DNA Demethylation

DNA 去甲基化
  • 文章类型: Journal Article
    AlkB同源物1(ALKBH1)是双加氧酶AlkB家族的成员,该酶依赖于Fe(II)和α-酮戊二酸。越来越多的证据表明,ALKBH1对各种底物表现出酶活性,包括N6-甲基腺苷(M6A),N1-甲基腺苷(m1A),N3-甲基胞苷(m3C),5-甲基胞嘧啶(m5C),N6-甲基腺嘌呤(N6-mA,6mA),H2A,表明它在不同的生物过程和参与人类疾病中的双重作用。到现在,关于ALKBH1的酶活性正在进行辩论。在这次审查中,我们对ALKBH1的最新研究进行了全面总结,包括其底物多样性和在各种人类疾病中的病理作用,其功能的潜在机制,和它的失调。我们还探讨了ALKBH1作为预后靶标的潜力。
    AlkB homolog 1 (ALKBH1) is a member of the AlkB family of dioxygenases that are dependent on Fe(II) and α-ketoglutarate. Mounting evidence demonstrates that ALKBH1 exhibits enzymatic activity against various substrates, including N6-methyladenosine (m6A), N1-methyladenosine (m1A), N3-methylcytidine (m3C), 5-methylcytosine (m5C), N6-methyladenine (N6-mA, 6mA), and H2A, indicating its dual roles in different biological processes and involvement in human diseases. Up to the present, there is ongoing debate regarding ALKBH1\'s enzymatic activity. In this review, we present a comprehensive summary of recent research on ALKBH1, including its substrate diversity and pathological roles in a wide range of human disorders, the underlying mechanisms of its functions, and its dysregulation. We also explored the potential of ALKBH1 as a prognostic target.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    TET家族以活跃的DNA去甲基化而闻名,在调节转录中起着重要作用,表观基因组和发育。然而,以前使用敲除(KD)或敲除(KO)模型研究TET功能的研究在区分其酶和非酶作用方面面临挑战,以及TET家庭成员之间的补偿效应,这使得人们对TET酶促作用的认识不够准确。为了解决这个问题,我们成功地产生了对特定Tet成员无催化活性的小鼠(Tetm/m)。我们观察到,与报道的KO小鼠相比,突变小鼠表现出明显的发育缺陷,包括生长迟缓,性别失衡,不孕症,和围产期致死性。值得注意的是,Tetm/m小鼠胚胎干细胞(mESCs)成功建立,但进入了受损的发育程序,证明了由异常DNA甲基化引起的外胚层分化的延长多能性和缺陷。有趣的是,Tet3,传统上被认为对mESC不太重要,因为它的表达水平较低,对全球羟甲基化产生了重大影响,基因表达,和mESCs的分化潜能。值得注意的是,在多能性调节中,Tet1和Tet3之间存在共同的调节区.总之,本研究为Tet羟甲基化酶活性在小鼠发育和ESC多能性调控中的作用机制提供了更准确的参考。
    The TET family is well known for active DNA demethylation and plays important roles in regulating transcription, the epigenome and development. Nevertheless, previous studies using knockdown (KD) or knockout (KO) models to investigate the function of TET have faced challenges in distinguishing its enzymatic and nonenzymatic roles, as well as compensatory effects among TET family members, which has made the understanding of the enzymatic role of TET not accurate enough. To solve this problem, we successfully generated mice catalytically inactive for specific Tet members (Tetm/m). We observed that, compared with the reported KO mice, mutant mice exhibited distinct developmental defects, including growth retardation, sex imbalance, infertility, and perinatal lethality. Notably, Tetm/m mouse embryonic stem cells (mESCs) were successfully established but entered an impaired developmental program, demonstrating extended pluripotency and defects in ectodermal differentiation caused by abnormal DNA methylation. Intriguingly, Tet3, traditionally considered less critical for mESCs due to its lower expression level, had a significant impact on the global hydroxymethylation, gene expression, and differentiation potential of mESCs. Notably, there were common regulatory regions between Tet1 and Tet3 in pluripotency regulation. In summary, our study provides a more accurate reference for the functional mechanism of Tet hydroxymethylase activity in mouse development and ESC pluripotency regulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    科学证据表明,糖尿病与乳腺癌的发病率更高、侵袭性增加有关;然而,对参与这一过程的众多监管机构的机械研究还不够彻底。晚期糖基化终末产物(AGEs)在糖尿病慢性并发症中发挥重要作用,但AGEs在乳腺癌中的作用机制在很大程度上还未被研究.在这项研究中,我们首先证明乳腺癌组织中的高AGEs水平与糖尿病状态和不良患者预后相关.此外,AGEs与AGEs受体(RAGE)相互作用,促进乳腺癌细胞迁移和侵袭。机械上,基于RNA测序(RNA-seq)分析,我们发现生长停滞和DNA损伤基因45α(GADD45α)是AGEs通过P53依赖性途径上调的重要蛋白。接下来,GADD45α募集胸腺嘧啶DNA糖基化酶(TDG)进行碱基切除修复,在MMP-9的启动子区域形成去甲基化复合物,并通过DNA去甲基化增强MMP-9的反式激活。总的来说,我们的研究结果表明AGEs在乳腺癌和糖尿病患者中具有重要的调节作用,并揭示了表观遗传修饰促进乳腺癌转移的新机制.
    Scientific evidence has linked diabetes to a higher incidence and increased aggressiveness of breast cancer; however, mechanistic studies of the numerous regulators involved in this process are insufficiently thorough. Advanced glycation end products (AGEs) play an important role in the chronic complications of diabetes, but the mechanisms of AGEs in breast cancer are largely unexplored. In this study, we first demonstrate that high AGE levels in breast cancer tissues are associated with the diabetic state and poor patient outcomes. Furthermore, AGEs interact with the receptor for AGEs (RAGE) to promote breast cancer cell migration and invasion. Mechanistically, based on RNA sequencing (RNA-seq) analysis, we reveal that growth arrest and DNA damage gene 45α (GADD45α) is a vital protein upregulated by AGEs through a P53-dependent pathway. Next, GADD45α recruits thymine DNA glycosylase for base excision repair to form the demethylation complex at the promoter region of MMP-9 and enhance MMP-9 transactivation through DNA demethylation. Overall, our results indicate a critical regulatory role of AGEs in patients with breast cancer and diabetes and reveal a novel mechanism of epigenetic modification in promoting breast cancer metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    自发获得或丢失的DNA甲基化发生在植物和动物基因组,和DNA甲基化变化可以导致产生可遗传表型多样性的减数分裂稳定的表观等位基因。然而,目前尚不清楚跨代表观遗传稳定性是否受任何细胞因子的调节.这里,我们检查了野生型和ros1突变拟南芥植物中自发发生的DNA甲基化变异,这些植物从单种子血统繁殖了十代。我们发现Ros1突变体,在活跃的DNA去甲基化中存在缺陷,表现出增加的跨代生殖率。在单个胞嘧啶中,ros1突变导致自发获得的甲基化比丢失的甲基化更多。与具有相似数量的自发获得和丢失甲基化胞嘧啶的野生型相比。始终如一,在ros1突变体中,跨代差异甲基化区域也偏向超甲基化.我们的结果揭示了ROS1DNA去甲基酶对跨代表观遗传稳定性的遗传贡献,并表明ROS1在防止跨代DNA甲基化增加方面可能具有意想不到的监测功能。
    Spontaneous gain or loss of DNA methylation occurs in plant and animal genomes, and DNA methylation changes can lead to meiotically stable epialleles that generate heritable phenotypic diversity. However, it is unclear whether transgenerational epigenetic stability may be regulated by any cellular factors. Here, we examined spontaneously occurring variations in DNA methylation in wild-type and ros1 mutant Arabidopsis plants that were propagated for ten generations from single-seed descent. We found that the ros1 mutant, which is defective in active DNA demethylation, showed an increased transgenerational epimutation rate. The ros1 mutation led to more spontaneously gained methylation than lost methylation at individual cytosines, compared to the wild type which had similar numbers of spontaneously gained and lost methylation cytosines. Consistently, transgenerational differentially methylated regions were also biased toward hypermethylation in the ros1 mutant. Our results reveal a genetic contribution of the ROS1 DNA demethylase to transgenerational epigenetic stability and suggest that ROS1 may have an unexpected surveillance function in preventing transgenerational DNA methylation increases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    早期的研究表明,双酚A(BPA)的发育暴露对成人的大脑生长和行为都有持续的影响。有人认为,BPA可能会阻碍大脑中基因的甲基化编码。在这项研究中,产前BPA暴露后,检测雄性幼鼠海马组织的甲基化变化。在妊娠和哺乳的整个过程中,用媒介物(生育酚剥离的玉米油)或BPA(4、40或400μg/kg·体重/天)处理怀孕的Sprague-Dawley大鼠。在3周龄时,雄性大鼠的后代被安乐死,解剖海马体进行分析。DNA甲基转移酶的表达水平(DNMT1,DNMT3A,和DNMT3B)和DNA去甲基酶(TET1,Gadd45a,Gadd45b,和Apobec1)通过定量实时聚合酶链反应和蛋白质印迹在海马中进行分析,分别。结果表明,产前暴露于BPA上调了雄性大鼠后代海马中与DNA甲基化和去甲基化过程相关的酶的表达。这些结果表明,产前暴露于低剂量的BPA可能潜在地破坏海马中甲基化和去甲基化的平衡。从而扰乱表观遗传修饰。这可能代表了BPA的神经毒性机制。
    Earlier research has demonstrated that developmental exposure to bisphenol A (BPA) has persistent impacts on both adult brain growth and actions. It has been suggested that BPA might obstruct the methylation coding of the genes in the brain. In this study, the methylation changes in the hippocampus tissue of male rat pups were examined following prenatal BPA exposure. Pregnant Sprague-Dawley rats were treated with either vehicle (tocopherol-stripped corn oil) or BPA (4, 40, or 400 μg/kg·body weight/day) throughout the entire duration of gestation and lactation. At 3 weeks of age, the male rat offspring were euthanized, and the hippocampus were dissected out for analysis. The expression levels of DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B) and DNA demethylases (TET1, Gadd45a, Gadd45b, and Apobec1) were analyzed in the hippocampus by means of quantitative real-time polymerase chain reaction and Western blotting, respectively. The results showed that prenatal exposure to BPA upregulated the expression of enzymes associated with DNA methylation and demethylation processes in the hippocampus of male rat offspring. These findings suggest that prenatal exposure to a low dose of BPA could potentially disrupt the balance of methylation and demethylation in the hippocampus, thereby perturbing epigenetic modifications. This may represent a neurotoxicity mechanism of BPA.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    由EB病毒(EBV)诱导的代谢重编程通常反映在癌细胞中观察到的代谢变化。越来越多的证据表明,裂解液的再激活在EBV相关的肿瘤发生中至关重要。这项研究的目的是探讨代谢物变化在EBV相关恶性肿瘤和病毒生命周期控制中的作用。我们首先揭示了EBV(LMP1)加速代谢产物D-2HG的分泌,血清D-2HG水平是NPC的潜在诊断生物标志物。EBV(LMP1)驱动的代谢物变化破坏了全局DNA甲基化和去甲基化的稳态,对活性DNA去甲基化和5hmC含量有明显的抑制作用。我们发现5hmC的丢失表明NPC患者的预后不良,5hmC修饰是EBV再激活的限制因素。我们证实了一种新的EBV再激活抑制剂,α-KG,通过增强5hmC修饰,抑制含有CpG的ZREs和潜伏裂解开关的EBV裂解基因的表达。我们的结果证明了由EBV驱动的代谢物异常通过表观遗传修饰控制病毒裂解再激活的新机制。这项研究提出了阻断EBV再激活的潜在策略,为鼻咽癌的诊断和治疗提供了潜在的靶点。
    Metabolic reprogramming induced by Epstein-Barr virus (EBV) often mirrors metabolic changes observed in cancer cells. Accumulating evidence suggests that lytic reactivation is crucial in EBV-associated oncogenesis. The aim of this study was to explore the role of metabolite changes in EBV-associated malignancies and viral life cycle control. We first revealed that EBV (LMP1) accelerates the secretion of the oncometabolite D-2HG, and serum D-2HG level is a potential diagnostic biomarker for NPC. EBV (LMP1)-driven metabolite changes disrupts the homeostasis of global DNA methylation and demethylation, which have a significantly inhibitory effect on active DNA demethylation and 5hmC content. We found that loss of 5hmC indicates a poor prognosis for NPC patients, and that 5hmC modification is a restriction factor of EBV reactivation. We confirmed a novel EBV reactivation inhibitor, α-KG, which inhibits the expression of EBV lytic genes with CpG-containing ZREs and the latent-lytic switch by enhancing 5hmC modification. Our results demonstrate a novel mechanism of which metabolite abnormality driven by EBV controls the viral lytic reactivation through epigenetic modification. This study presents a potential strategy for blocking EBV reactivation, and provides potential targets for the diagnosis and therapy of NPC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    作为最普遍的表观基因组修饰,N6-甲基腺苷(m6A)通过调控过程在多种疾病中显示出重要的作用,目标RNA的稳定性和翻译。然而,m6A对RNA功能的潜在贡献尚不清楚.这里,我们在肝细胞癌(HCC)中发现了一种功能和预后相关的m6A修饰RNASREBF2-AS1.通过RT-qPCR测量肝癌组织和细胞中SREBF2-AS1和SREBF2的表达。通过甲基化RNA免疫沉淀测定测量SREBF2-AS1的m6A修饰水平。通过增殖研究SREBF2-AS1在HCC进展和索拉非尼耐药中的作用,凋亡,迁移,和细胞活力测定。通过RNA纯化的染色质分离研究了SREBF2-AS1对SREBF2的调控机制,RNA免疫沉淀,CUT&RUN,和亚硫酸氢盐DNA测序分析。我们的研究结果表明,SREBF2-AS1在肝癌组织和细胞中的表达增加,并与HCC患者的不良生存率呈正相关。SREBF2-AS1的m6A修饰水平在HCC中也升高,并与HCC患者的不良预后呈正相关。METTL3和METTL14诱导的m6A修饰通过增加SREBF2-AS1转录物稳定性上调SREBF2-AS1表达。功能分析显示,只有m6A修饰,但未修饰的SREBF2-AS1不能促进HCC进展和索拉非尼耐药。机制研究表明,m6A修饰的SREBF2-AS1将m6A阅读器FXR1和DNA5-甲基胞嘧啶双加氧酶TET1结合并招募到SREBF2启动子,导致SREBF2启动子处的DNA去甲基化和SREBF2转录的上调。功能挽救试验表明,SREBF2是SREBF2-AS1在HCC中致癌作用的关键介质。一起,这项研究表明,m6A修饰的SREBF2-AS1通过诱导DNA去甲基化和SREBF2的转录激活在HCC中发挥致癌作用,并建议m6A修饰的SREBF2-AS1作为HCC的预后生物标志物和治疗靶标。
    As the most prevalent epitranscriptomic modification, N6-methyladenosine (m6A) shows important roles in a variety of diseases through regulating the processing, stability and translation of target RNAs. However, the potential contributions of m6A to RNA functions are unclear. Here, we identified a functional and prognosis-related m6A-modified RNA SREBF2-AS1 in hepatocellular carcinoma (HCC). The expression of SREBF2-AS1 and SREBF2 in HCC tissues and cells was measured by RT-qPCR. m6A modification level of SREBF2-AS1 was measured by methylated RNA immunoprecipitation assay. The roles of SREBF2-AS1 in HCC progression and sorafenib resistance were investigated by proliferation, apoptosis, migration, and cell viability assays. The regulatory mechanisms of SREBF2-AS1 on SREBF2 were investigated by Chromatin isolation by RNA purification, RNA immunoprecipitation, CUT&RUN, and bisulfite DNA sequencing assays. Our findings showed that the expression of SREBF2-AS1 was increased in HCC tissues and cells, and positively correlated with poor survival of HCC patients. m6A modification level of SREBF2-AS1 was also increased in HCC and positively correlated with poor prognosis of HCC patients. METTL3 and METTL14-induced m6A modification upregulated SREBF2-AS1 expression through increasing SREBF2-AS1 transcript stability. Functional assays showed that only m6A-modified, but not non-modified SREBF2-AS1 promoted HCC progression and sorafenib resistance. Mechanistic investigations revealed that m6A-modified SREBF2-AS1 bound and recruited m6A reader FXR1 and DNA 5-methylcytosine dioxygenase TET1 to SREBF2 promoter, leading to DNA demethylation at SREBF2 promoter and the upregulation of SREBF2 transcription. Functional rescue assays showed that SREBF2 was the critical mediator of the oncogenic roles of SREBF2-AS1 in HCC. Together, this study showed that m6A-modified SREBF2-AS1 exerted oncogenic roles in HCC through inducing DNA demethylation and transcriptional activation of SREBF2, and suggested m6A-modified SREBF2-AS1 as a prognostic biomarker and therapeutic target for HCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:Src同源磷酸酪氨酸磷酸酶2(SHP2)与几种癌症类型的进展有关。然而,其在子宫内膜癌(EC)中的功能尚不清楚。这里,我们报道了十11易位3(TET3)介导的DNA去甲基化修饰负责SHP2在EC中的致癌作用,并探讨了其详细机制。
    方法:使用生物信息学工具分析EC组织和对照组织之间的转录组差异,其次是蛋白质-蛋白质相互作用网络的建立。用靶向SHP2的shRNA单独处理EC细胞,或与异孕酮醇联合处理,表皮生长因子受体(EGFR)信号激活因子。使用细胞计数试剂盒-8检查细胞生物学行为,集落形成,流式细胞术,划痕试验,和transwell分析,并计算了醋酸甲羟孕酮/吉非替尼的中位抑制浓度值.证实了TET3与SHP2启动子的结合。选择具有TET3敲低并与SHP2过表达组合的EC细胞以在小鼠中构建肿瘤异种移植物。
    结果:TET3和SHP2在EC细胞中过表达。TET3与SHP2启动子结合,从而增加DNA羟甲基化修饰并激活SHP2以诱导EGFR/细胞外信号调节激酶(ERK)途径。TET3或SHP2的敲除抑制EC细胞恶性侵袭性并损害EGFR/ERK途径。TET3的沉默抑制了EC细胞的致瘤能力,SHP2或异孕酮烯醇的异位表达逆转了TET3敲低对EC细胞生物学活性的抑制作用。
    结论:TET3促进SHP2启动子的DNA去甲基化修饰并激活SHP2,从而激活EGFR/ERK通路并导致EC进展。
    OBJECTIVE: Src homology phosphotyrosin phosphatase 2 (SHP2) has been implicated in the progression of several cancer types. However, its function in endometrial cancer (EC) remains unclear. Here, we report that the ten-eleven translocation 3 (TET3)-mediated DNA demethylation modification is responsible for the oncogenic role of SHP2 in EC and explore the detailed mechanism.
    METHODS: The transcriptomic differences between EC tissues and control tissues were analyzed using bioinformatics tools, followed by protein-protein interaction network establishment. EC cells were treated with shRNA targeting SHP2 alone or in combination with isoprocurcumenol, an epidermal growth factor receptor (EGFR) signaling activator. The cell biological behavior was examined using cell counting kit-8, colony formation, flow cytometry, scratch assay, and transwell assays, and the median inhibition concentration values to medroxyprogesterone acetate/gefitinib were calculated. The binding of TET3 to the SHP2 promoter was verified. EC cells with TET3 knockdown and combined with SHP2 overexpression were selected to construct tumor xenografts in mice.
    RESULTS: TET3 and SHP2 were overexpressed in EC cells. TET3 bound to the SHP2 promoter, thereby increasing the DNA hydroxymethylation modification and activating SHP2 to induce the EGFR/extracellular signal-regulated kinase (ERK) pathway. Knockdown of TET3 or SHP2 inhibited EC cell malignant aggressiveness and impaired the EGFR/ERK pathway. Silencing of TET3 inhibited the tumorigenic capacity of EC cells, and ectopic expression of SHP2 or isoprocurcumenol reversed the inhibitory effect of TET3 knockdown on the biological activity of EC cells.
    CONCLUSIONS: TET3 promoted the DNA demethylation modification in the SHP2 promoter and activated SHP2, thus activating the EGFR/ERK pathway and leading to EC progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    DNA修饰为真核基因组增加了另一层复杂性,以调节基因表达,作为表观遗传标记发挥关键作用。在真核生物中,几十年来,DNA表观遗传修饰的研究一直局限于5mC及其衍生物。然而,在过去的几年中,快速发展的方法见证了DNA修饰库的扩张,包括6mA的识别,5gmC,4mC,和4acC在不同的生物体中。然而,这些DNA修饰是否作为表观遗传标记,需要仔细考虑。在这次审查中,我们试图呈现真核生物中所有DNA表观遗传修饰的全景,强调最近在鉴定新型DNA修饰方面的突破。总结了它们在转录调控中作为潜在表观遗传标记的作用。更重要的是,产生或消除这些DNA修饰的途径,以及所涉及的蛋白质被全面解剖。此外,我们简要讨论了潜在的挑战和前景,在研究新的DNA修饰时应该考虑到这一点。
    DNA modifications add another layer of complexity to the eukaryotic genome to regulate gene expression, playing critical roles as epigenetic marks. In eukaryotes, the study of DNA epigenetic modifications has been confined to 5mC and its derivatives for decades. However, rapid developing approaches have witnessed the expansion of DNA modification reservoirs during the past several years, including the identification of 6mA, 5gmC, 4mC, and 4acC in diverse organisms. However, whether these DNA modifications function as epigenetic marks requires careful consideration. In this review, we try to present a panorama of all the DNA epigenetic modifications in eukaryotes, emphasizing recent breakthroughs in the identification of novel DNA modifications. The characterization of their roles in transcriptional regulation as potential epigenetic marks is summarized. More importantly, the pathways for generating or eliminating these DNA modifications, as well as the proteins involved are comprehensively dissected. Furthermore, we briefly discuss the potential challenges and perspectives, which should be taken into account while investigating novel DNA modifications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    DNA去甲基化参与植物开花的调节,然而,潜在的分子机制在很大程度上仍未被探索。沉默1(ROS1)基因的释放,编码DNA去甲基转移酶,在许多发展过程中起着关键作用。在这项研究中,ROS1基因从菊花中分离,它在树叶中强烈表达,芽和花。ClROS1基因的过表达导致拟南芥的早期开花表型。对转基因植物的RNA-seq分析表明,差异表达的基因(DEG)在昼夜节律途径中显著富集,并且开花的正调节因子,康斯坦斯(CO),被上调了。此外,全基因组亚硫酸氢盐测序(WGBS),用酶McrBC甲基化依赖性消化后的PCR,和亚硫酸氢盐测序PCR(BSP)证实,AtCO启动子的甲基化水平降低,特别是在CG背景下。总的来说,我们的结果表明,ClROS1通过降低AtCO启动子的甲基化水平来加速开花。这些发现阐明了ClROS1介导的DNA去甲基化调节开花的表观遗传机制。
    DNA demethylation is involved in the regulation of flowering in plants, yet the underlying molecular mechanisms remain largely unexplored. The RELEASE OF SILENCING 1 (ROS1) gene, encoding a DNA demethyltransferase, plays key roles in many developmental processes. In this study, the ROS1 gene was isolated from Chrysanthemum lavandulifolium, where it was strongly expressed in the leaves, buds and flowers. Overexpression of the ClROS1 gene caused an early flowering phenotype in Arabidopsis thaliana. RNA-seq analysis of the transgenic plants revealed that differentially expressed genes (DEGs) were significantly enriched in the circadian rhythm pathway and that the positive regulator of flowering, CONSTANS (CO), was up-regulated. Additionally, whole-genome bisulphite sequencing (WGBS), PCR following methylation-dependent digestion with the enzyme McrBC, and bisulfite sequencing PCR (BSP) confirmed that the methylation level of the AtCO promoter was reduced, specifically in CG context. Overall, our results demonstrated that ClROS1 accelerates flowering by reducing the methylation level of the AtCO promoter. These findings clarify the epigenetic mechanism by which ClROS1-mediated DNA demethylation regulates flowering.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号