Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 147(6): 1283-94, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22153073

RESUMEN

Key regulatory genes, suppressed by Polycomb and H3K27me3, become active during normal differentiation and induced reprogramming. Using the well-characterized enhancer/promoter pair of MYOD1 as a model, we have identified a critical role for enhancers in reprogramming. We observed an unexpected nucleosome-depleted region (NDR) at the H3K4me1-enriched enhancer at which transcriptional regulators initially bind, leading to subsequent changes in the chromatin at the cognate promoter. Exogenous Myod1 activates its own transcription by binding first at the enhancer, leading to an NDR and transcription-permissive chromatin at the associated MYOD1 promoter. Exogenous OCT4 also binds first to the permissive MYOD1 enhancer but has a different effect on the cognate promoter, where the monovalent H3K27me3 marks are converted to the bivalent state characteristic of stem cells. Genome-wide, a high percentage of Polycomb targets are associated with putative enhancers in permissive states, suggesting that they may provide a widespread avenue for the initiation of cell-fate reprogramming.


Asunto(s)
Elementos de Facilitación Genéticos , Proteínas Represoras/metabolismo , Animales , Línea Celular , Epigenómica , Fibroblastos/metabolismo , Humanos , Ratones , Proteína MioD/genética , Nucleosomas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas
2.
Genome Res ; 25(4): 467-77, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25747664

RESUMEN

The holistic role of DNA methylation in the organization of the cancer epigenome is not well understood. Here we perform a comprehensive, high-resolution analysis of chromatin structure to compare the landscapes of HCT116 colon cancer cells and a DNA methylation-deficient derivative. The NOMe-seq accessibility assay unexpectedly revealed symmetrical and transcription-independent nucleosomal phasing across active, poised, and inactive genomic elements. DNA methylation abolished this phasing primarily at enhancers and CpG island (CGI) promoters, with little effect on insulators and non-CGI promoters. Abolishment of DNA methylation led to the context-specific reestablishment of the poised and active states of normal colon cells, which were marked in methylation-deficient cells by distinct H3K27 modifications and the presence of either well-phased nucleosomes or nucleosome-depleted regions, respectively. At higher-order genomic scales, we found that long, H3K9me3-marked domains had lower accessibility, consistent with a more compact chromatin structure. Taken together, our results demonstrate the nuanced and context-dependent role of DNA methylation in the functional, multiscale organization of cancer epigenomes.


Asunto(s)
Cromatina/genética , Neoplasias del Colon/genética , Metilación de ADN/genética , Línea Celular Tumoral , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , ADN (Citosina-5-)-Metiltransferasas/genética , Epigénesis Genética , Células HCT116 , Histonas/genética , Humanos , Nucleosomas/genética , Regiones Promotoras Genéticas/genética , ADN Metiltransferasa 3B
3.
Proc Biol Sci ; 285(1878)2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29720411

RESUMEN

Ethologists discovered over 100 years ago that some lifelong behavioural patterns were acquired exclusively during restricted developmental phases called critical periods (CPs). Developmental song learning in zebra finches is one of the most striking examples of a CP for complex learned behaviour. After post-hatch day 65, whether or not a juvenile male can memorize the song of a 'tutor' depends on his experiences in the month prior. If he experienced a tutor, he can no longer learn, but if he has been isolated from hearing a tutor the learning period is extended. We aimed to identify how tutor experience alters the brain and controls the ability to learn. Epigenetic landscapes are modulated by experience and are able to regulate the transcription of sets of genes, thereby affecting cellular function. Thus, we hypothesized that tutor experiences determine the epigenetic landscape in the auditory forebrain, a region required for tutor song memorization. Using ChIPseq, RNAseq and molecular biology, we provide evidence that naturalistic experiences associated with the ability to learn can induce epigenetic changes, and propose transcriptional plasticity as a mediator of CP learning potential.


Asunto(s)
Epigénesis Genética/fisiología , Aprendizaje , Pájaros Cantores/fisiología , Transcripción Genética , Vocalización Animal , Animales , Pinzones/genética , Pinzones/fisiología , Regulación del Desarrollo de la Expresión Génica , Masculino , Música , Pájaros Cantores/genética
4.
Mol Cell ; 39(6): 901-11, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20864037

RESUMEN

Profound chromatin changes occur during mitosis to allow for gene silencing and chromosome segregation followed by reactivation of memorized transcription states in daughter cells. Using genome-wide sequencing, we found H2A.Z-containing +1 nucleosomes of active genes shift upstream to occupy TSSs during mitosis, significantly reducing nucleosome-depleted regions. Single-molecule analysis confirmed nucleosome shifting and demonstrated that mitotic shifting is specific to active genes that are silenced during mitosis and, thus, is not seen on promoters, which are silenced by methylation or mitotically expressed genes. Using the GRP78 promoter as a model, we found H3K4 trimethylation is also maintained while other indicators of active chromatin are lost and expression is decreased. These key changes provide a potential mechanism for rapid silencing and reactivation of genes during the cell cycle.


Asunto(s)
Silenciador del Gen , Histonas/metabolismo , Mitosis/genética , Nucleosomas/metabolismo , Acetilación , Factor de Unión a CCAAT/metabolismo , Proteínas de Ciclo Celular/genética , División Celular/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Metilación de ADN/fisiología , ADN Polimerasa II/metabolismo , Chaperón BiP del Retículo Endoplásmico , Fase G1/genética , Expresión Génica/genética , Genes p16/fisiología , Proteínas de Choque Térmico/genética , Humanos , Proteínas de la Membrana/genética , Metilación , Modelos Genéticos , Fosforilación/fisiología , Regiones Promotoras Genéticas/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Fase de Descanso del Ciclo Celular/genética , Análisis de Secuencia de ADN , Proteína de Unión a TATA-Box/metabolismo , Sitio de Iniciación de la Transcripción/fisiología , Quinasa Tipo Polo 1
5.
Genome Res ; 24(9): 1421-32, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24916973

RESUMEN

It is well established that cancer-associated epigenetic repression occurs concomitant with CpG island hypermethylation and loss of nucleosomes at promoters, but the role of nucleosome occupancy and epigenetic reprogramming at distal regulatory elements in cancer is still poorly understood. Here, we evaluate the scope of global epigenetic alterations at enhancers and insulator elements in prostate and breast cancer cells using simultaneous genome-wide mapping of DNA methylation and nucleosome occupancy (NOMe-seq). We find that the genomic location of nucleosome-depleted regions (NDRs) is mostly cell type specific and preferentially found at enhancers in normal cells. In cancer cells, however, we observe a global reconfiguration of NDRs at distal regulatory elements coupled with a substantial reorganization of the cancer methylome. Aberrant acquisition of nucleosomes at enhancer-associated NDRs is associated with hypermethylation and epigenetic silencing marks, and conversely, loss of nucleosomes with demethylation and epigenetic activation. Remarkably, we show that nucleosomes remain strongly organized and phased at many facultative distal regulatory elements, even in the absence of a NDR as an anchor. Finally, we find that key transcription factor (TF) binding sites also show extensive peripheral nucleosome phasing, suggesting the potential for TFs to organize NDRs genome-wide and contribute to deregulation of cancer epigenomes. Together, our findings suggest that "decommissioning" of NDRs and TFs at distal regulatory elements in cancer cells is accompanied by DNA hypermethylation susceptibility of enhancers and insulator elements, which in turn may contribute to an altered genome-wide architecture and epigenetic deregulation in malignancy.


Asunto(s)
Metilación de ADN , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Elementos Aisladores , Nucleosomas/genética , Epigénesis Genética , Humanos , Células MCF-7 , Nucleosomas/metabolismo
6.
Genome Res ; 24(3): 454-66, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24299735

RESUMEN

Epigenetic information is available from contemporary organisms, but is difficult to track back in evolutionary time. Here, we show that genome-wide epigenetic information can be gathered directly from next-generation sequence reads of DNA isolated from ancient remains. Using the genome sequence data generated from hair shafts of a 4000-yr-old Paleo-Eskimo belonging to the Saqqaq culture, we generate the first ancient nucleosome map coupled with a genome-wide survey of cytosine methylation levels. The validity of both nucleosome map and methylation levels were confirmed by the recovery of the expected signals at promoter regions, exon/intron boundaries, and CTCF sites. The top-scoring nucleosome calls revealed distinct DNA positioning biases, attesting to nucleotide-level accuracy. The ancient methylation levels exhibited high conservation over time, clustering closely with modern hair tissues. Using ancient methylation information, we estimated the age at death of the Saqqaq individual and illustrate how epigenetic information can be used to infer ancient gene expression. Similar epigenetic signatures were found in other fossil material, such as 110,000- to 130,000-yr-old bones, supporting the contention that ancient epigenomic information can be reconstructed from a deep past. Our findings lay the foundation for extracting epigenomic information from ancient samples, allowing shifts in epialleles to be tracked through evolutionary time, as well as providing an original window into modern epigenomics.


Asunto(s)
Citosina/metabolismo , Metilación de ADN , Genoma Humano , Inuk/genética , Nucleosomas/genética , Animales , Mapeo Cromosómico , Epigénesis Genética , Epigenómica , Evolución Molecular , Expresión Génica , Regulación de la Expresión Génica , Humanos , Filogenia , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN
7.
Genome Res ; 22(12): 2497-506, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22960375

RESUMEN

DNA methylation and nucleosome positioning work together to generate chromatin structures that regulate gene expression. Nucleosomes are typically mapped using nuclease digestion requiring significant amounts of material and varying enzyme concentrations. We have developed a method (NOMe-seq) that uses a GpC methyltransferase (M.CviPI) and next generation sequencing to generate a high resolution footprint of nucleosome positioning genome-wide using less than 1 million cells while retaining endogenous DNA methylation information from the same DNA strand. Using a novel bioinformatics pipeline, we show a striking anti-correlation between nucleosome occupancy and DNA methylation at CTCF regions that is not present at promoters. We further show that the extent of nucleosome depletion at promoters is directly correlated to expression level and can accommodate multiple nucleosomes and provide genome-wide evidence that expressed non-CpG island promoters are nucleosome-depleted. Importantly, NOMe-seq obtains DNA methylation and nucleosome positioning information from the same DNA molecule, giving the first genome-wide DNA methylation and nucleosome positioning correlation at the single molecule, and thus, single cell level, that can be used to monitor disease progression and response to therapy.


Asunto(s)
Mapeo Cromosómico , Metilación de ADN , ADN/genética , Epigenómica/métodos , Nucleosomas/genética , Alelos , Línea Celular , Ensamble y Desensamble de Cromatina , Islas de CpG , Huella de ADN , Eliminación de Gen , Regulación de la Expresión Génica , Sitios Genéticos , Humanos , Metiltransferasas , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Alineación de Secuencia , Análisis de Secuencia de ADN
8.
Proc Natl Acad Sci U S A ; 108(35): 14497-502, 2011 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-21844352

RESUMEN

Recent epigenome-wide mapping studies describe nucleosome-depleted regions (NDRs) at transcription start sites and enhancers. However, these static maps do not address causality or the roles of NDRs in gene control, and their relationship to transcription factors and DNA methylation is not well understood. Using a high-resolution single-molecule mapping approach to simultaneously investigate endogenous DNA methylation and nucleosome occupancies on individual DNA molecules, we show that the unmethylated OCT4 distal enhancer has an NDR, whereas NANOG has a clear NDR at its proximal promoter. These NDRs are maintained by binding of OCT4 and are required for OCT4 and NANOG expression. Differentiation causes a rapid loss of both NDRs accompanied by nucleosome occupancy, which precedes de novo DNA methylation. NDRs can be restored by forced expression of OCT4 in somatic cells but only when there is no cytosine methylation. These data show the central role of the NDRs, established by OCT4, in ensuring the autoregulatory loop of pluripotency and, furthermore, that de novo methylation follows the loss of NDRs and stabilizes the suppressed state.


Asunto(s)
Epigénesis Genética , Nucleosomas/fisiología , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Diferenciación Celular , Células Cultivadas , Islas de CpG , Metilación de ADN , Regulación de la Expresión Génica , Proteínas de Homeodominio/fisiología , Humanos , Proteína Homeótica Nanog
9.
J Thromb Haemost ; 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38866247

RESUMEN

BACKGROUND: Neutrophils, the most abundant white blood cells in humans, play pivotal roles in innate immunity, rapidly migrating to sites of infection and inflammation to phagocytose, neutralize, and eliminate invading pathogens. Neutrophil extracellular trap (NET) formation is increasingly recognized as an essential rapid innate immune response, but when dysregulated, it contributes to pathogenesis of sepsis and immunothrombotic disease. OBJECTIVES: Current NETosis models are limited, routinely employing nonphysiological triggers that can bypass natural NET regulatory pathways. Models utilizing isolated neutrophils and immortalized cell lines do not reflect the complex biology underlying neutrophil activation and NETosis that occurs in whole blood. To our knowledge, we report the first human ex vivo model utilizing naturally occurring molecules to induce NETosis in whole blood. This approach could be used for drug screening and, importantly, inadvertent activators of NETosis. METHODS: Here we describe a novel, high-throughput ex vivo whole blood-induced NETosis model using combinatorial pooling of native NETosis-inducing factors in a more biologically relevant Synthetic-Sepsis model. RESULTS: We found different combinations of factors evoked distinct neutrophil responses in the rate of NET generation and/or magnitude of NETosis. Despite interdonor variability, similar sets of proinflammatory molecules induced consistent responses across donors. We found that at least 3 biological triggers were necessary to induce NETosis in our system including either tumor necrosis factor-α or lymphotoxin-α. CONCLUSION: These findings emphasize the importance of investigating neutrophil physiology in a biologically relevant context to enable a better understanding of disease pathology, risk factors, and therapeutic targets, potentially providing novel strategies for disease intervention and treatment.

10.
PLoS One ; 18(5): e0281796, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37163491

RESUMEN

BACKGROUND: Hematopoietic malignancies are extremely common in pet dogs and represent nearly 30% of the malignancies diagnosed in this population each year. Clinicians commonly use existing tools such as physical exam findings, radiographs, ultrasound and baseline blood work to monitor these patients for treatment response and remission. Circulating biomarkers, such as prostate specific antigen or carcinoembryonic antigen, can be useful tools for monitoring treatment response and remission status in human cancer patients. To date, there has a been a lack of useful circulating biomarkers available to veterinary oncology patients. METHODS: Circulating plasma nucleosome concentrations were evaluated at diagnosis, throughout treatment and during remission monitoring for 40 dogs with lymphoma, acute myelogenous leukemia and multiple myeloma. Additionally, C-reactive protein and thymidine kinase-1 levels were recorded. RESULTS: Plasma nucleosome concentrations were significantly higher at diagnosis and progressive disease than they were when dogs were in remission. All but two dogs had plasma nucleosome concentrations that returned to the low range during treatment. These two dogs had the shortest progression free and overall survival times. Dogs with the highest plasma nucleosome concentrations had a significantly shorter first progression free survival than dogs with lower plasma nucleosome concentrations at diagnosis. Plasma nucleosome concentrations correlated better with disease response and progression than either thymidine kinase or C reactive protein. CONCLUSIONS: Plasma nucleosome concentrations can be a useful tool for treatment monitoring and disease progression in dogs with hematopoietic malignancies.


Asunto(s)
Enfermedades de los Perros , Neoplasias Hematológicas , Neoplasias , Masculino , Humanos , Perros , Animales , Nucleosomas , Timidina Quinasa , Biomarcadores , Neoplasias Hematológicas/veterinaria , Proteína C-Reactiva , Enfermedades de los Perros/diagnóstico
11.
Carcinogenesis ; 31(1): 27-36, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19752007

RESUMEN

Epigenetic mechanisms are essential for normal development and maintenance of tissue-specific gene expression patterns in mammals. Disruption of epigenetic processes can lead to altered gene function and malignant cellular transformation. Global changes in the epigenetic landscape are a hallmark of cancer. The initiation and progression of cancer, traditionally seen as a genetic disease, is now realized to involve epigenetic abnormalities along with genetic alterations. Recent advancements in the rapidly evolving field of cancer epigenetics have shown extensive reprogramming of every component of the epigenetic machinery in cancer including DNA methylation, histone modifications, nucleosome positioning and non-coding RNAs, specifically microRNA expression. The reversible nature of epigenetic aberrations has led to the emergence of the promising field of epigenetic therapy, which is already making progress with the recent FDA approval of three epigenetic drugs for cancer treatment. In this review, we discuss the current understanding of alterations in the epigenetic landscape that occur in cancer compared with normal cells, the roles of these changes in cancer initiation and progression, including the cancer stem cell model, and the potential use of this knowledge in designing more effective treatment strategies.


Asunto(s)
Epigénesis Genética , Neoplasias/genética , Humanos , MicroARNs/genética , Neoplasias/patología , Células Madre Neoplásicas
12.
J Cell Biol ; 170(3): 413-27, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16061694

RESUMEN

Maternal embryonic leucine zipper kinase (MELK) was previously identified in a screen for genes enriched in neural progenitors. Here, we demonstrate expression of MELK by progenitors in developing and adult brain and that MELK serves as a marker for self-renewing multipotent neural progenitors (MNPs) in cultures derived from the developing forebrain and in transgenic mice. Overexpression of MELK enhances (whereas knockdown diminishes) the ability to generate neurospheres from MNPs, indicating a function in self-renewal. MELK down-regulation disrupts the production of neurogenic MNP from glial fibrillary acidic protein (GFAP)-positive progenitors in vitro. MELK expression in MNP is cell cycle regulated and inhibition of MELK expression down-regulates the expression of B-myb, which is shown to also mediate MNP proliferation. These findings indicate that MELK is necessary for proliferation of embryonic and postnatal MNP and suggest that it regulates the transition from GFAP-expressing progenitors to rapid amplifying progenitors in the postnatal brain.


Asunto(s)
Proliferación Celular , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Multipotentes/fisiología , Neuronas/fisiología , Proteínas Serina-Treonina Quinasas/biosíntesis , Animales , Astrocitos/metabolismo , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Proteína Ácida Fibrilar de la Glía/biosíntesis , Ratones , Ratones Transgénicos , Células Madre Multipotentes/metabolismo , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/biosíntesis , Transactivadores/metabolismo
13.
Chem Biol ; 14(9): 1019-30, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17884634

RESUMEN

High-throughput identification of small molecules that selectively modulate molecular, cellular, or systems-level properties of the mammalian brain is a significant challenge. Here we report the chemical genetic identification of the orphan ligand phosphoserine (P-Ser) as an enhancer of neurogenesis. P-Ser inhibits neural stem cell/progenitor proliferation and self-renewal, enhances neurogenic fate commitment, and improves neuronal survival. We further demonstrate that the effects of P-Ser are mediated by the group III metabotropic glutamate receptor 4 (mGluR4). siRNA-mediated knockdown of mGluR4 abolished the effects of P-Ser and increased neurosphere proliferation, at least in part through upregulation of mTOR pathway activity. We also found that P-Ser increases neurogenesis in human embryonic stem cell-derived neural progenitors. This work highlights the tremendous potential of developing effective small-molecule drugs for use in regenerative medicine or transplantation therapy.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Neuronas/citología , Fosfoserina/farmacología , Receptores de Glutamato Metabotrópico/fisiología , Células Madre/citología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Ligandos , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Fosfoserina/metabolismo , Proteínas Quinasas/metabolismo , Células Madre/efectos de los fármacos , Serina-Treonina Quinasas TOR
14.
Methods Mol Biol ; 1708: 267-284, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29224149

RESUMEN

Various methodologies are available to interrogate specific components of epigenetic mechanisms such as DNA methylation or nucleosome occupancy at both the locus-specific and the genome-wide level. It has become increasingly clear, however, that comprehension of the functional interactions between epigenetic mechanisms is critical for understanding how cellular transcription programs are regulated or deregulated during normal and disease development. The Nucleosome Occupancy and Methylome sequencing (NOMe-seq) assay allows us to directly measure the relationship between DNA methylation and nucleosome occupancy by taking advantage of the methyltransferase M.CviPI, which methylates unprotected GpC dinucleotides to create a footprint of chromatin accessibility. This assay generates dual nucleosome occupancy and DNA methylation information at a single-DNA molecule resolution using as little as 200,000 cells and in as short as 15 min reaction time. DNA methylation levels and nucleosome occupancy status of genomic regions of interest can be subsequently interrogated by cloning PCR-amplified bisulfite DNA and sequencing individual clones. Alternatively, NOMe-seq can be combined with next-generation sequencing in order to generate an integrated global map of DNA methylation and nucleosome occupancy, which allows for comprehensive examination as to how these epigenetic components correlate with each other.


Asunto(s)
Metilación de ADN , Nucleosomas/metabolismo , Análisis de Secuencia de ADN/métodos , Islas de CpG , Epigénesis Genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Metiltransferasas/metabolismo , Regiones Promotoras Genéticas
15.
PLoS One ; 11(11): e0166438, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27875550

RESUMEN

Chromatin immunoprecipitation and DNA sequencing (ChIP-seq) has been instrumental in inferring the roles of histone post-translational modifications in the regulation of transcription, chromatin compaction and other cellular processes that require modulation of chromatin structure. However, analysis of ChIP-seq data is challenging when the manipulation of a chromatin-modifying enzyme significantly affects global levels of histone post-translational modifications. For example, small molecule inhibition of the methyltransferase EZH2 reduces global levels of histone H3 lysine 27 trimethylation (H3K27me3). However, standard ChIP-seq normalization and analysis methods fail to detect a decrease upon EZH2 inhibitor treatment. We overcome this challenge by employing an alternative normalization approach that is based on the addition of Drosophila melanogaster chromatin and a D. melanogaster-specific antibody into standard ChIP reactions. Specifically, the use of an antibody that exclusively recognizes the D. melanogaster histone variant H2Av enables precipitation of D. melanogaster chromatin as a minor fraction of the total ChIP DNA. The D. melanogaster ChIP-seq tags are used to normalize the human ChIP-seq data from DMSO and EZH2 inhibitor-treated samples. Employing this strategy, a substantial reduction in H3K27me3 signal is now observed in ChIP-seq data from EZH2 inhibitor treated samples.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Histonas/metabolismo , Animales , Inmunoprecipitación de Cromatina , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/genética , Inhibidores Enzimáticos/farmacología , Estudio de Asociación del Genoma Completo , Histonas/genética , Humanos , Metilación/efectos de los fármacos , Análisis de Secuencia de ADN
16.
Genom Data ; 3: 94-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26484155

RESUMEN

DNA methylation and nucleosome positioning are two key mechanisms that contribute to the epigenetic control of gene expression. During carcinogenesis, the expression of many genes is altered alongside extensive changes in the epigenome, with repressed genes often being associated with local DNA hypermethylation and gain of nucleosomes at their promoters. However the spectrum of alterations that occur at distal regulatory regions has not been extensively studied. To address this we used Nucleosome Occupancy and Methylation sequencing (NOMe-seq) to compare the genome-wide DNA methylation and nucleosome occupancy profiles between normal and cancer cell line models of the breast and prostate. Here we describe the bioinformatic pipeline and methods that we developed for the processing and analysis of the NOMe-seq data published by (Taberlay et al., 2014 [1]) and deposited in the Gene Expression Omnibus with accession GSE57498.

17.
Cancer Cell ; 21(5): 655-667, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22624715

RESUMEN

Cancer cells typically exhibit aberrant DNA methylation patterns that can drive malignant transformation. Whether cancer cells are dependent on these abnormal epigenetic modifications remains elusive. We used experimental and bioinformatic approaches to unveil genomic regions that require DNA methylation for survival of cancer cells. First, we surveyed the residual DNA methylation profiles in cancer cells with highly impaired DNA methyltransferases. Then, we clustered these profiles according to their DNA methylation status in primary normal and tumor tissues. Finally, we used gene expression meta-analysis to identify regions that are dependent on DNA methylation-mediated gene silencing. We further showed experimentally that these genes must be silenced by DNA methylation for cancer cell survival, suggesting these are key epigenetic events associated with tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Metilación de ADN , Epigénesis Genética , Neoplasias/genética , Muerte Celular , Supervivencia Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Análisis por Conglomerados , Biología Computacional , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HCT116 , Humanos , Neoplasias/enzimología , Neoplasias/patología , Fenotipo , Interferencia de ARN , Reproducibilidad de los Resultados , Factores de Tiempo , Transfección
18.
Nat Biotechnol ; 28(10): 1069-78, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20944599

RESUMEN

Epigenetic modifications work in concert with genetic mechanisms to regulate transcriptional activity in normal tissues and are often dysregulated in disease. Although they are somatically heritable, modifications of DNA and histones are also reversible, making them good targets for therapeutic intervention. Epigenetic changes often precede disease pathology, making them valuable diagnostic indicators for disease risk or prognostic indicators for disease progression. Several inhibitors of histone deacetylation or DNA methylation are approved for hematological malignancies by the US Food and Drug Administration and have been in clinical use for several years. More recently, histone methylation and microRNA expression have gained attention as potential therapeutic targets. The presence of multiple epigenetic aberrations within malignant tissue and the abilities of cells to develop resistance suggest that epigenetic therapies are most beneficial when combined with other anticancer strategies, such as signal transduction inhibitors or cytotoxic treatments. A key challenge for future epigenetic therapies will be to develop inhibitors with specificity to particular regions of chromosomes, thereby potentially reducing side effects.


Asunto(s)
Antineoplásicos/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Histonas/metabolismo , Neoplasias/tratamiento farmacológico , Metilación de ADN/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Nucleósidos/uso terapéutico
19.
Curr Protoc Mol Biol ; Chapter 21: Unit 21.17.1-16, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20069538

RESUMEN

Methylation-sensitive single-molecule analysis of chromatin structure is a high-resolution method for studying nucleosome positioning. As described in this unit, this method allows for the analysis of the chromatin structure of unmethylated CpG islands or in vitro-remodeled nucleosomes by treatment with the CpG-specific DNA methyltransferase SssI (M.SssI), followed by bisulfite sequencing of individual progeny DNA molecules. Unlike nuclease-based approaches, this method allows each molecule to be viewed as an individual entity instead of an average population.


Asunto(s)
Cromatina/química , Metilación de ADN , Técnicas Genéticas , Animales , Biocatálisis , Islas de CpG , Humanos
20.
PLoS One ; 4(1): e4213, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19148290

RESUMEN

Neural stem cells (NSCs) can be isolated from different regions of the central nervous system. There has been controversy whether regional differences amongst stem and progenitor cells are cell intrinsic and whether these differences are maintained during expansion in culture. The identification of inherent regional differences has important implications for the use of these cells in neural repair. Here, we compared NSCs derived from the spinal cord and embryonic cortex. We found that while cultured cortical and spinal cord derived NSCs respond similarly to mitogens and are equally neuronogenic, they retain and maintain through multiple passages gene expression patterns indicative of the region from which they were isolated (e.g Emx2 and HoxD10). Further microarray analysis identified 229 genes that were differentially expressed between cortical and spinal cord derived neurospheres, including many Hox genes, Nuclear receptors, Irx3, Pace4, Lhx2, Emx2 and Ntrk2. NSCs in the cortex express LeX. However, in the embryonic spinal cord there are two lineally related populations of NSCs: one that expresses LeX and one that does not. The LeX negative population contains few markers of regional identity but is able to generate LeX expressing NSCs that express markers of regional identity. LeX positive cells do not give rise to LeX-negative NSCs. These results demonstrate that while both embryonic cortical and spinal cord NSCs have similar self-renewal properties and multipotency, they retain aspects of regional identity, even when passaged long-term in vitro. Furthermore, there is a population of a LeX negative NSC that is present in neurospheres derived from the embryonic spinal cord but not the cortex.


Asunto(s)
Encéfalo/citología , Neuronas/citología , Médula Espinal , Células Madre/citología , Animales , Linaje de la Célula , Embrión de Mamíferos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA