Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 32(6): 1010-1031, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36282542

ABSTRACT

Emery-Dreifuss muscular dystrophy (EDMD) is a genetically and clinically variable disorder. Previous attempts to use gene expression changes to find its pathomechanism were unavailing, so we engaged a functional pathway analysis. RNA-Seq was performed on cells from 10 patients diagnosed with an EDMD spectrum disease with different mutations in seven genes. Upon comparing to controls, the pathway analysis revealed that multiple genes involved in fibrosis, metabolism, myogenic signaling and splicing were affected in all patients. Splice variant analysis revealed alterations of muscle-specific variants for several important muscle genes. Deeper analysis of metabolic pathways revealed a reduction in glycolytic and oxidative metabolism and reduced numbers of mitochondria across a larger set of 14 EDMD spectrum patients and 7 controls. Intriguingly, the gene expression signatures segregated the patients into three subgroups whose distinctions could potentially relate to differences in clinical presentation. Finally, differential expression analysis of miRNAs changing in the patients similarly highlighted fibrosis, metabolism and myogenic signaling pathways. This pathway approach revealed a transcriptome profile that can both be used as a template for establishing a biomarker panel for EDMD and direct further investigation into its pathomechanism. Furthermore, the segregation of specific gene changes into distinct groups that appear to correlate with clinical presentation may template development of prognostic biomarkers, though this will first require their testing in a wider set of patients with more clinical information.


Subject(s)
Muscular Dystrophy, Emery-Dreifuss , Humans , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation , Fibrosis , Biomarkers
2.
Nat Commun ; 13(1): 321, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027552

ABSTRACT

Little is known about how the observed fat-specific pattern of 3D-spatial genome organisation is established. Here we report that adipocyte-specific knockout of the gene encoding nuclear envelope transmembrane protein Tmem120a disrupts fat genome organisation, thus causing a lipodystrophy syndrome. Tmem120a deficiency broadly suppresses lipid metabolism pathway gene expression and induces myogenic gene expression by repositioning genes, enhancers and miRNA-encoding loci between the nuclear periphery and interior. Tmem120a-/- mice, particularly females, exhibit a lipodystrophy syndrome similar to human familial partial lipodystrophy FPLD2, with profound insulin resistance and metabolic defects that manifest upon exposure to an obesogenic diet. Interestingly, similar genome organisation defects occurred in cells from FPLD2 patients that harbour nuclear envelope protein encoding LMNA mutations. Our data indicate TMEM120A genome organisation functions affect many adipose functions and its loss may yield adiposity spectrum disorders, including a miRNA-based mechanism that could explain muscle hypertrophy in human lipodystrophy.


Subject(s)
Genetic Loci , Ion Channels/deficiency , Lipodystrophy/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Body Weight , Carbohydrate Metabolism , Diet, High-Fat , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation , Glucose Tolerance Test , Humans , Insulin Resistance , Ion Channels/metabolism , Lamin Type B/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Development/genetics , Nuclear Envelope/metabolism , Obesity/genetics , Organ Specificity , Oxidation-Reduction , RNA/genetics , RNA/metabolism
3.
iScience ; 24(9): 103055, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34541469

ABSTRACT

STimulator of INterferon Genes (STING) is an adaptor for cytoplasmic DNA sensing by cGAMP/cGAS that helps trigger innate immune responses (IIRs). Although STING is mostly localized in the ER, we find a separate inner nuclear membrane pool of STING that increases mobility and redistributes to the outer nuclear membrane upon IIR stimulation by transfected dsDNA or dsRNA mimic poly(I:C). Immunoprecipitation of STING from isolated nuclear envelopes coupled with mass spectrometry revealed a distinct nuclear envelope-STING proteome consisting of known nuclear membrane proteins and enriched in DNA- and RNA-binding proteins. Seventeen of these nuclear envelope STING partners are known to bind direct interactors of IRF3/7 transcription factors, and testing a subset of these revealed STING partners SYNCRIP, MEN1, DDX5, snRNP70, RPS27a, and AATF as novel modulators of dsDNA-triggered IIRs. Moreover, we find that SYNCRIP is a novel antagonist of the RNA virus, influenza A, potentially shedding light on reports of STING inhibition of RNA viruses.

4.
Front Cell Dev Biol ; 8: 592573, 2020.
Article in English | MEDLINE | ID: mdl-33330474

ABSTRACT

Tissue-specific patterns of radial genome organization contribute to genome regulation and can be established by nuclear envelope proteins. Studies in this area often use cancer cell lines, and it is unclear how well such systems recapitulate genome organization of primary cells or animal tissues; so, we sought to investigate radial genome organization in primary liver tissue hepatocytes. Here, we have used a NET47/Tm7sf2-/- liver model to show that manipulating one of these nuclear membrane proteins is sufficient to alter tissue-specific gene positioning and expression. Dam-LaminB1 global profiling in primary liver cells shows that nearly all the genes under such positional regulation are related to/important for liver function. Interestingly, Tm7sf2 is a paralog of the HP1-binding nuclear membrane protein LBR that, like Tm7sf2, also has an enzymatic function in sterol reduction. Fmo3 gene/locus radial mislocalization could be rescued with human wild-type, but not TM7SF2 mutants lacking the sterol reductase function. One central pathway affected is the cholesterol synthesis pathway. Within this pathway, both Cyp51 and Msmo1 are under Tm7sf2 positional and expression regulation. Other consequences of the loss of Tm7sf2 included weight gain, insulin sensitivity, and reduced levels of active Akt kinase indicating additional pathways under its regulation, several of which are highlighted by mispositioning genes. This study emphasizes the importance for tissue-specific radial genome organization in tissue function and the value of studying genome organization in animal tissues and primary cells over cell lines.

5.
EBioMedicine ; 51: 102587, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31862442

ABSTRACT

BACKGROUND: As genome-wide approaches prove difficult with genetically heterogeneous orphan diseases, we developed a new approach to identify candidate genes. We applied this to Emery-Dreifuss muscular dystrophy (EDMD), characterised by early onset contractures, slowly progressive muscular wasting, and life-threatening heart conduction disturbances with wide intra- and inter-familial clinical variability. Roughly half of EDMD patients are linked to six genes encoding nuclear envelope proteins, but the disease mechanism remains unclear because the affected proteins function in both cell mechanics and genome regulation. METHODS: A primer library was generated to test for mutations in 301 genes from four categories: (I) all known EDMD-linked genes; (II) genes mutated in related muscular dystrophies; (III) candidates generated by exome sequencing in five families; (IV) functional candidates - other muscle nuclear envelope proteins functioning in mechanical/genome processes affected in EDMD. This was used to sequence 56 unlinked patients with EDMD-like phenotype. FINDINGS: Twenty-one patients could be clearly assigned: 18 with mutations in genes of similar muscular dystrophies; 3 with previously missed mutations in EDMD-linked genes. The other categories yielded novel candidate genes, most encoding nuclear envelope proteins with functions in gene regulation. INTERPRETATION: Our multi-pronged approach identified new disease alleles and many new candidate EDMD genes. Their known functions strongly argue the EDMD pathomechanism is from altered gene regulation and mechanotransduction due to connectivity of candidates from the nuclear envelope to the plasma membrane. This approach highlights the value of testing for related diseases using primer libraries and may be applied for other genetically heterogeneous orphan diseases. FUNDING: The Wellcome Trust, Muscular Dystrophy UK, Medical Research Council, European Community's Seventh Framework Programme "Integrated European -omics research project for diagnosis and therapy in rare neuromuscular and neurodegenerative diseases (NEUROMICS)".


Subject(s)
Alleles , Gene Expression Regulation , Muscular Dystrophy, Emery-Dreifuss/genetics , Sequence Analysis, DNA , Gene Ontology , Muscles/metabolism , Mutation/genetics , Exome Sequencing
6.
Front Cell Dev Biol ; 7: 18, 2019.
Article in English | MEDLINE | ID: mdl-30949476

ABSTRACT

Every living organism, from bacteria to humans, contains DNA encoding anything from a few hundred genes in intracellular parasites such as Mycoplasma, up to several tens of thousands in many higher organisms. The first observations indicating that the nucleus had some kind of organization were made over a hundred years ago. Understanding of its significance is both limited and aided by the development of techniques, in particular electron microscopy, fluorescence in situ hybridization, DamID and most recently HiC. As our knowledge about genome organization grows, it becomes apparent that the mechanisms are conserved in evolution, even if the individual players may vary. These mechanisms involve DNA binding proteins such as histones, and a number of architectural proteins, some of which are very much conserved, with some others having diversified and multiplied, acquiring specific regulatory functions. In this review we will look at the principles of genome organization in a hierarchical manner, from DNA packaging to higher order genome associations such as TADs, and the significance of radial positioning of genomic loci. We will then elaborate on the dynamics of genome organization during development, and how genome architecture plays an important role in cell fate determination. Finally, we will discuss how misregulation can be a factor in human disease.

7.
Methods ; 157: 88-99, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30445179

ABSTRACT

DamID, a method to identify DNA associating with a particular protein, was originally developed for use in immortalized tissue culture lines. The power of this technique has led to its adaptation for a number of additional systems. Here we report adaptations for its use in primary cells isolated from rodents with emphasis on the challenges this presents. Specifically, we present several modifications that allow the method to be performed in mouse acutely isolated primary hepatocytes while seemingly maintaining tissue genome architecture. We also describe the downstream bioinformatic analysis necessary to identify LADs and discuss some of the parameters and their effects with regards to the sensitivity of the method.


Subject(s)
Chromatin/genetics , DNA/isolation & purification , Lamin Type B/genetics , Primary Cell Culture/methods , Animals , DNA/genetics , DNA Methylation/genetics , Genome/genetics , Hepatocytes/metabolism , Lamin Type B/chemistry , Mice
8.
Genome Res ; 27(7): 1126-1138, 2017 07.
Article in English | MEDLINE | ID: mdl-28424353

ABSTRACT

The 3D organization of the genome changes concomitantly with expression changes during hematopoiesis and immune activation. Studies have focused either on lamina-associated domains (LADs) or on topologically associated domains (TADs), defined by preferential local chromatin interactions, and chromosome compartments, defined as higher-order interactions between TADs sharing functionally similar states. However, few studies have investigated how these affect one another. To address this, we mapped LADs using Lamin B1-DamID during Jurkat T-cell activation, finding significant genome reorganization at the nuclear periphery dominated by release of loci frequently important for T-cell function. To assess how these changes at the nuclear periphery influence wider genome organization, our DamID data sets were contrasted with TADs and compartments. Features of specific repositioning events were then tested by fluorescence in situ hybridization during T-cell activation. First, considerable overlap between TADs and LADs was observed with the TAD repositioning as a unit. Second, A1 and A2 subcompartments are segregated in 3D space through differences in proximity to LADs along chromosomes. Third, genes and a putative enhancer in LADs that were released from the periphery during T-cell activation became preferentially associated with A2 subcompartments and were constrained to the relative proximity of the lamina. Thus, lamina associations influence internal nuclear organization, and changes in LADs during T-cell activation may provide an important additional mode of gene regulation.


Subject(s)
Chromosomes, Human/metabolism , Enhancer Elements, Genetic , Lamin Type B/metabolism , Lymphocyte Activation , Neoplasm Proteins/metabolism , Nuclear Envelope/metabolism , T-Lymphocytes/metabolism , Chromosomes, Human/genetics , Humans , Jurkat Cells , Lamin Type B/genetics , Neoplasm Proteins/genetics , Nuclear Envelope/genetics , T-Lymphocytes/cytology
9.
Nat Commun ; 8: 14048, 2017 01 16.
Article in English | MEDLINE | ID: mdl-28091603

ABSTRACT

Repo-Man is a protein phosphatase 1 (PP1) targeting subunit that regulates mitotic progression and chromatin remodelling. After mitosis, Repo-Man/PP1 remains associated with chromatin but its function in interphase is not known. Here we show that Repo-Man, via Nup153, is enriched on condensed chromatin at the nuclear periphery and at the edge of the nucleopore basket. Repo-Man/PP1 regulates the formation of heterochromatin, dephosphorylates H3S28 and it is necessary and sufficient for heterochromatin protein 1 binding and H3K27me3 recruitment. Using a novel proteogenomic approach, we show that Repo-Man is enriched at subtelomeric regions together with H2AZ and H3.3 and that depletion of Repo-Man alters the peripheral localization of a subset of these regions and alleviates repression of some polycomb telomeric genes. This study shows a role for a mitotic phosphatase in the regulation of the epigenetic landscape and gene expression in interphase.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Heterochromatin/metabolism , Interphase , Nuclear Proteins/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Heterochromatin/genetics , Histones/genetics , Histones/metabolism , Humans , Nuclear Proteins/genetics , Phosphorylation
10.
Nucleus ; 8(1): 81-97, 2017 01 02.
Article in English | MEDLINE | ID: mdl-28045568

ABSTRACT

Different cell types exhibit distinct patterns of 3D genome organization that correlate with changes in gene expression in tissue and differentiation systems. Several tissue-specific nuclear envelope transmembrane proteins (NETs) have been found to influence the spatial positioning of genes and chromosomes that normally occurs during tissue differentiation. Here we study 3 such NETs: NET29, NET39, and NET47, which are expressed preferentially in fat, muscle and liver, respectively. We found that even when exogenously expressed in a heterologous system they can specify particular genome organization patterns and alter gene expression. Each NET affected largely different subsets of genes. Notably, the liver-specific NET47 upregulated many genes in HT1080 fibroblast cells that are normally upregulated in hepatogenesis, showing that tissue-specific NETs can favor expression patterns associated with the tissue where the NET is normally expressed. Similarly, global profiling of peripheral chromatin after exogenous expression of these NETs using lamin B1 DamID revealed that each NET affected the nuclear positioning of distinct sets of genomic regions with a significant tissue-specific component. Thus NET influences on genome organization can contribute to gene expression changes associated with differentiation even in the absence of other factors and overt cellular differentiation changes.


Subject(s)
Genome, Human/genetics , Membrane Proteins/genetics , Nuclear Envelope/metabolism , Cell Differentiation , Cell Line , Chromosomes, Human/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Liver/cytology , Organ Specificity
11.
Mol Cell ; 62(6): 834-847, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27264872

ABSTRACT

Whether gene repositioning to the nuclear periphery during differentiation adds another layer of regulation to gene expression remains controversial. Here, we resolve this by manipulating gene positions through targeting the nuclear envelope transmembrane proteins (NETs) that direct their normal repositioning during myogenesis. Combining transcriptomics with high-resolution DamID mapping of nuclear envelope-genome contacts, we show that three muscle-specific NETs, NET39, Tmem38A, and WFS1, direct specific myogenic genes to the nuclear periphery to facilitate their repression. Retargeting a NET39 fragment to nucleoli correspondingly repositioned a target gene, indicating a direct tethering mechanism. Being able to manipulate gene position independently of other changes in differentiation revealed that repositioning contributes ⅓ to ⅔ of a gene's normal repression in myogenesis. Together, these NETs affect 37% of all genes changing expression during myogenesis, and their combined knockdown almost completely blocks myotube formation. This unequivocally demonstrates that NET-directed gene repositioning is critical for developmental gene regulation.


Subject(s)
Chromosome Positioning , Gene Expression Regulation, Developmental , Ion Channels/genetics , Membrane Proteins/genetics , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , Animals , Cell Differentiation , Cell Line , Down-Regulation , Humans , Ion Channels/metabolism , Kinetics , Membrane Proteins/metabolism , Mice , Nuclear Proteins/metabolism , RNA Interference , Transfection
12.
PLoS One ; 10(5): e0127712, 2015.
Article in English | MEDLINE | ID: mdl-26024229

ABSTRACT

Recent work indicates that the nuclear envelope is a major signaling node for the cell that can influence tissue differentiation processes. Here we present two nuclear envelope trans-membrane proteins TMEM120A and TMEM120B that are paralogs encoded by the Tmem120A and Tmem120B genes. The TMEM120 proteins are expressed preferentially in fat and both are induced during 3T3-L1 adipocyte differentiation. Knockdown of one or the other protein altered expression of several genes required for adipocyte differentiation, Gata3, Fasn, Glut4, while knockdown of both together additionally affected Pparg and Adipoq. The double knockdown also increased the strength of effects, reducing for example Glut4 levels by 95% compared to control 3T3-L1 cells upon pharmacologically induced differentiation. Accordingly, TMEM120A and B knockdown individually and together impacted on adipocyte differentiation/metabolism as measured by lipid accumulation through binding of Oil Red O and coherent anti-Stokes Raman scattering microscopy (CARS). The nuclear envelope is linked to several lipodystrophies through mutations in lamin A; however, lamin A is widely expressed. Thus it is possible that the TMEM120A and B fat-specific nuclear envelope transmembrane proteins may play a contributory role in the tissue-specific pathology of this disorder or in the wider problem of obesity.


Subject(s)
Adipocytes/metabolism , Cell Differentiation , Membrane Proteins/metabolism , Nuclear Envelope/metabolism , Obesity/metabolism , 3T3-L1 Cells , Adiponectin/genetics , Adiponectin/metabolism , Animals , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Knockdown Techniques , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Membrane Proteins/genetics , Mice , Nuclear Envelope/genetics , Obesity/genetics , PPAR gamma/genetics , PPAR gamma/metabolism
13.
PLoS One ; 9(11): e111851, 2014.
Article in English | MEDLINE | ID: mdl-25386906

ABSTRACT

Changes in the peripheral distribution and amount of condensed chromatin are observed in a number of diseases linked to mutations in the lamin A protein of the nuclear envelope. We postulated that lamin A interactions with nuclear envelope transmembrane proteins (NETs) that affect chromatin structure might be altered in these diseases and so screened thirty-one NETs for those that promote chromatin compaction as determined by an increase in the number of chromatin clusters of high pixel intensity. One of these, NET23 (also called STING, MITA, MPYS, ERIS, Tmem173), strongly promoted chromatin compaction. A correlation between chromatin compaction and endogenous levels of NET23/STING was observed for a number of human cell lines, suggesting that NET23/STING may contribute generally to chromatin condensation. NET23/STING has separately been found to be involved in innate immune response signaling. Upon infection cells make a choice to either apoptose or to alter chromatin architecture to support focused expression of interferon genes and other response factors. We postulate that the chromatin compaction induced by NET23/STING may contribute to this choice because the cells expressing NET23/STING eventually apoptose, but the chromatin compaction effect is separate from this as the condensation was still observed when cells were treated with Z-VAD to block apoptosis. NET23/STING-induced compacted chromatin revealed changes in epigenetic marks including changes in histone methylation and acetylation. This indicates a previously uncharacterized nuclear role for NET23/STING potentially in both innate immune signaling and general chromatin architecture.


Subject(s)
Chromatin/metabolism , Membrane Proteins/metabolism , Nuclear Envelope/metabolism , Acetylation , Histones/metabolism , Humans , Immunity, Innate , Methylation
14.
Adv Exp Med Biol ; 773: 5-26, 2014.
Article in English | MEDLINE | ID: mdl-24563341

ABSTRACT

Cancer has been diagnosed for millennia, but its cellular nature only began to be understood in the mid-nineteenth century when advances in microscopy allowed detailed specimen observations. It was soon noted that cancer cells often possessed nuclei that were altered in size and/or shape. This became an important criterion for cancer diagnosis that continues to be used today. The mechanisms linking nuclear abnormalities and cancer only started to be understood in the second half of the twentieth century, with the discovery of nuclear lamina composition differences in cancer cells compared to normal cells. The nuclear envelope, rather than providing a mere physical barrier between the genetic material in the nucleus and the cytoplasm, is a very important functional hub for many cellular processes. In this review we give an overview of the links between cancer biology and nuclear envelope, from the early days of microscopy until the present day's understanding of some of the molecular mechanisms behind those links.


Subject(s)
Neoplasms/diagnosis , Nuclear Envelope/physiology , Humans , Image Processing, Computer-Assisted , Neoplasms/physiopathology
15.
Nucleus ; 4(6): 460-77, 2013.
Article in English | MEDLINE | ID: mdl-24213376

ABSTRACT

Nuclear envelope links to inherited disease gave the conundrum of how mutations in near-ubiquitous proteins can yield many distinct pathologies, each focused in different tissues. One conundrum-resolving hypothesis is that tissue-specific partner proteins mediate these pathologies. Such partner proteins may have now been identified with recent proteome studies determining nuclear envelope composition in different tissues. These studies revealed that the majority of the total nuclear envelope proteins are tissue restricted in their expression. Moreover, functions have been found for a number these tissue-restricted nuclear envelope proteins that fit with mechanisms proposed to explain how the nuclear envelope could mediate disease, including defects in mechanical stability, cell cycle regulation, signaling, genome organization, gene expression, nucleocytoplasmic transport, and differentiation. The wide range of functions to which these proteins contribute is consistent with not only their involvement in tissue-specific nuclear envelope disease pathologies, but also tissue evolution.


Subject(s)
Cell Nucleus/metabolism , Nuclear Envelope/chemistry , Organ Specificity/physiology , Active Transport, Cell Nucleus , Animals , Cell Cycle/physiology , Cell Differentiation/physiology , Evolution, Molecular , Humans , Membrane Proteins/metabolism , Models, Animal , Nuclear Proteins/metabolism , Proteome/metabolism , Signal Transduction
16.
Genome Biol ; 14(2): R14, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23414781

ABSTRACT

BACKGROUND: Different cell types have distinctive patterns of chromosome positioning in the nucleus. Although ectopic affinity-tethering of specific loci can be used to relocate chromosomes to the nuclear periphery, endogenous nuclear envelope proteins that control such a mechanism in mammalian cells have yet to be widely identified. RESULTS: To search for such proteins, 23 nuclear envelope transmembrane proteins were screened for their ability to promote peripheral localization of human chromosomes in HT1080 fibroblasts. Five of these proteins had strong effects on chromosome 5, but individual proteins affected different subsets of chromosomes. The repositioning effects were reversible and the proteins with effects all exhibited highly tissue-restricted patterns of expression. Depletion of two nuclear envelope transmembrane proteins that were preferentially expressed in liver each reduced the normal peripheral positioning of chromosome 5 in liver cells. CONCLUSIONS: The discovery of nuclear envelope transmembrane proteins that can modulate chromosome position and have restricted patterns of expression may enable dissection of the functional relevance of tissue-specific patterns of radial chromosome positioning.


Subject(s)
Cell Nucleus/metabolism , Chromosome Positioning , Membrane Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Chromosomes, Human/metabolism , Hep G2 Cells , Humans , Membrane Proteins/genetics , Nuclear Proteins/genetics , Organ Specificity
17.
Semin Cancer Biol ; 23(2): 125-37, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22311402

ABSTRACT

Although its properties have long been used for both typing and prognosis of various tumors, the nuclear envelope (NE) itself and its potential roles in tumorigenesis are only beginning to be understood. Historically viewed as merely a protective barrier, the nuclear envelope is now linked to a wide range of functions. Nuclear membrane proteins connect the nucleus to the cytoskeleton on one side and to chromatin on the other. Several newly identified nuclear envelope functions associated with these connections intersect with cancer pathways. For example, the nuclear envelope could affect genome stability by tethering chromatin. Some nuclear envelope proteins affect cell cycle regulation by directly binding to the master regulator pRb, others by interacting with TGF-ß and Smad signaling cascades, and others by affecting the mitotic spindle. Finally, the NE directly affects cytoskeletal organization and can also influence cell migration in metastasis. In this review we discuss the link between the nuclear envelope and cellular defects that are common in cancer cells, and we show that NE proteins are often aberrantly expressed in tumors. The NE represents a potential reservoir of diagnostic and prognostic markers in cancer.


Subject(s)
Neoplasms/etiology , Nuclear Envelope/physiology , Animals , Cell Nucleus/genetics , Cell Nucleus/pathology , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin/physiology , Genomic Instability/genetics , Genomic Instability/physiology , Humans , Lamins/genetics , Lamins/metabolism , Lamins/physiology , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Envelope/pathology , Organelle Shape/physiology
18.
Genome Res ; 21(1): 83-94, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21149390

ABSTRACT

LSH, a member of the SNF2 family of chromatin remodeling ATPases encoded by the Hells gene, is essential for normal levels of DNA methylation in the mammalian genome. While the role of LSH in the methylation of repetitive DNA sequences is well characterized, its contribution to the regulation of DNA methylation and the expression of protein-coding genes has not been studied in detail. In this report we investigate genome-wide patterns of DNA methylation at gene promoters in Hells(-/-) mouse embryonic fibroblasts (MEFs). We find that in the absence of LSH, DNA methylation is lost or significantly reduced at ∼20% of all normally methylated promoter sequences. As a consequence, a large number of genes are misexpressed in Hells(-/-) MEFs. Comparison of Hells(-/-) MEFs with wild-type MEFs and embryonic stem (ES) cells suggests that LSH is important for de novo DNA methylation events that accompany the establishment and differentiation of embryonic lineage cells. We further show that the generation of normal DNA methylation patterns and stable gene silencing at specific promoters require cooperation between LSH and the G9a/GLP complex of histone methylases. At such loci, G9a recruitment is compromised when LSH is absent or greatly reduced. Taken together, our data suggest a mechanism whereby LSH promotes binding of DNA methyltransferases and the G9a/GLP complex to specific loci and facilitates developmentally programmed DNA methylation and stable gene silencing during lineage commitment and differentiation.


Subject(s)
DNA Helicases/metabolism , DNA Methylation , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA Helicases/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Gene Silencing , Histone-Lysine N-Methyltransferase/genetics , Methyltransferases/genetics , Methyltransferases/metabolism , Mice
19.
Chromosoma ; 113(2): 84-91, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15300445

ABSTRACT

Centromeres are required for faithful segregation of chromosomes in cell division. It is not clear how centromere sites are specified on chromosomes in vertebrates. We have previously introduced a mini-chromosome, named ST1, into a variety of cell lines including human HT1080, mouse LA9 and chicken DT40. This mini-chromosome, segregating faithfully in these cells, contains mouse minor and major, and human Y alpha-satellite DNA repeats. In this study, after determining the organisation of the satellite repeats, we investigated the location of the centromere on the mini-chromosome by combined immunocytochemistry and fluorescence in situ hybridisation analysis. Centromeric proteins were consistently co-localised with the minor satellite repeats in all three cell lines. When chromatin fibres were highly stretched, centromeric proteins were only seen on a small portion of the minor satellite repeats. These results indicate that a fraction of the minor satellite repeats is competent in centromere function not only in mouse but also in human and chicken cells.


Subject(s)
Chromosomes, Artificial, Mammalian/physiology , DNA, Satellite/physiology , DNA-Binding Proteins/metabolism , Minisatellite Repeats/physiology , Animals , Cell Line , Centromere/genetics , Centromere/physiology , Chickens , Chromatin/genetics , Chromatin/physiology , Chromosomes, Artificial, Mammalian/genetics , DNA, Satellite/genetics , Humans , Mice , Minisatellite Repeats/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...