Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38260534

RESUMO

The linear DNA sequence of mammalian chromosomes is organized in large blocks of DNA with similar sequence properties, producing a pattern of dark and light staining bands on mitotic chromosomes. Cytogenetic banding is essentially invariant between people and cell-types and thus may be assumed unrelated to genome regulation. We investigate whether large blocks of Alu-rich R-bands and L1-rich G-bands provide a framework upon which functional genome architecture is built. We examine two models of large-scale chromatin condensation: X-chromosome inactivation and formation of senescence-associated heterochromatin foci (SAHFs). XIST RNA triggers gene silencing but also formation of the condensed Barr Body (BB), thought to reflect cumulative gene silencing. However, we find Alu-rich regions are depleted from the L1-rich BB, supporting it is a dense core but not the entire chromosome. Alu-rich bands are also gene-rich, affirming our earlier findings that genes localize at the outer periphery of the BB. SAHFs similarly form within each territory by coalescence of syntenic L1 regions depleted for highly Alu-rich DNA. Analysis of senescent cell Hi-C data also shows large contiguous blocks of G-band and R-band DNA remodel as a segmental unit. Entire dark-bands gain distal intrachromosomal interactions as L1-rich regions form the SAHF. Most striking is that sharp Alu peaks within R-bands resist these changes in condensation. We further show that Chr19, which is exceptionally Alu rich, fails to form a SAHF. Collective results show regulation of genome architecture corresponding to large blocks of DNA and demonstrate resistance of segments with high Alu to chromosome condensation.

2.
Cell Rep ; 42(7): 112686, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37384527

RESUMO

XIST RNA triggers chromosome-wide gene silencing and condenses an active chromosome into a Barr body. Here, we use inducible human XIST to examine early steps in the process, showing that XIST modifies cytoarchitecture before widespread gene silencing. In just 2-4 h, barely visible transcripts populate the large "sparse zone" surrounding the smaller "dense zone"; importantly, density zones exhibit different chromatin impacts. Sparse transcripts immediately trigger immunofluorescence for H2AK119ub and CIZ1, a matrix protein. H3K27me3 appears hours later in the dense zone, which enlarges with chromosome condensation. Genes examined are silenced after compaction of the RNA/DNA territory. Insights into this come from the findings that the A-repeat alone can silence genes and rapidly, but only where dense RNA supports sustained histone deacetylation. We propose that sparse XIST RNA quickly impacts architectural elements to condense the largely non-coding chromosome, coalescing RNA density that facilitates an unstable, A-repeat-dependent step required for gene silencing.


Assuntos
RNA Longo não Codificante , Inativação do Cromossomo X , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Cromatina , Inativação Gênica , Cromossomo X/metabolismo
3.
Cell Rep ; 21(13): 3691-3699, 2017 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-29281819

RESUMO

During female mouse embryogenesis, two forms of X chromosome inactivation (XCI) ensure dosage compensation from sex chromosomes. Beginning at the four-cell stage, imprinted XCI (iXCI) exclusively silences the paternal X (Xp), and this pattern is maintained in extraembryonic cell types. Epiblast cells, which give rise to the embryo proper, reactivate the Xp (XCR) and undergo a random form of XCI (rXCI) around implantation. Both iXCI and rXCI depend on the long non-coding RNA Xist. The ubiquitin ligase RLIM is required for iXCI in vivo and occupies a central role in current models of rXCI. Here, we demonstrate the existence of Rlim-dependent and Rlim-independent pathways for rXCI in differentiating female ESCs. Upon uncoupling these pathways, we find more efficient Rlim-independent XCI in ESCs cultured under physiological oxygen conditions. Our results revise current models of rXCI and suggest that caution must be taken when comparing XCI studies in ESCs and mice.


Assuntos
Células-Tronco Embrionárias Murinas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Inativação do Cromossomo X/genética , Animais , Técnicas de Cultura de Células , Feminino , Camundongos , Proteínas Mutantes/metabolismo
4.
Cell Rep ; 18(12): 2943-2956, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28329686

RESUMO

This study reveals that high-copy satellite II (HSATII) sequences in the human genome can bind and impact distribution of chromatin regulatory proteins and that this goes awry in cancer. In many cancers, master regulatory proteins form two types of cancer-specific nuclear bodies, caused by locus-specific deregulation of HSATII. DNA demethylation at the 1q12 mega-satellite, common in cancer, causes PRC1 aggregation into prominent Cancer-Associated Polycomb (CAP) bodies. These loci remain silent, whereas HSATII loci with reduced PRC1 become derepressed, reflecting imbalanced distribution of UbH2A on these and other PcG-regulated loci. Large nuclear foci of HSATII RNA form and sequester copious MeCP2 into Cancer-Associated Satellite Transcript (CAST) bodies. Hence, HSATII DNA and RNA have an exceptional capacity to act as molecular sponges and sequester chromatin regulatory proteins into abnormal nuclear bodies in cancer. The compartmentalization of regulatory proteins within nuclear structure, triggered by demethylation of "junk" repeats, raises the possibility that this contributes to further compromise of the epigenome and neoplastic progression.


Assuntos
Desmetilação do DNA , DNA Satélite/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Neoplasias/genética , Proteínas do Grupo Polycomb/metabolismo , RNA/metabolismo , Proteína BRCA1/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Loci Gênicos , Humanos , Modelos Biológicos , Complexo Repressor Polycomb 1/metabolismo , Agregados Proteicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
Elife ; 52016 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-27642011

RESUMO

Mammalian X-linked gene expression is highly regulated as female cells contain two and male one X chromosome (X). To adjust the X gene dosage between genders, female mouse preimplantation embryos undergo an imprinted form of X chromosome inactivation (iXCI) that requires both Rlim (also known as Rnf12) and the long non-coding RNA Xist. Moreover, it is thought that gene expression from the single active X is upregulated to correct for bi-allelic autosomal (A) gene expression. We have combined mouse genetics with RNA-seq on single mouse embryos to investigate functions of Rlim on the temporal regulation of iXCI and Xist. Our results reveal crucial roles of Rlim for the maintenance of high Xist RNA levels, Xist clouds and X-silencing in female embryos at blastocyst stages, while initial Xist expression appears Rlim-independent. We find further that X/A upregulation is initiated in early male and female preimplantation embryos.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Genes Ligados ao Cromossomo X , Ubiquitina-Proteína Ligases/metabolismo , Animais , Camundongos , RNA Longo não Codificante/metabolismo , Análise de Sequência de RNA , Inativação do Cromossomo X
6.
Nature ; 511(7507): 86-9, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24870238

RESUMO

In female mice, two forms of X-chromosome inactivation (XCI) ensure the selective silencing of female sex chromosomes during mouse embryogenesis. Beginning at the four-cell stage, imprinted XCI (iXCI) exclusively silences the paternal X chromosome. Later, around implantation, epiblast cells of the inner cell mass that give rise to the embryo reactivate the paternal X chromosome and undergo a random form of XCI (rXCI). Xist, a long non-coding RNA crucial for both forms of XCI, is activated by the ubiquitin ligase RLIM (also known as Rnf12). Although RLIM is required for triggering iXCI in mice, its importance for rXCI has been controversial. Here we show that RLIM levels are downregulated in embryonic cells undergoing rXCI. Using mouse genetics we demonstrate that female cells lacking RLIM from pre-implantation stages onwards show hallmarks of XCI, including Xist clouds and H3K27me3 foci, and have full embryogenic potential. These results provide evidence that RLIM is dispensable for rXCI, indicating that in mice an RLIM-independent mechanism activates Xist in the embryo proper.


Assuntos
Camadas Germinativas/embriologia , Camadas Germinativas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Inativação do Cromossomo X/genética , Animais , Regulação para Baixo , Implantação do Embrião , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Histonas/química , Histonas/metabolismo , Hibridização in Situ Fluorescente , Lisina/metabolismo , Metilação , Camundongos , Camundongos Knockout , RNA Longo não Codificante/genética , Ubiquitina-Proteína Ligases/genética
7.
Cell ; 156(5): 907-19, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24581492

RESUMO

Recent studies recognize a vast diversity of noncoding RNAs with largely unknown functions, but few have examined interspersed repeat sequences, which constitute almost half our genome. RNA hybridization in situ using C0T-1 (highly repeated) DNA probes detects surprisingly abundant euchromatin-associated RNA comprised predominantly of repeat sequences (C0T-1 RNA), including LINE-1. C0T-1-hybridizing RNA strictly localizes to the interphase chromosome territory in cis and remains stably associated with the chromosome territory following prolonged transcriptional inhibition. The C0T-1 RNA territory resists mechanical disruption and fractionates with the nonchromatin scaffold but can be experimentally released. Loss of repeat-rich, stable nuclear RNAs from euchromatin corresponds to aberrant chromatin distribution and condensation. C0T-1 RNA has several properties similar to XIST chromosomal RNA but is excluded from chromatin condensed by XIST. These findings impact two "black boxes" of genome science: the poorly understood diversity of noncoding RNA and the unexplained abundance of repetitive elements.


Assuntos
Cromossomos de Mamíferos/química , Eucromatina/química , Interfase , RNA não Traduzido/análise , Animais , Núcleo Celular/química , Humanos , Células Híbridas , Elementos Nucleotídeos Longos e Dispersos , Camundongos , RNA não Traduzido/genética , Sequências Repetitivas de Ácido Nucleico , Transcrição Gênica
8.
Science ; 341(6147): 789-92, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23907535

RESUMO

An inducible program of inflammatory gene expression is central to antimicrobial defenses. This response is controlled by a collaboration involving signal-dependent activation of transcription factors, transcriptional co-regulators, and chromatin-modifying factors. We have identified a long noncoding RNA (lncRNA) that acts as a key regulator of this inflammatory response. Pattern recognition receptors such as the Toll-like receptors induce the expression of numerous lncRNAs. One of these, lincRNA-Cox2, mediates both the activation and repression of distinct classes of immune genes. Transcriptional repression of target genes is dependent on interactions of lincRNA-Cox2 with heterogeneous nuclear ribonucleoprotein A/B and A2/B1. Collectively, these studies unveil a central role of lincRNA-Cox2 as a broad-acting regulatory component of the circuit that controls the inflammatory response.


Assuntos
Regulação da Expressão Gênica , Imunidade Inata/genética , Inflamação/genética , Macrófagos/imunologia , Macrófagos/metabolismo , RNA Longo não Codificante/genética , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Ciclo-Oxigenase 2/genética , Citocinas/genética , Citocinas/metabolismo , Citosol/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Ativação de Macrófagos , Camundongos , Modelos Imunológicos , Interferência de RNA , RNA Longo não Codificante/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional
9.
Nature ; 500(7462): 296-300, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23863942

RESUMO

Down's syndrome is a common disorder with enormous medical and social costs, caused by trisomy for chromosome 21. We tested the concept that gene imbalance across an extra chromosome can be de facto corrected by manipulating a single gene, XIST (the X-inactivation gene). Using genome editing with zinc finger nucleases, we inserted a large, inducible XIST transgene into the DYRK1A locus on chromosome 21, in Down's syndrome pluripotent stem cells. The XIST non-coding RNA coats chromosome 21 and triggers stable heterochromatin modifications, chromosome-wide transcriptional silencing and DNA methylation to form a 'chromosome 21 Barr body'. This provides a model to study human chromosome inactivation and creates a system to investigate genomic expression changes and cellular pathologies of trisomy 21, free from genetic and epigenetic noise. Notably, deficits in proliferation and neural rosette formation are rapidly reversed upon silencing one chromosome 21. Successful trisomy silencing in vitro also surmounts the major first step towards potential development of 'chromosome therapy'.


Assuntos
Cromossomos Humanos Par 21/genética , Mecanismo Genético de Compensação de Dose , Síndrome de Down/genética , RNA Longo não Codificante/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Metilação de DNA , Síndrome de Down/terapia , Inativação Gênica , Humanos , Células-Tronco Pluripotentes Induzidas , Masculino , Camundongos , Mutagênese Insercional , Neurogênese , RNA Longo não Codificante/genética , Cromatina Sexual/genética , Inativação do Cromossomo X/genética
10.
Curr Protoc Hum Genet ; Chapter 4: Unit 4.15, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23315927

RESUMO

Fluorescence in situ hybridization (FISH) is not a singular technique, but a battery of powerful and versatile tools for examining the distribution of endogenous genes and RNAs in precise context with each other and in relation to specific proteins or cell structures. This unit offers the details of highly sensitive and successful protocols that were initially developed largely in our lab and honed over a number of years. Our emphasis is on analysis of nuclear RNAs and DNA to address specific biological questions about nuclear structure, pre-mRNA metabolism, or the role of noncoding RNAs; however, cytoplasmic RNA detection is also discussed. Multifaceted molecular cytological approaches bring precise resolution and sensitive multicolor detection to illuminate the organization and functional roles of endogenous genes and their RNAs within the native structure of fixed cells. Solutions to several common technical pitfalls are discussed, as are cautions regarding the judicious use of digital imaging and the rigors of analyzing and interpreting complex molecular cytological results.


Assuntos
Núcleo Celular/genética , DNA/análise , Hibridização in Situ Fluorescente/métodos , RNA/análise , Animais , Sondas de DNA , Humanos , Indicadores e Reagentes , Sondas de Oligonucleotídeos
11.
Cell ; 149(3): 630-41, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22541433

RESUMO

In female mouse embryos, somatic cells undergo a random form of X chromosome inactivation (XCI), whereas extraembryonic trophoblast cells in the placenta undergo imprinted XCI, silencing exclusively the paternal X chromosome. Initiation of imprinted XCI requires a functional maternal allele of the X-linked gene Rnf12, which encodes the ubiquitin ligase Rnf12/RLIM. We find that knockout (KO) of Rnf12 in female mammary glands inhibits alveolar differentiation and milk production upon pregnancy, with alveolar cells that lack RLIM undergoing apoptosis as they begin to differentiate. Genetic analyses demonstrate that these functions are mediated primarily by the paternal Rnf12 allele due to nonrandom maternal XCI in mammary epithelial cells. These results identify paternal Rnf12/RLIM as a critical survival factor for milk-producing alveolar cells and, together with population models, reveal implications of transgenerational epigenetic inheritance.


Assuntos
Sobrevivência Celular , Glândulas Mamárias Animais/citologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Epigênese Genética , Feminino , Impressão Genômica , Masculino , Glândulas Mamárias Animais/fisiologia , Camundongos , Gravidez , Ubiquitina-Proteína Ligases/genética , Inativação do Cromossomo X
12.
Nat Struct Mol Biol ; 18(1): 107-14, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21131981

RESUMO

We developed a general approach that combines chromosome conformation capture carbon copy (5C) with the Integrated Modeling Platform (IMP) to generate high-resolution three-dimensional models of chromatin at the megabase scale. We applied this approach to the ENm008 domain on human chromosome 16, containing the α-globin locus, which is expressed in K562 cells and silenced in lymphoblastoid cells (GM12878). The models accurately reproduce the known looping interactions between the α-globin genes and their distal regulatory elements. Further, we find using our approach that the domain folds into a single globular conformation in GM12878 cells, whereas two globules are formed in K562 cells. The central cores of these globules are enriched for transcribed genes, whereas nontranscribed chromatin is more peripheral. We propose that globule formation represents a higher-order folding state related to clustering of transcribed genes around shared transcription machineries, as previously observed by microscopy.


Assuntos
Cromatina/química , Cromossomos Humanos Par 16/química , alfa-Globinas/genética , Cromatina/ultraestrutura , Cromossomos Humanos Par 16/metabolismo , Cromossomos Humanos Par 16/ultraestrutura , Humanos , Hibridização in Situ Fluorescente , Células K562 , Modelos Moleculares , Conformação de Ácido Nucleico , alfa-Globinas/química
13.
Nature ; 467(7318): 977-81, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20962847

RESUMO

Two forms of X-chromosome inactivation (XCI) ensure the selective silencing of female sex chromosomes during mouse embryogenesis. Imprinted XCI begins with the detection of Xist RNA expression on the paternal X chromosome (Xp) at about the four-cell stage of embryonic development. In the embryonic tissues of the inner cell mass, a random form of XCI occurs in blastocysts that inactivates either Xp or the maternal X chromosome (Xm). Both forms of XCI require the non-coding Xist RNA that coats the inactive X chromosome from which it is expressed. Xist has crucial functions in the silencing of X-linked genes, including Rnf12 (refs 3, 4) encoding the ubiquitin ligase RLIM (RING finger LIM-domain-interacting protein). Here we show, by targeting a conditional knockout of Rnf12 to oocytes where RLIM accumulates to high levels, that the maternal transmission of the mutant X chromosome (Δm) leads to lethality in female embryos as a result of defective imprinted XCI. We provide evidence that in Δm female embryos the initial formation of Xist clouds and Xp silencing are inhibited. In contrast, embryonic stem cells lacking RLIM are able to form Xist clouds and silence at least some X-linked genes during random XCI. These results assign crucial functions to the maternal deposit of Rnf12/RLIM for the initiation of imprinted XCI.


Assuntos
Cromossomos de Mamíferos/genética , Impressão Genômica , Mães , Proteínas Repressoras/metabolismo , Inativação do Cromossomo X/genética , Cromossomo X/genética , Animais , Animais Congênicos , Blastocisto/metabolismo , Linhagem Celular , Perda do Embrião/genética , Pai , Feminino , Inativação Gênica , Masculino , Camundongos , Camundongos Transgênicos , RNA Longo não Codificante , RNA não Traduzido/genética , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Ubiquitina-Proteína Ligases
14.
J Cell Biol ; 186(4): 491-507, 2009 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-19704020

RESUMO

How XIST RNA strictly localizes across the inactive X chromosome is unknown; however, prophase release of human XIST RNA provides a clue. Tests of inhibitors that mimic mitotic chromatin modifications implicated an indirect role of PP1 (protein phosphatase 1), potentially via its interphase repression of Aurora B kinase (AURKB), which phosphorylates H3 and chromosomal proteins at prophase. RNA interference to AURKB causes mitotic retention of XIST RNA, unlike other mitotic or broad kinase inhibitors. Thus, AURKB plays an unexpected role in regulating RNA binding to heterochromatin, independent of mechanics of mitosis. H3 phosphorylation (H3ph) was shown to precede XIST RNA release, whereas results exclude H1ph involvement. Of numerous Xi chromatin (chromosomal protein) hallmarks, ubiquitination closely follows XIST RNA retention or release. Surprisingly, H3S10ph staining (but not H3S28ph) is excluded from Xi and is potentially linked to ubiquitination. Results suggest a model of multiple distinct anchor points for XIST RNA. This study advances understanding of RNA chromosome binding and the roles of AURKB and demonstrates a novel approach to manipulate and study XIST RNA.


Assuntos
Cromatina/metabolismo , Cromossomos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA não Traduzido/metabolismo , RNA/metabolismo , Animais , Aurora Quinase B , Aurora Quinases , Ciclo Celular/fisiologia , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos/genética , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Feminino , Humanos , Indóis/metabolismo , Masculino , Complexo Mediador , Modelos Moleculares , Proteína Fosfatase 1/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , RNA/genética , Interferência de RNA , RNA Longo não Codificante , RNA não Traduzido/genética , Sulfonamidas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transgenes
15.
J Cell Physiol ; 216(2): 445-52, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18340642

RESUMO

The clinical and research value of human embryonic stem cells (hESC) depends upon maintaining their epigenetically naïve, fully undifferentiated state. Inactivation of one X chromosome in each cell of mammalian female embryos is a paradigm for one of the earliest steps in cell specialization through formation of facultative heterochromatin. Mouse ES cells are derived from the inner cell mass (ICM) of blastocyst stage embryos prior to X-inactivation, and cultured murine ES cells initiate this process only upon differentiation. Less is known about human X-inactivation during early development. To identify a human ES cell model for X-inactivation and study differences in the epigenetic state of hESC lines, we investigated X-inactivation in all growth competent, karyotypically normal, NIH approved, female hESC lines and several sublines. In the vast majority of undifferentiated cultures of nine lines examined, essentially all cells exhibit hallmarks of X-inactivation. However, subcultures of any hESC line can vary in X-inactivation status, comprising distinct sublines. Importantly, we identified rare sublines that have not yet inactivated Xi and retain competence to undergo X-inactivation upon differentiation. Other sublines exhibit defects in counting or maintenance of XIST expression on Xi. The few hESC sublines identified that have not yet inactivated Xi may reflect the earlier epigenetic state of the human ICM and represent the most promising source of NIH hESC for study of human X-inactivation. The many epigenetic anomalies seen indicate that maintenance of fully unspecialized cells, which have not formed Xi facultative heterochromatin, is a delicate epigenetic balance difficult to maintain in culture.


Assuntos
Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/fisiologia , Epigênese Genética , Inativação do Cromossomo X , Animais , Cromossomos Humanos X , Mecanismo Genético de Compensação de Dose , Células-Tronco Embrionárias/citologia , Feminino , Humanos , Camundongos , RNA Longo não Codificante , RNA não Traduzido/genética , RNA não Traduzido/metabolismo
16.
J Cell Biol ; 178(6): 951-64, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17846170

RESUMO

In myotonic dystrophy type 1 (DM1), triplet repeat expansion in the 3' untranslated region of dystrophia myotonica protein kinase (DMPK) causes the nuclear retention of mutant messenger RNA (mRNA). Although the DMPK gene locus positions precisely at the outer edge of a factor-rich SC-35 domain, the normal mRNA consistently accumulates within the domain, and this RNA is depleted upon transcriptional inhibition. In DM1, mutant transcripts detach from the gene but accumulate in granules that abut but do not enter SC-35 domains, suggesting that RNA entry into the domain is blocked. Despite their exclusion from these compartments, mutant transcripts are spliced. MBNL1 (muscleblind-like protein 1) is an alternative splicing factor that becomes highly concentrated with mutant RNA foci. Small interfering RNA-mediated knockdown of MBNL1 promotes the accumulation or entry of newly synthesized mutant transcripts in the SC-35 domain. Collectively, these data suggest that an initial step in the intranuclear path of some mRNAs is passage from the gene into an SC-35 domain and implicate these structures in postsplicing steps before export.


Assuntos
Distrofia Miotônica/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Splicing de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Células Cultivadas , Criança , Humanos , Masculino , Modelos Moleculares , Mutação , Mioblastos Esqueléticos/metabolismo , Distrofia Miotônica/genética , Miotonina Proteína Quinase , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Transporte de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Expansão das Repetições de Trinucleotídeos
17.
Anat Rec A Discov Mol Cell Evol Biol ; 288(7): 664-75, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16761280

RESUMO

Direct localization of specific genes, RNAs, and proteins has allowed the dissection of individual nuclear speckles in relation to the molecular biology of gene expression. Nuclear speckles (aka SC35 domains) are essentially ubiquitous structures enriched for most pre-mRNA metabolic factors, yet their relationship to gene expression has been poorly understood. Analyses of specific genes and their spliced or mature mRNA strongly support that SC35 domains are hubs of activity, not stores of inert factors detached from gene expression. We propose that SC35 domains are hubs that spatially link expression of specific pre-mRNAs to rapid recycling of copious RNA metabolic complexes, thereby facilitating expression of many highly active genes. In addition to increasing the efficiency of each step, sequential steps in gene expression are structurally integrated at each SC35 domain, consistent with other evidence that the biochemical machineries for transcription, splicing, and mRNA export are coupled. Transcription and splicing are subcompartmentalized at the periphery, with largely spliced mRNA entering the domain prior to export. In addition, new findings presented here begin to illuminate the structural underpinnings of a speckle by defining specific perturbations of phosphorylation that promote disassembly or assembly of an SC35 domain in relation to other components. Results thus far are consistent with the SC35 spliceosome assembly factor as an integral structural component. Conditions that disperse SC35 also disperse poly(A) RNA, whereas the splicing factor ASF/SF2 can be dispersed under conditions in which SC35 or SRm300 remain as intact components of a core domain.


Assuntos
Proteínas Nucleares/química , Proteínas Nucleares/fisiologia , Precursores de RNA/química , Precursores de RNA/metabolismo , Ribonucleoproteínas/química , Ribonucleoproteínas/fisiologia , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/genética , Núcleo Celular/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Proteínas Nucleares/genética , Estrutura Terciária de Proteína/genética , Precursores de RNA/biossíntese , Splicing de RNA/genética , Ribonucleoproteínas/genética , Fatores de Processamento de Serina-Arginina , Spliceossomos/química , Spliceossomos/genética , Spliceossomos/metabolismo
18.
Proc Natl Acad Sci U S A ; 103(20): 7688-93, 2006 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-16682630

RESUMO

We investigated whether genes escape X chromosome inactivation by positioning outside of the territory defined by XIST RNA. Results reveal an unanticipated higher order organization of genes and noncoding sequences. All 15 X-linked genes, regardless of activity, position on the border of the XIST RNA territory, which resides outside of the DAPI-dense Barr body. Although more strictly delineated on the inactive X chromosome (Xi), all genes localized predominantly to the outer rim of the Xi and active X chromosome. This outer rim is decorated only by X chromosome DNA paints and is excluded from both the XIST RNA and dense DAPI staining. The only DNA found well within the Barr body and XIST RNA territory was centromeric and Cot-1 DNA; hence, the core of the X chromosome essentially excludes genes and is composed primarily of noncoding repeat-rich DNA. Moreover, we show that this core of repetitive sequences is expressed throughout the nucleus yet is silenced throughout Xi, providing direct evidence for chromosome-wide regulation of "junk" DNA transcription. Collective results suggest that the Barr body, long presumed to be the physical manifestation of silenced genes, is in fact composed of a core of silenced noncoding DNA. Instead of acting at a local gene level, XIST RNA appears to interact with and silence core architectural elements to effectively condense and shut down the Xi.


Assuntos
Cromossomos Humanos X/genética , Inativação Gênica , Genes Ligados ao Cromossomo X , Inativação do Cromossomo X , Cromatina/genética , Cromatina/metabolismo , Cromossomos Humanos X/ultraestrutura , Feminino , Humanos , Hibridização In Situ , RNA Longo não Codificante , RNA não Traduzido/genética , Cromatina Sexual/genética , Transcrição Gênica
19.
Chromosoma ; 113(6): 324-35, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15616869

RESUMO

The inactive X chromosome (Xi) forms a heterochromatic structure in the nucleus that is known to have several modifications to specific histones involving acetylation or methylation. Using three different antibodies in four different cell lines, we demonstrate that the Xi in human and mouse cells is highly enriched in ubiquitinated protein(s), much of which is polyubiquitinated. This ubiquitination appears specific for the Xi as it was not observed for centromeres or other regions of heterochromatin. Results using an antibody specific to ubiquitinated H2A provide a clear link between H2A ubiquitination and gene repression, as visualized across an entire inactive chromosome. Interestingly, the ubiquitination of the chromosome persists into mitosis and can be seen in a reproducible banded pattern. This pattern matches that of Xist RNA which forms bands as it detaches from the mitotic X chromosome. Both ubiquitination and Xist RNA appear enriched in gene dense regions and depleted in gene poor bands, but do not correlate with L1 LINE elements which have been suggested as key to X-inactivation. These results provide evidence that ubiquitination along with Xist RNA plays an important role in the formation of facultative heterochromatin during X-inactivation.


Assuntos
Cromossomos Humanos X/metabolismo , Mecanismo Genético de Compensação de Dose , Histonas/metabolismo , RNA não Traduzido/metabolismo , Ubiquitinas/metabolismo , Cromossomo X/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Linhagem Celular , Cromossomos Humanos X/imunologia , Regulação para Baixo , Expressão Gênica , Histonas/análise , Histonas/imunologia , Humanos , Camundongos , Mitose/fisiologia , RNA Longo não Codificante , RNA não Traduzido/análise , Cromatina Sexual/química , Cromatina Sexual/imunologia , Cromatina Sexual/metabolismo , Ubiquitinas/análise , Ubiquitinas/imunologia , Cromossomo X/química , Cromossomo X/imunologia
20.
J Cell Biochem ; 93(6): 1282-96, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15503302

RESUMO

Definitive localization of c-Myc within the nucleus is important to fully understand the regulation and function of this oncoprotein. Studies of c-Myc distribution, however, have produced conflicting results. To overcome technical challenges inherent in c-Myc cytology, we use here three methods to visualize c-Myc and in addition examine the impact of proteasome inhibition. EYFP or HA-tagged Myc was reintroduced by stable transfection into myc null diploid rat fibroblasts, replacing endogenous Myc with tagged Myc expressed at or near normal levels. This tagged Myc is shown to functionally replace the endogenous Myc by restoration of normal cell morphology and growth rate. We were able to confirm key findings using antibodies to the endogenous c-Myc and/or its partner, Max. Contrary to some published reports, by all three methods the c-Myc protein in rat fibroblasts distributes predominantly throughout the nucleus in a dispersed granular pattern, avoiding the nucleolus. Importantly, however, several findings provide evidence for an unanticipated relationship between c-Myc and PML nuclear bodies, which is enhanced under conditions of proteasome inhibition. Evidence of Max concentration within PML bodies is shown both with and without proteasome inhibition, strengthening the relationship between PML bodies and Myc/Max. Some accumulation of Myc and Max in nucleoli upon proteasome inhibition is also observed, although co-localization of ubiquitin was only seen with PML bodies. This work provides a comprehensive study of c-Myc distribution and also presents the first evidence of a relationship between turnover of this oncoprotein and PML nuclear bodies, known to break down in certain cancers.


Assuntos
Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Corpos de Inclusão Intranuclear , Proteínas Proto-Oncogênicas c-myc/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fatores de Transcrição de Zíper de Leucina Básica , Nucléolo Celular/ultraestrutura , Núcleo Celular/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Diploide , Fibroblastos/metabolismo , Células HeLa , Heterozigoto , Homozigoto , Humanos , Leupeptinas/farmacologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares/fisiologia , Proteína da Leucemia Promielocítica , Inibidores de Proteassoma , Proteínas Proto-Oncogênicas c-myc/genética , Ratos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Transfecção , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA