Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Mutat Res Rev Mutat Res ; 790: 108440, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35970331

RESUMO

In higher eukaryotes, sophisticate regulation of genome function requires all chromosomes to be packed into a single nucleus. Micronucleus (MN), the dissociative nucleus-like structure frequently observed in aging and multiple disease settings, has critical, yet under-recognized, pathophysiological functions. Micronuclei (MNi) have recently emerged as major sources of cytosolic DNA that can activate the cGAS-STING axis in a cell-intrinsic manner. However, MNi induced from different genotoxic stressors display great heterogeneity in binding or activating cGAS and the signaling responses downstream of the MN-induced cGAS-STING axis have divergent outcomes including autoimmunity, autoinflammation, metastasis, or cell death. Thus, full characterization of molecular network underpinning the interplay of cGAS and MN is important to elucidate the pathophysiological roles of immunogenic MN and design improved drugs that selectively target cancer via boosting the MN-derived cGAS-STING axis. Here, we summarize our current understanding of the mechanisms for self-DNA discrimination by cGAS. We focus on discussing how MN immunogencity is dictated by multiple mechanisms including integrity of micronuclear envelope, state of nucleosome and DNA, competitive factors, damaged mitochondrial DNA and micronucleophagy. We also describe emerging links between immunogenic MN and human diseases including cancer, neurodegenerative diseases and COVID-19. Particularly, we explore the exciting concept of inducing immunogenic MN as a therapeutic approach in treating cancer. We propose a new theoretical framework to describe immunogenic MN as a biological sensor to modulate cellular processes in response to genotoxic stress and provide perspectives on developing novel experimental approaches to unravel the complexity of MN immunogenicity regulation and immunogenic MN pathophysiology.


Assuntos
Estruturas do Núcleo Celular , Proteínas de Membrana , Nucleotidiltransferases , Humanos , DNA/metabolismo , Imunidade Inata/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Estruturas do Núcleo Celular/genética , Estruturas do Núcleo Celular/metabolismo
2.
Cell Rep ; 37(2): 109824, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34644584

RESUMO

RAG1 and RAG2 form a tetramer nuclease to initiate V(D)J recombination in developing T and B lymphocytes. The RAG1 protein evolves from a transposon ancestor and possesses nuclease activity that requires interaction with RAG2. Here, we show that the human RAG1 aggregates in the nucleus in the absence of RAG2, exhibiting an extremely low V(D)J recombination activity. In contrast, RAG2 does not aggregate by itself, but it interacts with RAG1 to disrupt RAG1 aggregates and thereby activate robust V(D)J recombination. Moreover, RAG2 from mouse and zebrafish could not disrupt the aggregation of human RAG1 as efficiently as human RAG2 did, indicating a species-specific regulatory mechanism for RAG1 by RAG2. Therefore, we propose that RAG2 coevolves with RAG1 to release inert RAG1 from aggregates and thereby activate V(D)J recombination to generate diverse antigen receptors in lymphocytes.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fase G1 , Proteínas de Homeodomínio/metabolismo , Linfócitos/metabolismo , Proteínas Nucleares/metabolismo , Recombinação V(D)J , Linhagem Celular Tumoral , Estruturas do Núcleo Celular/genética , Proteínas de Ligação a DNA/genética , Células HEK293 , Proteínas de Homeodomínio/genética , Humanos , Proteínas Nucleares/genética , Agregados Proteicos , Especificidade da Espécie , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
Mol Biol Cell ; 32(9): 942-955, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33788585

RESUMO

The histone locus body (HLB) is an evolutionarily conserved nuclear body that regulates the transcription and processing of replication-dependent (RD) histone mRNAs, which are the only eukaryotic mRNAs lacking a poly-A tail. Many nuclear bodies contain distinct domains, but how internal organization is related to nuclear body function is not fully understood. Here, we demonstrate using structured illumination microscopy that Drosophila HLBs have a "core-shell" organization in which the internal core contains transcriptionally active RD histone genes. The N-terminus of Mxc, which contains a domain required for Mxc oligomerization, HLB assembly, and RD histone gene expression, is enriched in the HLB core. In contrast, the C-terminus of Mxc is enriched in the HLB outer shell as is FLASH, a component of the active U7 snRNP that cotranscriptionally cleaves RD histone pre-mRNA. Consistent with these results, we show biochemically that FLASH binds directly to the Mxc C-terminal region. In the rapid S-M nuclear cycles of syncytial blastoderm Drosophila embryos, the HLB disassembles at mitosis and reassembles the core-shell arrangement as histone gene transcription is activated immediately after mitosis. Thus, the core-shell organization is coupled to zygotic histone gene transcription, revealing a link between HLB internal organization and RD histone gene expression.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Histonas/metabolismo , Microscopia/métodos , Animais , Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Estruturas do Núcleo Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Mitose , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , Elementos Reguladores de Transcrição/genética , Ribonucleoproteína Nuclear Pequena U7/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Zigoto/metabolismo
4.
Semin Cell Dev Biol ; 90: 94-103, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30017905

RESUMO

Proteins and RNAs inside the cell nucleus are organized into distinct phases, also known as liquid-liquid phase separated (LLPS) droplet organelles or nuclear bodies. These regions exist within the spaces between chromatin-rich regions but their function is tightly linked to gene activity. They include major microscopically-observable structures such as the nucleolus, paraspeckle and Cajal body. The biochemical and assembly factors enriched inside these microenvironments regulate chromatin structure, transcription, and RNA processing, and other important cellular functions. Here, we describe published evidence that suggests nuclear bodies are bona fide LLPS droplet organelles and major regulators of the processes listed above. We also outline an updated "Supply or Sequester" model to describe nuclear body function, in which proteins or RNAs are supplied to surrounding genomic regions or sequestered away from their sites of activity. Finally, we describe recent evidence that suggests these microenvironments are both reflective and drivers of diverse pathophysiological states.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Núcleo Celular/química , Separação Celular , Epigênese Genética/genética , Extração Líquido-Líquido , Organelas/metabolismo , RNA/metabolismo , Núcleo Celular/metabolismo , Estruturas do Núcleo Celular/química , Estruturas do Núcleo Celular/genética , Humanos , Organelas/química , Organelas/genética , Tamanho da Partícula , RNA/genética , RNA/isolamento & purificação
5.
Chromosoma ; 127(4): 529-537, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30291421

RESUMO

Nuclear bodies are relatively immobile organelles. Here, we investigated the mechanisms underlying their movement using experimentally induced interphase prenucleolar bodies (iPNBs). Most iPNBs demonstrated constrained diffusion, exhibiting infrequent fusions with other iPNBs and nucleoli. Fusion events were actin-independent and appeared to be the consequence of stochastic collisions between iPNBs. Most iPNBs were surrounded by condensed chromatin, while fusing iPNBs were usually found in a single heterochromatin-delimited compartment ("cage"). The experimentally induced over-condensation of chromatin significantly decreased the frequency of iPNB fusion. Thus, the data obtained indicate that the mobility of nuclear bodies is restricted by heterochromatin.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Heterocromatina/metabolismo , Estruturas do Núcleo Celular/genética , Cromatina/metabolismo , Células HeLa , Humanos , Interfase , Imagem com Lapso de Tempo
6.
Proc Natl Acad Sci U S A ; 114(19): E3823-E3829, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28439026

RESUMO

Nuclear domain 10 (ND10) bodies are small (0.1-1 µM) nuclear structures containing both constant [e.g., promyelocytic leukemia protein (PML), SP100, death domain-associated protein (Daxx)] and variable proteins, depending on the function of the cells or the stress to which they are exposed. In herpes simplex virus (HSV)-infected cells, ND10 bodies assemble at the sites of DNA entering the nucleus after infection. In sequence, the ND10 bodies become viral replication compartments, and ICP0, a viral E3 ligase, degrades both PML and SP100. The amounts of PML and SP100 and the number of ND10 structures increase in cells exposed to IFN-ß. Earlier studies have shown that PML has three key functions. Thus, (i) the interaction of PML with viral components facilitates the initiation of replication compartments, (ii) viral replication is significantly less affected by IFN-ß in PML-/- cells than in parental PML+/+ cells, and (iii) viral yields are significantly lower in PML-/- cells exposed to low ratios of virus per cell compared with parental PML+/+ cells. This report focuses on the function of SP100. In contrast to PML-/- cells, SP100-/- cells retain the sensitivity of parental SP100+/+ cells to IFN-ß and support replication of the ΔICP0 virus. At low multiplicities of infection, wild-type virus yields are higher in SP100-/- cells than in parental HEp-2 cells. In addition, the number of viral replication compartments is significantly higher in SP100-/- cells than in parental SP100+/+ cells or in PML-/- cells.


Assuntos
Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Estruturas do Núcleo Celular/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Simplexvirus/fisiologia , Montagem de Vírus/fisiologia , Animais , Antígenos Nucleares/genética , Autoantígenos/genética , Estruturas do Núcleo Celular/genética , Estruturas do Núcleo Celular/virologia , Chlorocebus aethiops , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica/genética , Células Vero
7.
Biochim Biophys Acta ; 1862(9): 1513-20, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27239700

RESUMO

The human genetic disorders caused by CAG repeat expansions in the translated sequences of various genes are called polyglutamine (polyQ) diseases because of the cellular "toxicity" of the mutant proteins. The contribution of mutant transcripts to the pathogenesis of these diseases is supported by several observations obtained from cellular models of these disorders. Here, we show that the common feature of cell lines modeling polyQ diseases is the formation of nuclear CAG RNA foci. We performed qualitative and quantitative analyses of these foci in numerous cellular models endogenously and exogenously expressing mutant transcripts by fluorescence in situ hybridization (FISH). We compared the CAG RNA foci of polyQ diseases with the CUG foci of myotonic dystrophy type 1 and found substantial differences in their number and morphology. Smaller differences within the polyQ disease group were also revealed and included a positive correlation between the foci number and the CAG repeat length. We show that expanded CAA repeats, also encoding glutamine, did not trigger RNA foci formation and foci formation is independent of the presence of mutant polyglutamine protein. Using FISH combined with immunofluorescence, we demonstrated partial co-localization of CAG repeat foci with MBNL1 alternative splicing factor, which explains the mild deregulation of MBNL1-dependent genes. We also showed that foci reside within nuclear speckles in diverse cell types: fibroblasts, lymphoblasts, iPS cells and neuronal progenitors and remain dependent on integrity of these nuclear structures.


Assuntos
Estruturas do Núcleo Celular/genética , Estruturas do Núcleo Celular/metabolismo , Expansão das Repetições de Trinucleotídeos , Processamento Alternativo , Animais , Linhagem Celular , Estruturas do Núcleo Celular/patologia , Células HeLa , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/patologia , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Hibridização in Situ Fluorescente , Camundongos , Peptídeos/genética , Peptídeos/metabolismo , RNA/genética , RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica
8.
Nucleic Acids Res ; 43(17): 8435-51, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26286192

RESUMO

Telomerase, a unique ribonucleoprotein complex that contains the telomerase reverse transcriptase (TERT), the telomerase RNA component (TERC) and the TERC-binding protein dyskerin, is required for continued cell proliferation in stem cells and cancer cells. Here we identify SRSF11 as a novel TERC-binding protein that localizes to nuclear speckles, subnuclear structures that are enriched in pre-messenger RNA splicing factors. SRSF11 associates with active telomerase enzyme through an interaction with TERC and directs it to nuclear speckles specifically during S phase of the cell cycle. On the other hand, a subset of telomeres is shown to be constitutively present at nuclear speckles irrespective of cell cycle phase, suggesting that nuclear speckles could be the nuclear sites for telomerase recruitment to telomeres. SRSF11 also associates with telomeres through an interaction with TRF2, which facilitates translocation of telomerase to telomeres. Depletion of SRSF11 prevents telomerase from associating with nuclear speckles and disrupts telomerase recruitment to telomeres, thereby abrogating telomere elongation by telomerase. These findings suggest that SRSF11 acts as a nuclear speckle-targeting factor that is essential for telomerase association with telomeres through the interactions with TERC and TRF2, and provides a potential target for modulating telomerase activity in cancer.


Assuntos
Ciclo Celular , Estruturas do Núcleo Celular/enzimologia , Fatores de Processamento de Serina-Arginina/metabolismo , Telomerase/metabolismo , Telômero/enzimologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Estruturas do Núcleo Celular/genética , Células HeLa , Humanos , Domínios e Motivos de Interação entre Proteínas , RNA/metabolismo , Fatores de Processamento de Serina-Arginina/química , Telomerase/química , Homeostase do Telômero , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
9.
Methods Mol Biol ; 1206: 1-14, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25240882

RESUMO

Long noncoding RNAs are becoming increasingly appreciated as major players in gene regulation. They have been reported to play diverse roles in many biological processes. Here, we discuss their discovery, features, and known functions in cells. While not comprehensive, this chapter should serve to illustrate the power and promise of studying long noncoding RNAs.


Assuntos
Mamíferos/genética , RNA Longo não Codificante/fisiologia , Processamento Alternativo , Animais , Estruturas do Núcleo Celular/genética , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Impressão Genômica , Humanos , Neoplasias/genética , Doenças do Sistema Nervoso/genética , RNA Longo não Codificante/classificação , Inativação do Cromossomo X
10.
Biochem Biophys Res Commun ; 446(1): 119-24, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24569078

RESUMO

Nuclear speckles are subnuclear structures enriched with RNA processing factors and poly (A)(+) RNAs comprising mRNAs and poly (A)(+) non-coding RNAs (ncRNAs). Nuclear speckles are thought to be involved in post-transcriptional regulation of gene expression, such as pre-mRNA splicing. By screening 3585 culture extracts of actinomycetes with in situ hybridization using an oligo dT probe, we identified tubercidin, an analogue of adenosine, as an inhibitor of speckle formation, which induces the delocalization of poly (A)(+) RNA and dispersion of splicing factor SRSF1/SF2 from nuclear speckles in HeLa cells. Treatment with tubercidin also decreased steady-state MALAT1 long ncRNA, thought to be involved in the retention of SRSF1/SF2 in nuclear speckles. In addition, we found that tubercidin treatment promoted exon skipping in the alternative splicing of Clk1 pre-mRNA. These results suggest that nuclear speckles play a role in modulating the concentration of splicing factors in the nucleoplasm to regulate alternative pre-mRNA splicing.


Assuntos
Processamento Alternativo , Estruturas do Núcleo Celular/efeitos dos fármacos , Estruturas do Núcleo Celular/metabolismo , Precursores de RNA/metabolismo , Actinobacteria/química , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Estruturas do Núcleo Celular/genética , Avaliação Pré-Clínica de Medicamentos , Éxons , Células HeLa , Humanos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Marcação in Situ com Primers , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Precursores de RNA/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina , Tubercidina/isolamento & purificação , Tubercidina/farmacologia
11.
Curr Biol ; 23(23): R1045-7, 2013 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-24309281

RESUMO

How do nuclear components, apart from chromosomes, partition equally to daughter nuclei during mitosis? In Schizosaccharomyces japonicus, the conserved LEM-domain nuclear envelope protein Man1 ensures the formation of identical daughter nuclei by coupling nuclear pore complexes to the segregating chromosomes.


Assuntos
Divisão do Núcleo Celular/genética , Estruturas do Núcleo Celular/genética , Segregação de Cromossomos/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Schizosaccharomyces/genética , Cromossomos/genética , Mitose , Poro Nuclear/genética , Proteínas de Schizosaccharomyces pombe/genética
12.
PLoS One ; 8(8): e74885, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24015320

RESUMO

ERH is a small, highly evolutionarily conserved nuclear protein of unknown function. Its three-dimensional structure is absolutely unique and it can form a homodimer through a ß sheet surface. ERH has been shown to interact, among others, with PDIP46/SKAR and Ciz1. When coexpressed with the latter protein, ERH accumulates in replication foci in the nucleus of HeLa cells. Here, we report that when ERH is coexpressed with PDIP46/SKAR in HeLa cells, it is recruited to nuclear speckles, and identify amino acid residues critical for targeting ERH to both these subnuclear structures. ERH H3A Q9A shows a diminished recruitment to nuclear speckles but it is recruited to replication foci. ERH E37A T51A is very poorly recruited to replication foci while still accumulating in nuclear speckles. Consequently, ERH H3A Q9A E37A T51A is recruited neither to nuclear speckles nor to replication foci. The lack of interactions of these three ERH forms with PDIP46/SKAR and/or Ciz1 was further confirmed in vitro by GST pull-down assay. The residues whose substitutions interfere with the accumulation in nuclear speckles are situated on the ß sheet surface of ERH, indicating that only the monomer of ERH can interact with PDIP46/SKAR. Substitutions affecting the recruitment to replication foci map to the other side of ERH, near a long loop between the α1 and α2 helices, thus both the monomer and the dimer of ERH could interact with Ciz1. The construction of the ERH mutants not recruited to nuclear speckles or replication foci will facilitate further studies on ERH actions in these subnuclear structures.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Estruturas do Núcleo Celular/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Substituição de Aminoácidos , Proteínas de Ciclo Celular/genética , Estruturas do Núcleo Celular/genética , Células HeLa , Humanos , Mutação de Sentido Incorreto , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estrutura Secundária de Proteína , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/genética
13.
RNA Biol ; 10(3): 391-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23324608

RESUMO

The nucleolinus is a nuclear subcompartment long ago posited to play a role in cell division. In a recent study using surf clam oocytes, cytoplasmic foci containing a nucleolinar protein were shown to later recruit γ-tubulin, identifying them as centrosomal precursors. (1) We now demonstrate the presence of structural RNAs from the nucleolinus in these procentrosomes. They include the well-known but poorly understood rRNA-transcribed spacer regions. In situ hybridization revealed a specific and dynamic association of these structural RNAs with the cell division apparatus that extends through the early stages of meiosis. In addition to their bearing on the debate over the nature of centrosome- and spindle-associated RNAs, the observations also suggest that rRNA spacer regions are not simply waste products to be discarded immediately, but may be functional byproducts that play a role in formation of the cell division apparatus.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Centrossomo/fisiologia , RNA Ribossômico/genética , Spisula/genética , Tubulina (Proteína)/metabolismo , Animais , Estruturas do Núcleo Celular/genética , Citoplasma/metabolismo , DNA Espaçador Ribossômico/genética , Feminino , Meiose , Oócitos/fisiologia , RNA Ribossômico/metabolismo , Fuso Acromático/fisiologia , Spisula/metabolismo
14.
Eur J Hum Genet ; 21(3): 317-23, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22872099

RESUMO

A block of single-nucleotide polymorphisms within intron 1 of the FTO (fat mass and obesity associated) gene is associated with variation in body weight. Previous works suggest that increased expression of FTO, which encodes a 2-oxoglutarate-dependent nucleic acid demethylase, leads to increased body weight, although the underlying mechanism has remained unclear. To elucidate the function of FTO, we examined the consequences of altered FTO levels in cultured cells and murine brain. Here we show that a knockdown of FTO in HEK293 cells affects the transcripts levels of genes involved in the response to starvation, whereas overexpression of FTO affects the transcript levels of genes related to RNA processing and metabolism. Subcellular localization of FTO further strengthens the latter notion. Using immunocytochemistry and confocal laser scanning microscopy, we detected FTO in nuclear speckles and--to a lesser and varying extent--in the nucleoplasm and nucleoli of HEK293, HeLa and MCF-7 cells. Moreover, RNA modification analyses revealed that loss of Fto affects the 3-methyluridine/uridine and pseudouridine/uridine ratios in total brain RNA. We conclude that altered levels of FTO have multiple and diverse consequences on RNA modifications and the transcriptome.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Oxigenases de Função Mista/genética , Oxo-Ácido-Liases/genética , Proteínas/metabolismo , Processamento Pós-Transcricional do RNA , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Encéfalo/metabolismo , Linhagem Celular , Estruturas do Núcleo Celular/genética , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Knockout , Oxigenases de Função Mista/metabolismo , Oxo-Ácido-Liases/metabolismo , Proteínas/genética , RNA Mensageiro/metabolismo , Transcriptoma , Uridina/análogos & derivados , Uridina/metabolismo
15.
Mol Biochem Parasitol ; 173(1): 48-52, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20433875

RESUMO

Fluorescence in situ hybridization (FISH) has been used extensively in the study of nuclear organization and gene positioning in Plasmodium falciparum. While performing FISH with published protocols, we observed large variations in parasite nuclear morphology. We hypothesized that these inconsistencies might be due to the type of parasite preparation prior to FISH, which commonly involves air-drying, prompting us to develop a new fixation protocol. Here we show both qualitatively and quantitatively that compared to air-dried and briefly fixed parasites, longer fixation in suspension leads to improved conservation of nuclear structure and lower intra-population variation of nuclear shape as well as area after FISH development. While the fixation protocol per se does not cause detectable disruptions in nuclear morphology, it greatly influences the conservation of nuclear shape and size during the most stringent steps of FISH. The type of fixation used also influences the detection of telomeric clusters, and we show that the new fixation protocol permits improved conservation of the chromosome end cluster perinuclear distribution and higher colocalization indexes for two adjacent chromosome end probes, Rep20 and telomere. Overall, the results indicate that our alternative protocol dramatically improves conservation of the nuclear architecture compared to previously reported Plasmodium DNA-FISH protocols and highlights the necessity of carefully choosing the fixation protocol for FISH.


Assuntos
Estruturas do Núcleo Celular/química , Hibridização in Situ Fluorescente/métodos , Plasmodium falciparum/citologia , Fixação de Tecidos/métodos , Estruturas do Núcleo Celular/genética , Estruturas do Núcleo Celular/metabolismo , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Telômero/genética , Telômero/metabolismo
16.
Artigo em Inglês | MEDLINE | ID: mdl-21467138

RESUMO

The eukaryotic nucleus is a congested place, and macromolecular crowding is thought to have an important role in increasing the relative concentrations of nuclear proteins, thereby accelerating the rates of biochemical reactions. Crowding is also thought to provide the environment needed for formation of nuclear bodies/subcompartments, such as the Cajal body (CB) and the histone locus body (HLB), via self-organization. In this chapter, we contrast the theories of stochastic self-organization and hierarchical self-organization in their application to nuclear body assembly, using CBs and HLBs as paradigms. Genetic ablation studies in Drosophila on components of CBs and HLBs have revealed an order to the assembly of these structures that is suggestive of a hierarchical model of self-organization. These studies also show that functions attributed to the nuclear bodies are largely unaffected in their absence, reinforcing an emerging theme in the field that the purpose of these subdomains may be to enhance the efficiency and specificity of reactions.


Assuntos
Estruturas do Núcleo Celular/genética , Modelos Biológicos , Dinâmica não Linear , Animais , Evolução Biológica , Corpos Enovelados/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Histonas/metabolismo , Processamento Pós-Transcricional do RNA/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo
17.
Stud Health Technol Inform ; 147: 251-6, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19593063

RESUMO

Parameters of the genome architecture of cell nuclei like copy number changes of genes or numerical and structural aberrations of chromosomes displayed by changes of size, shape, form and geometric arrangement of the related territories and domains play an important role in tumour diagnosis and monitoring of tumour therapy. We have defined data structures for such parameters, accompanied by meta data describing cell biology and microscopy protocols, and developed algorithms to deduce geometric data from microscopic raw images of fluorescently labelled cell nuclei. The statistical evaluation of nucleus geometry and architecture data is a valuable aid for diagnostic decisions and monitoring of cancer development, as indicated by several research case studies. The algorithms and data storage devices are presently administrated by different operating systems. Unification of workflow is being achieved for a local cluster, but gridification is still subject to problems of licensing, monitoring, and administering systems, including data security.


Assuntos
Algoritmos , Estruturas do Núcleo Celular , Formação de Conceito , Bases de Dados Genéticas , Diagnóstico por Imagem , Armazenamento e Recuperação da Informação , Estruturas do Núcleo Celular/genética , Microscopia , Neoplasias/diagnóstico , Neoplasias/terapia
18.
PLoS Pathog ; 4(7): e1000100, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18617993

RESUMO

Herpesviruses are large, ubiquitous DNA viruses with complex host interactions, yet many of the proteins encoded by these viruses have not been functionally characterized. As a first step in functional characterization, we determined the subcellular localization of 234 epitope-tagged proteins from herpes simplex virus, cytomegalovirus, and Epstein-Barr virus. Twenty-four of the 93 proteins with nuclear localization formed subnuclear structures. Twelve of these localized to the nucleolus, and five at least partially localized with promyelocytic leukemia (PML) bodies, which are known to suppress viral lytic infection. In addition, two proteins disrupted Cajal bodies, and 19 of the nuclear proteins significantly decreased the number of PML bodies per cell, including six that were shown to be SUMO-modified. These results have provided the first functional insights into over 120 previously unstudied proteins and suggest that herpesviruses employ multiple strategies for manipulating nuclear bodies that control key cellular processes.


Assuntos
Citomegalovirus/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas Nucleares/metabolismo , Simplexvirus/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Virais/metabolismo , Estruturas do Núcleo Celular/genética , Estruturas do Núcleo Celular/metabolismo , Citomegalovirus/genética , Testes Genéticos , Genoma , Células Precursoras de Granulócitos/metabolismo , Herpesvirus Humano 4/genética , Humanos , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Simplexvirus/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Replicação Viral , Dedos de Zinco
19.
Science ; 318(5856): 1637-40, 2007 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-18006705

RESUMO

Cells respond to DNA double-strand breaks by recruiting factors such as the DNA-damage mediator protein MDC1, the p53-binding protein 1 (53BP1), and the breast cancer susceptibility protein BRCA1 to sites of damaged DNA. Here, we reveal that the ubiquitin ligase RNF8 mediates ubiquitin conjugation and 53BP1 and BRCA1 focal accumulation at sites of DNA lesions. Moreover, we establish that MDC1 recruits RNF8 through phosphodependent interactions between the RNF8 forkhead-associated domain and motifs in MDC1 that are phosphorylated by the DNA-damage activated protein kinase ataxia telangiectasia mutated (ATM). We also show that depletion of the E2 enzyme UBC13 impairs 53BP1 recruitment to sites of damage, which suggests that it cooperates with RNF8. Finally, we reveal that RNF8 promotes the G2/M DNA damage checkpoint and resistance to ionizing radiation. These results demonstrate how the DNA-damage response is orchestrated by ATM-dependent phosphorylation of MDC1 and RNF8-mediated ubiquitination.


Assuntos
Estruturas do Núcleo Celular/genética , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas Mutadas de Ataxia Telangiectasia , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Reparo do DNA , Proteínas de Ligação a DNA/química , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , RNA Interferente Pequeno , Transativadores/química , Transativadores/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação
20.
Cancer Res ; 67(15): 7072-7, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17652140

RESUMO

The linear chromosomes of vertebrates terminate in telomeres that consist of a tandemly repeated hexameric sequence, 5'TTAGGG3'. Telomeres form a protective loop structure (t-loop), which is thought to prevent them from being recognized as a double-strand break. Approximately 10% of human tumors prevent shortening of their telomeres by using a recombination-mediated alternative lengthening of telomeres (ALT) mechanism. ALT-positive human cells contain extrachromosomal telomere repeat (ECTR) DNA that may either be circular or linear. It has been proposed that ECTR may be generated by recombination events involving the t-loop. A proportion of the cells within ALT-positive cell populations contain promyelocytic leukemia (PML) nuclear bodies that contain telomeric DNA and telomere-binding proteins that are called ALT-associated PML bodies (APB). Although the presence of APBs is very useful for determining whether tumors and cell lines use the ALT mechanism, the function of APBs is unknown. It has previously been shown that telomeric DNA is particularly susceptible to damage by hydrogen peroxide and N-methyl-N'-nitro-N-nitrosoguanidine. We report here that these DNA-damaging agents induce both linear and circular ECTR DNA in ALT cells and increase the proportion of cells that contain APBs. We partially purified APBs and showed that the telomeric repeat DNA they contain is predominantly linear. We propose that a function of APBs is to sequester linear telomeric DNA.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Dano ao DNA/efeitos dos fármacos , Corpos de Inclusão Intranuclear/metabolismo , Telômero/metabolismo , Proteínas de Ciclo Celular/metabolismo , Estruturas do Núcleo Celular/genética , Cromossomos Humanos , Reparo do DNA , Eletroforese em Gel Bidimensional , Humanos , Peróxido de Hidrogênio/farmacologia , Corpos de Inclusão Intranuclear/genética , Corpos de Inclusão Intranuclear/ultraestrutura , Metilnitronitrosoguanidina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...