Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Cell Rep ; 42(12): 113564, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-38100350

RESUMEN

Triple-negative breast cancer (TNBC) is a heterogeneous disease with limited treatment options. To characterize TNBC heterogeneity, we defined transcriptional, epigenetic, and metabolic subtypes and subtype-driving super-enhancers and transcription factors by combining functional and molecular profiling with computational analyses. Single-cell RNA sequencing revealed relative homogeneity of the major transcriptional subtypes (luminal, basal, and mesenchymal) within samples. We found that mesenchymal TNBCs share features with mesenchymal neuroblastoma and rhabdoid tumors and that the PRRX1 transcription factor is a key driver of these tumors. PRRX1 is sufficient for inducing mesenchymal features in basal but not in luminal TNBC cells via reprogramming super-enhancer landscapes, but it is not required for mesenchymal state maintenance or for cellular viability. Our comprehensive, large-scale, multiplatform, multiomics study of both experimental and clinical TNBC is an important resource for the scientific and clinical research communities and opens venues for future investigation.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/patología , Factores de Transcripción/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo
3.
Nat Cell Biol ; 21(7): 879-888, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31263265

RESUMEN

Most human tumours are heterogeneous, composed of cellular clones with different properties present at variable frequencies. Highly heterogeneous tumours have poor clinical outcomes, yet the underlying mechanism remains poorly understood. Here, we show that minor subclones of breast cancer cells expressing IL11 and FIGF (VEGFD) cooperate to promote metastatic progression and generate polyclonal metastases composed of driver and neutral subclones. Expression profiling of the epithelial and stromal compartments of monoclonal and polyclonal primary and metastatic lesions revealed that this cooperation is indirect, mediated through the local and systemic microenvironments. We identified neutrophils as a leukocyte population stimulated by the IL11-expressing minor subclone and showed that the depletion of neutrophils prevents metastatic outgrowth. Single-cell RNA-seq of CD45+ cell populations from primary tumours, blood and lungs demonstrated that IL11 acts on bone-marrow-derived mesenchymal stromal cells, which induce pro-tumorigenic and pro-metastatic neutrophils. Our results indicate key roles for non-cell-autonomous drivers and minor subclones in metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia/patología , Neutrófilos/metabolismo , Microambiente Tumoral , Animales , Carcinogénesis/metabolismo , Progresión de la Enfermedad , Humanos , Pulmón/patología , Neoplasias Pulmonares/secundario , Células Madre Mesenquimatosas/citología
4.
PLoS Genet ; 15(3): e1008002, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30893315

RESUMEN

Mammary epithelial progenitors are the normal cell-of-origin of breast cancer. We previously defined a population of p27+ quiescent hormone-responsive progenitor cells in the normal human breast whose frequency associates with breast cancer risk. Here, we describe that deletion of the Cdkn1b gene encoding the p27 cyclin-dependent kinase inhibitor in the estrogen-induced mammary tumor-susceptible ACI rat strain leads to a decrease in the relative frequencies of Cd49b+ mammary luminal epithelial progenitors and pregnancy-related differentiation. We show by comprehensive gene expression profiling of purified progenitor and differentiated mammary epithelial cell populations that p27 deletion has the most pronounced effects on luminal progenitors. Cdkn1b-/- females have decreased fertility, but rats that are able to get pregnant had normal litter size and were able to nurse their pups implying that loss of p27 in ACI rats does not completely abrogate ovarian function and lactation. Reciprocal mammary gland transplantation experiments indicate that the p27-loss-induced changes in mammary epithelial cells are not only caused by alterations in their intrinsic properties, but are likely due to altered hormonal signaling triggered by the perturbed systemic endocrine environment observed in Cdkn1b-/- females. We also observed a decrease in the frequency of mammary epithelial cells positive for progesterone receptor (Pr) and FoxA1, known direct transcriptional targets of the estrogen receptor (Erα), and an increase in phospho-Stat5 positive cells commonly induced by prolactin (Prl). Characterization of genome-wide Pr chromatin binding revealed distinct binding patterns in mammary epithelial cells of Cdkn1b+/+ and Cdkn1b-/- females and enrichment in genes with known roles in Notch, ErbB, leptin, and Erα signaling and regulation of G1-S transition. Our data support a role for p27 in regulating the pool size of hormone-responsive luminal progenitors that could impact breast cancer risk.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Animales , Animales Modificados Genéticamente/genética , Neoplasias de la Mama/genética , Diferenciación Celular , Proliferación Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Endocrinas/fisiología , Células Epiteliales , Receptor alfa de Estrógeno , Estrógenos , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Integrina alfa1 , Glándulas Mamarias Animales , Glándulas Mamarias Humanas/crecimiento & desarrollo , Embarazo , Progesterona , Ratas , Ratas Endogámicas ACI , Ratas Sprague-Dawley , Receptores de Estrógenos , Receptores de Progesterona , Factores de Riesgo , Transducción de Señal , Células Madre
5.
Methods Mol Biol ; 1908: 219-228, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30649731

RESUMEN

High-throughput targeted repeat element bisulfite sequencing (HT-TREBS) is designed to assay the methylation level of individual retrotransposon loci of a targeted family, in a locus-specific manner, and on a genome-wide scale. Briefly, genomic DNA is sheared and ligated to Ion Torrent A adaptors (with methylated cytosines), followed by bisulfite-conversion, and amplification with primers designed to bind the targeted retrotransposon. Since the primers carry the Ion Torrent P1 adaptor as a 5'-extension, the amplified library is ready to be size-selected and sequenced on a next-generation sequencing platform. Once sequenced, each retrotransposon is mapped to a particular genomic locus, which is achieved through ensuring at least a 10-bp overlap with flanking unique sequence, followed by the calculation of methylation levels of the mapped retrotransposon using a BiQ Analyzer HT. A complete protocol for library construction as well as the bioinformatics for HT-TREBS is described in this chapter.


Asunto(s)
Metilación de ADN , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Retroelementos , Análisis de Secuencia de ADN/métodos , Epigenómica/métodos , Sitios Genéticos , Humanos , Sulfitos
6.
Cell Rep ; 25(5): 1255-1267.e5, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30380416

RESUMEN

Perturbed epigenomic programs play key roles in tumorigenesis, and chromatin modulators are candidate therapeutic targets in various human cancer types. To define singular and shared dependencies on DNA and histone modifiers and transcription factors in poorly differentiated adult and pediatric cancers, we conducted a targeted shRNA screen across 59 cell lines of 6 cancer types. Here, we describe the TRPS1 transcription factor as a strong breast cancer-specific hit, owing largely to lineage-restricted expression. Knockdown of TRPS1 resulted in perturbed mitosis, apoptosis, and reduced tumor growth. Integrated analysis of TRPS1 transcriptional targets, chromatin binding, and protein interactions revealed that TRPS1 is associated with the NuRD repressor complex. These findings uncover a transcriptional network that is essential for breast cancer cell survival and propagation.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Linaje de la Célula , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Línea Celular Tumoral , Supervivencia Celular/genética , Femenino , Células HEK293 , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Unión Proteica , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
7.
Cancer Res ; 77(20): 5479-5490, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28916652

RESUMEN

Androgen receptor (AR) signaling is a key driver of prostate cancer, and androgen-deprivation therapy (ADT) is a standard treatment for patients with advanced and metastatic disease. However, patients receiving ADT eventually develop incurable castration-resistant prostate cancer (CRPC). Here, we report that the chromatin modifier LSD1, an important regulator of AR transcriptional activity, undergoes epigenetic reprogramming in CRPC. LSD1 reprogramming in this setting activated a subset of cell-cycle genes, including CENPE, a centromere binding protein and mitotic kinesin. CENPE was regulated by the co-binding of LSD1 and AR to its promoter, which was associated with loss of RB1 in CRPC. Notably, genetic deletion or pharmacological inhibition of CENPE significantly decreases tumor growth. Our findings show how LSD1-mediated epigenetic reprogramming drives CRPC, and they offer a mechanistic rationale for its therapeutic targeting in this disease. Cancer Res; 77(20); 5479-90. ©2017 AACR.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Histona Demetilasas/genética , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata/embriología , Neoplasias de la Próstata/genética , Andrógenos/metabolismo , Animales , Línea Celular Tumoral , Reprogramación Celular/genética , Proteínas Cromosómicas no Histona/biosíntesis , Proteínas Cromosómicas no Histona/genética , Progresión de la Enfermedad , Epigénesis Genética , Xenoinjertos , Histona Demetilasas/metabolismo , Humanos , Masculino , Ratones , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal , Transfección
8.
Cancer Discov ; 7(10): 1098-1115, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28652380

RESUMEN

To investigate immune escape during breast tumor progression, we analyzed the composition of leukocytes in normal breast tissues, ductal carcinoma in situ (DCIS), and invasive ductal carcinomas (IDC). We found significant tissue and tumor subtype-specific differences in multiple cell types including T cells and neutrophils. Gene expression profiling of CD45+CD3+ T cells demonstrated a decrease in CD8+ signatures in IDCs. Immunofluorescence analysis showed fewer activated GZMB+CD8+ T cells in IDC than in DCIS, including in matched DCIS and recurrent IDC. T-cell receptor clonotype diversity was significantly higher in DCIS than in IDCs. Immune checkpoint protein TIGIT-expressing T cells were more frequent in DCIS, whereas high PD-L1 expression and amplification of CD274 (encoding PD-L1) was only detected in triple-negative IDCs. Coamplification of a 17q12 chemokine cluster with ERBB2 subdivided HER2+ breast tumors into immunologically and clinically distinct subtypes. Our results show coevolution of cancer cells and the immune microenvironment during tumor progression.Significance: The design of effective cancer immunotherapies requires the understanding of mechanisms underlying immune escape during tumor progression. Here we demonstrate a switch to a less active tumor immune environment during the in situ to invasive breast carcinoma transition, and identify immune regulators and genomic alterations that shape tumor evolution. Cancer Discov; 7(10); 1098-115. ©2017 AACR.See related commentary by Speiser and Verdeil, p. 1062This article is highlighted in the In This Issue feature, p. 1047.


Asunto(s)
Neoplasias de la Mama/inmunología , Carcinoma Ductal de Mama/inmunología , Carcinoma Intraductal no Infiltrante/inmunología , Perfilación de la Expresión Génica/métodos , Linfocitos T/inmunología , Antígeno B7-H1/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Complejo CD3/genética , Carcinoma Ductal de Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Antígenos Comunes de Leucocito/genética , Receptor ErbB-2/genética , Microambiente Tumoral
9.
Nature ; 529(7586): 413-417, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26735014

RESUMEN

Triple-negative breast cancer (TNBC) is a heterogeneous and clinically aggressive disease for which there is no targeted therapy. BET bromodomain inhibitors, which have shown efficacy in several models of cancer, have not been evaluated in TNBC. These inhibitors displace BET bromodomain proteins such as BRD4 from chromatin by competing with their acetyl-lysine recognition modules, leading to inhibition of oncogenic transcriptional programs. Here we report the preferential sensitivity of TNBCs to BET bromodomain inhibition in vitro and in vivo, establishing a rationale for clinical investigation and further motivation to understand mechanisms of resistance. In paired cell lines selected for acquired resistance to BET inhibition from previously sensitive TNBCs, we failed to identify gatekeeper mutations, new driver events or drug pump activation. BET-resistant TNBC cells remain dependent on wild-type BRD4, which supports transcription and cell proliferation in a bromodomain-independent manner. Proteomic studies of resistant TNBC identify strong association with MED1 and hyper-phosphorylation of BRD4 attributable to decreased activity of PP2A, identified here as a principal BRD4 serine phosphatase. Together, these studies provide a rationale for BET inhibition in TNBC and present mechanism-based combination strategies to anticipate clinical drug resistance.


Asunto(s)
Azepinas/farmacología , Azepinas/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Nucleares/antagonistas & inhibidores , Estructura Terciaria de Proteína/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Triazoles/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Unión Competitiva/efectos de los fármacos , Quinasa de la Caseína II/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Resistencia a Antineoplásicos/genética , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Humanos , Subunidad 1 del Complejo Mediador/metabolismo , Ratones , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Proteína Fosfatasa 2/metabolismo , Proteómica , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Genom Data ; 3: 87-89, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25554740

RESUMEN

DNA methylation is a major epigenetic mark associated with multiple aspects of retrotransposons within the mammalian genome. In order to study DNA methylation of a large number of retrotransposons on an individual-locus basis, we have developed a new protocol termed High-Throughput Targeted Repeat Element Bisulfite Sequencing (HT-TREBS) (Ekram and Kim, 2014 [1]). We have used this technique to characterize the locus-specific patterns of DNA methylation of 4799 members of the mouse IAP LTR (Intracisternal A Particle Long Terminal Repeat) retrotransposon family in embryonic stem, somatic and Neuro2A cells (Bakshi and Kim, 2014 [2]). Here we describe in detail the sample preparation and bioinformatics analyses used for these studies. The somatic cell data may be accessed under GEO accession number GSE49222. The ES and Neuro2A data are deposited under GEO accession number GSE60007.

11.
Genomics ; 105(2): 108-15, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25451679

RESUMEN

Aebp2 encodes an evolutionarily conserved zinc finger protein that has not been well studied so far, yet recent studies indicated that this gene is closely associated with the Polycomb Repressive Complex 2 (PRC2). Thus, the current study characterized the basic aspects of this gene, including alternative promoters and protein isoforms. According to the results, Aebp2 is controlled through three alternative promoters, deriving three different transcripts encoding the embryonic (32 kDa) and somatic (52 kDa) forms. Chromatin Immuno-Precipitation (ChIP) experiments revealed that AEBP2 binds to its own promoter as well as the promoters of Jarid2 and Snai2. While the embryonic form acts as a transcriptional repressor for Snai2, the somatic form functions as a transcriptional activator for Jarid2, Aebp2 and Snai2. Cell migration assays also demonstrated that the Aebp2 somatic form has an enhancing activity in cell migration. This is consistent with the functional association of Aebp2 with migratory neural crest cells. These results suggest that the two protein isoforms of AEBP2 may have opposite functions for the PcG target genes, and may play significant roles in cell migration during development.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Activación Transcripcional , Animales , Secuencia de Bases , Regulación del Desarrollo de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Cresta Neural/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Isoformas de Proteínas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
12.
PLoS One ; 9(7): e101683, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25003790

RESUMEN

In vertebrates, DNA methylation-mediated repression of retrotransposons is essential for the maintenance of genomic integrity. In the current study, we developed a technique termed HT-TREBS (High-Throughput Targeted Repeat Element Bisulfite Sequencing). This technique is designed to measure the DNA methylation levels of individual loci of any repeat families with next-generation sequencing approaches. To test the feasibility of HT-TREBS, we analyzed the DNA methylation levels of the IAP LTR family using a set of 12 different genomic DNA isolated from the brain, liver and kidney of 4 one-week-old littermates of the mouse strain C57BL/6N. This technique has successfully generated the CpG methylation data of 5,233 loci common in all the samples, representing more than 80% of the individual loci of the five targeted subtypes of the IAP LTR family. According to the results, approximately 5% of the IAP LTR loci have less than 80% CpG methylation levels with no genomic position preference. Further analyses of the IAP LTR loci also revealed the presence of extensive DNA methylation variations between different tissues and individuals. Overall, these data demonstrate the efficiency and robustness of the new technique, HT-TREBS, and also provide new insights regarding the genome-wide DNA methylation patterns of the IAP LTR repeat elements.


Asunto(s)
Metilación de ADN , Genes de Partícula A Intracisternal , Estudio de Asociación del Genoma Completo/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Animales , Biología Computacional , Islas de CpG , Femenino , Masculino , Ratones , Retroelementos
13.
Cancer Cell ; 25(6): 762-77, 2014 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-24937458

RESUMEN

Recurrent mutations in histone-modifying enzymes imply key roles in tumorigenesis, yet their functional relevance is largely unknown. Here, we show that JARID1B, encoding a histone H3 lysine 4 (H3K4) demethylase, is frequently amplified and overexpressed in luminal breast tumors and a somatic mutation in a basal-like breast cancer results in the gain of unique chromatin binding and luminal expression and splicing patterns. Downregulation of JARID1B in luminal cells induces basal genes expression and growth arrest, which is rescued by TGFß pathway inhibitors. Integrated JARID1B chromatin binding, H3K4 methylation, and expression profiles suggest a key function for JARID1B in luminal cell-specific expression programs. High luminal JARID1B activity is associated with poor outcome in patients with hormone receptor-positive breast tumors.


Asunto(s)
Neoplasias de la Mama/genética , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas Nucleares/genética , Oncogenes , Proteínas Represoras/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Factor de Unión a CCCTC , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Linaje de la Célula , Femenino , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células MCF-7 , Mutación , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Pirazoles/farmacología , Pirroles/farmacología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteínas Represoras/metabolismo , Transfección , Factor de Crecimiento Transformador beta/metabolismo
14.
Gene ; 512(2): 314-20, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23078764

RESUMEN

The Peg3 gene is expressed only from the paternally inherited allele located on proximal mouse chromosome 7. The PEG3 protein encoded by this imprinted gene is predicted to bind DNA based on its multiple zinc finger motifs and nuclear localization. In the current study, we demonstrated PEG3's DNA-binding ability by characterizing its binding motif and target genes. We successfully identified target regions bound by PEG3 from mouse brain extracts using chromatin immunoprecipitation analysis. PEG3 was demonstrated to bind these candidate regions through the consensus DNA-binding motif AGTnnCnnnTGGCT. In vitro promoter assays established that PEG3 controls the expression of a given gene through this motif. Consistent with these observations, the transcriptional levels of a subset of the target genes are also affected in a mutant mouse model with reduced levels of PEG3 protein. Overall, these results confirm PEG3 as a DNA-binding protein controlling specific target genes that are involved in distinct cellular functions.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Regulación de la Expresión Génica/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Elementos de Respuesta/fisiología , Animales , Cromosomas de los Mamíferos/genética , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Mutantes
15.
PLoS One ; 8(12): e83359, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24391757

RESUMEN

Peg3 (paternally expressed gene 3) is an imprinted gene encoding a DNA-binding protein. This gene plays important roles in controlling fetal growth rates and nurturing behaviors. In the current study, a new mutant mouse model has been generated to further characterize the functions of this DNA-binding protein. Besides known phenotypes, this new mutant model also revealed potential roles of Peg3 in mammalian reproduction. Female heterozygotes produce a much smaller number of mature oocytes than the wild-type littermates, resulting in reduced litter sizes. According to genome-wide expression analyses, several placenta-specific gene families are de-repressed in the brain of Peg3 heterozygous embryos, including prolactin, cathepsin and carcinoembryonic antigen cell adhesion molecule (Ceacam) families. The observed de-repression is more pronounced in females than in males. The de-repression of several members of these gene families is observed even in the adult brain, suggesting potential defects in epigenetic setting of the placenta-specific gene families in the Peg3 mutants. Overall, these results indicate that Peg3 likely controls the transcription of several placenta-specific gene families, and further suggest that this predicted transcriptional control by Peg3 might be mediated through unknown epigenetic mechanisms.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , Mutación , Placenta/metabolismo , Reproducción/genética , Animales , Animales Recién Nacidos , Animales Lactantes , Epigénesis Genética , Femenino , Expresión Génica , Impresión Genómica , Cabeza/embriología , Heterocigoto , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/fisiología , Tamaño de la Camada/genética , Tamaño de la Camada/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Familia de Multigenes , Embarazo , Reproducción/fisiología , Transcripción Genética
16.
Epigenetics ; 7(4): 370-82, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22415164

RESUMEN

Transcription of retrotransposons is usually repressed by DNA methylation, but a few elements, such as intracisternal A-particles (IAPs) associated with the Agouti and Axin-fused loci, partially escape this repression mechanism. The levels of this repression are also variable among individuals with an identical genome sequence, generating epigenetically different states of loci or 'epialleles.' In the current study, we tested the existence of additional retrotransposon-derived epialleles in the mouse genome. Using a series of bioinformatic approaches, 143 candidate epialleles were first identified from the mouse genome based on their promoter activity and association with active histone modification marks. Detailed analyses suggest that a subset of these elements showed variable levels of DNA methylation among the individual mice of an isogenic background, revealing their stochastic nature (metastability) of DNA methylation. The analyses also identified two opposite patterns of DNA methylation during development, progressive gaining vs. losing, confirming the dynamic nature of their DNA methylation patterns. qRT-PCR analyses demonstrated that the expression levels of these elements are indeed variable among the individual mice, suggesting functional consequences on their associated endogenous genes. Overall, these data confirm the presence of a number of new retrotransposon-derived epialleles with suggestions of the presence of more, and further identify retrotransposons as a major source of epigenetic variations in the mammalian genome.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Mamíferos/genética , Retroelementos , Alelos , Animales , Blastocisto/citología , Blastocisto/metabolismo , Biología Computacional , Islas de CpG , Bases de Datos de Ácidos Nucleicos , Femenino , Inestabilidad Genómica , Histonas/genética , Histonas/metabolismo , Masculino , Mamíferos/metabolismo , Ratones , Ratones Endogámicos C57BL , Oocitos/citología , Oocitos/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN/métodos , Espermatozoides/citología , Secuencias Repetidas Terminales
17.
Hum Mol Genet ; 21(12): 2677-87, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22394678

RESUMEN

The imprinting and transcription of the 500 kb genomic region surrounding the mouse Peg3 is predicted to be regulated by the Peg3-differentially methylated region (DMR). In the current study, this prediction was tested using a mutant mouse line lacking this potential imprinting control region (ICR). At the organismal level, paternal and maternal transmission of this knockout (KO) allele caused either reduced or increased growth rates in the mouse, respectively. In terms of the imprinting control, the paternal transmission of the KO allele resulted in bi-allelic expression of the normally maternally expressed Zim2, whereas the maternal transmission switched the transcriptionally dominant allele for Zfp264 (paternal to maternal). However, the allele-specific DNA methylation patterns of the DMRs of Peg3, Zim2 and Zim3 were not affected in the mice that inherited the KO allele either paternally or maternally. In terms of the transcriptional control, the paternal transmission caused a dramatic down-regulation in Peg3 expression, but overall up-regulation in the other nearby imprinted genes. Taken together, deletion of the Peg3-DMR caused global changes in the imprinting and transcription of the Peg3 domain, confirming that the Peg3-DMR is an ICR for this imprinted domain.


Asunto(s)
Metilación de ADN , Impresión Genómica , Factores de Transcripción de Tipo Kruppel/genética , Alelos , Animales , Animales Recién Nacidos , Sitios de Unión/genética , Western Blotting , Encéfalo/metabolismo , Cruzamientos Genéticos , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Perfilación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Dedos de Zinc/genética
18.
PLoS One ; 6(9): e25174, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949878

RESUMEN

Aebp2 is a potential targeting protein for the mammalian Polycomb Repression Complex 2 (PRC2). We generated a mutant mouse line disrupting the transcription of Aebp2 to investigate its in vivo roles. Aebp2-mutant homozygotes were embryonic lethal while heterozygotes survived to adulthood with fertility. In developing mouse embryos, Aebp2 is expressed mainly within cells of neural crest origin. In addition, many heterozygotes display a set of phenotypes, enlarged colon and hypopigmentation, similar to those observed in human patients with Hirschsprung's disease and Waardenburg syndrome. These phenotypes are usually caused by the absence of the neural crest-derived ganglia in hindguts and melanocytes. ChIP analyses demonstrated that the majority of the genes involved in the migration and development process of neural crest cells are downstream target genes of AEBP2 and PRC2. Furthermore, expression analyses confirmed that some of these genes are indeed affected in the Aebp2 heterozygotes. Taken together, these results suggest that Aebp2 may regulate the migration and development of the neural crest cells through the PRC2-mediated epigenetic mechanism.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Epigenómica , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/metabolismo , Proteínas Nucleares/fisiología , Proteínas Represoras/metabolismo , Animales , Southern Blotting , Western Blotting , Inmunoprecipitación de Cromatina , Femenino , Humanos , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cresta Neural/embriología , Proteínas del Grupo Polycomb , ARN Mensajero/genética , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Hum Mol Genet ; 20(7): 1353-62, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21233130

RESUMEN

Zfp42/Rex1 (reduced expression gene 1) is a well-known stem-cell marker that has been duplicated from YY1 in the eutherian lineage. In the current study, we characterized the in vivo roles of Rex1 using a mutant mouse line disrupting its transcription. In contrast to the ubiquitous expression of YY1, Rex1 is expressed only during spermatogenesis and early embryogenesis and also in a very limited area of the placenta. Yet, the gene dosage of Rex1 is very critical for the survival of the late-stage embryos and neonates. This delayed phenotypic consequence suggests potential roles for Rex1 in establishing and maintaining unknown epigenetic modifications. Consistently, Rex1-null blastocysts display hypermethylation in the differentially methylated regions (DMRs) of Peg3 and Gnas imprinted domains, which are known to contain YY1 binding sites. Further analyses confirmed in vivo binding of Rex1 only to the unmethylated allele of these two regions. Thus, Rex1 may function as a protector for these DMRs against DNA methylation. Overall, the functional connection of Rex1 to genomic imprinting represents another case where newly made genes have co-evolved with lineage-specific phenomena.


Asunto(s)
Alelos , Blastocisto/metabolismo , Metilación de ADN/fisiología , Impresión Genómica/fisiología , Factores de Transcripción/metabolismo , Animales , Blastocisto/citología , Cromograninas , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Mutantes , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...