Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nature ; 587(7832): 109-114, 2020 11.
Article in English | MEDLINE | ID: mdl-32908309

ABSTRACT

Despite its size and rigidity, the cell nucleus can be moved or reorganized by cytoskeletal filaments under various conditions (for example, during viral infection)1-11. Moreover, whereas chromatin organizes into non-random domains12, extensive heterogeneity at the single-cell level13 means that precisely how and why nuclei reorganize remains an area of intense investigation. Here we describe convolutional neural network-based automated cell classification and analysis pipelines, which revealed the extent to which human cytomegalovirus generates nuclear polarity through a virus-assembled microtubule-organizing centre. Acetylation of tubulin enables microtubules emanating from this centre to rotate the nucleus by engaging cytoplasmically exposed dynein-binding domains in the outer nuclear membrane protein nesprin-2G, which polarizes the inner nuclear membrane protein SUN1. This in turn creates intranuclear polarity in emerin, and thereby controls nuclear actin filaments that spatially segregate viral DNA from inactive histones and host DNA, maximizing virus replication. Our findings demonstrate the extent to which viruses can control the nucleus from the cytoplasm.


Subject(s)
Cell Nucleus/metabolism , Cell Polarity , Cytomegalovirus/physiology , Cytoplasm/metabolism , Cytoplasm/virology , Acetylation , Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Line , Cell Nucleus/chemistry , DNA, Viral/metabolism , Dyneins/metabolism , Histones/metabolism , Humans , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/chemistry , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Neural Networks, Computer , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Rotation , Tubulin/chemistry , Tubulin/metabolism , Virus Replication
2.
Proc Natl Acad Sci U S A ; 113(12): E1691-700, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26957603

ABSTRACT

The linear distribution of genes across chromosomes and the spatial localization of genes within the nucleus are related to their transcriptional regulation. The mechanistic consequences of linear gene order, and how it may relate to the functional output of genome organization, remain to be fully resolved, however. Here we tested the relationship between linear and 3D organization of gene regulation during myogenesis. Our analysis has identified a subset of topologically associated domains (TADs) that are significantly enriched for muscle-specific genes. These lineage-enriched TADs demonstrate an expression-dependent pattern of nuclear organization that influences the positioning of adjacent nonenriched TADs. Therefore, lineage-enriched TADs inform cell-specific genome organization during myogenesis. The reduction of allelic spatial distance of one of these domains, which contains Myogenin, correlates with reduced transcriptional variability, identifying a potential role for lineage-specific nuclear topology. Using a fusion-based strategy to decouple mitosis and myotube formation, we demonstrate that the cell-specific topology of syncytial nuclei is dependent on cell division. We propose that the effects of linear and spatial organization of gene loci on gene regulation are linked through TAD architecture, and that mitosis is critical for establishing nuclear topologies during cellular differentiation.


Subject(s)
Cell Lineage/genetics , Gene Expression Regulation, Developmental , Muscle Development/genetics , Alleles , Chromosome Mapping , Fibroblasts , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , Imaging, Three-Dimensional , In Situ Hybridization, Fluorescence , MyoD Protein/genetics , Myogenin/genetics , Protein Structure, Tertiary , Transcription, Genetic , Transduction, Genetic
3.
Differentiation ; 102: 19-26, 2018.
Article in English | MEDLINE | ID: mdl-29979997

ABSTRACT

The protein-DNA complexes that compose the end of mammalian chromosomes-telomeres-serve to stabilize linear genomic DNA and are involved in cellular and organismal aging. One mechanism that protects telomeres from premature degradation is the formation of structures called t-loops, in which the single-stranded 3' overhang present at the terminal end of telomeres loops back and invades medial double-stranded telomeric DNA. We identified looped structures formed between terminal chromosome ends and interstitial telomeric sequences (ITSs), which are found throughout the human genome, that we have termed interstitial telomeric loops (ITLs). While they form in a TRF2-dependent manner similar to t-loops, ITLs further require the physical interaction of TRF2 with the nuclear intermediate filament protein lamin A/C. Our findings suggest that interactions between telomeres and the nucleoskeleton broadly impact genomic integrity, including telomere stability, chromosome structure, and chromosome fragility. Here, we review the roles of TRF2 and lamin A/C in telomere biology and consider how their interaction may relate telomere homeostasis to cellular and organismal aging.


Subject(s)
Aging/genetics , Lamin Type A/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 2/genetics , Animals , DNA/metabolism , DNA-Binding Proteins/genetics , Humans , Telomere/genetics , Telomeric Repeat Binding Protein 2/metabolism
4.
Genes Dev ; 25(24): 2579-93, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22155925

ABSTRACT

Nuclear lamin B1 (LB1) is a major structural component of the nucleus that appears to be involved in the regulation of many nuclear functions. The results of this study demonstrate that LB1 expression in WI-38 cells decreases during cellular senescence. Premature senescence induced by oncogenic Ras also decreases LB1 expression through a retinoblastoma protein (pRb)-dependent mechanism. Silencing the expression of LB1 slows cell proliferation and induces premature senescence in WI-38 cells. The effects of LB1 silencing on proliferation require the activation of p53, but not pRb. However, the induction of premature senescence requires both p53 and pRb. The proliferation defects induced by silencing LB1 are accompanied by a p53-dependent reduction in mitochondrial reactive oxygen species (ROS), which can be rescued by growth under hypoxic conditions. In contrast to the effects of LB1 silencing, overexpression of LB1 increases the proliferation rate and delays the onset of senescence of WI-38 cells. This overexpression eventually leads to cell cycle arrest at the G1/S boundary. These results demonstrate the importance of LB1 in regulating the proliferation and senescence of human diploid cells through a ROS signaling pathway.


Subject(s)
Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Cell Cycle/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Proliferation , Cellular Senescence/genetics , Gene Expression Profiling , Gene Expression Regulation , Gene Silencing , Humans , Reactive Oxygen Species/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Telomere/metabolism , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism
5.
J Cell Sci ; 129(18): 3412-25, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27505898

ABSTRACT

Actin is abundant in the nucleus and it is clear that nuclear actin has important functions. However, mystery surrounds the absence of classical actin filaments in the nucleus. To address this question, we investigated how polymerizing nuclear actin into persistent nuclear actin filaments affected transcription by RNA polymerase II. Nuclear filaments impaired nuclear actin dynamics by polymerizing and sequestering nuclear actin. Polymerizing actin into stable nuclear filaments disrupted the interaction of actin with RNA polymerase II and correlated with impaired RNA polymerase II localization, dynamics, gene recruitment, and reduced global transcription and cell proliferation. Polymerizing and crosslinking nuclear actin in vitro similarly disrupted the actin-RNA-polymerase-II interaction and inhibited transcription. These data rationalize the general absence of stable actin filaments in mammalian somatic nuclei. They also suggest a dynamic pool of nuclear actin is required for the proper localization and activity of RNA polymerase II.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Nucleus/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , Actins/metabolism , Animals , COS Cells , Cell Proliferation , Chlorocebus aethiops , Cross-Linking Reagents/metabolism , HeLa Cells , Humans , Polymerization , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Nucleic Acids Res ; 44(7): 3082-94, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26673704

ABSTRACT

Higher order chromatin structure establishes domains that organize the genome and coordinate gene expression. However, the molecular mechanisms controlling transcription of individual loci within a topological domain (TAD) are not fully understood. The cystic fibrosis transmembrane conductance regulator (CFTR) gene provides a paradigm for investigating these mechanisms.CFTR occupies a TAD bordered by CTCF/cohesin binding sites within which are cell-type-selective cis-regulatory elements for the locus. We showed previously that intronic and extragenic enhancers, when occupied by specific transcription factors, are recruited to the CFTR promoter by a looping mechanism to drive gene expression. Here we use a combination of CRISPR/Cas9 editing of cis-regulatory elements and siRNA-mediated depletion of architectural proteins to determine the relative contribution of structural elements and enhancers to the higher order structure and expression of the CFTR locus. We found the boundaries of the CFTRTAD are conserved among diverse cell types and are dependent on CTCF and cohesin complex. Removal of an upstream CTCF-binding insulator alters the interaction profile, but has little effect on CFTR expression. Within the TAD, intronic enhancers recruit cell-type selective transcription factors and deletion of a pivotal enhancer element dramatically decreases CFTR expression, but has minor effect on its 3D structure.


Subject(s)
Chromatin/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Insulator Elements , CCCTC-Binding Factor , Caco-2 Cells , Cell Cycle Proteins/metabolism , Cell Line , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Genetic Loci , Humans , Repressor Proteins/metabolism , Cohesins
7.
Proc Natl Acad Sci U S A ; 111(14): 5260-5, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706864

ABSTRACT

α-Catenin (α-cat) is an actin-binding protein required for cell-cell cohesion. Although this adhesive function for α-cat is well appreciated, cells contain a substantial amount of nonjunctional α-cat that may be used for other functions. We show that α-cat is a nuclear protein that can interact with ß-catenin (ß-cat) and T-cell factor (TCF) and that the nuclear accumulation of α-cat depends on ß-cat. Using overexpression, knockdown, and chromatin immunoprecipitation approaches, we show that α-cat attenuates Wnt/ß-cat-responsive genes in a manner that is downstream of ß-cat/TCF loading on promoters. Both ß-cat- and actin-binding domains of α-cat are required to inhibit Wnt signaling. A nuclear-targeted form of α-cat induces the formation of nuclear filamentous actin, whereas cells lacking α-cat show altered nuclear actin properties. Formation of nuclear actin filaments correlates with reduced RNA synthesis and altered chromatin organization. Conversely, nuclear extracts made from cells lacking α-cat show enhanced general transcription in vitro, an activity that can be partially rescued by restoring the C-terminal actin-binding region of α-cat. These data demonstrate that α-cat may limit gene expression by affecting nuclear actin organization.


Subject(s)
Transcription, Genetic/physiology , alpha Catenin/physiology , Cell Line, Tumor , Humans , Signal Transduction
8.
Nucleic Acids Res ; 42(15): 9612-22, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25081205

ABSTRACT

Higher order chromatin structures across the genome are maintained in part by the architectural proteins CCCTC binding factor (CTCF) and the cohesin complex, which co-localize at many sites across the genome. Here, we examine the role of these proteins in mediating chromatin structure at the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CFTR encompasses nearly 200 kb flanked by CTCF-binding enhancer-blocking insulator elements and is regulated by cell-type-specific intronic enhancers, which loop to the promoter in the active locus. SiRNA-mediated depletion of CTCF or the cohesin component, RAD21, showed that these two factors have distinct roles in regulating the higher order organization of CFTR. CTCF mediates the interactions between CTCF/cohesin binding sites, some of which have enhancer-blocking insulator activity. Cohesin shares this tethering role, but in addition stabilizes interactions between the promoter and cis-acting intronic elements including enhancers, which are also dependent on the forkhead box A1/A2 (FOXA1/A2) transcription factors (TFs). Disruption of the three-dimensional structure of the CFTR gene by depletion of CTCF or RAD21 increases gene expression, which is accompanied by alterations in histone modifications and TF occupancy across the locus, and causes internalization of the gene from the nuclear periphery.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gene Expression Regulation , Repressor Proteins/metabolism , Binding Sites , CCCTC-Binding Factor , Caco-2 Cells , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/physiology , Cell Line , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Genetic Loci , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/physiology , Cohesins
9.
Biochim Biophys Acta ; 1839(3): 178-90, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412853

ABSTRACT

The spatial organization of the nucleus results in a compartmentalized structure that affects all aspects of nuclear function. This compartmentalization involves genome organization as well as the formation of nuclear bodies and plays a role in many functions, including gene regulation, genome stability, replication, and RNA processing. Here we review the recent findings associated with the spatial organization of the nucleus and reveal that a common theme for nuclear proteins is their ability to participate in a variety of functions and pathways. We consider this multiplicity of function in terms of Crowdsourcing, a recent phenomenon in the world of information technology, and suggest that this model provides a novel way to synthesize the many intersections between nuclear organization and function. This article is part of a Special Issue entitled: Chromatin and epigenetic regulation of animal development.


Subject(s)
Cell Nucleus/metabolism , DNA Replication/physiology , Gene Expression Regulation/physiology , Genomic Instability/physiology , RNA Processing, Post-Transcriptional/physiology , Animals , Cell Nucleus/genetics , Humans
10.
Chromosoma ; 122(5): 351-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23749207

ABSTRACT

Bacterial artificial chromosomes (BACs) are widely used in transgenesis, particularly for the humanization of animal models. Moreover, due to their extensive capacity, BACs provide attractive tools to study distal regulatory elements associated with large gene loci. However, despite their widespread use, little is known about the integration dynamics of these large transgenes in mammalian cells. Here, we investigate the post-integration structure of a ~260 kb BAC carrying the cystic fibrosis transmembrane conductance regulator (CFTR) locus following delivery by bacterial invasion and compare this to the outcome of a more routine lipid-based delivery method. We find substantial variability in integrated copy number and expression levels of the BAC CFTR transgene after bacterial invasion-mediated delivery. Furthermore, we frequently observed variation in the representation of different regions of the CFTR transgene within individual cell clones, indicative of BAC fragmentation. Finally, using fluorescence in situ hybridization, we observed that the integrated BAC forms extended megabase-scale structures in some clones that are apparently stably maintained at cell division. These data demonstrate that the utility of large BACs to investigate cis-regulatory elements in the genomic context may be limited by recombination events that complicate their use.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gene Transfer Techniques , Transgenes/genetics , Animals , Genetic Vectors , Humans , In Situ Hybridization, Fluorescence , Regulatory Sequences, Nucleic Acid/genetics
12.
Proc Natl Acad Sci U S A ; 106(16): 6679-84, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19276122

ABSTRACT

Although the importance of chromosome organization during mitosis is clear, it remains to be determined whether the nucleus assumes other functionally relevant chromosomal topologies. We have previously shown that homologous chromosomes have a tendency to associate during hematopoiesis according to their distribution of coregulated genes, suggesting cell-specific nuclear organization. Here, using the mathematical approaches of distance matrices and coupled oscillators, we model the dynamic relationship between gene expression and chromosomal associations during the differentiation of a multipotential hematopoietic progenitor. Our analysis reveals dramatic changes in total genomic order: Commitment of the progenitor results in an initial increase in entropy at both the level of gene coregulation and chromosomal organization, which we suggest represents a phase transition, followed by a progressive decline in entropy during differentiation. The stabilization of a highly ordered state in the differentiated cell types results in lineage-specific chromosomal topologies and is related to the emergence of coherence-or self-organization-between chromosomal associations and coordinate gene regulation. We discuss how these observations may be generally relevant to cell fate decisions encountered by progenitor/stem cells.


Subject(s)
Cell Lineage/genetics , Chromosomes/genetics , Gene Expression Regulation , Cell Differentiation/genetics
13.
Mol Syst Biol ; 6: 395, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20664641

ABSTRACT

The nuclei of differentiating cells exhibit several fundamental principles of self-organization. They are composed of many dynamical units connected physically and functionally to each other--a complex network--and the different parts of the system are mutually adapted and produce a characteristic end state. A unique cell-specific signature emerges over time from complex interactions among constituent elements that delineate coordinate gene expression and chromosome topology. Each element itself consists of many interacting components, all dynamical in nature. Self-organizing systems can be simplified while retaining complex information using approaches that examine the relationship between elements, such as spatial relationships and transcriptional information. These relationships can be represented using well-defined networks. We hypothesize that during the process of differentiation, networks within the cell nucleus rewire according to simple rules, from which a higher level of order emerges. Studying the interaction within and among networks provides a useful framework for investigating the complex organization and dynamic function of the nucleus.


Subject(s)
Cell Differentiation/genetics , Cell Nucleus/physiology , Chromosomes , Gene Regulatory Networks , Signal Transduction/genetics , Systems Biology , Animals , Gene Expression Regulation , Humans , Models, Theoretical
14.
PLoS Biol ; 5(11): e309, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18031200

ABSTRACT

Gene loci are found in nuclear subcompartments that are related to their expression status. For instance, silent genes are often localized to heterochromatin and the nuclear periphery, whereas active genes tend to be found in the nuclear center. Evidence also suggests that chromosomes may be specifically positioned within the nucleus; however, the nature of this organization and how it is achieved are not yet fully understood. To examine whether gene regulation is related to a discernible pattern of genomic organization, we analyzed the linear arrangement of co-regulated genes along chromosomes and determined the organization of chromosomes during the differentiation of a hematopoietic progenitor to erythroid and neutrophil cell types. Our analysis reveals that there is a significant tendency for co-regulated genes to be proximal, which is related to the association of homologous chromosomes and the spatial juxtaposition of lineage-specific gene domains. We suggest that proximity in the form of chromosomal gene distribution and homolog association may be the basis for organizing the genome for coordinate gene regulation during cellular differentiation.


Subject(s)
Chromosome Positioning/genetics , Gene Expression Regulation, Developmental , Genome , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation , Cell Nucleus/genetics , Cells, Cultured , Erythroid Cells/cytology , Hematopoietic Stem Cells/cytology , Mice , Neutrophils/cytology , Neutrophils/physiology , Oligonucleotide Array Sequence Analysis
15.
Dev Cell ; 2(6): 690-2, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12062080

ABSTRACT

The interchromosome domain (ICD) model proposes that genes are selectively positioned at the surfaces of chromosome territories to facilitate their regulation. A paper in the May 13 issue of the Journal of Cell Biology provides evidence that supports a reinterpretation of this model.


Subject(s)
Chromosomes , Gene Expression Regulation , Transcription, Genetic/genetics , Cell Nucleus/genetics , Centromere/chemistry , Chromosome Mapping , Chromosome Painting , Chromosomes/genetics , Euchromatin/genetics , Heterochromatin/chemistry , Interphase , Models, Biological
16.
Sci Rep ; 9(1): 8011, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142749

ABSTRACT

The three-dimensional organization of the genome in mammalian interphase nuclei is intrinsically linked to the regulation of gene expression. Whole chromosome territories and their encoded gene loci occupy preferential positions within the nucleus that changes according to the expression profile of a given cell lineage or stage. To further illuminate the relationship between chromosome organization, epigenetic environment, and gene expression, here we examine the functional organization of chromosome X and corresponding X-linked genes in a variety of healthy human and disease state X diploid (XX) cells. We observe high frequencies of homologous chromosome X colocalization (or coalescence), typically associated with initiation of X-chromosome inactivation, occurring in XX cells outside of early embryogenesis. Moreover, during chromosome X coalescence significant changes in Xist, H3K27me3, and X-linked gene expression occur, suggesting the potential exchange of gene regulatory information between the active and inactive X chromosomes. We also observe significant differences in chromosome X coalescence in disease-implicated lymphocytes isolated from systemic lupus erythematosus (SLE) patients compared to healthy controls. These results demonstrate that X chromosomes can functionally interact outside of embryogenesis when X inactivation is initiated and suggest a potential gene regulatory mechanism aberration underlying the increased frequency of autoimmunity in XX individuals.


Subject(s)
Dosage Compensation, Genetic/genetics , Lupus Erythematosus, Systemic/genetics , RNA, Long Noncoding/genetics , X Chromosome/genetics , Animals , Cell Nucleus/genetics , Diploidy , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental/genetics , Genes, X-Linked , Humans , Lupus Erythematosus, Systemic/pathology , Male , X Chromosome Inactivation/genetics
18.
Gend Genome ; 2(1): 2-7, 2018 Jan.
Article in English | MEDLINE | ID: mdl-30899898

ABSTRACT

Sex chromosome gene dosage compensation is required to ensure equivalent levels of X-linked gene expression between males (46, XY) and females (46, XX). To achieve similar expression, X-chromosome inactivation (XCI) is initiated in female cells during early stages of embryogenesis. Within each cell, either the maternal or paternal X chromosome is selected for whole chromosome transcriptional silencing, which is initiated and maintained by epigenetic and chromatin conformation mechanisms. With the emergence of small-molecule epigenetic inhibitors for the treatment of disease, such as cancer, the epigenetic mechanism underlying XCI may be inadvertently targeted. Here, we test 2 small-molecule epigenetic inhibitors being used clinically, GSK126 (a histone H3 lysine 27 methyltransferase inhibitor) and suberoylanilide hydroxamic acid (a histone deacetylase inhibitor), on their effects of the inactive X (Xi) in healthy human female fibroblasts. The combination of these modifiers, at subcancer therapeutic levels, leads to the inability to detect the repressive H3K27me3 modification characteristic of XCI in the majority of the cells. Importantly, genes positioned near the X-inactivation center (Xic), where inactivation is initiated, exhibit robust expression with treatment of the inhibitors, while genes located near the distal ends of the X chromosome intriguingly exhibit significant downregulation. These results demonstrate that small-molecule epigenetic inhibitors can have profound consequences on XCI in human cells, and they underscore the importance of considering gender when developing and clinically testing small-molecule epigenetic inhibitors, in particular those that target the well-characterized mechanisms of X inactivation.

19.
Nucleus ; 6(3): 172-8, 2015.
Article in English | MEDLINE | ID: mdl-25961132

ABSTRACT

Ever since the first demonstration of their repetitive sequence and unique replication pathway, telomeres have beguiled researchers with how they function in protecting chromosome ends. Of course much has been learned over the years, and we now appreciate that telomeres are comprised of the multimeric protein/DNA shelterin complex and that the formation of t-loops provides protection from DNA damage machinery. Deriving their name from D-loops, t-loops are generated by the insertion of the 3' overhang into telomeric repeats facilitated by the binding of TRF2. Recent studies have uncovered novel forms of chromosome end-structure that may implicate telomere organization in cellular processes beyond its essential role in telomere protection and homeostasis. In particular, we have recently described that t-loops form in a TRF2-dependent manner at interstitial telomere repeat sequences, which we termed interstitial telomere loops (ITLs). These structures are also dependent on association of lamin A/C, a canonical component of the nucleoskeleton that is mutated in myriad human diseases, including human segmental progeroid syndromes. Since ITLs are associated with telomere stability and require functional lamin A/C, our study suggests a mechanistic link between cellular aging (replicative senescence induced by telomere shortening) and organismal aging (modeled by Hutchinson Gilford Progeria Syndrome). Here we speculate on other potential ramifications of ITL formation, from gene expression to genome stability to chromosome structure.


Subject(s)
DNA/chemistry , Progeria/genetics , Telomere Shortening , Telomere/chemistry , Telomeric Repeat Binding Protein 2/genetics , Cell Division , DNA/metabolism , Gene Expression Regulation , Genomic Instability , Heterochromatin/chemistry , Heterochromatin/metabolism , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Nucleic Acid Conformation , Progeria/metabolism , Progeria/pathology , Shelterin Complex , Signal Transduction , Telomerase/genetics , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 2/metabolism
20.
Nat Commun ; 5: 5467, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25399868

ABSTRACT

Telomeres protect the ends of linear genomes, and the gradual loss of telomeres is associated with cellular ageing. Telomere protection involves the insertion of the 3' overhang facilitated by telomere repeat-binding factor 2 (TRF2) into telomeric DNA, forming t-loops. We present evidence suggesting that t-loops can also form at interstitial telomeric sequences in a TRF2-dependent manner, forming an interstitial t-loop (ITL). We demonstrate that TRF2 association with interstitial telomeric sequences is stabilized by co-localization with A-type lamins (lamin A/C). We also find that lamin A/C interacts with TRF2 and that reduction in levels of lamin A/C or mutations in LMNA that cause an autosomal dominant premature ageing disorder--Hutchinson Gilford Progeria Syndrome (HGPS)-lead to reduced ITL formation and telomere loss. We propose that cellular and organismal ageing are intertwined through the effects of the interaction between TRF2 and lamin A/C on chromosome structure.


Subject(s)
Chromosomes, Human/physiology , Lamin Type A/physiology , TATA Box Binding Protein-Like Proteins/physiology , Cellular Senescence/physiology , Fibroblasts/physiology , Humans , In Situ Hybridization, Fluorescence , Progeria/etiology , Telomere/physiology
SELECTION OF CITATIONS
SEARCH DETAIL