Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genes Cells ; 25(1): 54-64, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31733167

RESUMO

Monoallelic gene expression occurs in various mammalian cells and can be regulated genetically, epigenetically and/or stochastically. We identified 145 monoallelically expressed genes (MoEGs), including seven known imprinted genes, in mouse embryonic stem cells (ESCs) derived from reciprocal F1 hybrid blastocysts and cultured in 2i/LIF. As all MoEGs except for the imprinted genes were expressed in a genetic-origin-dependent manner, we focused on this class of MoEGs for mechanistic studies. We showed that a majority of the genetic-origin-dependent MoEGs identified in 2i/LIF ESCs remain monoallelically expressed in serum/LIF ESCs, but become more relaxed or even biallelically expressed upon differentiation. These MoEGs and their regulatory regions were highly enriched for single nucleotide polymorphisms. In addition, some MoEGs were associated with retrotransposon insertions/deletions, consistent with the fact that certain retrotransposons act as regulatory elements in pluripotent stem cells. Interestingly, most MoEGs showed allelic differences in enrichment of histone H3K27me and H3K4me marks, linking allelic epigenetic differences and monoallelic expression. In contrast, there was little or no allelic difference in CpG methylation or H3K9me. Taken together, our study highlights the impact of genetic variation including single nucleotide polymorphisms and retrotransposon insertions/deletions on monoallelic epigenetic marks and expression in ESCs.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Embrionárias Murinas/metabolismo , Transcriptoma/genética , Alelos , Animais , Diferenciação Celular/genética , Linhagem Celular , Metilação de DNA/genética , Células-Tronco Embrionárias/metabolismo , Epigênese Genética/genética , Epigenômica/métodos , Feminino , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Impressão Genômica/genética , Masculino , Camundongos , Camundongos Endogâmicos , Células-Tronco Pluripotentes/metabolismo
2.
J Biochem ; 165(4): 323-334, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30590713

RESUMO

Metabolites are sensitive indicators of moment-to-moment cellular status and activity. Expecting that tissue-specific metabolic signatures unveil a unique function of the tissue, we examined metabolomes of mouse liver and testis and found that an unusual metabolite, 2-hydroxyglutarate (2-HG), was abundantly accumulated in the testis. 2-HG can exist as D- or L-enantiomer, and both enantiomers interfere with the activities of 2-oxoglutarate (2-OG)-dependent dioxygenases, such as the Jumonji family of histone demethylases. Whereas D-2-HG is produced by oncogenic mutants of isocitrate dehydrogenases (IDH) and known as an oncometabolite, L-2-HG was the major enantiomer detected in the testis, suggesting that a distinct mechanism underlies the testicular production of this metabolite. We clarified that lactate dehydrogenase C (LDHC), a testis-specific lactate dehydrogenase, is responsible for L-2-HG accumulation by generating and analysing Ldhc-deficient mice. Although the inhibitory effects of 2-HG on 2-OG-dependent dioxygenases were barely observed in the testis, the LDHA protein level was remarkably decreased in Ldhc-deficient sperm, indicating that LDHC is required for LDHA expression in the sperm. This unique functional interaction between LDH family members supports lactate dehydrogenase activity in the sperm. The severely impaired motility of Ldhc-deficient sperm suggests a substantial contribution of glycolysis to energy production for sperm motility.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , L-Lactato Desidrogenase/biossíntese , L-Lactato Desidrogenase/metabolismo , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/enzimologia , Animais , Isoenzimas/biossíntese , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Desidrogenase/genética , Lactato Desidrogenase 5 , Masculino , Camundongos , Camundongos Knockout
3.
Clin Epigenetics ; 9: 79, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28785370

RESUMO

BACKGROUND: Frozen-thawed embryo transfer (FET) is increasingly available for the improvement of the success rate of assisted reproductive technologies other than fresh embryo transfer (ET). There have been numerous findings that FET provides better obstetric and perinatal outcomes. However, the birth weight of infants conceived using FET is heavier than that of those conceived via ET. In addition, some reports have suggested that FET is associated with perinatal diseases such as placenta accreta and pregnancy-induced hypertension (PIH). RESULTS: In this study, we compared the microRNA (miRNA) expression profiles in term placentae derived from FET, ET, and spontaneous pregnancy (SP). We identified four miRNAs, miR-130a-3p, miR-149-5p, miR-423-5p, and miR-487b-3p, that were significantly downregulated in FET placentae compared with those from SP and ET. We found that DNA methylation of MEG3-DMR, not but IG-DMR, was associated with miRNA expression of the DLK1-DIO3 imprinted domain in the human placenta. In functional analyses, GO terms and signaling pathways related to positive regulation of gene expression, growth, development, cell migration, and type II diabetes mellitus (T2DM) were enriched. CONCLUSIONS: This study supports the hypothesis that the process of FET may increase exposure of epigenome to external influences.


Assuntos
Peso ao Nascer/genética , Diabetes Mellitus Tipo 2/genética , Transferência Embrionária/efeitos adversos , Perfilação da Expressão Gênica/métodos , MicroRNAs/genética , Placenta/química , Adulto , Criopreservação , Metilação de DNA , Feminino , Estudo de Associação Genômica Ampla , Impressão Genômica , Humanos , Lactente , Idade Materna , Gravidez
4.
Am J Hum Genet ; 99(5): 1045-1058, 2016 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-27843122

RESUMO

DNA methylation is globally reprogrammed after fertilization, and as a result, the parental genomes have similar DNA-methylation profiles after implantation except at the germline differentially methylated regions (gDMRs). We and others have previously shown that human blastocysts might contain thousands of transient maternally methylated gDMRs (transient mDMRs), whose maternal methylation is lost in embryonic tissues after implantation. In this study, we performed genome-wide allelic DNA methylation analyses of purified trophoblast cells from human placentas and, surprisingly, found that more than one-quarter of the transient-in-embryo mDMRs maintained their maternally biased DNA methylation. RNA-sequencing-based allelic expression analyses revealed that some of the placenta-specific mDMRs were associated with expression of imprinted genes (e.g., TIGAR, SLC4A7, PROSER2-AS1, and KLHDC10), and three imprinted gene clusters were identified. This approach also identified some X-linked gDMRs. Comparisons of the data with those from other mammals revealed that genomic imprinting in the placenta is highly variable. These findings highlight the incomplete erasure of germline DNA methylation in the human placenta; understanding this erasure is important for understanding normal placental development and the pathogenesis of developmental disorders with imprinting effects.


Assuntos
Alelos , Perfilação da Expressão Gênica , Impressão Genômica , Placenta/metabolismo , Proteínas Reguladoras de Apoptose , Blastocisto/citologia , Blastocisto/metabolismo , Metilação de DNA , Exoma , Feminino , Genes Ligados ao Cromossomo X , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Anotação de Sequência Molecular , Monoéster Fosfórico Hidrolases , Placenta/citologia , Polimorfismo de Nucleotídeo Único , Gravidez , Análise de Sequência de RNA , Simportadores de Sódio-Bicarbonato/genética , Simportadores de Sódio-Bicarbonato/metabolismo , Trofoblastos/citologia , Trofoblastos/metabolismo
5.
PLoS One ; 11(11): e0167127, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27880848

RESUMO

DNA methylation plays important roles in the production and functioning of spermatozoa. Recent studies have suggested that DNA methylation patterns in spermatozoa can change with age, but the regions susceptible to age-related methylation changes remain to be fully elucidated. In this study, we conducted genome-scale DNA methylation profiling of spermatozoa obtained from C57BL/6N mice at 8 weeks (8w), 18 weeks (18w) and 17 months of age (17m). There was no substantial difference in the global DNA methylation patterns between 18w and 17m samples except for a slight increase of methylation levels in long interspersed nuclear elements in the 17m samples. We found that maternally methylated imprinting control regions (mICRs) and spermatogenesis-related gene promoters had 5-10% higher methylation levels in 8w samples than in 18w or 17m samples. Analysis of individual sequence reads suggested that these regions were fully methylated (80-100%) in a subset of 8w spermatozoa. These regions are also known to be highly methylated in a subset of postnatal spermatogonia, which might be the source of the increased DNA methylation in 8w spermatozoa. Another possible source was contamination by somatic cells. Although we carefully purified the spermatozoa, it was difficult to completely exclude the possibility of somatic cell contamination. Further studies are needed to clarify the source of the small increase in DNA methylation in the 8w samples. Overall, our findings suggest that DNA methylation patterns in mouse spermatozoa are relatively stable throughout reproductive life.


Assuntos
Envelhecimento/fisiologia , Metilação de DNA/fisiologia , Impressão Genômica/fisiologia , Espermatozoides/metabolismo , Animais , Masculino , Camundongos , Espermatozoides/citologia
6.
Congenit Anom (Kyoto) ; 55(3): 133-44, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26212350

RESUMO

The most common form of male infertility is a low sperm count, known as oligozoospermia. Studies suggest that oligozoospermia is associated with epigenetic alterations. Epigenetic alterations in sperm, which may arise due to the exposure of gametes to environmental factors or those that pre-exist in the sperm of infertile individuals, may contribute to the increased incidence of normally rare imprinting disorders in babies conceived after assisted reproductive technology using the sperm of infertile men. Genomic imprinting is an important developmental process whereby the allelic activity of certain genes is regulated by DNA methylation established during gametogenesis. The aberrant expression of several imprinted genes has been linked to various diseases, malignant tumors, lifestyle and mental disorders in humans. Understanding how infertility and environmental factors such as reproductive toxicants, certain foods, and drug exposures during gametogenesis contribute to the origins of these disorders via defects in sperm is of paramount importance. In this review, we discuss the association of epigenetic alterations with abnormal spermatogenesis and the evidence that epigenetic processes, including those required for genomic imprinting, may be sensitive to environmental exposures during gametogenesis, fertilization and early embryonic development. In addition, we review imprinting diseases and their relationships with environmental factors. While the plasticity of epigenetic marks may make these more susceptible to modification by the environment, this also suggests that aberrant epigenetic marks may be reversible. A greater understanding of this process and the function of epidrugs may lead to the development of new treatment methods for many adult diseases in the future.


Assuntos
Epigênese Genética , Impressão Genômica , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Espermatozoides/química , Adulto , Metilação de DNA , Humanos , Masculino
7.
PLoS Genet ; 10(12): e1004868, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25501653

RESUMO

DNA methylation is globally reprogrammed during mammalian preimplantation development, which is critical for normal development. Recent reduced representation bisulfite sequencing (RRBS) studies suggest that the methylome dynamics are essentially conserved between human and mouse early embryos. RRBS is known to cover 5-10% of all genomic CpGs, favoring those contained within CpG-rich regions. To obtain an unbiased and more complete representation of the methylome during early human development, we performed whole genome bisulfite sequencing of human gametes and blastocysts that covered>70% of all genomic CpGs. We found that the maternal genome was demethylated to a much lesser extent in human blastocysts than in mouse blastocysts, which could contribute to an increased number of imprinted differentially methylated regions in the human genome. Global demethylation of the paternal genome was confirmed, but SINE-VNTR-Alu elements and some other tandem repeat-containing regions were found to be specifically protected from this global demethylation. Furthermore, centromeric satellite repeats were hypermethylated in human oocytes but not in mouse oocytes, which might be explained by differential expression of de novo DNA methyltransferases. These data highlight both conserved and species-specific regulation of DNA methylation during early mammalian development. Our work provides further information critical for understanding the epigenetic processes underlying differentiation and pluripotency during early human development.


Assuntos
Metilação de DNA , Adulto , Blastocisto/fisiologia , Ilhas de CpG , Técnicas de Cultura Embrionária , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Estudo de Associação Genômica Ampla , Impressão Genômica , Humanos , Oócitos/fisiologia , Análise de Sequência de DNA , Sequências de Repetição em Tandem
8.
Reprod Med Biol ; 13(4): 193-202, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25298744

RESUMO

There has been an increase in incidence reports of rare imprinting disorders associated with assisted reproductive technology (ART). ART, including in vitro fertilization and intracytoplasmic sperm injections, is an important treatment for infertile people of reproductive age and increasingly produces children. The identification of epigenetic changes at imprinted loci in ART infants has led to the suggestion that ART techniques themselves may predispose embryos to acquire imprinting errors and diseases. In this review, we note that the particular steps of ART may be prone to induction of imprinting methylation errors during gametogenesis, fertilization and early embryonic development. In addition, we explain imprint-associated diseases and their causes. Moreover, from a Japanese nationwide epidemiological study of imprint-associated diseases, we determine their associations with ART. Epigenetic studies will be required to understand the pathogenesis, ART-related risk factor(s) and what precautions can be taken to prevent the occurrence of input methylation errors. We hope that the constitution of children born after each ART procedure will reveal the safest and most ethical approach to use, which will be invaluable for the future development of standard ART.

9.
Hum Mol Genet ; 23(4): 992-1001, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24105465

RESUMO

Animals cloned by somatic cell nuclear transfer (SCNT) provide a unique model for understanding the mechanisms of nuclear epigenetic reprogramming to a state of totipotency. Though many phenotypic abnormalities have been demonstrated in cloned animals, the underlying mechanisms are not well understood. In this study, we performed transcriptome-wide allelic expression analyses in brain and placental tissues of cloned mice. We found that Gab1, Sfmbt2 and Slc38a4 showed loss of imprinting in all cloned mice analyzed, which might be involved in placentomegaly of cloned mice. These three genes did not require de novo DNA methylation in growing oocytes for the establishment of imprinting, implying the involvement of a de novo DNA methylation-independent mechanism. Loss of Dlk1-Dio3 imprinting was also observed in nearly half of cloned mouse embryos and showed a strong correlation with embryonic lethality. Our findings are essential to understand the underlying mechanisms of developmental abnormalities of cloned animals. We also emphasize that particular attention should be paid to specific imprinted genes for therapeutic and agricultural applications of SCNT.


Assuntos
Clonagem de Organismos , Impressão Genômica , Proteínas Adaptadoras de Transdução de Sinal , Sistema A de Transporte de Aminoácidos/genética , Animais , Sequência de Bases , Encéfalo/metabolismo , Feminino , Iodeto Peroxidase/genética , Camundongos , Fosfoproteínas/genética , Placenta/metabolismo , Gravidez , Proteínas Repressoras , Análise de Sequência de RNA , Fatores de Transcrição/genética , Transcriptoma
10.
Pediatr Int ; 55(5): 542-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23919517

RESUMO

There have been increased incident reports of rare imprinting disorders associated with assisted reproductive technology (ART). ART is an important treatment for infertile people of reproductive age and is increasingly common. The identification of epigenetic changes at imprinted loci in ART infants has led to the suggestion that the techniques themselves may predispose embryos to acquisition of imprinting errors and disease. It is still unknown, however, at what point(s) these imprinting errors arise, or the risk factors. In this review it was hypothesized that the particular steps of the ART process may be prone to induction of imprinting methylation errors during gametogenesis, fertilization and early embryonic development. In addition, imprinting diseases and their causes are explained. Moreover, using a Japanese nationwide epidemiological study of imprinting diseases, their association with ART is determined. Epigenetic studies are required to understand the pathogenesis of this association; the ART-related risk factor(s); and the precautions that can be taken to prevent the occurrence of these syndromes. It is hoped that the constitution of children born after ART will indicate the safest and most ethical approach to use, which will be invaluable for the future development of standard ART treatment.


Assuntos
Metilação de DNA , Epigênese Genética , Impressão Genômica/genética , Infertilidade/terapia , Técnicas de Reprodução Assistida , Humanos , Infertilidade/genética
11.
PLoS Genet ; 9(4): e1003439, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23637617

RESUMO

DNA methylation is an epigenetic modification that plays a crucial role in normal mammalian development, retrotransposon silencing, and cellular reprogramming. Although methylation mainly occurs on the cytosine in a CG site, non-CG methylation is prevalent in pluripotent stem cells, brain, and oocytes. We previously identified non-CG methylation in several CG-rich regions in mouse germinal vesicle oocytes (GVOs), but the overall distribution of non-CG methylation and the enzymes responsible for this modification are unknown. Using amplification-free whole-genome bisulfite sequencing, which can be used with minute amounts of DNA, we constructed the base-resolution methylome maps of GVOs, non-growing oocytes (NGOs), and mutant GVOs lacking the DNA methyltransferase Dnmt1, Dnmt3a, Dnmt3b, or Dnmt3L. We found that nearly two-thirds of all methylcytosines occur in a non-CG context in GVOs. The distribution of non-CG methylation closely resembled that of CG methylation throughout the genome and showed clear enrichment in gene bodies. Compared to NGOs, GVOs were over four times more methylated at non-CG sites, indicating that non-CG methylation accumulates during oocyte growth. Lack of Dnmt3a or Dnmt3L resulted in a global reduction in both CG and non-CG methylation, showing that non-CG methylation depends on the Dnmt3a-Dnmt3L complex. Dnmt3b was dispensable. Of note, lack of Dnmt1 resulted in a slight decrease in CG methylation, suggesting that this maintenance enzyme plays a role in non-dividing oocytes. Dnmt1 may act on CG sites that remain hemimethylated in the de novo methylation process. Our results provide a basis for understanding the mechanisms and significance of non-CG methylation in mammalian oocytes.


Assuntos
Metilação de DNA , Oócitos , Animais , Ilhas de CpG , DNA/metabolismo , Genoma , Camundongos , Oócitos/metabolismo , Oogênese/genética
12.
Hum Mol Genet ; 21(3): 548-58, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22025075

RESUMO

Within the vertebrate groups, only mammals are subject to a specialized epigenetic process termed genomic imprinting in which genes are preferentially expressed from one parental allele. Imprinted expression has been reported for >100 mouse genes and, for approximately one-quarter of these genes, the imprinted expression is specific to the placenta (or extraembryonic tissues). This seemingly placenta-specific imprinted expression has garnered much attention, as has the apparent lack of conserved imprinting between the human and mouse placenta. In this study, we used a novel approach to re-investigate the placenta-specific expression using embryo transfer and trophoblast stem cells. We analyzed 20 genes previously reported to show maternal allele-specific expression in the placenta, and only 8 genes were confirmed to be imprinted. Other genes were likely to be falsely identified as imprinted due to their relatively high expression in contaminating maternal cells. Next, we performed a genome-wide transcriptome assay and identified 133 and 955 candidate imprinted genes with paternal allele- and maternal allele-specific expression. Of those we analyzed in detail, 1/6 (Gab1) of the candidates for paternal allele-specific expression and only 1/269 (Ano1) candidates for maternal allele-specific expression were authentically imprinted genes. Imprinting of Ano1 and Gab1 was specific to the placenta and neither gene displayed allele-specific promoter DNA methylation. Imprinting of ANO1, but not GAB1, was conserved in the human placenta. Our findings impose a considerable revision of the current views of placental imprinting.


Assuntos
Canais de Cloreto/genética , Impressão Genômica , Placenta/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Alelos , Animais , Anoctamina-1 , Canais de Cloreto/metabolismo , Decídua/metabolismo , Transferência Embrionária , Feminino , Perfilação da Expressão Gênica , Humanos , Camundongos , Fosfoproteínas/genética , Gravidez , Proteínas Repressoras , Análise de Sequência de RNA , Células-Tronco/citologia , Fatores de Transcrição/genética , Trofoblastos/citologia
13.
Nat Genet ; 42(12): 1093-100, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21057502

RESUMO

DNA methylation is extensively reprogrammed during the early phases of mammalian development, yet genomic targets of this process are largely unknown. We optimized methylated DNA immunoprecipitation for low numbers of cells and profiled DNA methylation during early development of the mouse embryonic lineage in vivo. We observed a major epigenetic switch during implantation at the transition from the blastocyst to the postimplantation epiblast. During this period, DNA methylation is primarily targeted to repress the germline expression program. DNA methylation in the epiblast is also targeted to promoters of lineage-specific genes such as hematopoietic genes, which are subsequently demethylated during terminal differentiation. De novo methylation during early embryogenesis is catalyzed by Dnmt3b, and absence of DNA methylation leads to ectopic gene activation in the embryo. Finally, we identify nonimprinted genes that inherit promoter DNA methylation from parental gametes, suggesting that escape of post-fertilization DNA methylation reprogramming is prevalent in the mouse genome.


Assuntos
Metilação de DNA/genética , Desenvolvimento Embrionário/genética , Regiões Promotoras Genéticas/genética , Animais , Linhagem da Célula/genética , Ilhas de CpG/genética , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Hematopoese/genética , Padrões de Herança/genética , Camundongos , Camundongos Endogâmicos C57BL , Anotação de Sequência Molecular , Mutação/genética , Especificidade de Órgãos/genética , Células-Tronco Pluripotentes/metabolismo , DNA Metiltransferase 3B
14.
Genes Cells ; 15(3): 169-79, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20132320

RESUMO

In the male and female germ-lines of mice, both of the two de novo DNA methyltransferases Dnmt3a and Dnmt3b are expressed. By the conditional knockout experiments using the Tnap-Cre gene, we previously showed that deletion of Dnmt3a in primordial germ cells disrupts paternal and maternal imprinting, however, Dnmt3b mutants did not show any defect. Here, we have knocked out Dnmt3a after birth in growing oocytes by using the Zp3-Cre gene and obtained genetic evidence that de novo methylation by Dnmt3a during the oocyte growth stage is indispensable for maternal imprinting. We also carried out DNA methylation analysis in the mutant oocytes and embryos and found that hypomethylation of imprinted genes in Dnmt3a-deficient oocytes was directly inherited to the embryos, but repetitive elements were re-methylated during development. Furthermore, we show that Dnmt3b-deficient cells can contribute to the male and female germ-lines in chimeric mice and can produce normal progeny, establishing that Dnmt3b is dispensable for mouse gametogenesis and imprinting. Finally, Dnmt3-related protein Dnmt3L is not only essential for methylation of imprinted genes but also enhances de novo methylation of repetitive elements in growing oocytes.


Assuntos
Quimera/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , Proteínas do Ovo/genética , Impressão Genômica , Integrases/genética , Glicoproteínas de Membrana/genética , Oócitos/crescimento & desenvolvimento , Receptores de Superfície Celular/genética , Animais , Quimera/embriologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Embrião de Mamíferos/metabolismo , Feminino , Sequências Repetitivas Dispersas , Masculino , Camundongos , Camundongos Transgênicos , Oócitos/metabolismo , Glicoproteínas da Zona Pelúcida , DNA Metiltransferase 3B
15.
Genesis ; 46(12): spc one, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19105216

RESUMO

Mouse blastocyst stage embryo stained for histone H3 lysine-27 trimethylation (red) and DNA (blue). H3K27me3 marks the inactive X chromosome. The study by Chiba et al. in this issue suggests that de novo DNA methyltransferases are dispensable for setting the imprint on the maternally-derived X chromsome in growing oocytes. See Chiba et al. in this issue.


Assuntos
Metilação de DNA/genética , Impressão Genômica , Oócitos/fisiologia , Cromossomo X/metabolismo , Animais , Camundongos , Cromossomo X/genética
16.
Genesis ; 46(12): 768-74, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18932249

RESUMO

In female mouse embryos, the paternal X chromosome (Xp) is preferentially inactivated during preimplantation development and trophoblast differentiation. This imprinted X-chromosome inactivation (XCI) is partly due to an activating imprint on the maternal X chromosome (Xm), which is set during oocyte growth. However, the nature of this imprint is unknown. DNA methylation is one candidate, and therefore we examined whether disruptions of the two de novo DNA methyltransferases in growing oocytes affect imprinted XCI. We found that accumulation of histone H3 lysine-27 trimethylation, a hallmark of XCI, occurs normally on the Xp, and not on the Xm, in female blastocysts developed from the mutant oocytes. Furthermore, the allelic expression patterns of X-linked genes including Xist and Tsix were unchanged in preimplantation embryos and also in the trophoblast. These results show that a maternal disruption of the DNA methyltransferases has no effect on imprinted XCI and argue that de novo DNA methylation is dispensable for Xm imprinting. This underscores the difference between imprinted XCI and autosomal imprinting.


Assuntos
Metilação de DNA/genética , Impressão Genômica , Oócitos/fisiologia , Cromossomo X/metabolismo , Alelos , Animais , Blastocisto/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Lisina/química , Lisina/metabolismo , Masculino , Camundongos , Transgenes/genética , Trofoblastos/metabolismo , Cromossomo X/genética
17.
Genes Dev ; 22(12): 1607-16, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559477

RESUMO

Parental origin-specific DNA methylation regulates the monoallelic expression of the mammalian imprinted genes. The methylation marks or imprints are established in the parental germline and maintained throughout embryonic development. However, it is unclear how the methylation imprints are maintained through extensive demethylation in cleavage-stage preimplantation embryos. Previous reports suggested that DNA methyltransferase(s) other than Dnmt1 is involved in the maintenance of the imprints during cleavage. Here we demonstrate, by using conditional knockout mice, that the other known DNA methyltransferases Dnmt3a and Dnmt3b are dispensable for the maintenance of the methylation marks at most imprinted loci. We further demonstrate that a lack of both maternal and zygotic Dnmt1 results in complete demethylation of all imprinted loci examined in blastocysts. Consistent with these results we find that zygotic Dnmt1 is expressed in the preimplantation embryo. Thus, contrary to the previous reports, Dnmt1 alone is sufficient to maintain the methylation marks of the imprinted genes.


Assuntos
DNA (Citosina-5-)-Metiltransferases/fisiologia , Metilação de DNA , Desenvolvimento Embrionário/genética , Impressão Genômica , Zigoto/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica/genética , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Oócitos/metabolismo , Gravidez , RNA Mensageiro Estocado/fisiologia , DNA Metiltransferase 3B
18.
Nature ; 453(7194): 539-43, 2008 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-18404146

RESUMO

RNA interference (RNAi) is a mechanism by which double-stranded RNAs (dsRNAs) suppress specific transcripts in a sequence-dependent manner. dsRNAs are processed by Dicer to 21-24-nucleotide small interfering RNAs (siRNAs) and then incorporated into the argonaute (Ago) proteins. Gene regulation by endogenous siRNAs has been observed only in organisms possessing RNA-dependent RNA polymerase (RdRP). In mammals, where no RdRP activity has been found, biogenesis and function of endogenous siRNAs remain largely unknown. Here we show, using mouse oocytes, that endogenous siRNAs are derived from naturally occurring dsRNAs and have roles in the regulation of gene expression. By means of deep sequencing, we identify a large number of both approximately 25-27-nucleotide Piwi-interacting RNAs (piRNAs) and approximately 21-nucleotide siRNAs corresponding to messenger RNAs or retrotransposons in growing oocytes. piRNAs are bound to Mili and have a role in the regulation of retrotransposons. siRNAs are exclusively mapped to retrotransposons or other genomic regions that produce transcripts capable of forming dsRNA structures. Inverted repeat structures, bidirectional transcription and antisense transcripts from various loci are sources of the dsRNAs. Some precursor transcripts of siRNAs are derived from expressed pseudogenes, indicating that one role of pseudogenes is to adjust the level of the founding source mRNA through RNAi. Loss of Dicer or Ago2 results in decreased levels of siRNAs and increased levels of retrotransposon and protein-coding transcripts complementary to the siRNAs. Thus, the RNAi pathway regulates both protein-coding transcripts and retrotransposons in mouse oocytes. Our results reveal a role for endogenous siRNAs in mammalian oocytes and show that organisms lacking RdRP activity can produce functional endogenous siRNAs from naturally occurring dsRNAs.


Assuntos
Oócitos/metabolismo , Interferência de RNA , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Animais , Proteínas Argonautas , Fator de Iniciação 2 em Eucariotos/deficiência , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Biblioteca Gênica , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Oócitos/crescimento & desenvolvimento , Reação em Cadeia da Polimerase , Pseudogenes/genética , RNA Mensageiro/metabolismo , Retroelementos/genética , Ribonuclease III/deficiência , Ribonuclease III/genética , Ribonuclease III/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...