首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 453 毫秒
1.
Active DNA demethylation occurs after a sperm enters an egg. However, the mechanisms for the active DNA demethylation remain poorly understood. Ten-eleven translocation enzymes were recently shown to catalyze the conversion of 5-methylcytosine to 5-hydroxymethylcytosine (5hmC). Thus, we decided to investigate the role of 5hmC in active demethylation. We analyzed the methylation and hydroxymethylation status in metaphase II oocytes as well as 1-cell stage and cleavage stage embryos. In zygotes, 5hmC was mainly detected in the paternal pronucleus and it increased from the pronuclear-2 (PN2) to PN5 stages, an indication that 5hmC was involved in paternal genomic DNA demethylation. Bisulfite-sequencing PCR and qGluMS-PCR (DNA glucosylation and digestion before quantitative PCR) results showed that a large reduction of methylcytosine and hydroxymethylcytosine in LINE1 (long interspersed nuclear element 1) occurred between the 4- and 8-cell stages, which indicates that demethylation potentially occurred after the 4-cell stage. We then microinjected mouse zygote with plasmids that were methylated in vitro by SssI methylase and analyzed for the hydroxymethylation status of the plasmids promoter region. We found that the rapid onset of expression of the unmethylated plasmids in mouse embryos happened in <12 h, but the expression of methylated plasmids was delayed until 50 h when most embryos were at the 8-cell stage. Quantitative GluMS-PCR results suggested that 5hmC was present in the plasmid's promoter region at the MspI site where the active demethylation occurred. Our results demonstrate that 5hmC is involved in active demethylation in mice.  相似文献   

2.
DNA active demethylation is an important epigenetic phenomenon observed in porcine zygotes, yet its molecular origins are unknown. Our results show that 5-methylcytosine (5mC) converts into 5-hydroxymethylcytosine (5hmC) during the first cell cycle in porcine in vivo fertilization (IVV), IVF, and SCNT embryos, but not in parthenogenetically activated embryos. Expression of Ten-Eleven Translocation 1 (TET1) correlates with this conversion. Expression of 5mC gradually decreases until the morula stage; it is only expressed in the inner cell mass, but not trophectoderm regions of IVV and IVF blastocysts. Expression of 5mC in SCNT embryos is ectopically distinct from that observed in IVV and IVF embryos. In addition, 5hmC expression was similar to that of 5mC in IVV cleavage-stage embryos. Expression of 5hmC remained constant in IVF and SCNT embryos, and was evenly distributed among the inner cell mass and trophectoderm regions derived from IVV, IVF, and SCNT blastocysts. Ten-Eleven Translocation 3 was highly expressed in two-cell embryos, whereas TET1 and TET2 were highly expressed in blastocysts. These data suggest that TET1-catalyzed 5hmC may be involved in active DNA demethylation in porcine early embryos. In addition, 5mC, but not 5hmC, participates in the initial cell lineage specification in porcine IVV and IVF blastocysts. Last, SCNT embryos show aberrant 5mC and 5hmC expression during early porcine embryonic development.  相似文献   

3.
4.
5.
6.
7.
8.
Inoue A  Shen L  Dai Q  He C  Zhang Y 《Cell research》2011,21(12):1670-1676
One of the recent advances in the epigenetic field is the demonstration that the Tet family of proteins are capable of catalyzing conversion of 5-methylcytosine (5mC) of DNA to 5-hydroxymethylcytosine (5hmC). Interestingly, recent studies have shown that 5hmC can be further oxidized by Tet proteins to generate 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC), which can be removed by thymine DNA glycosylase (TDG). To determine whether Tet-catalyzed conversion of 5mC to 5fC and 5caC occurs in vivo in zygotes, we generated antibodies specific for 5fC and 5caC. By immunostaining, we demonstrate that loss of 5mC in the paternal pronucleus is concurrent with the appearance of 5fC and 5caC, similar to that of 5hmC. Importantly, instead of being quickly removed through an enzyme-catalyzed process, both 5fC and 5caC exhibit replication-dependent dilution during mouse preimplantation development. These results not only demonstrate the conversion of 5mC to 5fC and 5caC in zygotes, but also indicate that both 5fC and 5caC are relatively stable and may be functional during preimplantation development. Together with previous studies, our study suggests that Tet-catalyzed conversion of 5mC to 5hmC/5fC/5caC followed by replication-dependent dilution accounts for paternal DNA demethylation during preimplantation development.  相似文献   

9.
10.
The TET family of dioxygenases (TET1/2/3) can convert 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC) and has been shown to be involved in active and passive DNA demethylation. Here, we demonstrate that altering TET dioxygenase levels within physiological range can affect DNA methylation dynamics of HEK293 cells. Overexpression of TET1 increased global 5hmC levels and was accompanied by mild DNA demethylation of promoters, gene bodies and CpG islands. Conversely, the simultaneous knockdown of TET1, TET2, and TET3 led to decreased global 5hmC levels and mild DNA hypermethylation of above-mentioned regions. The methylation changes observed in the overexpression and knockdown studies were mostly non-reciprocal and occurred with different preference depending on endogenous methylation and gene expression levels. Single-nucleotide 5hmC profiling performed on a genome-wide scale revealed that TET1 overexpression induced 5mC oxidation without a distribution bias among genetic elements and structures. Detailed analysis showed that this oxidation was related to endogenous 5hmC levels. In addition, our results support the notion that the effects of TET1 overexpression on gene expression are generally unrelated to its catalytic activity.  相似文献   

11.
12.
DNA methylation has been proven to be a critical epigenetic mark important for various cellular processes. Here, we report that redox-active quinones, a ubiquitous class of chemicals found in natural products, cancer therapeutics and environment, stimulate the conversion of 5mC to 5hmC in vivo, and increase 5hmC in 5751 genes in cells. 5hmC increase is associated with significantly altered gene expression of 3414 genes. Interestingly, in quinone-treated cells, labile iron-sensitive protein ferritin light chain showed a significant increase at both mRNA and protein levels indicating a role of iron regulation in stimulating Tet-mediated 5mC oxidation. Consistently, the deprivation of cellular labile iron using specific chelator blocked the 5hmC increase, and a delivery of labile iron increased the 5hmC level. Moreover, both Tet1/Tet2 knockout and dimethyloxalylglycine-induced Tet inhibition diminished the 5hmC increase. These results suggest an iron-regulated Tet-dependent DNA demethylation mechanism mediated by redox-active biomolecules.  相似文献   

13.
Cytosine methylation is the major epigenetic modification of metazoan DNA. Although there is strong evidence that active DNA demethylation occurs in animal cells, the molecular details of this process are unknown. The recent discovery of the TET protein family (TET1–3) 5-methylcytosine hydroxylases has provided a new entry point to reveal the identity of the long-sought DNA demethylase. Here, we review the recent progress in understanding the function of TET proteins and 5-hydroxymethylcytosine (5hmC) through various biochemical and genomic approaches, the current evidence for a role of 5hmC as an early intermediate in active DNA demethylation and the potential functions of TET proteins and 5hmC beyond active DNA demethylation. We also discuss how future studies can extend our knowledge of this novel epigenetic modification.Key words: TET1, 5-hydroxymethylcytosine, active DNA demethylation, epigenetic, DNA methylation, hippocampus, electroconvulsive stimulation, Gadd45b, BER  相似文献   

14.
15.
Although epigenetic modulation is critical for a variety of cellular activities, its role in erythropoiesis remains poorly understood. Ten-eleven translocation (TET) molecules participate in methylcytosine (5mC) hydroxylation, which results in DNA demethylation in several biological processes. In this research, the role of TETs in erythropoiesis was investigated by using the zebrafish model, where three TET homologs were identified. These homologs share conserved structural domains with their mammalian counterparts. Zebrafish TETs mediate the conversion of 5mC to hydroxymethylcytosine (5hmC) in zebrafish embryos, and the deletion of TET2 inhibits erythropoiesis by suppressing the expression of the scl, gata-1, and cmyb genes. TET2-upregulated lineage-specific genes and erythropoiesis are closely associated with the occurrence of 5hmC and demethylation in the intermediate CpG promoters (ICPs) of scl, gata-1, cmyb, which frequently occur at specific regions or CpG sites of these ICPs. Moreover, TET2 regulates the formation and differentiation of erythroid progenitors, and deletion of TET2 leads to erythrocyte dysplasia and anemia. Here, we preliminarily proved that TET2 plays an essential role in erythrocyte development by regulating lineage-specific genes via DNA oxidative demethylation. This report is anticipated to broaden current information on hematopoiesis and pathogenesis of hematopoiesis-related diseases.  相似文献   

16.
In zygotes, a global loss of DNA methylation occurs selectively in the paternal pronucleus before the first cell division, concomitantly with the appearance of modified forms of 5-methylcytosine. The adjacent maternal pronucleus and certain paternally-imprinted loci are protected from this process. Nakamura et al. recently clarified the molecular mechanism involved: PGC7/Stella/Dppa3 binds to dimethylated histone 3 lysine 9 (H3K9me2), thereby blocking the activity of the Tet3 methylcytosine oxidase in the maternal genome as well as at certain imprinted loci in the paternal genome.DNA methylation is a crucial epigenetic modification that regulates imprinting (differential silencing of maternal or paternal alleles) and repression of retrotransposons and other parasitic DNA, as well as possibly X-chromosome inactivation and cellular differentiation. DNA methylation needs to be faithfully maintained throughout the life cycle, since loss of DNA methylation can result in gene dosage problems, dysregulation of gene expression, and genomic instability due to retrotransposon reactivation1. Nevertheless, genome-wide loss of DNA methylation has been observed during germ cell development2 and in the paternal pronucleus soon after fertilization3.For almost a decade, the global decrease of DNA methylation observed in the paternal genome within a few hours of fertilization was ascribed to an “active”, replication-independent process3. The maternal pronucleus is spared and instead undergoes “passive”, replication-dependent demethylation during early embryogenesis, arising from inhibition of the DNA maintenance methyltransferase Dnmt1 (Dnmt1 is normally recruited to newly-replicated DNA because of the high affinity of its obligate partner, UHRF1, for hemi-methylated DNA strands, which are produced from symmetrically-methylated CpG dinucleotides as a result of DNA replication). The basis for active and passive demethylation of the paternal and maternal genomes remained a mystery until proteins of the TET family – TET1, TET2 and TET3 in humans – were discovered to be Fe(II)- and 2-oxoglutarate-dependent enzymes capable of oxidizing 5-methylcytosine (5mC) in DNA4,5,6. TET enzymes serially convert 5mC into 5-hydroxymethyl-cytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxycytosine (5caC)5,7,8.With the generation of specific antibodies to 5hmC, it became clear that the supposed “active demethylation” of the paternal pronucleus in mouse zygote after fertilization was due to the inability of anti-5mC antibodies to recognize 5hmC and other 5mC oxidation products9,10. The enzyme responsible for 5mC oxidation was shown to be Tet3, which unlike Tet1 and Tet2 is highly expressed in mouse oocytes and zygotes. RNAi-mediated depletion of Tet3 decreased the staining of the paternal pronucleus with 5hmC, suggesting that immediately after fertilization, Tet3 in the zygote selectively oxidizes 5mC in the paternal genome to 5hmC9,10.How is the maternal pronucleus protected from Tet3 activity? Nakamura et al.11 previously showed that zygotes lacking PGC7/Stella/Dppa3 lose asymmetric regulation of DNA methylation, instead showing global loss of 5mC staining in both paternal and maternal pronuclei. This was correlated with hypomethylation at several maternally-imprinted loci (Peg1, Peg3, Peg10) in PGC7-deficient zygotes, as judged by bisulfite sequencing. Further, certain paternally-imprinted loci (H19, Rasgrf1), which are normally protected from global loss of methylation in the paternal genome, also became hypomethylated in PGC7-deficient zygotes. These data suggested that PGC7 protects the maternal genome, as well as certain paternally imprinted loci, from loss of 5mC.In their recent publication, Nakamura et al.12 elegantly extended these findings to address the mechanism involved. Based on the fact that a major difference between maternal and paternal genomes is that the maternal genome contains histones, whereas the DNA of the entering sperm is tightly packaged with protamine, they asked whether PGC7 recognizes specific histone marks. Indeed, the maternal genome harbors considerable levels of the histone mark H3K9me211, leading them to examine whether PGC7 distinguishes maternal and paternal genomes by recognizing H3K9me2 in the maternal genome. Using wild-type (WT) ES cells and ES cells deficient in the G9a lysine methyltransferase which generates H3K9me2 mark, they showed that PGC7 associated loosely with nucleosomes and chromatin lacking H3K9me2, but tightly if H3K9me2 was present. The binding was recapitulated using recombinant bacterially-expressed PGC7 and histone tail peptides, indicating a direct interaction of PGC7 with the H3K9me2 mark. In agreement, genomic loci enriched with H3K9me2 recruited PGC7 as judged by chromatin immunoprecipitation (ChIP), but this recruitment was abrogated in G9a-deficient ES cells. These data indicated that PGC7 targets genomic regions occupied by nucleosomes containing H3K9me2 (Figure 1); an interesting extension would be to ask whether loss of maternal G9a also results in 5hmC conversion in the maternal pronucleus in zygotes.Open in a separate windowFigure 1Schematic view of paternal (left) and maternal (right) genomes soon after fertilization. Paternal and maternal pronuclei are indicated with immunostaining results in the boxes. PGC7 binds H3K9me2 in the maternal pronucleus and at certain paternally-imprinted loci (H19, Rasgrf1) in the paternal pronucleus, thereby potentially regulating chromatin organization to interfere with Tet3 accessibility.Next, Nakamura et al.12 tested by immunocytochemistry whether PGC7 in zygotes also required H3K9me2. It is known that H3K9me2 staining is concentrated in the maternal but not the paternal pronucleus13. Using conventional staining methods in which the cells are first fixed and then permeabilized to allow antibodies to enter the cell, the authors observed in their earlier study that PGC7 bound to both pronuclei11. Remarkably, by simply reversing the order of the fixation and permeabilization steps – permeabilizing first to allow the loss of loosely bound proteins by dissociation, then fixing and staining – they found that PGC7 associated much more tightly with the maternal pronucleus that bears H3K9me2 mark. Injection of mRNA encoding Jhdm2a (an H3K9me1/ me2-specific demethylase) into zygotes eliminated staining for H3K9me2 as well as PGC7 in the maternal pronucleus, and concomitantly caused loss of 5mC and acquisition of 5hmC. Taken together, these data strongly suggested that PGC7 was selectively recruited to the maternal pronucleus through binding H3K9me2, and that this binding protected zygotic maternal DNA from oxidation of 5mC to 5hmC and beyond (Figure 1).These findings led Nakamura et al. to investigate how PGC7 controls Tet3 activity in zygotes. They showed (in cells that were permeabilized before fixation and immunocytochemistry) that Tet3 was tightly associated only with the paternal pronucleus in WT zygotes, but was present in both pronuclei in PGC7-deficient zygotes. When PGC7 was prevented from binding to the maternal pronucleus by injection of Jhdm2a mRNA, Tet3 became tightly associated with both pronuclei. In other words, loss of PGC7 or loss of H3K9me2 that recruits PGC7 had the same effect – eliminating selective association of Tet3 with the paternal genome. The implication is that PGC7 – which preferentially binds the maternal genome – somehow promotes the selective binding of Tet3 to the paternal genome, thus permitting rapid 5mC oxidation in paternal but not maternal DNA (Figure 1).PGC7 is a small protein (150 amino acids (aa) in the mouse, 159 aa in humans) whose sequence is only moderately conserved. Nakamura et al.12 showed that the binding of PGC7 to H3K9me2 required the N-terminal half of PGC7, whereas its ability to exclude Tet3 from the maternal pronucleus required the C-terminal half. It is unclear how Tet3 exclusion is mediated. One possibility is that the C-terminal region of PGC7 sterically excludes Tet3 from binding, either to DNA or to a chromatin mark; another is that the C-terminal region of PGC7 is capable of altering chromatin configuration to prevent the binding of Tet3 to chromatin. In support of the latter hypothesis, the rate with which micrococcal nuclease (MNase) digested high-molecular weight chromatin was significantly slower in WT ES cells in which PGC7 was present, compared to PGC7−/− and G9a−/− ES cells in which PGC7 was either absent or not recruited to DNA because of the loss of H3K9me2 mark. In contrast, DNA methylation did not alter the chromatin association of PGC7 or its ability to protect high-molecular weight chromatin from MNase digestion, as shown by using Dnmt1−/−Dnmt3a−/−Dnmt3b−/− triple knockout ES cells that completely lack DNA methylation.How does PGC7 protect paternally-imprinted loci from Tet3-mediated 5mC oxidation? Although the haploid sperm genome is mostly packaged with protamine, a genome-wide analysis revealed that 4% of the genome of mature human sperm bears nucleosomes located at developmental and imprinted genes14. Nakamura et al.12 found that among paternally-imprinted differentially methylated regions (DMRs), the H19 and Rasgrf1 DMRs contained H3K9me2 whereas the Meg3 DMR did not, consistent with their previous finding that in PGC7-deficient zygotes, the H19 and Rasgrf1 DMRs were hypomethylated but the Meg3 DMR was unaffected11. Therefore, PGC7 may be recruited to paternally-imprinted loci through H3K9me2-containing nucleosomes that pre-exist in the sperm haploid genome upon fertilization. Alternatively, Nakamura et al. point out that protamine in the sperm is replaced soon after fertilization by the histone H3.3 variant, which in somatic cells does not bear H3K9me2 mark.In conclusion, Nakamura et al.12 demonstrate unambiguously that PGC7 specifically binds to H3K9me2 in the maternal genome in zygotes, where its global occupancy excludes Tet3 and inhibits Tet3-mediated 5mC oxidation. This novel finding provides new insights into the global alterations of DNA methylation status that occur during early embryogenesis. Follow-up questions abound. First, can PGC7 protect other methylated loci such as transposable elements and the X-chromosome? It would be interesting to assess H3K9me2 at these loci. Second, how does the N-terminal half of PGC7 recognize H3K9me2? Structural characterization of this interaction may elucidate a novel epigenetic “reader” domain specific for H3K9me2. Third, PGC7 is a marker for cells of the inner cell mass, and is co-expressed with Tet1 and Tet2 rather than Tet3 in ESCs15. Does PGC7 also antagonize Tet1 and Tet2 and protect imprinted loci in ESCs? Fourth, how does PGC7 inhibit the access of Tet3 to chromatin? Considering that PGC7 is small and is not equipped with known enzymatic domains, it is likely that PGC-interacting proteins, rather than PGC7 itself, function to regulate chromatin status. Fifth, how is Tet3 recruited to paternal chromatin – are there specific histone or other epigenetic marks that facilitate Tet3 recruitment? Finally, while technically challenging, it seems imperative to identify the target genes of PGC7 and Tet3, by profiling the genomic location of 5hmC and other 5mC oxidation products in the paternal and maternal genomes of zygotes from WT, Tet3-deficient and PGC7-deficient mice.  相似文献   

17.
In mammalian zygotes, the 5‐methyl‐cytosine (5mC) content of paternal chromosomes is rapidly changed by a yet unknown but presumably active enzymatic mechanism. Here, we describe the developmental dynamics and parental asymmetries of DNA methylation in relation to the presence of DNA strand breaks, DNA repair markers and a precise timing of zygotic DNA replication. The analysis shows that distinct pre‐replicative (active) and replicative (active and passive) phases of DNA demethylation can be observed. These phases of DNA demethylation are concomitant with the appearance of DNA strand breaks and DNA repair markers such as γH2A.X and PARP‐1, respectively. The same correlations are found in cloned embryos obtained after somatic cell nuclear transfer. Together, the data suggest that (1) DNA‐methylation reprogramming is more complex and extended as anticipated earlier and (2) the DNA demethylation, particularly the rapid loss of 5mC in paternal DNA, is likely to be linked to DNA repair mechanisms.  相似文献   

18.
19.
5-methyl-C (5mC) and 5-hydroxymethyl-C (5hmC) are epigenetic marks with well-known and putative roles in gene regulation, respectively. These two DNA covalent modifications cannot be distinguished by bisulfite sequencing or restriction digestion, the standard methods of 5mC detection. The methylated CpG island recovery assay (MIRA), however, specifically detects 5mC but not 5hmC. We further developed MIRA for the analysis of allele-specific CpG methylation at differentially methylated regions (DMRs) of imprinted genes. MIRA specifically distinguished between the parental alleles by capturing the paternally methylated H19/Igf2 DMR and maternally methylated KvDMR1 in mouse embryo fibroblasts (MEFs) carrying paternal and maternal duplication of mouse distal Chr7, respectively. MIRA in combination with multiplex single nucleotide primer extension (SNuPE) assays specifically captured the methylated parental allele from normal cells at a set of maternally and paternally methylated DMRs. The assay correctly recognized aberrant biallelic methylation in a case of loss of imprinting. The MIRA-SNuPE assays revealed that placenta exhibited less DNA methylation bias at DMRs compared to yolk sac, amnion, brain, heart, kidney, liver and muscle. This method should be useful for the analysis of allele-specific methylation events related to genomic imprinting, X chromosome inactivation and for verifying and screening haplotype-associated methylation differences in the human population.Key words: epigenetics, imprinting, DMR, MIRA, MBD, DNA methylation, SNuPE  相似文献   

20.
In mammalian development, epigenetic modifications, including DNA methylation patterns, play a crucial role in defining cell fate but also represent epigenetic barriers that restrict developmental potential. At two points in the life cycle, DNA methylation marks are reprogrammed on a global scale, concomitant with restoration of developmental potency. DNA methylation patterns are subsequently re-established with the commitment towards a distinct cell fate. This reprogramming of DNA methylation takes place firstly on fertilization in the zygote, and secondly in primordial germ cells (PGCs), which are the direct progenitors of sperm or oocyte. In each reprogramming window, a unique set of mechanisms regulates DNA methylation erasure and re-establishment. Recent advances have uncovered roles for the TET3 hydroxylase and passive demethylation, together with base excision repair (BER) and the elongator complex, in methylation erasure from the zygote. Deamination by AID, BER and passive demethylation have been implicated in reprogramming in PGCs, but the process in its entirety is still poorly understood. In this review, we discuss the dynamics of DNA methylation reprogramming in PGCs and the zygote, the mechanisms involved and the biological significance of these events. Advances in our understanding of such natural epigenetic reprogramming are beginning to aid enhancement of experimental reprogramming in which the role of potential mechanisms can be investigated in vitro. Conversely, insights into in vitro reprogramming techniques may aid our understanding of epigenetic reprogramming in the germline and supply important clues in reprogramming for therapies in regenerative medicine.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号