Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 29(7): 2281-2293, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-33744470

RESUMEN

Abnormal cholesterol/lipid homeostasis is linked to neurodegenerative conditions such as age-related macular degeneration (AMD), which is a leading cause of blindness in the elderly. The most prevalent form, termed "dry" AMD, is characterized by pathological cholesterol accumulation beneath the retinal pigment epithelial (RPE) cell layer and inflammation-linked degeneration in the retina. We show here that the cholesterol-regulating microRNA miR-33 was elevated in the RPE of aging mice. Expression of the miR-33 target ATP-binding cassette transporter (ABCA1), a cholesterol efflux pump genetically linked to AMD, declined reciprocally in the RPE with age. In accord, miR-33 modulated ABCA1 expression and cholesterol efflux in human RPE cells. Subcutaneous delivery of miR-33 antisense oligonucleotides (ASO) to aging mice and non-human primates fed a Western-type high fat/cholesterol diet resulted in increased ABCA1 expression, decreased cholesterol accumulation, and reduced immune cell infiltration in the RPE cell layer, accompanied by decreased pathological changes to RPE morphology. These findings suggest that miR-33 targeting may decrease cholesterol deposition and ameliorate AMD initiation and progression.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Colesterol/metabolismo , Inflamación/terapia , Degeneración Macular/terapia , MicroARNs/antagonistas & inhibidores , Fenotipo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Inflamación/etiología , Inflamación/patología , Macaca fascicularis , Degeneración Macular/etiología , Degeneración Macular/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Oligonucleótidos Antisentido/genética
2.
Cell ; 183(3): 684-701.e14, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33058756

RESUMEN

Positive selection in Europeans at the 2q21.3 locus harboring the lactase gene has been attributed to selection for the ability of adults to digest milk to survive famine in ancient times. However, the 2q21.3 locus is also associated with obesity and type 2 diabetes in humans, raising the possibility that additional genetic elements in the locus may have contributed to evolutionary adaptation to famine by promoting energy storage, but which now confer susceptibility to metabolic diseases. We show here that the miR-128-1 microRNA, located at the center of the positively selected locus, represents a crucial metabolic regulator in mammals. Antisense targeting and genetic ablation of miR-128-1 in mouse metabolic disease models result in increased energy expenditure and amelioration of high-fat-diet-induced obesity and markedly improved glucose tolerance. A thrifty phenotype connected to miR-128-1-dependent energy storage may link ancient adaptation to famine and modern metabolic maladaptation associated with nutritional overabundance.


Asunto(s)
Enfermedades Metabólicas/genética , MicroARNs/genética , Adipocitos Marrones/patología , Adiposidad , Alelos , Animales , Diferenciación Celular , Línea Celular , Células Cultivadas , Dieta Alta en Grasa , Metabolismo Energético , Epigénesis Genética , Sitios Genéticos , Glucosa/metabolismo , Homeostasis , Humanos , Hipertrofia , Resistencia a la Insulina , Leptina/deficiencia , Leptina/metabolismo , Masculino , Mamíferos/genética , Ratones Endogámicos C57BL , Ratones Obesos , MicroARNs/metabolismo , Obesidad/genética , Oligonucleótidos/metabolismo , Especificidad de la Especie
3.
Cell ; 177(3): 722-736.e22, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30955890

RESUMEN

Insulin receptor (IR) signaling is central to normal metabolic control and dysregulated in prevalent chronic diseases. IR binds insulin at the cell surface and transduces rapid signaling via cytoplasmic kinases. However, mechanisms mediating long-term effects of insulin remain unclear. Here, we show that IR associates with RNA polymerase II in the nucleus, with striking enrichment at promoters genome-wide. The target genes were highly enriched for insulin-related functions including lipid metabolism and protein synthesis and diseases including diabetes, neurodegeneration, and cancer. IR chromatin binding was increased by insulin and impaired in an insulin-resistant disease model. Promoter binding by IR was mediated by coregulator host cell factor-1 (HCF-1) and transcription factors, revealing an HCF-1-dependent pathway for gene regulation by insulin. These results show that IR interacts with transcriptional machinery at promoters and identify a pathway regulating genes linked to insulin's effects in physiology and disease.


Asunto(s)
Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Receptor de Insulina/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factor C1 de la Célula Huésped/antagonistas & inhibidores , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/metabolismo , Humanos , Insulina/metabolismo , Insulina/farmacología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Unión Proteica , Subunidades de Proteína/metabolismo , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño/metabolismo , Receptor de Insulina/química , Transducción de Señal/efectos de los fármacos
4.
Nucleic Acid Ther ; 28(5): 273-284, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30133337

RESUMEN

The Ebola virus is a zoonotic pathogen that can cause severe hemorrhagic fever in humans, with up to 90% lethality. The deadly 2014 Ebola outbreak quickly made an unprecedented impact on human lives. While several vaccines and therapeutics are under development, current approaches contain several limitations, such as virus mutational escape, need for formulation or refrigeration, poor scalability, long lead-time, and high cost. To address these challenges, we developed locked nucleic acid (LNA)-modified antisense oligonucleotides (ASOs) to target critical Ebola viral proteins and the human intracellular host protein Niemann-Pick C1 (NPC1), required for viral entry into infected cells. We generated noninfectious viral luciferase reporter assays to identify LNA ASOs that inhibit translation of Ebola viral proteins in vitro and in human cells. We demonstrated specific inhibition of key Ebola genes VP24 and nucleoprotein, which inhibit a proper immune response and promote Ebola virus replication, respectively. We also identified LNA ASOs targeting human host factor NPC1 and demonstrated reduced infection by chimeric vesicular stomatitis virus harboring the Ebola glycoprotein, which directly binds to NPC1 for viral infection. These results support further in vivo testing of LNA ASOs in infectious Ebola virus disease animal models as potential therapeutic modalities for treatment of Ebola.


Asunto(s)
Fiebre Hemorrágica Ebola/genética , Proteína Niemann-Pick C1/genética , Oligonucleótidos Antisentido/genética , Proteínas Virales/genética , Animales , Modelos Animales de Enfermedad , Ebolavirus/genética , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/terapia , Fiebre Hemorrágica Ebola/virología , Humanos , Inmunidad Innata/genética , Ratones , Proteína Niemann-Pick C1/antagonistas & inhibidores , Nucleoproteínas/antagonistas & inhibidores , Nucleoproteínas/genética , Oligonucleótidos/genética , Oligonucleótidos/uso terapéutico , Oligonucleótidos Antisentido/uso terapéutico , Primates/virología , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/genética
5.
Biochim Biophys Acta ; 1861(12 Pt B): 2047-2052, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26968099

RESUMEN

In the past decade, microRNAs (miRNAs) have emerged as key regulators of circulating levels of lipoproteins. Specifically, recent work has uncovered the role of miRNAs in controlling the levels of atherogenic low-density lipoprotein LDL (LDL)-cholesterol by post-transcriptionally regulating genes involved in very low-density lipoprotein (VLDL) secretion, cholesterol biosynthesis, and hepatic LDL receptor (LDLR) expression. Interestingly, several of these miRNAs are located in genomic loci associated with abnormal levels of circulating lipids in humans. These findings reinforce the interest of targeting this subset of non-coding RNAs as potential therapeutic avenues for regulating plasma cholesterol and triglyceride (TAG) levels. In this review, we will discuss how these new miRNAs represent potential pre-disposition factors for cardiovascular disease (CVD), and putative therapeutic targets in patients with cardiometabolic disorders. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.


Asunto(s)
LDL-Colesterol/genética , MicroARNs/genética , Animales , Enfermedades Cardiovasculares/genética , Humanos , Enfermedades Metabólicas/genética , Receptores de LDL/genética , Triglicéridos/genética
6.
Nat Med ; 21(11): 1290-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26501192

RESUMEN

Genome-wide association studies (GWASs) have linked genes to various pathological traits. However, the potential contribution of regulatory noncoding RNAs, such as microRNAs (miRNAs), to a genetic predisposition to pathological conditions has remained unclear. We leveraged GWAS meta-analysis data from >188,000 individuals to identify 69 miRNAs in physical proximity to single-nucleotide polymorphisms (SNPs) associated with abnormal levels of circulating lipids. Several of these miRNAs (miR-128-1, miR-148a, miR-130b, and miR-301b) control the expression of key proteins involved in cholesterol-lipoprotein trafficking, such as the low-density lipoprotein (LDL) receptor (LDLR) and the ATP-binding cassette A1 (ABCA1) cholesterol transporter. Consistent with human liver expression data and genetic links to abnormal blood lipid levels, overexpression and antisense targeting of miR-128-1 or miR-148a in high-fat diet-fed C57BL/6J and Apoe-null mice resulted in altered hepatic expression of proteins involved in lipid trafficking and metabolism, and in modulated levels of circulating lipoprotein-cholesterol and triglycerides. Taken together, these findings support the notion that altered expression of miRNAs may contribute to abnormal blood lipid levels, predisposing individuals to human cardiometabolic disorders.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Dieta Alta en Grasa , Dislipidemias/genética , MicroARNs/genética , Receptores de LDL/metabolismo , Triglicéridos/metabolismo , Animales , Apolipoproteínas E/genética , Colesterol/metabolismo , Estudio de Asociación del Genoma Completo , Homeostasis/genética , Humanos , Lipoproteínas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple
7.
Nat Med ; 21(11): 1280-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26437365

RESUMEN

The hepatic low-density lipoprotein receptor (LDLR) pathway is essential for clearing circulating LDL cholesterol (LDL-C). Whereas the transcriptional regulation of LDLR is well characterized, the post-transcriptional mechanisms that govern LDLR expression are just beginning to emerge. Here we develop a high-throughput genome-wide screening assay to systematically identify microRNAs (miRNAs) that regulate LDLR activity in human hepatic cells. From this screen we identified and characterized miR-148a as a negative regulator of LDLR expression and activity and defined a sterol regulatory element-binding protein 1 (SREBP1)-mediated pathway through which miR-148a regulates LDL-C uptake. In mice, inhibition of miR-148a increased hepatic LDLR expression and decreased plasma LDL-C. Moreover, we found that miR-148a regulates hepatic expression of ATP-binding cassette, subfamily A, member 1 (ABCA1) and circulating high-density lipoprotein cholesterol (HDL-C) levels in vivo. These studies uncover a role for miR-148a as a key regulator of hepatic LDL-C clearance through direct modulation of LDLR expression and demonstrate the therapeutic potential of inhibiting miR-148a to ameliorate an elevated LDL-C/HDL-C ratio, a prominent risk factor for cardiovascular disease.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , MicroARNs/genética , Receptores de LDL/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Regulación de la Expresión Génica , Células Hep G2 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN , Receptores de LDL/metabolismo , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
8.
Cell ; 150(6): 1147-57, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22980978

RESUMEN

Transcription elongation is increasingly recognized as an important mechanism of gene regulation. Here, we show that microprocessor controls gene expression in an RNAi-independent manner. Microprocessor orchestrates the recruitment of termination factors Setx and Xrn2, and the 3'-5' exoribonuclease, Rrp6, to initiate RNAPII pausing and premature termination at the HIV-1 promoter through cleavage of the stem-loop RNA, TAR. Rrp6 further processes the cleavage product, which generates a small RNA that is required to mediate potent transcriptional repression and chromatin remodeling at the HIV-1 promoter. Using chromatin immunoprecipitation coupled to high-throughput sequencing (ChIP-seq), we identified cellular gene targets whose transcription is modulated by microprocessor. Our study reveals RNAPII pausing and premature termination mediated by the co-operative activity of ribonucleases, Drosha/Dgcr8, Xrn2, and Rrp6, as a regulatory mechanism of RNAPII-dependent transcription elongation.


Asunto(s)
Exorribonucleasas/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Regulación Viral de la Expresión Génica , VIH-1/genética , ARN Helicasas/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética , Secuencia de Bases , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , ADN Helicasas , Duplicado del Terminal Largo de VIH , Humanos , Datos de Secuencia Molecular , Enzimas Multifuncionales , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Viral/química , ARN Viral/genética , Factores de Transcripción/metabolismo
9.
Retrovirology ; 6: 26, 2009 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-19272132

RESUMEN

The rate of HIV-1 gene expression is a key step that determines the kinetics of virus spread and AIDS progression. Viral entry and gene expression were described to be the key determinants for cell permissiveness to HIV. Recent reports highlighted the involvement of miRNA in regulating HIV-1 replication post-transcriptionally. In this study we explored the role of cellular factors required for miRNA-mediated mRNA translational inhibition in regulating HIV-1 gene expression. Here we show that HIV-1 mRNAs associate and co-localize with components of the RNA Induced Silencing Complex (RISC), and we characterize some of the proteins required for miRNA-mediated silencing (miRNA effectors). RCK/p54, GW182, LSm-1 and XRN1 negatively regulate HIV-1 gene expression by preventing viral mRNA association with polysomes. Interestingly, knockdown of RCK/p54 or DGCR8 resulted in virus reactivation in PBMCs isolated from HIV infected patients treated with suppressive HAART.


Asunto(s)
Regulación Viral de la Expresión Génica , VIH-1/fisiología , MicroARNs/metabolismo , ARN Viral/metabolismo , Replicación Viral/fisiología , Proteínas Argonautas , Línea Celular , Células Cultivadas , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Polirribosomas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Latencia del Virus/fisiología
10.
Genome Res ; 18(8): 1270-81, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18480470

RESUMEN

Genomic imprinting is a developmentally important mechanism that involves both differential DNA methylation and allelic histone modifications. Through detailed comparative characterization, a large imprinted domain mapping to chromosome 7q21 in humans and proximal chromosome 6 in mice was redefined. This domain is organized around a maternally methylated CpG island comprising the promoters of the adjacent PEG10 and SGCE imprinted genes. Examination of Dnmt3l(-/+) conceptuses shows that imprinted expression for all genes of the cluster depends upon the germline methylation at this putative "imprinting control region" (ICR). Similarly as for other ICRs, we find its DNA-methylated allele to be associated with trimethylation of lysine 9 on histone H3 (H3K9me3) and trimethylation of lysine 20 on histone H4 (H4K20me3), whereas the transcriptionally active paternal allele is enriched in H3K4me2 and H3K9 acetylation. Our study reveals a novel placenta-specific transcript, TFPI2, which is expressed from the maternal allele in both humans and mice. Deficiency for the histone methyltransferase EHMT2 (also known as G9A) or for the Polycomb group protein EED, involved in repressive H3K9me2 and H3K27me3 respectively, leads to biallelic expression of Tfpi2 in the extra-embryonic lineages, whereas the other genes in the cluster maintain correct imprinting. Apart from the putative ICR, however, no other promoter regions within the domain exhibited allele-specific repressive histone modifications. This unexpected general lack of repressive histone modifications suggests that this domain may utilize a different silencing mechanism as compared to other imprinted domains.


Asunto(s)
Cromosomas Humanos Par 7 , Silenciador del Gen , Impresión Genómica , Glicoproteínas/genética , N-Metiltransferasa de Histona-Lisina/fisiología , Proteínas Represoras/fisiología , Alelos , Animales , Proteínas Reguladoras de la Apoptosis , Cromosomas de los Mamíferos , Metilación de ADN , Proteínas de Unión al ADN , Femenino , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/metabolismo , Complejo Represivo Polycomb 2 , Embarazo , Proteínas/genética , Proteínas de Unión al ARN , Proteínas Represoras/genética
11.
Mol Cell Biol ; 28(3): 1104-13, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18039842

RESUMEN

Whereas DNA methylation is essential for genomic imprinting, the importance of histone methylation in the allelic expression of imprinted genes is unclear. Imprinting control regions (ICRs), however, are marked by histone H3-K9 methylation on their DNA-methylated allele. In the placenta, the paternal silencing along the Kcnq1 domain on distal chromosome 7 also correlates with the presence of H3-K9 methylation, but imprinted repression at these genes is maintained independently of DNA methylation. To explore which histone methyltransferase (HMT) could mediate the allelic H3-K9 methylation on distal chromosome 7, and at ICRs, we generated mouse conceptuses deficient for the SET domain protein G9a. We found that in the embryo and placenta, the differential DNA methylation at ICRs and imprinted genes is maintained in the absence of G9a. Accordingly, in embryos, imprinted gene expression was unchanged at the domains analyzed, in spite of a global loss of H3-K9 dimethylation (H3K9me2). In contrast, the placenta-specific imprinting of genes on distal chromosome 7 is impaired in the absence of G9a, and this correlates with reduced levels of H3K9me2 and H3K9me3. These findings provide the first evidence for the involvement of an HMT and suggest that histone methylation contributes to imprinted gene repression in the trophoblast.


Asunto(s)
Impresión Genómica , N-Metiltransferasa de Histona-Lisina/fisiología , Histonas/metabolismo , Placenta/enzimología , Animales , Embrión de Mamíferos , Femenino , Silenciador del Gen , Histona Metiltransferasas , Metilación , Ratones , Proteína Metiltransferasas , Trofoblastos/metabolismo
12.
CSH Protoc ; 2007: pdb.prot4767, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21357105

RESUMEN

INTRODUCTIONIn cells and tissues, the histone proteins that constitute the nucleosomes can present multiple post-translational modifications, such as lysine acetylation, lysine and arginine methylation, serine phosphorylation, and lysine ubiquitination. On their own, or in combination, these covalent modifications on the core histones are thought to play essential roles in chromatin organization and gene expression in eukaryotes. Importantly, patterns of histone modifications may be somatically conserved and can, thereby, maintain locus-specific repression/activity in defined lineages, or throughout development. Indirect immunofluorescence studies on cultured cells have been pivotal in unraveling the roles of histone modifications. However, to address in detail what happens at specific sites in vivo, chromatin immunoprecipitation (ChIP) is the method of choice. Here, we describe how ChIP can be performed on non-fixed chromatin from animal cells or tissues (fresh or frozen) to analyze histone modifications at specific chromosomal sites. These protocols are suitable only for analyzing histones and their modifications. For other applications, chromatin immunoprecipitation should be performed on cross-linked chromatin.

13.
CSH Protoc ; 2007: pdb.prot4768, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21357106

RESUMEN

INTRODUCTIONAfter chromatin immunoprecipitation (ChIP), different PCR-based approaches can be used to determine how much DNA is precipitated at a locus of interest. Real-time PCR amplification is often the preferred technique. One can also use duplex PCR amplification, which is the coamplification of a fragment from the region of interest and a control fragment (e.g., the actin gene, or the tubulin gene). This approach allows for estimating relative levels of specific histone modifications along chromosomal domains. For allele-specific studies (for instance, on dosage-compensation mechanisms or on genomic imprinting), electrophoretic detection of single-strand conformation polymorphisms (SSCP) or similar strategies such as hot-stop PCR can differentiate PCR products that represent the silent allele from those amplified from the active allele. If a polymorphic restriction site is present in one allele and absent in the other, the method of choice is hot-stop PCR. If no polymorphic restriction sites are available, but there are single nucleotide polymorphisms (SNPs) that distinguish the alleles of interest, the best approach is to separate the PCR products derived from the two different alleles using SSCP. In SSCP, it is possible to discriminate denatured PCR products derived from one allele or the other because the secondary structure of each single strand will be directly dependent on the sequence itself. Hence, in nondenaturing gel conditions, each single strand will migrate differently. These four PCR-based methodologies to analyze immunoprecipitated chromatin (real-time PCR, duplex PCR, hot-stop PCR, and SSCP) are presented here.

14.
Bioessays ; 28(5): 453-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16615080

RESUMEN

Human chromosome 11p15 comprises two imprinted domains important in the control of fetal and postnatal growth. Novel studies establish that imprinting at one of these, the IGF2-H19 domain, is epigenetically deregulated (with loss of DNA methylation) in Silver-Russell Syndrome (SRS), a congenital disease of growth retardation and asymmetry. Previously, the exact opposite epigenetic alteration (gain of DNA methylation) had been detected at the domain's 'imprinting control region' (ICR) in patients with Beckwith-Wiedemann Syndrome (BWS), a complex disorder of fetal overgrowth. However, more frequently, BWS is caused by loss of DNA methylation at the ICR that regulates the second imprinted domain at 11p15. Interestingly, a similar epigenetic alteration (with loss of methylation) at a putative ICR on human chromosome 6q24, is involved in transient neonatal diabetes mellitus (TNDM), a congenital disease with intrauterine growth retardation and a transient lack of insulin. Thus, fetal and postnatal growth is epigenetically controlled by different ICRs, at 11p15 and other chromosomal regions.


Asunto(s)
Epigénesis Genética , Impresión Genómica , Trastornos del Crecimiento/genética , Síndrome de Beckwith-Wiedemann/genética , Cromosomas Humanos Par 11/genética , Metilación de ADN , Diabetes Mellitus/congénito , Diabetes Mellitus/genética , Femenino , Trastornos del Crecimiento/congénito , Humanos , Recién Nacido , Masculino , Modelos Genéticos , Embarazo , Síndrome
15.
Cell ; 125(2): 315-26, 2006 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-16630819

RESUMEN

The most highly conserved noncoding elements (HCNEs) in mammalian genomes cluster within regions enriched for genes encoding developmentally important transcription factors (TFs). This suggests that HCNE-rich regions may contain key regulatory controls involved in development. We explored this by examining histone methylation in mouse embryonic stem (ES) cells across 56 large HCNE-rich loci. We identified a specific modification pattern, termed "bivalent domains," consisting of large regions of H3 lysine 27 methylation harboring smaller regions of H3 lysine 4 methylation. Bivalent domains tend to coincide with TF genes expressed at low levels. We propose that bivalent domains silence developmental genes in ES cells while keeping them poised for activation. We also found striking correspondences between genome sequence and histone methylation in ES cells, which become notably weaker in differentiated cells. These results highlight the importance of DNA sequence in defining the initial epigenetic landscape and suggest a novel chromatin-based mechanism for maintaining pluripotency.


Asunto(s)
Cromatina/química , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Conformación de Ácido Nucleico , Células Madre/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Perfilación de la Expresión Génica , Histonas/química , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre/citología
16.
Nat Genet ; 36(12): 1296-300, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15516932

RESUMEN

Imprinted genes are clustered in domains, and their allelic repression is mediated by imprinting control regions. These imprinting control regions are marked by DNA methylation, which is essential to maintain imprinting in the embryo. To explore how imprinting is regulated in placenta, we studied the Kcnq1 domain on mouse distal chromosome 7. This large domain is controlled by an intronic imprinting control region and comprises multiple genes that are imprinted in placenta, without the involvement of promoter DNA methylation. We found that the paternal repression along the domain involves acquisition of trimethylation at Lys27 and dimethylation at Lys9 of histone H3. Eed-Ezh2 Polycomb complexes are recruited to the paternal chromosome and potentially regulate its repressive histone methylation. Studies on embryonic stem cells and early embryos support our proposal that chromatin repression is established early in development and is maintained in the placenta. In the embryo, however, imprinting is stably maintained only at genes that have promoter DNA methylation. These data underscore the importance of histone methylation in placental imprinting and identify mechanistic similarities with X-chromosome inactivation in extraembryonic tissues, suggesting that the two epigenetic mechanisms are evolutionarily linked.


Asunto(s)
Cromosomas de los Mamíferos/genética , Epigénesis Genética/genética , Impresión Genómica/genética , Histonas/metabolismo , Ratones/genética , Placenta , Canales de Potasio con Entrada de Voltaje/genética , Animales , Cromatina/genética , Cruzamientos Genéticos , Femenino , Expresión Génica , Histonas/genética , Inmunoprecipitación , Canales de Potasio KCNQ , Canal de Potasio KCNQ1 , Metilación , Modelos Biológicos , Proteínas del Grupo Polycomb , Polimorfismo Conformacional Retorcido-Simple , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA