Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Reprod Biomed Online ; 47(4): 103285, 2023 10.
Article in English | MEDLINE | ID: mdl-37573752

ABSTRACT

RESEARCH QUESTION: Can embryos harbouring cell exclusion and their reproductive outcomes be classified based on morphokinetic profiles? DESIGN: A total of 469 time-lapse videos of embryos transferred between 2013 and 2019 from a single clinic were analysed. Videos were assessed and grouped according to the presence or absence of one or more excluded cells before compaction. Cell division timings, intervals between subsequent cell divisions and dynamic intervals were analysed to determine the morphokinetic profiles of embryos with cell exclusion (CE+), compared with fully compacted embryos without cell exclusion or extrusion (CE-). RESULTS: Transfer of CE+ embryos resulted in lower proportions of fetal heartbeat (FHB) and live birth compared with CE- embryos (both, P < 0.001). CE+ embryos were associated with delays in t2 (P = 0.030), t6 (P = 0.018), t7 (P < 0.001), t8 (P = 0.001), tSC (P < 0.001) and tM (P < 0.001). Earlier timings for t3 (P = 0.014) and t5 (P < 0.001) were positively associated with CE+; CE+ embryos indicated prolonged S2, S3, ECC3, cc2 and cc4. Logistic regression analysis revealed that t5, tM, S2 and ECC3 were the strongest predictive indicators of cell exclusion. Timings for S2 and ECC3 were useful in identifying increased odds of FHB when a cell exclusion event was present. CONCLUSION: Embryos harbouring cell exclusion indicated altered morphokinetic profiles. Their overall lower reproductive success was associated with two morphokinetic parameters. Morphokinetic profiles could be used as adjunct indicators for reproductive success during cycles producing few, low-quality embryos. This may allow more objective identification of cell exclusion and refinement of embryo ranking procedures before transfer.


Subject(s)
Embryo, Mammalian , Embryonic Development , Humans , Reproduction , Time-Lapse Imaging , Retrospective Studies , Embryo Culture Techniques , Blastocyst
2.
Bioessays ; 43(10): e2100038, 2021 10.
Article in English | MEDLINE | ID: mdl-34423467

ABSTRACT

Although the promyelocytic leukemia (PML) protein is renowned for regulating a wide range of cellular processes and as an essential component of PML nuclear bodies (PML-NBs), the mechanisms through which it exerts its broad physiological impact are far from fully elucidated. Here, we review recent studies supporting an emerging view that PML's pleiotropic effects derive, at least partially, from its role in regulating histone H3.3 chromatin assembly, a critical epigenetic mechanism. These studies suggest that PML maintains heterochromatin organization by restraining H3.3 incorporation. Examination of PML's contribution to H3.3 chromatin assembly in the context of the cell cycle and PML-NB assembly suggests that PML represses heterochromatic H3.3 deposition during S phase and that transcription and SUMOylation regulate PML's recruitment to heterochromatin. Elucidating PML' s contributions to H3.3-mediated epigenetic regulation will provide insight into PML's expansive influence on cellular physiology and open new avenues for studying oncogenesis linked to PML malfunction.


Subject(s)
Chromatin Assembly and Disassembly , Histones , Chromatin Assembly and Disassembly/genetics , Epigenesis, Genetic/genetics , Histones/genetics , Histones/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism
3.
Nat Genet ; 51(5): 835-843, 2019 05.
Article in English | MEDLINE | ID: mdl-31011212

ABSTRACT

Genomic information is selectively used to direct spatial and temporal gene expression during differentiation. Interactions between topologically associating domains (TADs) and between chromatin and the nuclear lamina organize and position chromosomes in the nucleus. However, how these genomic organizers together shape genome architecture is unclear. Here, using a dual-lineage differentiation system, we report long-range TAD-TAD interactions that form constitutive and variable TAD cliques. A differentiation-coupled relationship between TAD cliques and lamina-associated domains suggests that TAD cliques stabilize heterochromatin at the nuclear periphery. We also provide evidence of dynamic TAD cliques during mouse embryonic stem-cell differentiation and somatic cell reprogramming and of inter-TAD associations in single-cell high-resolution chromosome conformation capture (Hi-C) data. TAD cliques represent a level of four-dimensional genome conformation that reinforces the silencing of repressed developmental genes.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Adipogenesis/genetics , Animals , Cell Lineage/genetics , Chromatin/ultrastructure , Chromatin Assembly and Disassembly , Gene Expression , Genome , Genome, Human , Humans , Mice , Models, Genetic , Mouse Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis/genetics , Nuclear Lamina/genetics , Stem Cells/cytology
4.
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
5.
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
6.
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
7.
Genome Biol ; 18(1): 21, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28137286

ABSTRACT

Current three-dimensional (3D) genome modeling platforms are limited by their inability to account for radial placement of loci in the nucleus. We present Chrom3D, a user-friendly whole-genome 3D computational modeling framework that simulates positions of topologically-associated domains (TADs) relative to each other and to the nuclear periphery. Chrom3D integrates chromosome conformation capture (Hi-C) and lamin-associated domain (LAD) datasets to generate structure ensembles that recapitulate radial distributions of TADs detected in single cells. Chrom3D reveals unexpected spatial features of LAD regulation in cells from patients with a laminopathy-causing lamin mutation. Chrom3D is freely available on github.


Subject(s)
Chromatin/genetics , Computational Biology/methods , Nuclear Lamina/genetics , Adult , Female , Genome , HeLa Cells , Humans , Male , Models, Genetic , Young Adult
8.
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
9.
Biochem Biophys Res Commun ; 460(2): 348-53, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25783056

ABSTRACT

Replication-independent histone variant H3.3 is incorporated into distinct genomic regions including promoters. However topology of promoter-associated H3.3 in relation to chromatin modifications and transcriptional outcome is not known, providing no insight on any distinction between H3.3-containing active and inactive promoters. Here, we report algorithms providing information on gene expression status as a function of density and position of multiple chromatin marks on promoters. We identify a relationship between promoter enrichment in epitope-tagged H3.3 or its canonical isoform H3.2 and corresponding transcriptional outcomes, as a function of sub-promoter positioning of DNA methylation and H3K4me3, H3K9me3 and H3K27me3. We identify a low-frequency configuration of H3.3 and H3K9me3 co-occupancy associated with transcriptional activity, while H3.3 and H3K27me3 promoters are invariably inactive. We unveil H3.3 and DNA methylated promoters whose transcription outcome depends on H3.3 sub-promoter positioning. Our results indicate how spatially restricted positioning of H3.3 may add another layer of transcription regulation.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Promoter Regions, Genetic , Transcription, Genetic , Chromatin Immunoprecipitation , DNA Methylation , Humans
10.
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
11.
Hum Mol Genet ; 23(5): 1151-62, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24108105

ABSTRACT

The nuclear lamina is implicated in the regulation of various nuclear functions. Several laminopathy-causing mutations in the LMNA gene, notably the p.R482W substitution linked to familial partial lipodystrophy type 2 (FPLD2), are clustered in the immunoglobulin fold of lamin A. We report a functional association between lamin A and fragile X-related protein 1 (FXR1P), a protein of the fragile X-related family involved in fragile X syndrome. Searching for proteins differentially interacting with the immunoglobulin fold of wild-type and R482W mutant lamin A, we identify FXR1P as a novel component of the lamin A protein network. The p.R482W mutation abrogates interaction of FXR1P with lamin A. Fibroblasts from FPLD2 patients display elevated levels of FXR1P and delocalized FXR1P. In human adipocyte progenitors, deregulation of lamin A expression leads to FXR1P up-regulation, impairment of adipogenic differentiation and induction of myogenin expression. FXR1P overexpression also stimulates a myogenic gene expression program in these cells. Our results demonstrate a cross-talk between proteins hitherto implicated in two distinct mesodermal pathologies. We propose a model where the FPLD2 lamin A p.R482W mutation elicits, through up-regulation of FXR1P, a remodeling of an adipogenic differentiation program into a myogenic program.


Subject(s)
Gene Expression Regulation , Lamin Type A/genetics , Lamin Type A/metabolism , Muscle Development/genetics , Mutation , RNA-Binding Proteins/genetics , Adipocytes/cytology , Adipocytes/metabolism , Adult , Amino Acid Sequence , Amino Acid Substitution , Cell Differentiation/genetics , Codon , Female , Fibroblasts/metabolism , Humans , Lamin Type A/chemistry , Lipodystrophy/genetics , Lipodystrophy/metabolism , Lipodystrophy, Familial Partial/genetics , Lipodystrophy, Familial Partial/metabolism , Molecular Sequence Data , Nuclear Envelope/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Binding , RNA Processing, Post-Transcriptional , Stem Cells/cytology , Stem Cells/metabolism
12.
Genome Res ; 23(10): 1580-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23861385

ABSTRACT

The nuclear lamina is implicated in the organization of the eukaryotic nucleus. Association of nuclear lamins with the genome occurs through large chromatin domains including mostly, but not exclusively, repressed genes. How lamin interactions with regulatory elements modulate gene expression in different cellular contexts is unknown. We show here that in human adipose tissue stem cells, lamin A/C interacts with distinct spatially restricted subpromoter regions, both within and outside peripheral and intra-nuclear lamin-rich domains. These localized interactions are associated with distinct transcriptional outcomes in a manner dependent on local chromatin modifications. Down-regulation of lamin A/C leads to dissociation of lamin A/C from promoters and remodels repressive and permissive histone modifications by enhancing transcriptional permissiveness, but is not sufficient to elicit gene activation. Adipogenic differentiation resets a large number of lamin-genome associations globally and at subpromoter levels and redefines associated transcription outputs. We propose that lamin A/C acts as a modulator of local gene expression outcome through interaction with adjustable sites on promoters, and that these position-dependent transcriptional readouts may be reset upon differentiation.


Subject(s)
Adipose Tissue/cytology , Chromatin/metabolism , Lamin Type A/metabolism , Promoter Regions, Genetic , Stem Cells/metabolism , Transcription, Genetic , Adipogenesis , Adipose Tissue/metabolism , Cells, Cultured , Gene Expression Profiling , Genetic Loci , Humans , Lamin Type A/genetics , Nuclear Lamina/genetics , Nuclear Lamina/metabolism , Stem Cells/cytology , Transcriptional Activation
13.
Genome Res ; 23(3): 440-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23222847

ABSTRACT

Replication-independent chromatin deposition of histone variant H3.3 is mediated by several chaperones. We report a multistep targeting of newly synthesized epitope-tagged H3.3 to chromatin via PML bodies. H3.3 is recruited to PML bodies in a DAXX-dependent manner, a process facilitated by ASF1A. DAXX is required for enrichment of ATRX, but not ASF1A or HIRA, with PML. Nonetheless, the chaperones colocalize with H3.3 at PML bodies and are found in one or more complexes with PML. Both DAXX and PML are necessary to prevent accumulation of a soluble, nonincorporated pool of H3.3. H3.3 targeting to PML is enhanced with an (H3.3-H4)2 tetramerization mutant of H3.3, suggesting H3.3 recruitment to PML as an (H3.3-H4) dimer rather than as a tetramer. Our data support a model of DAXX-mediated recruitment of (H3.3-H4) dimers to PML bodies, which may function as triage centers for H3.3 deposition into chromatin by distinct chaperones.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Histones/genetics , Nuclear Proteins/genetics , Nucleosomes/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Co-Repressor Proteins , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication , HeLa Cells , Histone Chaperones/genetics , Histone Chaperones/metabolism , Histones/metabolism , Humans , Molecular Chaperones , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , X-linked Nuclear Protein
14.
FEBS Lett ; 584(14): 2999-3004, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20580717

ABSTRACT

Mutations in the lamin A/C (LMNA) gene that cause Hutchinson-Gilford progeria syndrome (HGPS) lead to expression of a protein called progerin with 50 amino acids deleted from the tail of prelamin A. In cells from patients with HGPS, both the amount and distribution of heterochromatin are altered. We designed in vitro assays to ask whether such alterations might reflect changes in chromatin, DNA and/or histone binding properties of progerin compared to wild-type lamin C-terminal tails. We show that progerin tail has a reduced DNA/chromatin binding capacity and modified trimethylated H3K27 binding pattern, offering a molecular mechanism for heterochromatin alterations related to HGPS.


Subject(s)
Progeria/genetics , Progeria/metabolism , Animals , Binding Sites/genetics , Chromatin/metabolism , DNA/genetics , DNA/metabolism , Genotype , Heterochromatin/metabolism , Humans , Lamin Type A , Lamins/genetics , Lamins/metabolism , Mice , Mice, Transgenic , Mutation , Nuclear Proteins , Protein Precursors , Sequence Deletion
15.
Mol Biol Cell ; 21(11): 1872-84, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20375147

ABSTRACT

In contrast to canonical histones, histone variant H3.3 is incorporated into chromatin in a replication-independent manner. Posttranslational modifications of H3.3 have been identified; however, the epigenetic environment of incorporated H3.3 is unclear. We have investigated the genomic distribution of epitope-tagged H3.3 in relation to histone modifications, DNA methylation, and transcription in mesenchymal stem cells. Quantitative imaging at the nucleus level shows that H3.3, relative to replicative H3.2 or canonical H2B, is enriched in chromatin domains marked by histone modifications of active or potentially active genes. Chromatin immunoprecipitation of epitope-tagged H3.3 and array hybridization identified 1649 H3.3-enriched promoters, a fraction of which is coenriched in H3K4me3 alone or together with H3K27me3, whereas H3K9me3 is excluded, corroborating nucleus-level imaging data. H3.3-enriched promoters are predominantly CpG-rich and preferentially DNA methylated, relative to the proportion of methylated RefSeq promoters in the genome. Most but not all H3.3-enriched promoters are transcriptionally active, and coenrichment of H3.3 with repressive H3K27me3 correlates with an enhanced proportion of expressed genes carrying this mark. H3.3-target genes are enriched in mesodermal differentiation and signaling functions. Our data suggest that in mesenchymal stem cells, H3.3 targets lineage-priming genes with a potential for activation facilitated by H3K4me3 in facultative association with H3K27me3.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/chemistry , DNA/metabolism , Genome , Histones/chemistry , DNA/genetics , DNA Methylation , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Humans , Microarray Analysis , Promoter Regions, Genetic , Protein Processing, Post-Translational , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
Exp Cell Res ; 314(8): 1869-80, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18396274

ABSTRACT

Autosomal dominant Emery-Dreifuss muscular dystrophy (EDMD) is characterized by muscle wasting and is caused by mutations in the LMNA gene encoding A-type lamins. Overexpression of the EDMD lamin A R453W mutation in C2C12 myoblasts impairs myogenic differentiation. We show here the influence of stable expression of the R453W and of the Dunnigan-type partial lipodystrophy R482W mutation of lamin A in C2C12 cells on transcription and epigenetic regulation of the myogenin (Myog) gene and on global chromatin organization. Expression of R453W-, but not R482W-lamin A, impairs activation of Myog and maintains a repressive chromatin state on the Myog promoter upon induction of differentiation, marked by H3 lysine (K) 9 dimethylation and failure to hypertrimethylate H3K4. Cells expressing WT-LaA also fail to hypertrimethylate H3K4. No defect occurs at the level of Myog promoter DNA methylation in any of the clones. Expression of R453W-lamin A and to a lesser extent R482W-lamin A in undifferentiated C2C12 cells redistributes H3K9me3 from pericentric heterochromatin. R453W-lamin A also elicits a redistribution of H3K27me3 from inactive X (Xi) and partial decondensation of Xi, but maintains Xist expression and coating of Xi, indicating that Xi remains inactivated. Our results argue that gene-specific and genome-wide chromatin rearrangements may constitute a molecular basis for laminopathies.


Subject(s)
Epigenesis, Genetic , Lamin Type A/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation, Missense , Myoblasts/metabolism , Myogenin/genetics , Amino Acid Substitution , Animals , Arginine/genetics , Cell Differentiation , Cell Line , Cell Nucleus/enzymology , DNA Methylation , Histones/chemistry , Histones/metabolism , Humans , Lamin Type A/metabolism , Methylation , Mice , Myoblasts/cytology , Myogenin/biosynthesis , Promoter Regions, Genetic , Tryptophan/genetics , Up-Regulation , X Chromosome/enzymology
17.
Exp Cell Res ; 314(6): 1392-405, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18294630

ABSTRACT

Mutation R453W in A-type lamins, that are major nuclear envelope proteins, generates Emery-Dreifuss muscular dystrophy. We previously showed that mouse myoblasts expressing R453W-lamin A incompletely exit the cell cycle and differentiate into myocytes with a low level of multinucleation. Here we attempted to improve differentiation by treating these cells with a mixture of PD98059, an extracellular-regulated kinase (ERK) kinase (also known as mitogen-activated kinase, MEK) inhibitor, and insulin-like growth factor-II, an activator of phosphoinositide 3-kinase. We show that mouse myoblasts expressing R453W-lamin A were sensitive to the drug treatment as shown by (i) an increase in multinucleation, (ii) downregulation of proliferation markers (cyclin D1, hyperphosphorylated Rb), (iii) upregulation of myogenin, and (iv) sustained activation of p21 and cyclin D3. However, nuclear matrix anchorage of p21 and cyclin D3 in a complex with hypophosphorylated Rb that is critical to trigger cell cycle arrest and myogenin induction was deficient and incompletely restored by drug treatment. As the turn-over of R453W-lamin A at the nuclear envelope was greatly enhanced, we propose that R453W-lamin A impairs the capacity of the nuclear lamina to serve as scaffold for substrates of the MEK-ERK pathway and for MyoD-induced proteins that play a role in the differentiation process.


Subject(s)
Cell Differentiation , Lamin Type A/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscular Dystrophy, Emery-Dreifuss/enzymology , Myoblasts/cytology , Myoblasts/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Antigens, CD1/metabolism , CD3 Complex/metabolism , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Green Fluorescent Proteins/metabolism , Insulin-Like Growth Factor II/pharmacology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Muscular Dystrophy, Emery-Dreifuss/pathology , Mutant Proteins/metabolism , Mutation/genetics , Myoblasts/drug effects , Myogenin/metabolism , Nuclear Lamina/drug effects , Nuclear Lamina/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Retinoblastoma Protein/metabolism
18.
BMC Cell Biol ; 7: 23, 2006 May 23.
Article in English | MEDLINE | ID: mdl-16719903

ABSTRACT

BACKGROUND: There exists abundant molecular and ultra-structural evidence to suggest that cytoplasmic actin can physically interact with the nuclear envelope (NE) membrane system. However, this interaction has yet to be characterised in living interphase cells. RESULTS: Using a fluorescent conjugate of the actin binding drug cytochalasin D (CD-BODIPY) we provide evidence that polymerising actin accumulates in vicinity to the NE. In addition, both transiently expressed fluorescent actin and cytoplasmic micro-injection of fluorescent actin resulted in accumulation of actin at the NE-membrane. Consistent with the idea that the cytoplasmic phase of NE-membranes can support this novel pool of perinuclear actin polymerisation we show that isolated, intact, differentiated primary hepatocyte nuclei support actin polymerisation in vitro. Further this phenomenon was inhibited by treatments hindering steric access to outer-nuclear-membrane proteins (e.g. wheat germ agglutinin, anti-nesprin and anti-nucleoporin antibodies). CONCLUSION: We conclude that actin polymerisation occurs around interphase nuclei of living cells at the cytoplasmic phase of NE-membranes.


Subject(s)
Actins/chemistry , Nuclear Envelope/chemistry , Animals , Binding Sites , Biopolymers , Boron Compounds/analysis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Circular Dichroism , Cytochalasin D/pharmacology , Depsipeptides/pharmacology , Fluorescent Dyes/analysis , HeLa Cells , Humans , Liver/ultrastructure , Rabbits , Rats , Thiazoles/pharmacology , Thiazolidines
19.
Hum Mol Genet ; 15(7): 1113-22, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16481358

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a dominant autosomal premature aging syndrome caused by the expression of a truncated prelamin A designated progerin (Pgn). A-type and B-type lamins are intermediate filament proteins that polymerize to form the nuclear lamina network apposed to the inner nuclear membrane of vertebrate somatic cells. It is not known if in vivo both type of lamins assemble independently or co-assemble. The blebbing and disorganization of the nuclear envelope and adjacent heterochromatin in cells from patients with HGPS is a hallmark of the disease, and the ex vivo reversal of this phenotype is considered important for the development of therapeutic strategies. Here, we investigated the alterations in the lamina structure that may underlie the disorganization caused in nuclei by Pgn expression. We studied the polymerization of enhanced green fluorescent protein- and red fluorescent protein-tagged wild-type and mutated lamins in the nuclear envelope of living cells by measuring fluorescence resonance energy transfer (FRET) that occurs between the two fluorophores when tagged lamins interact. Using time domain fluorescence lifetime imaging microscopy that allows a quantitative analysis of FRET signals, we show that wild-type lamins A and B1 polymerize in distinct homopolymers that further interact in the lamina. In contrast, expressed Pgn co-assembles with lamin B1 and lamin A to form a mixed heteropolymer in which A-type and B-type lamin segregation is lost. We propose that such structural lamina alterations may be part of the primary mechanisms leading to HGPS, possibly by impairing functions specific for each lamin type such as nuclear membrane biogenesis, signal transduction, nuclear compartmentalization and gene regulation.


Subject(s)
Lamin Type A/metabolism , Lamin Type B/metabolism , Nuclear Proteins/metabolism , Progeria/metabolism , Protein Precursors/metabolism , Animals , Cells, Cultured , Fluorescence Resonance Energy Transfer/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lamin Type A/genetics , Lamin Type B/genetics , Mice , Microscopy, Fluorescence/methods , Models, Biological , Models, Genetic , Myoblasts/cytology , Myoblasts/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , Protein Precursors/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...