Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Res Commun ; 4(9): 2427-2443, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39028932

RESUMO

An in-depth multiomic molecular characterization of PARP inhibitors revealed a distinct poly-pharmacology of niraparib (Zejula) mediated by its interaction with lanosterol synthase (LSS), which is not observed with other PARP inhibitors. Niraparib, in a similar way to the LSS inhibitor Ro-48-8071, induced activation of the 24,25-epoxysterol shunt pathway, which is a regulatory signaling branch of the cholesterol biosynthesis pathway. Interestingly, the combination of an LSS inhibitor with a PARP inhibitor that does not bind to LSS, such as olaparib, had an additive effect on killing cancer cells to levels comparable with niraparib as a single agent. In addition, the combination of PARP inhibitors and statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, an enzyme catalyzing the rate-limiting step in the mevalonate pathway, had a synergistic effect on tumor cell killing in cell lines and patient-derived ovarian tumor organoids. These observations suggest that concomitant inhibition of the cholesterol biosynthesis pathway and PARP activity might result in stronger efficacy of these inhibitors against tumor types highly dependent on cholesterol metabolism. SIGNIFICANCE: The presented data indicate, to our knowledge, for the first time, the potential benefit of concomitant modulation of cholesterol biosynthesis pathway and PARP inhibition and highlight the need for further investigation to assess its translational relevance.


Assuntos
Colesterol , Sinergismo Farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases , Humanos , Colesterol/biossíntese , Colesterol/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Linhagem Celular Tumoral , Feminino , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia
2.
Genome Biol ; 23(1): 8, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34991671

RESUMO

While it is established that the functional impact of genetic variation can vary across cell types and states, capturing this diversity remains challenging. Current studies using bulk sequencing either ignore this heterogeneity or use sorted cell populations, reducing discovery and explanatory power. Here, we develop scDALI, a versatile computational framework that integrates information on cellular states with allelic quantifications of single-cell sequencing data to characterize cell-state-specific genetic effects. We apply scDALI to scATAC-seq profiles from developing F1 Drosophila embryos and scRNA-seq from differentiating human iPSCs, uncovering heterogeneous genetic effects in specific lineages, developmental stages, or cell types.


Assuntos
Células-Tronco Pluripotentes Induzidas , Análise de Célula Única , Regulação da Expressão Gênica
3.
Genome Res ; 31(9): 1573-1581, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34266978

RESUMO

Inter-species comparisons of both morphology and gene expression within a phylum have revealed a period in the middle of embryogenesis with more similarity between species compared with earlier and later time points. This "developmental hourglass" pattern has been observed in many phyla, yet the evolutionary constraints on gene expression, as well as the underlying mechanisms of how this is regulated, remain elusive. Moreover, the role of positive selection on gene regulation in the more diverged earlier and later stages of embryogenesis remains unknown. Here, using DNase-seq to identify regulatory regions in two distant Drosophila species (D. melanogaster and D. virilis), we assessed the evolutionary conservation and adaptive evolution of enhancers throughout multiple stages of embryogenesis. This revealed a higher proportion of conserved enhancers at the phylotypic period, providing a regulatory basis for the hourglass expression pattern. Using an in silico mutagenesis approach, we detect signatures of positive selection on developmental enhancers at early and late stages of embryogenesis, with a depletion at the phylotypic period, suggesting positive selection as one evolutionary mechanism underlying the hourglass pattern of animal evolution.


Assuntos
Drosophila melanogaster , Evolução Molecular , Animais , Drosophila/genética , Drosophila melanogaster/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Sequências Reguladoras de Ácido Nucleico
4.
Dev Cell ; 55(5): 648-664.e9, 2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33171098

RESUMO

Enhancers are essential drivers of cell states, yet the relationship between accessibility, regulatory activity, and in vivo lineage commitment during embryogenesis remains poorly understood. Here, we measure chromatin accessibility in isolated neural and mesodermal lineages across a time course of Drosophila embryogenesis. Promoters, including tissue-specific genes, are often constitutively open, even in contexts where the gene is not expressed. In contrast, the majority of distal elements have dynamic, tissue-specific accessibility. Enhancer priming appears rarely within a lineage, perhaps reflecting the speed of Drosophila embryogenesis. However, many tissue-specific enhancers are accessible in other lineages early on and become progressively closed as embryogenesis proceeds. We demonstrate the usefulness of this tissue- and time-resolved resource to definitively identify single-cell clusters, to uncover predictive motifs, and to identify many regulators of tissue development. For one such predicted neural regulator, l(3)neo38, we generate a loss-of-function mutant and uncover an essential role for neuromuscular junction and brain development.


Assuntos
Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Desenvolvimento Embrionário/genética , Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , Animais , Linhagem da Célula/genética , Cromatina , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/embriologia , Músculos/embriologia , Neurônios/citologia , Especificidade de Órgãos/genética , Ligação Proteica , Análise de Célula Única , Fatores de Tempo , Fatores de Transcrição/metabolismo
5.
Genome Biol Evol ; 11(7): 1813-1828, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31114856

RESUMO

Transcription factor (TF) binding is determined by sequence as well as chromatin accessibility. Although the role of accessibility in shaping TF-binding landscapes is well recorded, its role in evolutionary divergence of TF binding, which in turn can alter cis-regulatory activities, is not well understood. In this work, we studied the evolution of genome-wide binding landscapes of five major TFs in the core network of mesoderm specification, between Drosophila melanogaster and Drosophila virilis, and examined its relationship to accessibility and sequence-level changes. We generated chromatin accessibility data from three important stages of embryogenesis in both Drosophila melanogaster and Drosophila virilis and recorded conservation and divergence patterns. We then used multivariable models to correlate accessibility and sequence changes to TF-binding divergence. We found that accessibility changes can in some cases, for example, for the master regulator Twist and for earlier developmental stages, more accurately predict binding change than is possible using TF-binding motif changes between orthologous enhancers. Accessibility changes also explain a significant portion of the codivergence of TF pairs. We noted that accessibility and motif changes offer complementary views of the evolution of TF binding and developed a combined model that captures the evolutionary data much more accurately than either view alone. Finally, we trained machine learning models to predict enhancer activity from TF binding and used these functional models to argue that motif and accessibility-based predictors of TF-binding change can substitute for experimentally measured binding change, for the purpose of predicting evolutionary changes in enhancer activity.


Assuntos
Cromatina/metabolismo , Proteínas de Drosophila/metabolismo , Fatores de Transcrição/metabolismo , Animais , Cromatina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Evolução Molecular , Ligação Proteica , Fatores de Transcrição/genética
6.
Nature ; 555(7697): 538-542, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29539636

RESUMO

Understanding how gene regulatory networks control the progressive restriction of cell fates is a long-standing challenge. Recent advances in measuring gene expression in single cells are providing new insights into lineage commitment. However, the regulatory events underlying these changes remain unclear. Here we investigate the dynamics of chromatin regulatory landscapes during embryogenesis at single-cell resolution. Using single-cell combinatorial indexing assay for transposase accessible chromatin with sequencing (sci-ATAC-seq), we profiled chromatin accessibility in over 20,000 single nuclei from fixed Drosophila melanogaster embryos spanning three landmark embryonic stages: 2-4 h after egg laying (predominantly stage 5 blastoderm nuclei), when each embryo comprises around 6,000 multipotent cells; 6-8 h after egg laying (predominantly stage 10-11), to capture a midpoint in embryonic development when major lineages in the mesoderm and ectoderm are specified; and 10-12 h after egg laying (predominantly stage 13), when each of the embryo's more than 20,000 cells are undergoing terminal differentiation. Our results show that there is spatial heterogeneity in the accessibility of the regulatory genome before gastrulation, a feature that aligns with future cell fate, and that nuclei can be temporally ordered along developmental trajectories. During mid-embryogenesis, tissue granularity emerges such that individual cell types can be inferred by their chromatin accessibility while maintaining a signature of their germ layer of origin. Analysis of the data reveals overlapping usage of regulatory elements between cells of the endoderm and non-myogenic mesoderm, suggesting a common developmental program that is reminiscent of the mesendoderm lineage in other species. We identify 30,075 distal regulatory elements that exhibit tissue-specific accessibility. We validated the germ-layer specificity of a subset of these predicted enhancers in transgenic embryos, achieving an accuracy of 90%. Overall, our results demonstrate the power of shotgun single-cell profiling of embryos to resolve dynamic changes in the chromatin landscape during development, and to uncover the cis-regulatory programs of metazoan germ layers and cell types.


Assuntos
Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Análise de Célula Única , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Cromatina/genética , Cromatina/metabolismo , Drosophila melanogaster/genética , Endoderma/citologia , Endoderma/metabolismo , Elementos Facilitadores Genéticos/genética , Feminino , Gastrulação/genética , Genoma de Inseto/genética , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Especificidade de Órgãos/genética , Organismos Geneticamente Modificados/citologia , Organismos Geneticamente Modificados/genética , Reprodutibilidade dos Testes
7.
Methods Mol Biol ; 1094: 243-58, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24162993

RESUMO

The discovery of 5-hydroxymethylcytosine (5hmC) as an abundant base in mammalian genomes has excited the field of epigenetics, and stimulated an intense period of research activity aimed at decoding its biological significance. However, initial research efforts were hampered by a lack of assays capable of specifically detecting 5hmC. Consequently, the last 3 years have seen the development of a plethora of new techniques designed to detect both global levels and locus-specific profiles of 5hmC in mammalian genomes. This research effort has culminated in the recent publication of two complementary techniques for quantitative, base-resolution mapping of 5hmC in mammalian genomes, the first true mammalian hydroxymethylomes. Here, we review the techniques currently available to researchers studying 5hmC, discuss their advantages and disadvantages, and explore the technical hurdles which remain to be overcome.


Assuntos
Citosina/análogos & derivados , Biologia Molecular/métodos , 5-Metilcitosina/análogos & derivados , Animais , Sequência de Bases , Citosina/metabolismo , Metilação de DNA , Loci Gênicos , Mamíferos , Dados de Sequência Molecular
8.
Bioessays ; 36(2): 134-40, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24277643

RESUMO

DNA methylation is a repressive epigenetic mark vital for normal development. Recent studies have uncovered an unexpected role for the DNA methylome in ensuring the correct targeting of the Polycomb repressive complexes throughout the genome. Here, we discuss the implications of these findings for cancer, where DNA methylation patterns are widely reprogrammed. We speculate that cancer-associated reprogramming of the DNA methylome leads to an altered Polycomb binding landscape, influencing gene expression by multiple modes. As the Polycomb system is responsible for the regulation of genes with key roles in cell fate decisions and cell cycle regulation, DNA methylation induced Polycomb mis-targeting could directly drive carcinogenesis and disease progression.


Assuntos
Metilação de DNA/fisiologia , Neoplasias/genética , Proteínas do Grupo Polycomb/metabolismo , Animais , Metilação de DNA/genética , Epigênese Genética/genética , Epigênese Genética/fisiologia , Humanos , Proteínas do Grupo Polycomb/genética
9.
Biochem J ; 451(1): 13-23, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23489368

RESUMO

Methylation of the cytosine base in DNA, DNA methylation, is an essential epigenetic mark in mammals that contributes to the regulation of transcription. Several advances have been made in this area in recent years, leading to a leap forward in our understanding of how this pathway contributes to gene regulation during embryonic development, and the functional consequences of its perturbation in human disease. Critical to these advances is a comprehension of the genomic distribution of modified cytosine bases in unprecedented detail, drawing attention to genomic regions beyond gene promoters. In addition, we have a more complete understanding of the multifactorial manner by which DNA methylation influences gene regulation at the molecular level, and which genes rely directly on the DNA methylome for their normal transcriptional regulation. It is becoming apparent that a major role of DNA modification is to act as a relatively stable, and mitotically heritable, template that contributes to the establishment and maintenance of chromatin states. In this regard, interplay is emerging between DNA methylation and the PcG (Polycomb group) proteins, which act as evolutionarily conserved mediators of cell identity. In the present paper we review these aspects of DNA methylation, and discuss how a multifunctional view of DNA modification as an integral part of chromatin organization is influencing our understanding of this epigenetic mark's contribution to transcriptional regulation.


Assuntos
Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Genoma Humano/fisiologia , Animais , Cromatina/genética , Cromatina/metabolismo , Humanos , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo
10.
Genome Biol ; 14(3): R25, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-23531360

RESUMO

BACKGROUND: DNA methylation and the Polycomb repression system are epigenetic mechanisms that play important roles in maintaining transcriptional repression. Recent evidence suggests that DNA methylation can attenuate the binding of Polycomb protein components to chromatin and thus plays a role in determining their genomic targeting. However, whether this role of DNA methylation is important in the context of transcriptional regulation is unclear. RESULTS: By genome-wide mapping of the Polycomb Repressive Complex 2-signature histone mark, H3K27me3, in severely DNA hypomethylated mouse somatic cells, we show that hypomethylation leads to widespread H3K27me3 redistribution, in a manner that reflects the local DNA methylation status in wild-type cells. Unexpectedly, we observe striking loss of H3K27me3 and Polycomb Repressive Complex 2 from Polycomb target gene promoters in DNA hypomethylated cells, including Hox gene clusters. Importantly, we show that many of these genes become ectopically expressed in DNA hypomethylated cells, consistent with loss of Polycomb-mediated repression. CONCLUSIONS: An intact DNA methylome is required for appropriate Polycomb-mediated gene repression by constraining Polycomb Repressive Complex 2 targeting. These observations identify a previously unappreciated role for DNA methylation in gene regulation and therefore influence our understanding of how this epigenetic mechanism contributes to normal development and disease.


Assuntos
Metilação de DNA/genética , Histonas/metabolismo , Lisina/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Proteínas Repressoras/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/deficiência , DNA (Citosina-5-)-Metiltransferases/metabolismo , Embrião de Mamíferos/citologia , Epigênese Genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Genes Homeobox , Camundongos , Modelos Genéticos , Família Multigênica , Regiões Promotoras Genéticas
11.
Hum Mol Genet ; 22(9): 1791-806, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23364048

RESUMO

DNA methylation plays an important role in suppressing retrotransposon activity in mammalian genomes, yet there are stages of mammalian development where global hypomethylation puts the genome at risk of retrotransposition-mediated genetic instability. Hypomethylated primordial germ cells appear to limit this risk by expressing a cohort of retrotransposon-suppressing genome-defence genes whose silencing depends on promoter DNA methylation. Here, we investigate whether similar mechanisms operate in hypomethylated trophectoderm-derived components of the mammalian placenta to couple expression of genome-defence genes to the potential for retrotransposon activity. We show that the hypomethylated state of the mouse placenta results in activation of only one of the hypomethylation-sensitive germline genome-defence genes: Tex19.1. Tex19.1 appears to play an important role in placenta function as Tex19.1(-/-) mouse embryos exhibit intra-uterine growth retardation and have small placentas due to a reduction in the number of spongiotrophoblast, glycogen trophoblast and sinusoidal trophoblast giant cells. Furthermore, we show that retrotransposon mRNAs are derepressed in Tex19.1(-/-) placentas and that protein encoded by the LINE-1 retrotransposon is upregulated in hypomethylated trophectoderm-derived cells that normally express Tex19.1. This study suggests that post-transcriptional genome-defence mechanisms are operating in the placenta to protect the hypomethylated cells in this tissue from retrotransposons and suggests that imbalances between retrotransposon activity and genome-defence mechanisms could contribute to placenta dysfunction and disease.


Assuntos
Retardo do Crescimento Fetal/genética , Elementos Nucleotídeos Longos e Dispersos , Proteínas Nucleares/genética , Placenta/metabolismo , Animais , Metilação de DNA , Feminino , Retardo do Crescimento Fetal/patologia , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Placenta/citologia , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Análise de Sequência de DNA , Trofoblastos/metabolismo , Regulação para Cima
12.
Development ; 139(19): 3623-32, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22949617

RESUMO

Mouse primordial germ cells (PGCs) erase global DNA methylation (5mC) as part of the comprehensive epigenetic reprogramming that occurs during PGC development. 5mC plays an important role in maintaining stable gene silencing and repression of transposable elements (TE) but it is not clear how the extensive loss of DNA methylation impacts on gene expression and TE repression in developing PGCs. Using a novel epigenetic disruption and recovery screen and genetic analyses, we identified a core set of germline-specific genes that are dependent exclusively on promoter DNA methylation for initiation and maintenance of developmental silencing. These gene promoters appear to possess a specialised chromatin environment that does not acquire any of the repressive H3K27me3, H3K9me2, H3K9me3 or H4K20me3 histone modifications when silenced by DNA methylation. Intriguingly, this methylation-dependent subset is highly enriched in genes with roles in suppressing TE activity in germ cells. We show that the mechanism for developmental regulation of the germline genome-defence genes involves DNMT3B-dependent de novo DNA methylation. These genes are then activated by lineage-specific promoter demethylation during distinct global epigenetic reprogramming events in migratory (~E8.5) and post-migratory (E10.5-11.5) PGCs. We propose that genes involved in genome defence are developmentally regulated primarily by promoter DNA methylation as a sensory mechanism that is coupled to the potential for TE activation during global 5mC erasure, thereby acting as a failsafe to ensure TE suppression and maintain genomic integrity in the germline.


Assuntos
Metilação de DNA/fisiologia , Desenvolvimento Embrionário/genética , Epigênese Genética , Genoma , Células Germinativas/metabolismo , Regiões Promotoras Genéticas , Animais , Células Cultivadas , Montagem e Desmontagem da Cromatina/genética , Citoproteção/genética , Dano ao DNA/genética , Embrião de Mamíferos , Epigênese Genética/fisiologia , Genoma/genética , Células Germinativas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Regiões Promotoras Genéticas/fisiologia
13.
Biochem J ; 441(3): 803-12, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22004688

RESUMO

PABP1 [poly(A)-binding protein 1] is a central regulator of mRNA translation and stability and is required for miRNA (microRNA)-mediated regulation and nonsense-mediated decay. Numerous protein, as well as RNA, interactions underlie its multi-functional nature; however, it is unclear how its different activities are co-ordinated, since many partners interact via overlapping binding sites. In the present study, we show that human PABP1 is subject to elaborate post-translational modification, identifying 14 modifications located throughout the functional domains, all but one of which are conserved in mouse. Intriguingly, PABP1 contains glutamate and aspartate methylations, modifications of unknown function in eukaryotes, as well as lysine and arginine methylations, and lysine acetylations. The latter dramatically alter the pI of PABP1, an effect also observed during the cell cycle, suggesting that different biological processes/stimuli can regulate its modification status, although PABP1 also probably exists in differentially modified subpopulations within cells. Two lysine residues were differentially acetylated or methylated, revealing that PABP1 may be the first example of a cytoplasmic protein utilizing a 'methylation/acetylation switch'. Modelling using available structures implicates these modifications in regulating interactions with individual PAM2 (PABP-interacting motif 2)-containing proteins, suggesting a direct link between PABP1 modification status and the formation of distinct mRNP (messenger ribonucleoprotein) complexes that regulate mRNA fate in the cytoplasm.


Assuntos
Proteína I de Ligação a Poli(A)/química , Proteína I de Ligação a Poli(A)/metabolismo , Proteína I de Ligação a Poli(A)/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Arginina/metabolismo , Células Cultivadas , Células HeLa , Humanos , Cinética , Metilação , Camundongos , Modelos Moleculares , Proteína I de Ligação a Poli(A)/genética , Proteínas Metiltransferases/metabolismo , Proteínas Metiltransferases/fisiologia , Processamento de Proteína Pós-Traducional/genética , Relação Estrutura-Atividade , Distribuição Tecidual
14.
Genome Res ; 22(3): 467-77, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22106369

RESUMO

The discovery of substantial amounts of 5-hydroxymethylcytosine (5hmC), formed by the oxidation of 5-methylcytosine (5mC), in various mouse tissues and human embryonic stem (ES) cells has necessitated a reevaluation of our knowledge of 5mC/5hmC patterns and functions in mammalian cells. Here, we investigate the tissue specificity of both the global levels and locus-specific distribution of 5hmC in several human tissues and cell lines. We find that global 5hmC content of normal human tissues is highly variable, does not correlate with global 5mC content, and decreases rapidly as cells from normal tissue adapt to cell culture. Using tiling microarrays to map 5hmC levels in DNA from normal human tissues, we find that 5hmC patterns are tissue specific; unsupervised hierarchical clustering based solely on 5hmC patterns groups independent biological samples by tissue type. Moreover, in agreement with previous studies, we find 5hmC associated primarily, but not exclusively, with the body of transcribed genes, and that within these genes 5hmC levels are positively correlated with transcription levels. However, using quantitative 5hmC-qPCR, we find that the absolute levels of 5hmC for any given gene are primarily determined by tissue type, gene expression having a secondary influence on 5hmC levels. That is, a gene transcribed at a similar level in several different tissues may have vastly different levels of 5hmC (>20-fold) dependent on tissue type. Our findings highlight tissue type as a major modifier of 5hmC levels in expressed genes and emphasize the importance of using quantitative analyses in the study of 5hmC levels.


Assuntos
Citosina/análogos & derivados , DNA/química , Regulação da Expressão Gênica , Transcrição Gênica , 5-Metilcitosina/análogos & derivados , Animais , Linhagem Celular , Células Cultivadas , Mapeamento Cromossômico , Análise por Conglomerados , Citosina/análise , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Perfilação da Expressão Gênica , Loci Gênicos , Humanos , Camundongos , Proteínas Nucleares/genética , Especificidade de Órgãos/genética , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA
16.
Cancers (Basel) ; 3(2): 1798-820, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24212783

RESUMO

Epigenetic mechanisms assist in maintaining gene expression patterns and cellular properties in developing and adult tissues. The molecular pathology of disease states frequently includes perturbation of DNA and histone methylation patterns, which can activate apoptotic pathways associated with maintenance of genome integrity. This perspective focuses on the pathways linking DNA methyltransferases and methyl-CpG binding proteins to apoptosis, and includes new bioinformatic analyses to characterize the evolutionary origin of two G/T mismatch-specific thymine DNA glycosylases, MBD4 and TDG.

17.
Development ; 136(5): 723-7, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19158184

RESUMO

We demonstrate that a direct interaction between the methyl-CpG-dependent transcription repressor Kaiso and xTcf3, a transducer of the Wnt signalling pathway, results in their mutual disengagement from their respective DNA-binding sites. Thus, the transcription functions of xTcf3 can be inhibited by overexpression of Kaiso in cell lines and Xenopus embryos. The interaction of Kaiso with xTcf3 is highly conserved and is dependent on its zinc-finger domains (ZF1-3) and the corresponding HMG DNA-binding domain of TCF3/4 factors. Our data rule out a model suggesting that xKaiso is a direct repressor of Wnt signalling target genes in early Xenopus development via binding to promoter-proximal CTGCNA sequences as part of a xTcf3 repressor complex. Instead, we propose that mutual inhibition by Kaiso/TCF3 of their DNA-binding functions may be important in developmental or cancer contexts and acts as a regulatory node that integrates epigenetic and Wnt signalling pathways.


Assuntos
Proteínas Repressoras/metabolismo , Fatores de Transcrição TCF/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Sítios de Ligação/genética , DNA/genética , DNA/metabolismo , Epigênese Genética , Camundongos , Modelos Biológicos , Modelos Genéticos , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Transdução de Sinais , Fatores de Transcrição TCF/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição , Xenopus/embriologia , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/genética
18.
Development ; 136(5): 729-38, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19158185

RESUMO

Mammalian forms of the transcription repressor, Kaiso, can reportedly bind methylated DNA and non-methylated CTGCNA motifs. Here we compare the DNA-binding properties of Kaiso from frog, fish and chicken and demonstrate that only the methyl-CpG-binding function of Kaiso is evolutionarily conserved. We present several independent experimental lines of evidence that the phenotypic abnormalities associated with xKaiso-depleted Xenopus laevis embryos are independent of the putative CTGCNA-dependent DNA-binding function of xKaiso. Our analysis suggests that xKaiso does not play a role in the regulation of either xWnt11 or Siamois, key signalling molecules in the Wnt pathway during X. laevis gastrulation. The major phenotypic defects associated with xKaiso depletion are premature transcription activation before the mid-blastula transition and concomitant activation of a p53-dependent cell-death pathway.


Assuntos
DNA/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/metabolismo , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Apoptose , Sequência de Bases , Sítios de Ligação/genética , Galinhas , Sequência Conservada , Ilhas de CpG , DNA/genética , Metilação de DNA , Gastrulação/genética , Gastrulação/fisiologia , Proteínas de Homeodomínio/metabolismo , Humanos , Fenótipo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais , Especificidade da Espécie , Takifugu , Fatores de Transcrição/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Xenopus/deficiência , Proteínas de Xenopus/genética , Xenopus laevis/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA