MeCP2E2

  • 文章类型: Journal Article
    表观遗传因子甲基-CpG-结合蛋白2(MeCP2)是结合甲基化DNA分子(5-甲基胞嘧啶和5-羟甲基胞嘧啶)并控制基因转录的核蛋白。MeCP2是一种重要的转录因子,在大脑中以剂量依赖的方式起作用;因此,其在脑细胞中的最佳表达水平是重要的。因此,它的表达失调,以及功能的增益或丧失突变,导致神经发育受损,脑细胞的结构和功能受损,特别是在神经元中。其他人和我们的研究已经表征了两种公认的MeCP2同工型:MeCP2E1和MeCP2E2。我们已经报道了在Daoy髓母细胞瘤脑细胞中,MeCP2E2过表达导致MeCP2E1蛋白降解。MeCP2同种型是否调节Mecp2启动子调节元件仍未探索。我们之前证明了在Daoy细胞中,二甲双胍(一种抗糖尿病药物)诱导MECP2E1转录本。然而,未研究二甲双胍对Mecp2启动子活性的可能影响。这里,我们产生了稳定转导的Daoy细胞报告基因,以表达由Mecp2启动子驱动的EGFP。将转导的细胞分成4个EGFP表达组(R4-至-R7),它们具有不同的EGFP表达强度。我们的结果证实Mecp2启动子在Daoy细胞中具有活性,任何一种同种型的过表达都会抑制Mecp2启动子的活性,通过流式细胞术和荧光素酶报告基因检测。有趣的是,二甲双胍部分缓解了MeCP2E1对Mecp2启动子的抑制作用,流式细胞仪检测。一起来看,我们的数据为MeCP2同工型在启动子水平的调控提供了重要的见解,这可能与大脑的神经生物学具有生物学相关性。
    The epigenetic factor Methyl-CpG-Binding Protein 2 (MeCP2) is a nuclear protein that binds methylated DNA molecules (both 5-methylcytosine and 5-hydroxymethylcytosine) and controls gene transcription. MeCP2 is an important transcription factor that acts in a dose-dependent manner in the brain; thus, its optimal expression level in brain cells is important. As such, its deregulated expression, as well as gain- or loss-of-function mutation, lead to impaired neurodevelopment, and compromised structure and function of brain cells, particularly in neurons. Studies from others and us have characterized two well-recognized MeCP2 isoforms: MeCP2E1 and MeCP2E2. We have reported that in Daoy medulloblastoma brain cells, MeCP2E2 overexpression leads to MeCP2E1 protein degradation. Whether MeCP2 isoforms regulate the Mecp2 promoter regulatory elements remains unexplored. We previously showed that in Daoy cells, metformin (an anti-diabetic drug) induces MECP2E1 transcripts. However, possible impact of metformin on the Mecp2 promoter activity was not studied. Here, we generated stably transduced Daoy cell reporters to express EGFP driven by the Mecp2 promoter. Transduced cells were sorted into four EGFP-expressing groups (R4-to-R7) with different intensities of EGFP expression. Our results confirm that the Mecp2 promoter is active in Daoy cells, and that overexpression of either isoform inhibits the Mecp2 promoter activity, as detected by flow cytometry and luciferase reporter assays. Interestingly, metformin partially relieved the inhibitory effect of MeCP2E1 on the Mecp2 promoter, detected by flow cytometry. Taken together, our data provide important insight towards the regulation of MeCP2 isoforms at the promoter level, which might have biological relevance to the neurobiology of the brain.
    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
    Methyl CpG binding protein 2 (MeCP2) is the main DNA methyl-binding protein in the brain that binds to 5-methylcytosine and 5-hydroxymethyl cytosine. MECP2 gene mutations are the main origin of Rett Syndrome (RTT), a neurodevelopmental disorder in young females. The disease has no existing cure, however, metabolic drugs such as metformin and statins have recently emerged as potential therapeutic candidates. In addition, induced MECP2-BDNF homeostasis regulation has been suggested as a therapy avenue. Here, we analyzed nascent RNA synthesis versus steady state total cellular RNA to study the transcriptional effects of metformin (an anti-diabetic drug) on MECP2 isoforms (E1 and E2) and BNDF in a human brain cell line. Additionally, we investigated the impact of simvastatin (a cholesterol lowering drug) on transcriptional regulation of MECP2E1/E2-BDNF. Metformin was capable of post-transcriptionally inducing BDNF and/or MECP2E1, while transcriptionally inhibiting MECP2E2. In contrast simvastatin significantly inhibited BDNF transcription without significantly impacting MECP2E2 transcripts. Further analysis of ribosomal RNA transcripts confirmed that the drug neither individually nor in combination affected these fundamentally important transcripts. Experimental analysis was completed in conditions of the presence or absence of serum starvation that showed minimal impact for serum deprival, although significant inhibition of steady state MECP2E1 by simvastatin was only detected in non-serum starved cells. Taken together, our results suggest that metformin controls MECP2E1/E2-BDNF transcriptionally and/or post-transcriptionally, and that simvastatin is a potent transcriptional inhibitor of BDNF. The transcriptional effect of these drugs on MECP2E1/E2-BDNF were not additive under these tested conditions, however, either drug may have potential application for related disorders.
    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
    Rett综合征(RTT)是一种严重的,罕见,和进行性发育障碍,患者表现出神经系统消退和自闭症谱系特征。受影响的个体主要是年轻女性,并且超过95%的患者在甲基-CpG-结合蛋白2(MECP2)基因中携带从头突变。虽然大多数RTT患者有MECP2突变(经典RTT),一小部分患者(非典型RTT)可能携带其他基因的基因突变,如细胞周期蛋白依赖性激酶样5(CDKL5)和FOXG1.由于RTT症状的神经学基础,MeCP2功能最初是在神经细胞(神经元)中研究的。然而,后来的研究强调了它在包括神经胶质在内的其他脑细胞类型中的重要性。在这方面,科学家们受益于使用许多不同的细胞系统和具有功能缺失或获得突变的转基因小鼠对疾病进行建模。此外,在人类死后脑组织中的有限研究在RTT病理生物学和疾病机制方面提供了宝贵的发现。MeCP2在大脑中的表达受到严格调控,它的表达改变导致大脑功能异常,在某些自闭症谱系障碍病例中涉及MeCP2。在某些疾病条件下,MeCP2稳态控制受损,在啮齿动物中,其调节涉及调节microRNA(miR132)和脑源性神经营养因子(BDNF)。这里,我们将概述了解RTT疾病的潜在机制和MECP2基因的相关基因突变以及疾病的病理生物学的最新进展,两种研究最多的蛋白质变体(MeCP2E1和MeCP2E2亚型)的作用,以及控制大脑中MeCP2稳态网络的调节机制,包括BDNF和miR132。
    Rett Syndrome (RTT) is a severe, rare, and progressive developmental disorder with patients displaying neurological regression and autism spectrum features. The affected individuals are primarily young females, and more than 95% of patients carry de novo mutation(s) in the Methyl-CpG-Binding Protein 2 (MECP2) gene. While the majority of RTT patients have MECP2 mutations (classical RTT), a small fraction of the patients (atypical RTT) may carry genetic mutations in other genes such as the cyclin-dependent kinase-like 5 (CDKL5) and FOXG1. Due to the neurological basis of RTT symptoms, MeCP2 function was originally studied in nerve cells (neurons). However, later research highlighted its importance in other cell types of the brain including glia. In this regard, scientists benefitted from modeling the disease using many different cellular systems and transgenic mice with loss- or gain-of-function mutations. Additionally, limited research in human postmortem brain tissues provided invaluable findings in RTT pathobiology and disease mechanism. MeCP2 expression in the brain is tightly regulated, and its altered expression leads to abnormal brain function, implicating MeCP2 in some cases of autism spectrum disorders. In certain disease conditions, MeCP2 homeostasis control is impaired, the regulation of which in rodents involves a regulatory microRNA (miR132) and brain-derived neurotrophic factor (BDNF). Here, we will provide an overview of recent advances in understanding the underlying mechanism of disease in RTT and the associated genetic mutations in the MECP2 gene along with the pathobiology of the disease, the role of the two most studied protein variants (MeCP2E1 and MeCP2E2 isoforms), and the regulatory mechanisms that control MeCP2 homeostasis network in the brain, including BDNF and miR132.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Rett Syndrome (RTT) is a rare and progressive neurodevelopmental disorder that is caused by de novo mutations in the X-linked Methyl CpG binding protein 2 (MECP2) gene and is subjected to X-chromosome inactivation. RTT is commonly associated with neurological regression, autistic features, motor control impairment, seizures, loss of speech and purposeful hand movements, mainly affecting females. Different animal and cellular model systems have tremendously contributed to our current knowledge about MeCP2 and RTT. However, the majority of these findings remain unexamined in the brain of RTT patients. Based on previous studies in rodent brains, the highly conserved neuronal microRNA \"miR132\" was suggested to be an inhibitor of MeCP2 expression. The neuronal miR132 itself is induced by Brain Derived Neurotrophic Factor (BDNF), a neurotransmitter modulator, which in turn is controlled by MeCP2. This makes the basis of the MECP2-BDNF-miR132 feedback regulatory loop in the brain. Here, we studied the components of this feedback regulatory network in humans, and its possible impairment in the brain of RTT patients. In this regard, we evaluated the transcript and protein levels of MECP2/MeCP2E1 and E2 isoforms, BDNF/BDNF, and miR132 (both 3p and 5p strands) by real time RT-PCR, Western blot, and ELISA in four different regions of the human RTT brains and their age-, post-mortem delay-, and sex-matched controls. The transcript level of the studied elements was significantly compromised in RTT patients, even though the change was not identical in different parts of the brain. Our data indicates that MeCP2E1/E2-BDNF protein levels did not follow their corresponding transcript trends. Correlational studies suggested that the MECP2E1/E2-BDNF-miR132 homeostasis regulation might not be similarly controlled in different parts of the human brain. Despite challenges in evaluating autopsy samples in rare diseases, our findings would help to shed some light on RTT pathobiology, and obscurities caused by limited studies on MeCP2 regulation in the human brain.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号