, explaining its presence in transcriptionally active euchromatic regions. Accordingly, H3.Y nucleosomes are enriched in the transcription-promoting FACT complex and depleted of repressive post-translational histone modifications. H3.Y mutational gain-of-function screens reveal an unexpected combinatorial amino acid sequence requirement for histone H3.3 interaction with DAXX but not HIRA, and for H3.3 recruitment to PML nuclear bodies. We demonstrate the importance and necessity of specific H3.3 core and C-terminal amino acids in discriminating between distinct chaperone complexes. Further, chromatin immunoprecipitation sequencing experiments reveal that in contrast to euchromatic HIRA-dependent deposition sites, human DAXX/ATRX-dependent regions of histone H3 variant incorporation are enriched in heterochromatic H3K9me3 and simple repeat sequences. These data demonstrate that H3.Y's unique amino acids allow a functional distinction between HIRA and DAXX binding and its consequent deposition into open chromatin.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Histone Chaperones/genetics , Histone Code , Histones/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Chromatin/chemistry , Chromatin/metabolism , Co-Repressor Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , HeLa Cells , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microsatellite Repeats , Molecular Chaperones , Nuclear Proteins/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Primary Cell Culture , Protein Binding , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
16.
Genome Res ; 25(12): 1825-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26359231

ABSTRACT

Dynamic interactions of nuclear lamins with chromatin through lamin-associated domains (LADs) contribute to spatial arrangement of the genome. Here, we provide evidence for prepatterning of differentiation-driven formation of lamin A/C LADs by domains of histone H2B modified on serine 112 by the nutrient sensor O-linked N-acetylglucosamine (H2BS112GlcNAc), which we term GADs. We demonstrate a two-step process of lamin A/C LAD formation during in vitro adipogenesis, involving spreading of lamin A/C-chromatin interactions in the transition from progenitor cell proliferation to cell-cycle arrest, and genome-scale redistribution of these interactions through a process of LAD exchange within hours of adipogenic induction. Lamin A/C LADs are found both in active and repressive chromatin contexts that can be influenced by cell differentiation status. De novo formation of adipogenic lamin A/C LADs occurs nonrandomly on GADs, which consist of megabase-size intergenic and repressive chromatin domains. Accordingly, whereas predifferentiation lamin A/C LADs are gene-rich, post-differentiation LADs harbor repressive features reminiscent of lamin B1 LADs. Release of lamin A/C from genes directly involved in glycolysis concurs with their transcriptional up-regulation after adipogenic induction, and with downstream elevations in H2BS112GlcNAc levels and O-GlcNAc cycling. Our results unveil an epigenetic prepatterning of adipogenic LADs by GADs, suggesting a coupling of developmentally regulated lamin A/C-genome interactions to a metabolically sensitive chromatin modification.


Subject(s)
Cell Differentiation , Chromatin/metabolism , Histones/metabolism , Lamin Type A/metabolism , Acetylation , Adipogenesis , Chromatin/genetics , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Gene Expression Regulation , Glycolysis/genetics , Glycosylation , High-Throughput Nucleotide Sequencing , Histones/chemistry , Humans , Protein Binding , Protein Interaction Domains and Motifs
17.
Hum Mol Genet ; 24(7): 2096-109, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25524705

ABSTRACT

Nuclear lamins are involved in many cellular functions due to their ability to bind numerous partners including chromatin and transcription factors, and affect their properties. Dunnigan type familial partial lipodystrophy (FPLD2; OMIM#151660) is caused in most cases by the A-type lamin R482W mutation. We report here that the R482W mutation affects the regulatory activity of sterol response element binding protein 1 (SREBP1), a transcription factor that regulates hundreds of genes involved in lipid metabolism and adipocyte differentiation. Using in situ proximity ligation assays (PLA), reporter assays and biochemical and transcriptomic approaches, we show that interactions of SREBP1 with lamin A and lamin C occur at the nuclear periphery and in the nucleoplasm. These interactions involve the Ig-fold of A-type lamins and are favored upon SREBP1 binding to its DNA target sequences. We show that SREBP1, LMNA and sterol response DNA elements form ternary complexes in vitro. In addition, overexpression of A-type lamins reduces transcriptional activity of SREBP1. In contrast, both overexpression of LMNA R482W in primary human preadipocytes and endogenous expression of A-type lamins R482W in FPLD2 patient fibroblasts, reduce A-type lamins-SREBP1 in situ interactions and upregulate a large number of SREBP1 target genes. As this LMNA mutant was previously shown to inhibit adipogenic differentiation, we propose that deregulation of SREBP1 by mutated A-type lamins constitutes one underlying mechanism of the physiopathology of FPLD2. Our data suggest that SREBP1 targeting molecules could be considered in a therapeutic context.


Subject(s)
Amino Acid Substitution , Lamin Type A/genetics , Lipodystrophy, Familial Partial/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Adult , Female , Humans , Lamin Type A/metabolism , Lipodystrophy, Familial Partial/genetics , Male , Middle Aged , Mutation, Missense , Protein Binding , Sterol Regulatory Element Binding Protein 1/genetics , Young Adult
18.
Genome Res ; 24(10): 1584-94, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25049225

ABSTRACT

Histone variant H3.3 is deposited in chromatin at active sites, telomeres, and pericentric heterochromatin by distinct chaperones, but the mechanisms of regulation and coordination of chaperone-mediated H3.3 loading remain largely unknown. We show here that the chromatin-associated oncoprotein DEK regulates differential HIRA- and DAAX/ATRX-dependent distribution of H3.3 on chromosomes in somatic cells and embryonic stem cells. Live cell imaging studies show that nonnucleosomal H3.3 normally destined to PML nuclear bodies is re-routed to chromatin after depletion of DEK. This results in HIRA-dependent widespread chromatin deposition of H3.3 and H3.3 incorporation in the foci of heterochromatin in a process requiring the DAXX/ATRX complex. In embryonic stem cells, loss of DEK leads to displacement of PML bodies and ATRX from telomeres, redistribution of H3.3 from telomeres to chromosome arms and pericentric heterochromatin, induction of a fragile telomere phenotype, and telomere dysfunction. Our results indicate that DEK is required for proper loading of ATRX and H3.3 on telomeres and for telomeric chromatin architecture. We propose that DEK acts as a "gatekeeper" of chromatin, controlling chromatin integrity by restricting broad access to H3.3 by dedicated chaperones. Our results also suggest that telomere stability relies on mechanisms ensuring proper histone supply and routing.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Oncogene Proteins/metabolism , Telomere/metabolism , Animals , Cell Line , Chromatin Assembly and Disassembly , Embryonic Stem Cells/metabolism , Humans , Mice , Poly-ADP-Ribose Binding Proteins
19.
Nucleic Acids Res ; 43(5): 2603-14, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25690891

ABSTRACT

Human ALT cancers show high mutation rates in ATRX and DAXX. Although it is well known that the absence of ATRX/DAXX disrupts H3.3 deposition at heterochromatin, its impact on H3.3 deposition and post-translational modification in the global genome remains unclear. Here, we explore the dynamics of phosphorylated H3.3 serine 31 (H3.3S31ph) in human ALT cancer cells. While H3.3S31ph is found only at pericentric satellite DNA repeats during mitosis in most somatic human cells, a high level of H3.3S31ph is detected on the entire chromosome in ALT cells, attributable to an elevated CHK1 activity in these cells. Drug inhibition of CHK1 activity during mitosis and expression of mutant H3.3S31A in these ALT cells result in a decrease in H3.3S31ph levels accompanied with increased levels of phosphorylated H2AX serine 139 on chromosome arms and at the telomeres. Furthermore, the inhibition of CHK1 activity in these cells also reduces cell viability. Our findings suggest a novel role of CHK1 as an H3.3S31 kinase, and that CHK1-mediated H3.3S31ph plays an important role in the maintenance of chromatin integrity and cell survival in ALT cancer cells.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Histones/metabolism , Protein Kinases/metabolism , Blotting, Western , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/genetics , Checkpoint Kinase 1 , Chromatin/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , HT29 Cells , HeLa Cells , Histones/genetics , Humans , Microscopy, Fluorescence , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Protein Kinases/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Serine/genetics , Serine/metabolism , Telomere/genetics , Telomere/metabolism , X-linked Nuclear Protein
20.
Nucleic Acids Res ; 43(21): 10227-37, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26304540

ABSTRACT

In addition to being a hallmark at active genes, histone variant H3.3 is deposited by ATRX at repressive chromatin regions, including the telomeres. It is unclear how H3.3 promotes heterochromatin assembly. We show that H3.3 is targeted for K9 trimethylation to establish a heterochromatic state enriched in trimethylated H3.3K9 at telomeres. In H3f3a(-/-) and H3f3b(-/-) mouse embryonic stem cells (ESCs), H3.3 deficiency results in reduced levels of H3K9me3, H4K20me3 and ATRX at telomeres. The H3f3b(-/-) cells show increased levels of telomeric damage and sister chromatid exchange (t-SCE) activity when telomeres are compromised by treatment with a G-quadruplex (G4) DNA binding ligand or by ASF1 depletion. Overexpression of wild-type H3.3 (but not a H3.3K9 mutant) in H3f3b(-/-) cells increases H3K9 trimethylation level at telomeres and represses t-SCE activity induced by a G4 ligand. This study demonstrates the importance of H3.3K9 trimethylation in heterochromatin formation at telomeres. It provides insights into H3.3 function in maintaining integrity of mammalian constitutive heterochromatin, adding to its role in mediating transcription memory in the genome.


Subject(s)
Heterochromatin/metabolism , Histone Code , Histones/metabolism , Lysine/metabolism , Telomere/metabolism , Animals , Cells, Cultured , DNA Damage , Gene Deletion , Histones/chemistry , Histones/genetics , Methylation , Mice , Sister Chromatid Exchange , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL