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











Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(3): e0265027, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35255108

RESUMO

Epigenetic mechanisms regulate osteogenic lineage differentiation of mesenchymal stromal cells. Histone methylation is controlled by multiple lysine demethylases and is an important step in controlling local chromatin structure and gene expression. Here, we show that the lysine-specific histone demethylase Kdm1A/Lsd1 is abundantly expressed in osteoblasts and that its suppression impairs osteoblast differentiation and bone nodule formation in vitro. Although Lsd1 knockdown did not affect global H3K4 methylation levels, genome-wide ChIP-Seq analysis revealed high levels of Lsd1 at gene promoters and its binding was associated with di- and tri-methylation of histone 3 at lysine 4 (H3K4me2 and H3K4me3). Lsd1 binding sites in osteoblastic cells were enriched for the Runx2 consensus motif suggesting a functional link between the two proteins. Importantly, inhibition of Lsd1 activity decreased osteoblast activity in vivo. In support, mesenchymal-targeted knockdown of Lsd1 led to decreased osteoblast activity and disrupted primary spongiosa ossification and reorganization in vivo. Together, our studies demonstrate that Lsd1 occupies Runx2-binding cites at H3K4me2 and H3K4me3 and its activity is required for proper bone formation.


Assuntos
Histonas , Osteogênese , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Osteoblastos/metabolismo , Osteogênese/genética
2.
Cell Rep ; 38(9): 110457, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35235780

RESUMO

Bifurcation of cellular fates, a critical process in development, requires histone 3 lysine 27 methylation (H3K27me3) marks propagated by the polycomb repressive complex 2 (PRC2). However, precise chromatin loci of functional H3K27me3 marks are not yet known. Here, we identify critical PRC2 functional sites at high resolution. We fused a computationally designed protein, EED binder (EB), which competes with EZH2 and thereby inhibits PRC2 function, to dCas9 (EBdCas9) to allow for PRC2 inhibition at a precise locus using gRNA. Targeting EBdCas9 to four different genes (TBX18, p16, CDX2, and GATA3) results in precise H3K27me3 and EZH2 reduction, gene activation, and functional outcomes in the cell cycle (p16) or trophoblast transdifferentiation (CDX2 and GATA3). In the case of TBX18, we identify a PRC2-controlled, functional TATA box >500 bp upstream of the TBX18 transcription start site (TSS) using EBdCas9. Deletion of this TATA box eliminates EBdCas9-dependent TATA binding protein (TBP) recruitment and transcriptional activation. EBdCas9 technology may provide a broadly applicable tool for epigenomic control of gene regulation.


Assuntos
Histonas , Complexo Repressor Polycomb 2 , Cromatina , Computadores , Histonas/metabolismo , Complexo Repressor Polycomb 2/metabolismo , TATA Box
3.
Blood Adv ; 5(4): 1122-1135, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33620431

RESUMO

Base editors are capable of installing precise genomic alterations without creating double-strand DNA breaks. In this study, we targeted critical motifs regulating γ-globin reactivation with base editors delivered via HDAd5/35++ vectors. Through optimized design, we successfully produced a panel of cytidine and adenine base editor (ABE) vectors targeting the erythroid BCL11A enhancer or recreating naturally occurring hereditary persistence of fetal hemoglobin (HPFH) mutations in the HBG1/2 promoter. All 5 tested vectors efficiently installed target base conversion and led to γ-globin reactivation in human erythroid progenitor cells. We observed ~23% γ-globin protein production over ß-globin, when using an ABE vector (HDAd-ABE-sgHBG-2) specific to the -113A>G HPFH mutation. In a ß-YAC mouse model, in vivo hematopoietic progenitor/stem cell (HSPC) transduction with HDAd-ABE-sgHBG-2 followed by in vivo selection resulted in >40% γ-globin+ erythrocytes in the peripheral blood. This result corresponded to 21% γ-globin production over human ß-globin. The average -113A>G conversion in total bone marrow cells was 20%. No alterations in hematological parameters, erythropoiesis, and bone marrow cellular composition were observed after treatment. No detectable editing was found at top-scoring, off-target genomic sites. Bone marrow lineage-negative cells from primary mice were capable of reconstituting secondary transplant-recipient mice with stable γ-globin expression. Importantly, the advantage of base editing over CRISPR/Cas9 was reflected by the markedly lower rates of intergenic HBG1/2 deletion and the absence of detectable toxicity in human CD34+ cells. Our observations suggest that HDAd-vectorized base editors represent a promising strategy for precise in vivo genome engineering for the treatment of ß-hemoglobinopathies.


Assuntos
Hemoglobina Fetal , gama-Globinas , Animais , Hemoglobina Fetal/genética , Terapia Genética , Células-Tronco Hematopoéticas , Camundongos , Globinas beta/genética , gama-Globinas/genética
4.
Mol Ther ; 27(12): 2195-2212, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31494053

RESUMO

Our goal is the development of in vivo hematopoietic stem cell (HSC) transduction technology with targeted integration. To achieve this, we modified helper-dependent HDAd5/35++ vectors to express a CRISPR/Cas9 specific to the "safe harbor" adeno-associated virus integration site 1 (AAVS1) locus and to provide a donor template for targeted integration through homology-dependent repair. We tested the HDAd-CRISPR + HDAd-donor vector system in AAVS1 transgenic mice using a standard ex vivo HSC gene therapy approach as well as a new in vivo HSC transduction approach that involves HSC mobilization and intravenous HDAd5/35++ injections. In both settings, the majority of treated mice had transgenes (GFP or human γ-globin) integrated into the AAVS1 locus. On average, >60% of peripheral blood cells expressed the transgene after in vivo selection with low-dose O6BG/bis-chloroethylnitrosourea (BCNU). Ex vivo and in vivo HSC transduction and selection studies with HDAd-CRISPR + HDAd-globin-donor resulted in stable γ-globin expression at levels that were significantly higher (>20% γ-globin of adult mouse globin) than those achieved in previous studies with a SB100x-transposase-based HDAd5/35++ system that mediates random integration. The ability to achieve therapeutically relevant transgene expression levels after in vivo HSC transduction and selection and targeted integration make our HDAd5/35++-based vector system a new tool in HSC gene therapy.


Assuntos
Adenoviridae/genética , Dependovirus/genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Transdução Genética , Transgenes/fisiologia , Integração Viral , Animais , Sistemas CRISPR-Cas , Feminino , Genes Reporter , Terapia Genética , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , gama-Globinas/antagonistas & inibidores , gama-Globinas/genética
5.
Semin Cell Dev Biol ; 96: 32-43, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31112800

RESUMO

Recent developments in the nucleic acid editing technologies have provided a powerful tool to precisely engineer the genome and epigenome for studying many aspects of immune cell differentiation and development as well as several immune mediated diseases (IMDs) including autoimmunity and cancer. Here, we discuss the recent technological achievements of the CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)-based RNA-guided genome and epigenome editing toolkit and provide an insight into how CRISPR/Cas9 (CRISPR Associated Protein 9) toolbox could be used to examine genetic and epigenetic mechanisms underlying IMDs. In addition, we will review the progress in CRISPR/Cas9-based genome-wide genome and epigenome screens in various cell types including immune cells. Finally, we will discuss the potential of CRISPR/Cas9 in defining the molecular function of disease associated SNPs overlapping gene regulatory elements.


Assuntos
Sistemas CRISPR-Cas/genética , Epigênese Genética/genética , Engenharia Genética , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/genética , Animais , Humanos
6.
Blood ; 131(26): 2915-2928, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29789357

RESUMO

Disorders involving ß-globin gene mutations, primarily ß-thalassemia and sickle cell disease, represent a major target for hematopoietic stem/progenitor cell (HSPC) gene therapy. This includes CRISPR/Cas9-mediated genome editing approaches in adult CD34+ cells aimed toward the reactivation of fetal γ-globin expression in red blood cells. Because models involving erythroid differentiation of CD34+ cells have limitations in assessing γ-globin reactivation, we focused on human ß-globin locus-transgenic (ß-YAC) mice. We used a helper-dependent human CD46-targeting adenovirus vector expressing CRISPR/Cas9 (HDAd-HBG-CRISPR) to disrupt a repressor binding region within the γ-globin promoter. We transduced HSPCs from ß-YAC/human CD46-transgenic mice ex vivo and subsequently transplanted them into irradiated recipients. Furthermore, we used an in vivo HSPC transduction approach that involves HSPC mobilization and the intravenous injection of HDAd-HBG-CRISPR into ß-YAC/CD46-transgenic mice. In both models, we demonstrated efficient target site disruption, resulting in a pronounced switch from human ß- to γ-globin expression in red blood cells of adult mice that was maintained after secondary transplantation of HSPCs. In long-term follow-up studies, we did not detect hematological abnormalities, indicating that HBG promoter editing does not negatively affect hematopoiesis. This is the first study that shows successful in vivo HSPC genome editing by CRISPR/Cas9.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes/métodos , Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Globinas beta/genética , gama-Globinas/genética , Animais , Eritrócitos/metabolismo , Feminino , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas
7.
Proc Natl Acad Sci U S A ; 114(38): 10125-10130, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28864533

RESUMO

The polycomb repressive complex 2 (PRC2) histone methyltransferase plays a central role in epigenetic regulation in development and in cancer, and hence to interrogate its role in a specific developmental transition, methods are needed for disrupting function of the complex with high temporal and spatial precision. The catalytic and substrate recognition functions of PRC2 are coupled by binding of the N-terminal helix of the Ezh2 methylase to an extended groove on the EED trimethyl lysine binding subunit. Disrupting PRC2 function can in principle be achieved by blocking this single interaction, but there are few approaches for blocking specific protein-protein interactions in living cells and organisms. Here, we describe the computational design of proteins that bind to the EZH2 interaction site on EED with subnanomolar affinity in vitro and form tight and specific complexes with EED in living cells. Induction of the EED binding proteins abolishes H3K27 methylation in human embryonic stem cells (hESCs) and at all but the earliest stage blocks self-renewal, pinpointing the first critical repressive H3K27me3 marks in development.


Assuntos
Simulação por Computador , Histonas/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Histonas/química , Células-Tronco Embrionárias Humanas/citologia , Humanos , Metilação , Complexo Repressor Polycomb 2/química
8.
Cell Rep ; 20(6): 1448-1462, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28793267

RESUMO

We currently lack a comprehensive understanding of the mechanisms underlying neural tube formation and their contributions to neural tube defects (NTDs). Developing a model to study such a complex morphogenetic process, especially one that models human-specific aspects, is critical. Three-dimensional, human embryonic stem cell (hESC)-derived neural rosettes (NRs) provide a powerful resource for in vitro modeling of human neural tube formation. Epigenomic maps reveal enhancer elements unique to NRs relative to 2D systems. A master regulatory network illustrates that key NR properties are related to their epigenomic landscapes. We found that folate-associated DNA methylation changes were enriched within NR regulatory elements near genes involved in neural tube formation and metabolism. Our comprehensive regulatory maps offer insights into the mechanisms by which folate may prevent NTDs. Lastly, our distal regulatory maps provide a better understanding of the potential role of neurological-disorder-associated SNPs.


Assuntos
Células-Tronco Embrionárias/citologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Defeitos do Tubo Neural/genética , Tubo Neural/embriologia , Linhagem Celular , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Elementos Facilitadores Genéticos , Humanos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese
9.
Stem Cell Reports ; 9(3): 999-1015, 2017 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-28844656

RESUMO

Human bone marrow stromal cells, or mesenchymal stem cells (BM-MSCs), need expansion prior to use as cell-based therapies in immunological and tissue repair applications. Aging and expansion of BM-MSCs induce epigenetic changes that can impact therapeutic outcomes. By applying sequencing-based methods, we reveal that the breadth of DNA methylation dynamics associated with aging and expansion is greater than previously reported. Methylation changes are enriched at known distal transcription factor binding sites such as enhancer elements, instead of CpG-rich regions, and are associated with changes in gene expression. From this, we constructed hypo- and hypermethylation-specific regulatory networks, including a sub-network of BM-MSC master regulators and their predicted target genes, and identified putatively disrupted signaling pathways. Our genome-wide analyses provide a broader overview of age- and expansion-induced DNA methylation changes and a better understanding of the extent to which these changes alter gene expression and functionality of human BM-MSCs.


Assuntos
Células da Medula Óssea/metabolismo , Metilação de DNA/genética , Células-Tronco Mesenquimais/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Sítios de Ligação , Células Cultivadas , Ilhas de CpG/genética , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Genoma Humano , Humanos , Pessoa de Meia-Idade , Análise de Sequência de DNA , Fatores de Transcrição/metabolismo , Adulto Jovem
10.
Nat Commun ; 8: 14454, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28195574

RESUMO

Gene replacement therapies utilizing adeno-associated viral (AAV) vectors hold great promise for treating Duchenne muscular dystrophy (DMD). A related approach uses AAV vectors to edit specific regions of the DMD gene using CRISPR/Cas9. Here we develop multiple approaches for editing the mutation in dystrophic mdx4cv mice using single and dual AAV vector delivery of a muscle-specific Cas9 cassette together with single-guide RNA cassettes and, in one approach, a dystrophin homology region to fully correct the mutation. Muscle-restricted Cas9 expression enables direct editing of the mutation, multi-exon deletion or complete gene correction via homologous recombination in myogenic cells. Treated muscles express dystrophin in up to 70% of the myogenic area and increased force generation following intramuscular delivery. Furthermore, systemic administration of the vectors results in widespread expression of dystrophin in both skeletal and cardiac muscles. Our results demonstrate that AAV-mediated muscle-specific gene editing has significant potential for therapy of neuromuscular disorders.


Assuntos
Sistemas CRISPR-Cas/genética , Distrofina/genética , Edição de Genes/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Animais , Proteínas de Bactérias/genética , Proteína 9 Associada à CRISPR , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Dependovirus/genética , Modelos Animais de Doenças , Endonucleases/genética , Terapia Genética/métodos , Vetores Genéticos , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/terapia , Mutação , Miocárdio , Doenças Neuromusculares/terapia , RNA Guia de Cinetoplastídeos , Deleção de Sequência
11.
Genome Med ; 8(1): 66, 2016 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-27287041

RESUMO

Patterns in expression data conserved across multiple independent disease studies are likely to represent important molecular events underlying the disease. We present the INSPIRE method to infer modules of co-expressed genes and the dependencies among the modules from multiple expression datasets that may contain different sets of genes. We show that INSPIRE infers more accurate models than existing methods to extract low-dimensional representation of expression data. We demonstrate that applying INSPIRE to nine ovarian cancer datasets leads to a new marker and potential driver of tumor-associated stroma, HOPX, followed by experimental validation. The implementation of INSPIRE is available at http://inspire.cs.washington.edu .


Assuntos
Biomarcadores Tumorais/genética , Biologia Computacional/métodos , Proteínas de Homeodomínio/genética , Neoplasias Ovarianas/genética , Proteínas Supressoras de Tumor/genética , Bases de Dados Genéticas , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas Supressoras de Tumor/metabolismo , Aprendizado de Máquina não Supervisionado
12.
Sci Rep ; 6: 22190, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26911679

RESUMO

Epigenomic regulation is likely to be important in the maintenance of genomic integrity of human pluripotent stem cells, however, the mechanisms are unknown. We explored the epigenomes and transcriptomes of human pluripotent stem cells before and after spontaneous transformation to abnormal karyotypes and in correlation to cancer cells. Our results reveal epigenetic silencing of Catalase, a key regulator of oxidative stress and DNA damage control in abnormal cells. Our findings provide novel insight into the mechanisms associated with spontaneous transformation of human pluripotent stem cells towards malignant fate. The same mechanisms may control the genomic stability of cells in somatic tissues.


Assuntos
Cariótipo Anormal , Catalase/genética , Inativação Gênica , Células-Tronco Pluripotentes/metabolismo , Neoplasias Testiculares/genética , Estudos de Casos e Controles , Catalase/metabolismo , Linhagem Celular , Humanos , Masculino , Estresse Oxidativo , Células-Tronco Pluripotentes/enzimologia , Neoplasias Testiculares/metabolismo , Transcriptoma
13.
Nat Cell Biol ; 17(12): 1523-35, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26571212

RESUMO

For nearly a century developmental biologists have recognized that cells from embryos can differ in their potential to differentiate into distinct cell types. Recently, it has been recognized that embryonic stem cells derived from both mice and humans exhibit two stable yet epigenetically distinct states of pluripotency: naive and primed. We now show that nicotinamide N-methyltransferase (NNMT) and the metabolic state regulate pluripotency in human embryonic stem cells (hESCs).  Specifically, in naive hESCs, NNMT and its enzymatic product 1-methylnicotinamide are highly upregulated, and NNMT is required for low S-adenosyl methionine (SAM) levels and the H3K27me3 repressive state. NNMT consumes SAM in naive cells, making it unavailable for histone methylation that represses Wnt and activates the HIF pathway in primed hESCs. These data support the hypothesis that the metabolome regulates the epigenetic landscape of the earliest steps in human development.


Assuntos
Diferenciação Celular , Epigênese Genética/genética , Células-Tronco Embrionárias Humanas/metabolismo , Metaboloma , Animais , Western Blotting , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Perfilação da Expressão Gênica/métodos , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Lisina/metabolismo , Espectrometria de Massas , Metabolômica/métodos , Metilação , Camundongos , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Nicotinamida N-Metiltransferase/genética , Nicotinamida N-Metiltransferase/metabolismo , Proteômica/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , S-Adenosilmetionina/metabolismo , Transdução de Sinais
14.
Nat Biotechnol ; 33(6): 646-55, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25798938

RESUMO

Chromosomal deletions associated with human diseases, such as cancer, are common, but synteny issues complicate modeling of these deletions in mice. We use cellular reprogramming and genome engineering to functionally dissect the loss of chromosome 7q (del(7q)), a somatic cytogenetic abnormality present in myelodysplastic syndromes (MDS). We derive del(7q)- and isogenic karyotypically normal induced pluripotent stem cells (iPSCs) from hematopoietic cells of MDS patients and show that the del(7q) iPSCs recapitulate disease-associated phenotypes, including impaired hematopoietic differentiation. These disease phenotypes are rescued by spontaneous dosage correction and can be reproduced in karyotypically normal cells by engineering hemizygosity of defined chr7q segments in a 20-Mb region. We use a phenotype-rescue screen to identify candidate haploinsufficient genes that might mediate the del(7q)- hematopoietic defect. Our approach highlights the utility of human iPSCs both for functional mapping of disease-associated large-scale chromosomal deletions and for discovery of haploinsufficient genes.


Assuntos
Deleção Cromossômica , Engenharia Genética , Células-Tronco Pluripotentes Induzidas/citologia , Síndromes Mielodisplásicas/genética , Animais , Cromossomos Humanos Par 7/genética , Humanos , Cariotipagem , Camundongos , Síndromes Mielodisplásicas/terapia
15.
Proc Natl Acad Sci U S A ; 111(12): 4484-9, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24623855

RESUMO

The naïve pluripotent state has been shown in mice to lead to broad and more robust developmental potential relative to primed mouse epiblast cells. The human naïve ES cell state has eluded derivation without the use of transgenes, and forced expression of OCT4, KLF4, and KLF2 allows maintenance of human cells in a naïve state [Hanna J, et al. (2010) Proc Natl Acad Sci USA 107(20):9222-9227]. We describe two routes to generate nontransgenic naïve human ES cells (hESCs). The first is by reverse toggling of preexisting primed hESC lines by preculture in the histone deacetylase inhibitors butyrate and suberoylanilide hydroxamic acid, followed by culture in MEK/ERK and GSK3 inhibitors (2i) with FGF2. The second route is by direct derivation from a human embryo in 2i with FGF2. We show that human naïve cells meet mouse criteria for the naïve state by growth characteristics, antibody labeling profile, gene expression, X-inactivation profile, mitochondrial morphology, microRNA profile and development in the context of teratomas. hESCs can exist in a naïve state without the need for transgenes. Direct derivation is an elusive, but attainable, process, leading to cells at the earliest stage of in vitro pluripotency described for humans. Reverse toggling of primed cells to naïve is efficient and reproducible.


Assuntos
Células-Tronco Embrionárias/citologia , Animais , Linhagem da Célula , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Transgenes , Inativação do Cromossomo X
16.
Cell ; 153(5): 1134-48, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23664764

RESUMO

Epigenetic mechanisms have been proposed to play crucial roles in mammalian development, but their precise functions are only partially understood. To investigate epigenetic regulation of embryonic development, we differentiated human embryonic stem cells into mesendoderm, neural progenitor cells, trophoblast-like cells, and mesenchymal stem cells and systematically characterized DNA methylation, chromatin modifications, and the transcriptome in each lineage. We found that promoters that are active in early developmental stages tend to be CG rich and mainly engage H3K27me3 upon silencing in nonexpressing lineages. By contrast, promoters for genes expressed preferentially at later stages are often CG poor and primarily employ DNA methylation upon repression. Interestingly, the early developmental regulatory genes are often located in large genomic domains that are generally devoid of DNA methylation in most lineages, which we termed DNA methylation valleys (DMVs). Our results suggest that distinct epigenetic mechanisms regulate early and late stages of ES cell differentiation.


Assuntos
Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Epigenômica , Regulação da Expressão Gênica no Desenvolvimento , Animais , Diferenciação Celular , Cromatina/metabolismo , Ilhas de CpG , Células-Tronco Embrionárias/citologia , Histonas/metabolismo , Humanos , Metilação , Neoplasias/genética , Regiões Promotoras Genéticas , Peixe-Zebra/embriologia
17.
Genome Res ; 22(2): 246-58, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22156296

RESUMO

While genetic mutation is a hallmark of cancer, many cancers also acquire epigenetic alterations during tumorigenesis including aberrant DNA hypermethylation of tumor suppressors, as well as changes in chromatin modifications as caused by genetic mutations of the chromatin-modifying machinery. However, the extent of epigenetic alterations in cancer cells has not been fully characterized. Here, we describe complete methylome maps at single nucleotide resolution of a low-passage breast cancer cell line and primary human mammary epithelial cells. We find widespread DNA hypomethylation in the cancer cell, primarily at partially methylated domains (PMDs) in normal breast cells. Unexpectedly, genes within these regions are largely silenced in cancer cells. The loss of DNA methylation in these regions is accompanied by formation of repressive chromatin, with a significant fraction displaying allelic DNA methylation where one allele is DNA methylated while the other allele is occupied by histone modifications H3K9me3 or H3K27me3. Our results show a mutually exclusive relationship between DNA methylation and H3K9me3 or H3K27me3. These results suggest that global DNA hypomethylation in breast cancer is tightly linked to the formation of repressive chromatin domains and gene silencing, thus identifying a potential epigenetic pathway for gene regulation in cancer cells.


Assuntos
Neoplasias da Mama/genética , Montagem e Desmontagem da Cromatina , Metilação de DNA , Inativação Gênica , Alelos , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Análise por Conglomerados , Epigênese Genética , Feminino , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Humanos , Modelos Genéticos , Sequências Repetitivas de Ácido Nucleico , Transcrição Gênica
18.
PLoS One ; 2(6): e525, 2007 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-17565378

RESUMO

Loss of inner ear sensory hair cells (HC) is a leading cause of human hearing loss and balance disorders. Unlike mammals, many lower vertebrates can regenerate these cells. We used cross-species microarrays to examine this process in the avian inner ear. Specifically, changes in expression of over 1700 transcription factor (TF) genes were investigated in hair cells of auditory and vestibular organs following treatment with two different damaging agents and regeneration in vitro. Multiple components of seven distinct known signaling pathways were clearly identifiable: TGFbeta, PAX, NOTCH, WNT, NFKappaB, INSULIN/IGF1 and AP1. Numerous components of apoptotic and cell cycle control pathways were differentially expressed, including p27(KIP) and TFs that regulate its expression. A comparison of expression trends across tissues and treatments revealed identical patterns of expression that occurred at identical times during regenerative proliferation. Network analysis of the patterns of gene expression in this large dataset also revealed the additional presence of many components (and possible network interactions) of estrogen receptor signaling, circadian rhythm genes and parts of the polycomb complex (among others). Equal numbers of differentially expressed genes were identified that have not yet been placed into any known pathway. Specific time points and tissues also exhibited interesting differences: For example, 45 zinc finger genes were specifically up-regulated at later stages of cochlear regeneration. These results are the first of their kind and should provide the starting point for more detailed investigations of the role of these many pathways in HC recovery, and for a description of their possible interactions.


Assuntos
Biomarcadores/metabolismo , Galinhas/genética , Células Epiteliais/fisiologia , Perfilação da Expressão Gênica , Células Ciliadas Auditivas Internas/fisiologia , Regeneração/fisiologia , Animais , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Hum Mol Genet ; 12(11): 1261-72, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12761041

RESUMO

The sensory receptors for hearing and balance are the hair cells of the cochlea and vestibular organs of the inner ear. Permanent hearing and balance deficits can be triggered by genetic susceptibilities or environmental factors such as infection. Unlike mammalian hair cells that have a limited capacity for regeneration, the vestibular organ of the avian ear is constantly undergoing hair cell regeneration, whereas the avian cochlea undergoes regeneration only when hair cells are damaged. In order to gain insights into the genetic programs that govern the regenerative capacity of hair cells, we interrogated custom human cDNA microarrays with sensory epithelial cell targets from avian inner ears. The arrays contained probes from conserved regions of approximately 400 genes expressed primarily in the inner ear and approximately 1500 transcription factors (TF). Highly significant differences were observed for 20 inner-ear genes and more than 80 TFs. Genes up-regulated in the cochlea included BMP4, GATA3, GSN, FOXF1 and PRDM7. Genes up-regulated in the utricle included SMAD2, KIT, beta-AMYLOID, LOC51637, HMG20B and CRIP2. Many of the highly significant changes were validated by Q-PCR and in situ methods. Some of the observed changes implicated a number of known biochemical pathways including the c-kit pathway previously observed in melanogenesis. Twenty differentially expressed TFs map to chromosomal regions harboring uncloned human deafness loci, and represent novel candidates for hearing loss. The approach described here also illustrates the power of utilizing conserved human cDNA probes for cross-species comparisons.


Assuntos
Galinhas/genética , Células Epiteliais/fisiologia , Expressão Gênica , Células Ciliadas Auditivas/fisiologia , Fatores de Transcrição/genética , Animais , DNA Complementar , Orelha Interna/fisiologia , Biblioteca Gênica , Transtornos da Audição/genética , Humanos , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Reação em Cadeia da Polimerase/métodos , Regeneração/genética , Transtornos de Sensação/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA