Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Biol Reprod ; 90(5): 98, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24571989

RESUMO

Preterm birth involves the interaction of societal and environmental factors potentially modulating the length of gestation via the epigenome. An established form of epigenetic regulation is DNA methylation where promoter hypermethylation is associated with gene repression. We hypothesized we would find differences in DNA methylation in the myometrium of women with preterm labor of different phenotypes versus normal term labor. Myometrial tissue was obtained at cesarean section at term with or without labor, preterm without labor, idiopathic preterm labor, and twin gestations with labor. Genomic DNA was isolated, and samples in each group were combined and analyzed on a NimbleGen 2.1M human DNA methylation array. Differences in methylation from -8 to +3 kb of transcription start sites of 22 contraction-associated genes were determined. Cytosine methylation was not present in CpG islands of any gene but was present outside of CpG islands in shores and shelves in 19 genes. No differential methylation was found across the tissue groups for six genes (PTGES3L, PTGER2, PTGER4, PTGFRN, ESR2, and GJA1). For 13 genes, differential methylation occurred in several patterns between tissue groups. We find a correlation between hypomethylation and increased mRNA expression of PTGES/mPGES-1, indicating potential functional relevance of methylation, but no such correlation for PTGS2/COX-2, suggesting other regulatory mechanisms for PTGS2 at labor. The majority of differential DNA methylation of myometrial contraction-associated genes with different labor phenotypes occurs outside of CpG islands in gene promoters, suggesting that the entirety of DNA methylation across the genome should be considered.


Assuntos
Metilação de DNA , Miométrio/fisiologia , Trabalho de Parto Prematuro/genética , Contração Uterina/genética , DNA/química , DNA/genética , Epigênese Genética , Feminino , Humanos , Imunoprecipitação , Recém-Nascido , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Regiões Promotoras Genéticas , RNA/química , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Nature ; 450(7171): 908-12, 2007 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-17994007

RESUMO

DNA methyltransferase (cytosine-5) 1 (Dnmt1) is the principal enzyme responsible for maintenance of CpG methylation and is essential for the regulation of gene expression, silencing of parasitic DNA elements, genomic imprinting and embryogenesis. Dnmt1 is needed in S phase to methylate newly replicated CpGs occurring opposite methylated ones on the mother strand of the DNA, which is essential for the epigenetic inheritance of methylation patterns in the genome. Despite an intrinsic affinity of Dnmt1 for such hemi-methylated DNA, the molecular mechanisms that ensure the correct loading of Dnmt1 onto newly replicated DNA in vivo are not understood. The Np95 (also known as Uhrf1 and ICBP90) protein binds methylated CpG through its SET and RING finger-associated (SRA) domain. Here we show that localization of mouse Np95 to replicating heterochromatin is dependent on the presence of hemi-methylated DNA. Np95 forms complexes with Dnmt1 and mediates the loading of Dnmt1 to replicating heterochromatic regions. By using Np95-deficient embryonic stem cells and embryos, we show that Np95 is essential in vivo to maintain global and local DNA methylation and to repress transcription of retrotransposons and imprinted genes. The link between hemi-methylated DNA, Np95 and Dnmt1 thus establishes key steps of the mechanism for epigenetic inheritance of DNA methylation.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA/metabolismo , Epigênese Genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT , Ilhas de CpG/genética , DNA/química , DNA (Citosina-5-)-Metiltransferase 1 , Replicação do DNA , Células-Tronco Embrionárias/metabolismo , Impressão Genômica , Células HeLa , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Camundongos , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Estrutura Terciária de Proteína , Retroelementos/genética , Transcrição Gênica , Ubiquitina-Proteína Ligases
3.
Nat Genet ; 36(12): 1291-5, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15516931

RESUMO

Imprinted genes are expressed from only one of the parental chromosomes and are marked epigenetically by DNA methylation and histone modifications. The imprinting center 2 (IC2) on mouse distal chromosome 7 is flanked by several paternally repressed genes, with the more distant ones imprinted exclusively in the placenta. We found that most of these genes lack parent-specific DNA methylation, and genetic ablation of methylation does not lead to loss of their imprinting in the trophoblast (placenta). The silent paternal alleles of the genes are marked in the trophoblast by repressive histone modifications (dimethylation at Lys9 of histone H3 and trimethylation at Lys27 of histone H3), which are disrupted when IC2 is deleted, leading to reactivation of the paternal alleles. Thus, repressive histone methylation is recruited by IC2 (potentially through a noncoding antisense RNA) to the paternal chromosome in a region of at least 700 kb and maintains imprinting in this cluster in the placenta, independently of DNA methylation. We propose that an evolutionarily older imprinting mechanism limited to extraembryonic tissues was based on histone modifications, and that this mechanism was subsequently made more stable for use in embryonic lineages by the recruitment of DNA methylation.


Assuntos
Cromossomos de Mamíferos/genética , Epigênese Genética/genética , Impressão Genômica/genética , Camundongos/genética , Modelos Biológicos , Placenta , Animais , Southern Blotting , Cromatina/genética , Ilhas de CpG/genética , Metilação de DNA , Feminino , Histonas/metabolismo , Imunoprecipitação , Metilação , Camundongos Mutantes , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Cell Rep ; 42(9): 113067, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37659081

RESUMO

Tumor-associated macrophages (TAMs) are integral to the development of complex tumor microenvironments (TMEs) and can execute disparate cellular programs in response to extracellular cues. However, upstream signaling processes underpinning this phenotypic plasticity remain to be elucidated. Here, we report that concordant AXL-STAT3 signaling in TAMs is triggered by lung cancer cells or cancer-associated fibroblasts in the cytokine milieu. This paracrine action drives TAM differentiation toward a tumor-promoting "M2-like" phenotype with upregulation of CD163 and putative mesenchymal markers, contributing to TAM heterogeneity and diverse cellular functions. One of the upregulated markers, CD44, mediated by AXL-IL-11-pSTAT3 signaling cascade, enhances macrophage ability to interact with endothelial cells and facilitate formation of primitive vascular networks. We also found that AXL-STAT3 inhibition can impede the recruitment of TAMs in a xenograft mouse model, thereby suppressing tumor growth. These findings suggest the potential application of AXL-STAT3-related markers to quantitatively assess metastatic potential and inform therapeutic strategies in lung cancer.


Assuntos
Neoplasias Pulmonares , Macrófagos Associados a Tumor , Humanos , Animais , Camundongos , Células Endoteliais , Transdução de Sinais , Diferenciação Celular , Microambiente Tumoral , Linhagem Celular Tumoral
5.
Cell Rep ; 38(2): 110220, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021081

RESUMO

The epigenome delineates lineage-specific transcriptional programs and restricts cell plasticity to prevent non-physiological cell fate transitions. Although cell diversification fosters tumor evolution and therapy resistance, upstream mechanisms that regulate the stability and plasticity of the cancer epigenome remain elusive. Here we show that 2-hydroxyglutarate (2HG) not only suppresses DNA repair but also mediates the high-plasticity chromatin landscape. A combination of single-cell epigenomics and multi-omics approaches demonstrates that 2HG disarranges otherwise well-preserved stable nucleosome positioning and promotes cell-to-cell variability. 2HG induces loss of motif accessibility to the luminal-defining transcriptional factors FOXA1, FOXP1, and GATA3 and a shift from luminal to basal-like gene expression. Breast tumors with high 2HG exhibit enhanced heterogeneity with undifferentiated epigenomic signatures linked to adverse prognosis. Further, ascorbate-2-phosphate (A2P) eradicates heterogeneity and impairs growth of high 2HG-producing breast cancer cells. These findings suggest 2HG as a key determinant of cancer plasticity and provide a rational strategy to counteract tumor cell evolution.


Assuntos
Cromatina/metabolismo , Glutaratos/metabolismo , Oxirredutases do Álcool/metabolismo , Ácido Ascórbico/análogos & derivados , Ácido Ascórbico/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Reparo do DNA/fisiologia , Epigenoma/genética , Fatores de Transcrição Forkhead/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Humanos , Isocitrato Desidrogenase/genética , Neoplasias/genética , Neoplasias/metabolismo , Nucleossomos/metabolismo , Proteínas Repressoras/genética
6.
Cancers (Basel) ; 12(9)2020 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-32971831

RESUMO

The interplay between glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) is central to maintain energy homeostasis. It remains to be determined whether there is a mechanism governing metabolic fluxes based on substrate availability in microenvironments. Here we show that menin is a key transcription factor regulating the expression of OXPHOS and glycolytic genes in cancer cells and primary tumors with poor prognosis. A group of menin-associated proteins (MAPs), including KMT2A, MED12, WAPL, and GATA3, is found to restrain menin's full function in this transcription regulation. shRNA knockdowns of menin and MAPs result in reduced ATP production with proportional alterations of cellular energy generated through glycolysis and OXPHOS. When shRNA knockdown cells are exposed to metabolic stress, the dual functionality can clearly be distinguished among these metabolic regulators. A MAP can negatively counteract the regulatory mode of menin for OXPHOS while the same protein positively influences glycolysis. A close-proximity interaction between menin and MAPs allows transcriptional regulation for metabolic adjustment. This coordinate regulation by menin and MAPs is necessary for cells to rapidly adapt to fluctuating microenvironments and to maintain essential metabolic functions.

7.
BMC Med Genomics ; 13(1): 69, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-32408897

RESUMO

BACKGROUND: Chromothripsis is an event of genomic instability leading to complex chromosomal alterations in cancer. Frequent long-range chromatin interactions between transcription factors (TFs) and targets may promote extensive translocations and copy-number alterations in proximal contact regions through inappropriate DNA stitching. Although studies have proposed models to explain the initiation of chromothripsis, few discussed how TFs influence this process for tumor progression. METHODS: This study focused on genomic alterations in amplification associated regions within chromosome 17. Inter-/intra-chromosomal rearrangements were analyzed using whole genome sequencing data of breast tumors in the Cancer Genome Atlas (TCGA) cohort. Common ERα binding sites were defined based on MCF-7, T47D, and MDA-MB-134 breast cancer cell lines using univariate K-means clustering methods. Nanopore sequencing technology was applied to validate frequent rearrangements detected between ATC loci on 17q23 and an ERα hub on 20q13. The efficacy of pharmacological inhibition of a potentially druggable target gene on 17q23 was evaluated using breast cancer cell lines and patient-derived circulating breast tumor cells. RESULTS: There are five adjoining regions from 17q11.1 to 17q24.1 being hotspots of chromothripsis. Inter-/intra-chromosomal rearrangements of these regions occurred more frequently in ERα-positive tumors than in ERα-negative tumors. In addition, the locations of the rearrangements were often mapped within or close to dense ERα binding sites localized on these five 17q regions or other chromosomes. This chromothriptic event was linked to concordant upregulation of 96 loci that predominantly regulate cell-cycle machineries in advanced luminal tumors. Genome-editing analysis confirmed that an ERα hub localized on 20q13 coordinately regulates a subset of these loci localized on 17q23 through long-range chromosome interactions. One of these loci, Tousled Like Kinase 2 (TLK2) known to participate in DNA damage checkpoint control, is an actionable target using phenothiazine antipsychotics (PTZs). The antiproliferative effect of PTZs was prominent in high TLK2-expressing cells, compared to low expressing cells. CONCLUSION: This study demonstrates a new approach for identifying tumorigenic drivers from genomic regions highly susceptible to ERα-related chromothripsis. We found a group of luminal breast tumors displaying 17q-related chromothripsis for which antipsychotics can be repurposed as treatment adjuncts.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/genética , Cromossomos Humanos Par 17/genética , Cromotripsia , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica , Biomarcadores Tumorais/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular , Proliferação de Células , Receptor alfa de Estrogênio/genética , Feminino , Humanos , Prognóstico , Taxa de Sobrevida , Transcrição Gênica , Células Tumorais Cultivadas , Sequenciamento do Exoma , Sequenciamento Completo do Genoma
8.
Mol Cell Biol ; 24(13): 5650-6, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15199123

RESUMO

Igf2 and H19 are reciprocally imprinted genes on mouse distal chromosome 7. They share several regulatory elements, including a differentially methylated region (DMR) upstream of H19 that is paternally methylated throughout development. The cis-acting sequence requirements for targeting DNA methylation to the DMR remain unknown; however, it has been suggested that direct tandem repeats near DMRs could be involved. Previous studies of the imprinted Rasgrf1 locus demonstrate indeed that a direct repeat element adjacent to a DMR is responsible for establishing paternal allele-specific methylation at the DMR and therefore allelic expression of the Rasgrf1 transcript. We identified a prominent and conserved direct tandem repeat 1 kb upstream of the H19 DMR and proposed that it played a similar role in imprinted regulation of H19. To test our hypothesis, we generated mice harboring a 1.7-kb targeted deletion of the direct repeat element and analyzed fetal growth, allelic expression, and methylation within the Igf2-H19 region. Surprisingly the deletion had no effect on imprinting. These results together with deletions of other repeats close to imprinted genes suggest that direct repeats may not be important for the targeting of methylation at the majority of imprinted loci and that the Rasgrf1 locus may be an exception to this rule.


Assuntos
Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , RNA não Traduzido/genética , Sequências de Repetição em Tandem/fisiologia , Animais , Células Cultivadas , Mapeamento Cromossômico , Sequência Conservada , Metilação de DNA , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário e Fetal/genética , Camundongos , Camundongos Knockout , Proteínas/genética , RNA Longo não Codificante , Sequências Reguladoras de Ácido Nucleico , Deleção de Sequência , Células-Tronco , Sequências de Repetição em Tandem/genética
9.
PLoS One ; 12(10): e0186115, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29045485

RESUMO

The inflammatory and metabolic derangements of obesity in pregnant women generate an adverse intrauterine environment, increase pregnancy complications and adverse fetal outcomes and program the fetus for obesity and metabolic syndrome in later life. We hypothesized that epigenetic modifications in placenta including altered DNA methylation/hydroxymethylation may mediate these effects. Term placental villous tissue was collected following cesarean section from lean (prepregnancy BMI<25) or obese (BMI>30) women. Genomic DNA was isolated, methylated and hydroxymethylated DNA immunoprecipitated and hybridized to the NimbleGen 2.1M human DNA methylation array. Intermediate metabolites in placental tissues were measured by HPLC-ESI-MS, ascorbate levels by reverse phase HPLC and gene expression by RT-PCR. Differentially methylated and hydroxymethylated regions occurred across the genome, with a 21% increase in methylated but a 31% decrease in hydroxymethylated regions in obese vs lean groups. Whereas increased methylation and decreased methylation was evident around transcription start sites of multiple genes in the GH/CSH and PSG gene clusters on chromosomes 17 and 19 in other areas there was no relationship. Increased methylation was associated with decreased expression only for some genes in these clusters. Biological pathway analysis revealed the 262 genes which showed reciprocal differential methylation/ hydroxymethylation were enriched for pregnancy, immune response and cell adhesion-linked processes. We found a negative relationship for maternal BMI but a positive relationship for ascorbate with α-ketoglutarate a metabolite that regulates ten eleven translocase (TET) which mediates DNA methylation. We provide evidence for the obese maternal metabolic milieu being linked to an altered DNA methylome that may affect placental gene expression in relation to adverse outcomes.


Assuntos
Metilação de DNA/genética , Obesidade/genética , Obesidade/metabolismo , Placenta/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Adiposidade/genética , Índice de Massa Corporal , Epigênese Genética , Feminino , Genoma Humano , Humanos , Metaboloma/genética , Família Multigênica , Gravidez , Magreza/genética
10.
Science ; 332(6031): 848-52, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21566194

RESUMO

Genomic imprinting causes parental origin-specific monoallelic gene expression through differential DNA methylation established in the parental germ line. However, the mechanisms underlying how specific sequences are selectively methylated are not fully understood. We have found that the components of the PIWI-interacting RNA (piRNA) pathway are required for de novo methylation of the differentially methylated region (DMR) of the imprinted mouse Rasgrf1 locus, but not other paternally imprinted loci. A retrotransposon sequence within a noncoding RNA spanning the DMR was targeted by piRNAs generated from a different locus. A direct repeat in the DMR, which is required for the methylation and imprinting of Rasgrf1, served as a promoter for this RNA. We propose a model in which piRNAs and a target RNA direct the sequence-specific methylation of Rasgrf1.


Assuntos
Metilação de DNA , Impressão Genômica , RNA Interferente Pequeno/genética , RNA não Traduzido/genética , ras-GRF1/genética , Animais , Proteínas Argonautas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Modelos Genéticos , Mutação , Fosfolipase D/genética , Fosfolipase D/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA Interferente Pequeno/metabolismo , RNA não Traduzido/metabolismo , Sequências Repetitivas de Ácido Nucleico , Retroelementos , Espermatogônias/metabolismo , Testículo/embriologia , Testículo/metabolismo , Transcrição Gênica
11.
Nucleic Acids Symp Ser (Oxf) ; (51): 125-6, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18029618

RESUMO

Biological responses to environmental effects are mediated through epigenetic changes such as chemical modifications of the histone tails and DNA (5-cytosine) methylation. We report that dietary protein restriction in pregnant mice can alter histone modifications in the promoter region of the Igf2 gene and cause a two-fold repression in promoter specific Igf2 transcription in the liver of the fetus. Suppression of Igf2 is accompanied with low birth weight of the pups born to the protein-restricted dams. Our results provide new information about the epigenetic aspects of early life programming and will improve our understanding about the developmental origins of adult diseases.


Assuntos
Epigênese Genética , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Histonas/química , Fator de Crescimento Insulin-Like II/genética , Animais , Feminino , Peso Fetal , Privação de Alimentos , Histonas/metabolismo , Fator de Crescimento Insulin-Like II/biossíntese , Camundongos , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo
12.
J Hum Genet ; 50(3): 124-132, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15744456

RESUMO

Human chromosome 15q11-q13 involves a striking imprinted gene cluster of more than 2 Mb that is concomitant with multiple neurological disorders manifested by Prader-Willi syndrome (PWS) and Angelman syndrome (AS). PWS and AS patients with imprinting mutation have microdeletions, which share a 4.3 kb short region of overlap (SRO) at the 5' end of the paternal SNURF-SNRPN gene in PWS, or on the maternal allele, which shares a 880 bp SRO located at the 35 kb upstream of the SNURF-SNRPN promoter in AS. Recent studies have revealed an essential role of PWS-SRO in the postzygotic maintenance of the appropriate epigenotype on the paternal chromosome. For AS-SRO, however, there is insufficient experimental evidence exists to determine the direct functions. Here we show that the complete deletion of AS-SRO does not cause any anomalies of imprinted gene expression or DNA methylation on the mutated human chromosome 15, further supporting the idea that AS-SRO is dispensable for post implantation imprint maintenance. This implies that AS-SRO is not essential for the robust epigenotype preservation in somatic cells.


Assuntos
Síndrome de Angelman/genética , Cromossomos Humanos Par 15/genética , Regulação da Expressão Gênica , Impressão Genômica/genética , Proteínas Nucleares/genética , Animais , Southern Blotting , Células Cultivadas , Análise Citogenética , Metilação de DNA , Primers do DNA , Deleção de Genes , Técnicas de Transferência de Genes , Humanos , Camundongos , Mutação/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Philos Trans R Soc Lond B Biol Sci ; 358(1436): 1403-9; discussion 1409, 2003 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-14511488

RESUMO

Epigenetic asymmetry between parental genomes and embryonic lineages exists at the earliest stages of mammalian development. The maternal genome in the zygote is highly methylated in both its DNA and its histones and most imprinted genes have maternal germline methylation imprints. The paternal genome is rapidly remodelled with protamine removal, addition of acetylated histones, and rapid demethylation of DNA before replication. A minority of imprinted genes have paternal germline methylation imprints. Methylation and chromatin reprogramming continues during cleavage divisions, but at the blastocyst stage lineage commitment to inner cell mass (ICM) or trophectoderm (TE) fate is accompanied by a dramatic increase in DNA and histone methylation, predominantly in the ICM. This may set up major epigenetic differences between embryonic and extraembryonic tissues, including in X-chromosome inactivation and perhaps imprinting. Maintaining epigenetic asymmetry appears important for development as asymmetry is lost in cloned embryos, most of which have developmental defects, and in particular an imbalance between extraembryonic and embryonic tissue development.


Assuntos
Blastocisto/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Metilação de DNA , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Impressão Genômica/fisiologia , Animais , Blastocisto/citologia , Cromatina/fisiologia , Mecanismo Genético de Compensação de Dose , Feminino , Histonas/fisiologia , Masculino , Mamíferos
14.
Hum Mol Genet ; 12(23): 3123-32, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14532328

RESUMO

Genomic imprinting in mammals marks the parental alleles in gametes, resulting in differential gene expression in offspring. A number of epigenetic features are associated with imprinted genes. These include differential DNA methylation, histone acetylation and methylation, subnuclear localization and DNA replication timing. While DNA methylation has been shown to be necessary both for establishment and maintenance of imprinting, the connections with the other types of epigenetic marking systems are not clear. Specifically, it is not known whether the other marking systems, either on their own or in conjunction with DNA methylation, are required for imprinting. Here we show that in the mouse mutant Minute (Mnt) the Igf2-H19 locus acquires a paternal methylation imprint in the maternal germline. DNA methylation of the H19 DMR is established in oogenesis, maintained during postzygotic development on the maternal allele, and erased in primordial germ cells. The fact that a paternal type methylation imprint can also be established in the maternal germline indicates that trans-acting factors that target methylation to this imprinted region are likely to be the same in both germlines. Surprisingly, however, asynchrony of DNA replication of the locus is maintained despite the altered expression and methylation imprint of Igf2 and H19. These results show clearly that replication asynchrony of this region is neither the determinant factor for, nor a consequence of, epigenetic modifications that are critical for genomic imprinting. Replication asynchrony may thus be regulated differently from methylation imprints and have a separate function.


Assuntos
Período de Replicação do DNA , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , RNA não Traduzido/genética , Alelos , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Metilação de DNA , Epigênese Genética , Feminino , Células Germinativas/citologia , Masculino , Camundongos , RNA Longo não Codificante , Proteínas Repressoras/genética
15.
Genomics ; 83(3): 402-12, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14962666

RESUMO

MEST is one of the imprinted genes in human. With the assistance of our integration map and the complete sequence in the registry, we mapped a total of 16 genes/transcripts at the 1.5-Mb MEST-flanking region at 7q32. This region has been suggested to form an imprinted gene cluster, because MEST and its three flanking genes/transcripts (MESTIT1, CPA4, and COPG2IT1) were reported to be imprinted, although two (TSGA14 and COPG2) were shown to escape imprinting. In this study, 10 other genes/transcripts were examined for their imprinting status in human fetal tissues. The results indicated that 8 genes/transcripts (NRF1, UBE2H, HSPC216, KIAA0265, FLJ14803, CPA2, CPA1, and DKFZp667F0312) were expressed biallelically. The imprinting status of two (TSGA13 and CPA5) was not conclusive, because of their weak and/or tissue-specific expression and inconstant results. These findings provided evidence that only 4 of the 16 genes/transcripts located to the region show monoallelic expression, while others are not involved in imprinting. Therefore, it is less likely that the MEST-flanking 7q32 region forms a large imprinted domain.


Assuntos
Cromossomos Humanos Par 7 , Genes , Impressão Genômica , Proteínas/genética , Adulto , Alelos , Animais , Composição de Bases , Feminino , Perfilação da Expressão Gênica , Genoma Humano , Humanos , Células Híbridas , Masculino , Camundongos , Família Multigênica , Mapeamento Físico do Cromossomo , Enzimas de Conjugação de Ubiquitina/genética
16.
Hum Mol Genet ; 11(15): 1743-56, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12095916

RESUMO

Imprinted gene(s) on human chromosome 7 are thought to be involved in Russell-Silver syndrome (RSS), based on the fact that approximately 10% of patients have maternal uniparental disomy of chromosome 7. However, involvement of the known imprinted genes (GRB10 at 7p12, PEG10 at 7q21.3 and MEST at 7q32) in RSS has yet to be established. To screen for new imprinted genes, we are initially using somatic cell hybrids containing a paternal or maternal human chromosome 7. Transcripts located between D7S530 and D7S649 (a 1.5 Mb interval encompassing MEST ) were subjected to RT-PCR analysis using somatic cell hybrids. One transcript named MESTIT1 (for MEST intronic transcript 1) reproducibly showed paternal-specific expression. Upon further analysis, we found MESTIT1 to be (1) paternally (and not maternally) expressed in all fetal tissues and fibroblasts examined, (2) to be located in an intron of one of the two isoforms of MEST but transcribed in the opposite direction, (3) to be composed of at least two exons without any significant open reading frame, and (4) to exist as a 4.2 kb transcript in many fetal and adult tissues. We could also identify two isoforms of the mouse Mest gene as observed in humans, but it is still unknown if a murine ortholog of MESTIT1 exists. We also examined the imprinting status of MEST isoforms as a first step in assessing whether MESTIT1 might influence the allelic expression pattern of the sense transcript. MEST isoform 1 was determined to be exclusively expressed from the paternal allele in all fetal tissues and cell lines examined, whereas MEST isoform 2 was only preferentially expressed from the paternal allele in a tissue/cell-type-specific manner. Our results suggest that MESTIT1 is a paternally expressed non-coding RNA that may be involved in the regulation of MEST expression during development. MESTIT1 (also known as PEG1-AS) is now the third independent transcript (with MEST and COPG2IT1) identified at human chromosome 7q32 demonstrating paternal chromosome-specific expression.


Assuntos
Cromossomos Humanos Par 7 , Impressão Genômica , Proteínas/genética , RNA Antissenso , Northern Blotting , Linhagem Celular , DNA Complementar , Fibroblastos , Humanos , Dados de Sequência Molecular , Isoformas de Proteínas , Análise de Sequência de DNA
17.
Genomics ; 79(2): 186-96, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11829489

RESUMO

Russell-Silver syndrome (RSS) is a form of congenital short stature characterized by severe growth retardation and variable dysmorphic features. In some RSS individuals, alterations in imprinted genes may be involved because approximately 7% of sporadic patients have been observed to have maternal uniparental disomy (mUPD) of chromosome 7. RSS patients with structural abnormalities of chromosome 7 have also been described. In these individuals the chromosome rearrangement could disrupt the balance of imprinted genes, contribute to a recessive form of RSS, or lead to haploinsufficiency of a crucial developmental gene product. Because the mechanism and molecular defects on chromosome 7 causing RSS are still unknown, we tested our collection of 77 RSS families for mUPD7 and were able to identify three new cases. We also characterized two RSS patients with de novo cytogenetic abnormalities involving the short arm of chromosome 7. One had a partial duplication [46, XX, dup(7)(p12 p14)] and the second contained a paracentric inversion [46, XY, inv(7)(p14 p21)]. Fluorescence in situ hybridization (FISH) mapping revealed that the breakpoints on 7p14 were localized to the same novel gene, C7orf10, which encompasses >700 kb of DNA. We also identified other transcription units from this immediate region, but all seem to be biallelically expressed when using a somatic cell hybrid assay.


Assuntos
Anormalidades Múltiplas/genética , Cromossomos Humanos Par 7 , Sequência de Aminoácidos , Animais , Estatura/genética , Aberrações Cromossômicas , Análise Citogenética , Fácies , Humanos , Células Híbridas , Hibridização in Situ Fluorescente , Camundongos , Dados de Sequência Molecular , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Síndrome
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa