MECP2

MECP2
  • 文章类型: Journal Article
    罕见的神经系统疾病包括大量具有外周和/或中枢神经系统原发性损害的异质性综合征。这种罕见的疾病可能有重叠的表型,尽管他们独特的遗传病因。罕见神经系统疾病的一个独特方面是它们与改变的表观遗传机制的潜在共同关联。表观遗传机制包括控制基因表达和细胞表型而不改变相应DNA序列组成的调节过程。表观遗传因素包括三种类型的蛋白质,“读者”,作家,DNA和DNA结合蛋白的橡皮擦。因此,许多神经系统疾病的表观遗传损伤可能导致其病理和表现表型。这里,我们的目的是对某些罕见神经系统疾病的一般病因进行全面审查,包括Rett综合征,Prader-Willi综合征,Rubinstein-Taybi综合征,亨廷顿病,和Angelman综合征,关于它们相关的异常表观遗传机制。
    Rare neurological diseases include a vast group of heterogenous syndromes with primary impairment(s) in the peripheral and/or central nervous systems. Such rare disorders may have overlapping phenotypes, despite their distinct genetic etiology. One unique aspect of rare neurological diseases is their potential common association with altered epigenetic mechanisms. Epigenetic mechanisms include regulatory processes that control gene expression and cellular phenotype without changing the composition of the corresponding DNA sequences. Epigenetic factors include three types of proteins, the \"readers, writers, and erasers\" of DNA and DNA-bound proteins. Thus, epigenetic impairments of many neurological diseases may contribute to their pathology and manifested phenotypes. Here, we aim to provide a comprehensive review on the general etiology of selected rare neurological diseases, that include Rett Syndrome, Prader-Willi Syndrome, Rubinstein-Taybi Syndrome, Huntington\'s disease, and Angelman syndrome, with respect to their associated aberrant epigenetic mechanisms.
    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)是由MECP2突变引起的神经发育障碍,它编码甲基CpG结合蛋白2,是许多基因的转录调节因子,包括脑源性神经营养因子(BDNF)。在Mecp2缺陷小鼠的多个脑区,BDNF水平较低,通过实验增加BDNF水平可以改善Mecp2突变小鼠的非典型表型。由于BDNF本身的血脑屏障通透性较低,我们测试了LM22A-4的效果,一种脑渗透剂,BDNF受体TrkB的小分子配体(由Ntrk2编码),对雌性Mecp2杂合(HET)小鼠的海马锥体神经元的树突棘密度和形态以及行为表型的影响。用LM22A-4对Mecp2HET小鼠进行为期4周的全身治疗,将MeCP2表达神经元的脊柱体积恢复到野生型(WT)水平,而缺乏MeCP2的神经元的脊柱体积仍与雌性WT小鼠的神经元相当。雌性Mecp2HET小鼠比WT小鼠更有攻击行为,通过4周的LM22A-4治疗,其水平降低至WT水平。这些数据为新疗法不仅对RTT而且对其他BDNF相关疾病的潜在有用性提供了额外的支持。
    Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in MECP2, which encodes methyl-CpG-binding protein 2, a transcriptional regulator of many genes, including brain-derived neurotrophic factor (BDNF). BDNF levels are lower in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of LM22A-4, a brain-penetrant, small-molecule ligand of the BDNF receptor TrkB (encoded by Ntrk2), on dendritic spine density and form in hippocampal pyramidal neurons and on behavioral phenotypes in female Mecp2 heterozygous (HET) mice. A 4-week systemic treatment of Mecp2 HET mice with LM22A-4 restored spine volume in MeCP2-expressing neurons to wild-type (WT) levels, whereas spine volume in MeCP2-lacking neurons remained comparable to that in neurons from female WT mice. Female Mecp2 HET mice engaged in aggressive behaviors more than WT mice, the levels of which were reduced to WT levels by the 4-week LM22A-4 treatment. These data provide additional support to the potential usefulness of novel therapies not only for RTT but also to other BDNF-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
    结构变体(SV)的检测目前偏向于改变拷贝数的那些。倒位对遗传疾病的相对贡献尚不清楚。在这项研究中,我们分析了来自100,000基因组计划的33,924个罕见疾病家庭的基因组测序数据.从托管超过5亿个SV的数据库中,我们专注于351个基因,其中单倍体功能不全是已确认的疾病机制,并确定了47个超罕见重排,包括倒置(24bp至36.4Mb,20/47从头)。验证使用了许多正交方法,包括回顾性外显子组分析。RNA-seq数据支持六名参与者的各自诊断。表型混合在四个先证中很明显。诊断异常是一个共同的主题(一个人>50年),和特定基因的有针对性的分析已经进行了30%的这些个体,但没有发现。我们为基因内MSH2反演提供了欧洲创始人的正式确认。对于两个具有涉及MECP2突变热点的复杂SV的个体,使用长读数测序解决了模糊的SV结构,影响临床解释。在一个患有Kantaputra型中膜发育不良的家庭中发现了HOXD11-13的从头倒置。最后,一个复杂的易位干扰APC并涉及9个重排的节段,证实了3个家庭成员的临床诊断,并解决了一个患有单个息肉的兄弟姐妹的难题.总的来说,倒置在罕见疾病中起着很小但值得注意的作用,可能解释了大约1/750个家庭在不同临床队列中的病因。
    Detection of structural variants (SVs) is currently biased toward those that alter copy number. The relative contribution of inversions toward genetic disease is unclear. In this study, we analyzed genome sequencing data for 33,924 families with rare disease from the 100,000 Genomes Project. From a database hosting >500 million SVs, we focused on 351 genes where haploinsufficiency is a confirmed disease mechanism and identified 47 ultra-rare rearrangements that included an inversion (24 bp to 36.4 Mb, 20/47 de novo). Validation utilized a number of orthogonal approaches, including retrospective exome analysis. RNA-seq data supported the respective diagnoses for six participants. Phenotypic blending was apparent in four probands. Diagnostic odysseys were a common theme (>50 years for one individual), and targeted analysis for the specific gene had already been performed for 30% of these individuals but with no findings. We provide formal confirmation of a European founder origin for an intragenic MSH2 inversion. For two individuals with complex SVs involving the MECP2 mutational hotspot, ambiguous SV structures were resolved using long-read sequencing, influencing clinical interpretation. A de novo inversion of HOXD11-13 was uncovered in a family with Kantaputra-type mesomelic dysplasia. Lastly, a complex translocation disrupting APC and involving nine rearranged segments confirmed a clinical diagnosis for three family members and resolved a conundrum for a sibling with a single polyp. Overall, inversions play a small but notable role in rare disease, likely explaining the etiology in around 1/750 families across heterogeneous clinical cohorts.
    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
    静止(G0)的维持和退出对于哺乳动物的组织稳态和再生至关重要。这里,我们表明,甲基-CpG结合蛋白2(Mecp2)的表达是细胞周期依赖性的,并且在培养的细胞和损伤诱导的肝再生小鼠模型中负调节静止退出。具体来说,随着Mecp2缺失加速,Mecp2的急性减少是有效的静止退出所必需的,而Mecp2的过表达延迟了静止退出,Mecp2条件性敲除后强制表达Mecp2可挽救细胞周期重新进入。E3连接酶Nedd4介导Mecp2的泛素化和降解,从而促进静止退出。一项全基因组研究揭示了Mecp2在通过转录激活代谢基因同时抑制增殖相关基因来防止静止退出中的双重作用。特别是两个核受体的破坏,Rara或Nr1h3,加速静止出口,模仿Mecp2耗竭表型。我们的研究揭示了Mecp2作为静止退出和组织再生的重要调节剂的先前未被认可的作用。
    Quiescence (G0) maintenance and exit are crucial for tissue homeostasis and regeneration in mammals. Here, we show that methyl-CpG binding protein 2 (Mecp2) expression is cell cycle-dependent and negatively regulates quiescence exit in cultured cells and in an injury-induced liver regeneration mouse model. Specifically, acute reduction of Mecp2 is required for efficient quiescence exit as deletion of Mecp2 accelerates, while overexpression of Mecp2 delays quiescence exit, and forced expression of Mecp2 after Mecp2 conditional knockout rescues cell cycle reentry. The E3 ligase Nedd4 mediates the ubiquitination and degradation of Mecp2, and thus facilitates quiescence exit. A genome-wide study uncovered the dual role of Mecp2 in preventing quiescence exit by transcriptionally activating metabolic genes while repressing proliferation-associated genes. Particularly disruption of two nuclear receptors, Rara or Nr1h3, accelerates quiescence exit, mimicking the Mecp2 depletion phenotype. Our studies unravel a previously unrecognized role for Mecp2 as an essential regulator of quiescence exit and tissue regeneration.
    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)是一种无法治愈的进行性X连锁神经发育障碍。RTT患者在18月龄内表现出疾病相关症状,包括发育退化,逐渐失去有用的手部动作,呼吸困难,伴随着神经损伤,癫痫发作,震颤,和精神残疾。Rett综合征也与代谢异常有关,抗糖尿病药物二甲双胍被认为是一种潜在的选择药物,副作用低或无副作用。以前,我们发现二甲双胍在人脑细胞系中的体外暴露诱导MECP2E1转录本,大脑中MECP2基因的显性同工型,导致RTT的突变。这里,我们报道了二甲双胍对小鼠的分子影响。通过免疫印迹对雄性和雌性小鼠的特定脑区进行蛋白质分析,表明二甲双胍在海马中诱导MeCP2,以性别依赖的方式。其他实验证实,二甲双胍对MeCP2靶标“BDNF”的调节作用是大脑区域依赖性和性别特异性的。核糖体蛋白S6(磷酸化和非磷酸化形式)的测量证实了二甲双胍在肝脏中的性别依赖性作用。我们的结果可以帮助更好地理解二甲双胍在雄性和雌性成年小鼠不同脑区的分子影响。同时为其在Rett综合征治疗策略中的潜力提供了一些见解。
    Rett Syndrome (RTT) is a progressive X-linked neurodevelopmental disorder with no cure. RTT patients show disease-associated symptoms within 18 months of age that include developmental regression, progressive loss of useful hand movements, and breathing difficulties, along with neurological impairments, seizures, tremor, and mental disability. Rett Syndrome is also associated with metabolic abnormalities, and the anti-diabetic drug metformin is suggested to be a potential drug of choice with low or no side-effects. Previously, we showed that in vitro exposure of metformin in a human brain cell line induces MECP2E1 transcripts, the dominant isoform of the MECP2 gene in the brain, mutations in which causes RTT. Here, we report the molecular impact of metformin in mice. Protein analysis of specific brain regions in the male and female mice by immunoblotting indicated that metformin induces MeCP2 in the hippocampus, in a sex-dependent manner. Additional experiments confirm that the regulatory role of metformin on the MeCP2 target \"BDNF\" is brain region-dependent and sex-specific. Measurement of the ribosomal protein S6 (in both phosphorylated and unphosphorylated forms) confirms the sex-dependent role of metformin in the liver. Our results can help foster a better understanding of the molecular impact of metformin in different brain regions of male and female adult mice, while providing some insight towards its potential in therapeutic strategies for the treatment of Rett Syndrome.
    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
    简介:Rett综合征(RTT,MIM#312750)是一种罕见的遗传性疾病,可导致发育退化和严重残疾,由MECP2基因的致病性变异引起。RTT的诊断基于临床特征,根据资源和访问,关于分子确认。拉丁美洲患者的分子诊断信息很少,主要是由于基因组测试的可用性和覆盖范围有限。这项初步研究旨在实施基因组测试,并在一组具有RTT临床诊断的智利患者中表征临床和分子发现。方法:28例疑似RTT患者通过SOPHiAGenetics使用临床外显子组溶液TMCES_V2进行表型表征和分子检测。使用商业生物信息学平台分析数据,SOPHIADDMTM。一个由34个基因组成的虚拟小组,包括MECP2和其他在RTT鉴别诊断中的基因,用于优先考虑初始分析,然后评估完整的外显子组序列数据。结果:12例患者(42.8%的参与者)有MECP2变异,其中11例(39.2%)被解释为致病性/可能致病性(P/LP)。从而证实了RTT在其中的诊断。另外8名患者(28.5%)在其他9个基因中携带10个变异。其中四个变体被解释为P/LP(14.2%)(GRIN2B,MADD,TRPM3和ZEB2)导致替代神经发育诊断,六个被认为具有不确定的意义。8例患者未发现明显的候选变异。讨论:这项研究允许一半的参与者得到诊断。超过三分之一的人证实了RTT的诊断,而其他人则被发现患有替代性神经发育障碍。需要进一步评估以确定结果为阴性或不确定的原因。这些信息对患者很有用,家庭,和临床医生指导临床管理,自RTT新疗法的开发以来,更是如此。我们还展示了在资源有限的环境中实施逐步方法进行基因组测试的可行性。
    Introduction: Rett syndrome (RTT, MIM #312750) is a rare genetic disorder that leads to developmental regression and severe disability and is caused by pathogenic variants in the MECP2 gene. The diagnosis of RTT is based on clinical features and, depending on resources and access, on molecular confirmation. There is scarce information on molecular diagnosis from patients in Latin America, mostly due to limited availability and coverage of genomic testing. This pilot study aimed to implement genomic testing and characterize clinical and molecular findings in a group of Chilean patients with a clinical diagnosis of RTT. Methods: Twenty-eight patients with suspected RTT underwent characterization of phenotypic manifestations and molecular testing using Clinical Exome SolutionTM CES_V2 by SOPHiA Genetics. Data was analyzed using the commercial bioinformatics platform, SOPHiA DDMTM. A virtual panel of 34 genes, including MECP2 and other genes that are in the differential diagnosis of RTT, was used to prioritize initial analyses, followed by evaluation of the complete exome sequence data. Results: Twelve patients (42.8% of participants) had variants in MECP2, of which 11 (39.2%) were interpreted as pathogenic/likely pathogenic (P/LP), thus confirming the diagnosis of RTT in them. Eight additional patients (28.5%) harbored ten variants in nine other genes. Four of these variants were interpreted as P/LP (14.2%) (GRIN2B, MADD, TRPM3 and ZEB2) resulting in alternative neurodevelopmental diagnoses, and six were considered of uncertain significance. No evident candidate variant was found for eight patients. Discussion: This study allowed to reach a diagnosis in half of the participants. The diagnosis of RTT was confirmed in over a third of them, while others were found to have alternative neurodevelopmental disorders. Further evaluation is needed to identify the cause in those with negative or uncertain results. This information is useful for the patients, families, and clinicians to guide clinical management, even more so since the development of novel therapies for RTT. We also show the feasibility of implementing a step-wide approach to genomic testing in a setting with limited resources.
    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
    系统性红斑狼疮(SLE)影响中枢神经系统(CNS),导致严重的神经和精神表现,称为神经精神狼疮(NPSLE)。NPSLE临床表现的复杂性和异质性阻碍了对患者疾病病因的直接调查。为NPSLE开发的现有小鼠模型的局限性阻碍了对这种疾病的全面了解。因此,需要识别一个稳健的NPSLE小鼠模型。
    人MeCP2转基因的C57BL/6小鼠(B6。Mecp2Tg1)进行表型分析,包括通过抗原阵列的自身抗体谱分析,通过流式细胞术分析脾免疫细胞的细胞性和活化,和蛋白尿的测量。进行行为测试以探索其神经精神功能。免疫荧光分析用于揭示改变的神经发生和脑部炎症。使用蛋白质印迹检查了与狼疮发病机理有关的各种信号分子。
    B6。Mecp2Tg1表现出升高的蛋白尿和自身抗体的整体增加,尤其是女性B6。Mecp2Tg1小鼠。观察到转基因小鼠中CD3+CD4+T细胞的增加,以及激活的生发中心细胞和激活的CD11b+F4/80+巨噬细胞。此外,转基因小鼠表现出降低的运动活性,焦虑和抑郁加剧,和短期记忆受损。免疫荧光分析显示转基因小鼠的肾脏和大脑中IgG沉积和免疫细胞浸润,以及改变的神经发生,激活的小胶质细胞,和受损的血脑屏障(BBB)。此外,发现各种关键信号分子的蛋白质水平在MeCP2过表达时受到差异调节,包括GFAP,BDNF,白蛋白,NCoR1,mTOR,NLRP3。
    集体,这项工作证明了B6。Mecp2Tg1小鼠表现出狼疮样表型以及强大的中枢神经系统功能障碍,表明它作为一种新的NPSLE动物模型的实用性。
    Systemic Lupus Erythematosus (SLE) impacts the central nervous system (CNS), leading to severe neurological and psychiatric manifestations known as neuropsychiatric lupus (NPSLE). The complexity and heterogeneity of clinical presentations of NPSLE impede direct investigation of disease etiology in patients. The limitations of existing mouse models developed for NPSLE obstruct a comprehensive understanding of this disease. Hence, the identification of a robust mouse model of NPSLE is desirable.
    C57BL/6 mice transgenic for human MeCP2 (B6.Mecp2Tg1) were phenotyped, including autoantibody profiling through antigen array, analysis of cellularity and activation of splenic immune cells through flow cytometry, and measurement of proteinuria. Behavioral tests were conducted to explore their neuropsychiatric functions. Immunofluorescence analyses were used to reveal altered neurogenesis and brain inflammation. Various signaling molecules implicated in lupus pathogenesis were examined using western blotting.
    B6.Mecp2Tg1 exhibits elevated proteinuria and an overall increase in autoantibodies, particularly in female B6.Mecp2Tg1 mice. An increase in CD3+CD4+ T cells in the transgenic mice was observed, along with activated germinal center cells and activated CD11b+F4/80+ macrophages. Moreover, the transgenic mice displayed reduced locomotor activity, heightened anxiety and depression, and impaired short-term memory. Immunofluorescence analysis revealed IgG deposition and immune cell infiltration in the kidneys and brains of transgenic mice, as well as altered neurogenesis, activated microglia, and compromised blood-brain barrier (BBB). Additionally, protein levels of various key signaling molecules were found to be differentially modulated upon MeCP2 overexpression, including GFAP, BDNF, Albumin, NCoR1, mTOR, and NLRP3.
    Collectively, this work demonstrates that B6.Mecp2Tg1 mice exhibit lupus-like phenotypes as well as robust CNS dysfunctions, suggesting its utility as a new animal model for NPSLE.
    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综合征是一种获得性进行性神经发育障碍,由X连锁MECP2基因的从头突变引起,该基因编码一种多效性蛋白,该蛋白充当全局转录调节因子和染色质修饰因子。Rett综合征主要影响杂合雌性,而受影响的雄性半合子很少存活。Rett综合征的基因疗法已被证明是具有挑战性的,因为需要对表达进行严格的调节,而过表达或过表达不足都是有毒的。MECP2与调控miRNA靶序列的异位表达已取得一定的成功,但是这种方法的持久性仍然未知。在这里,我们评估了基于无核酸酶同源重组(HR)的基因组编辑策略,以纠正MECP2基因中的突变。干细胞衍生的AAVHSC先前已被证明介导无缝和精确的基于HR的基因组编辑。我们测试了基于HR的基因组编辑纠正MECP2基因外显子3和4中致病性突变的能力,并恢复野生型序列,同时保留与MECP2表达相关的所有天然基因组调控元件。因此,有可能解决Rett综合征基因治疗中的一个重要问题。此外,因为突变是直接在基因组水平上编辑的,对于继承编辑基因的子代细胞,预计校正将是持久的。AAVHSCMECP2编辑载体被设计为与靶MECP2区域完全同源,并在外显子4的末端插入无启动子的Venus报道基因。在外显子3和4中携带突变的一组Rett细胞系中对AAVHSC编辑的评估表明成功校正和挽救编辑的MECP2基因的表达。编辑的Rett细胞的序列分析揭示了外显子3和外显子4两者中的突变的成功且准确的校正,并且允许HR交叉事件的定位。只有当交叉事件在5'和3'末端侧翼突变时才观察到成功的校正,但不是当两个交叉都发生在突变的上游或下游。重要的是,我们得出的结论是,在分析的每个Rett细胞系中,致病性突变都得到了成功纠正,证明了基于HR的基因组编辑的治疗潜力。
    Rett syndrome is an acquired progressive neurodevelopmental disorder caused by de novo mutations in the X-linked MECP2 gene which encodes a pleiotropic protein that functions as a global transcriptional regulator and a chromatin modifier. Rett syndrome predominantly affects heterozygous females while affected male hemizygotes rarely survive. Gene therapy of Rett syndrome has proven challenging due to a requirement for stringent regulation of expression with either over- or under-expression being toxic. Ectopic expression of MECP2 in conjunction with regulatory miRNA target sequences has achieved some success, but the durability of this approach remains unknown. Here we evaluated a nuclease-free homologous recombination (HR)-based genome editing strategy to correct mutations in the MECP2 gene. The stem cell-derived AAVHSCs have previously been shown to mediate seamless and precise HR-based genome editing. We tested the ability of HR-based genome editing to correct pathogenic mutations in Exons 3 and 4 of the MECP2 gene and restore the wild type sequence while preserving all native genomic regulatory elements associated with MECP2 expression, thus potentially addressing a significant issue in gene therapy for Rett syndrome. Moreover, since the mutations are edited directly at the level of the genome, the corrections are expected to be durable with progeny cells inheriting the edited gene. The AAVHSC MECP2 editing vector was designed to be fully homologous to the target MECP2 region and to insert a promoterless Venus reporter at the end of Exon 4. Evaluation of AAVHSC editing in a panel of Rett cell lines bearing mutations in Exons 3 and 4 demonstrated successful correction and rescue of expression of the edited MECP2 gene. Sequence analysis of edited Rett cells revealed successful and accurate correction of mutations in both Exons 3 and 4 and permitted mapping of HR crossover events. Successful correction was observed only when the mutations were flanked at both the 5\' and 3\' ends by crossover events, but not when both crossovers occurred either exclusively upstream or downstream of the mutation. Importantly, we concluded that pathogenic mutations were successfully corrected in every Rett line analyzed, demonstrating the therapeutic potential of HR-based genome editing.
    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
    背景:脑卒中后,抑制因子1[RE1]-沉默转录因子通过结合脑启动子(Br)上的RE1位点抑制神经元中Na+/Ca2+交换因子-1(Ncx1)的转录。我们在Ncx1心脏启动子(Ht)序列(Ht-RE1)中确定了一个新的推定RE1位点,该位点参与神经元Ncx1转录。因为REST在不同的神经元基因上招募DNA-甲基转移酶-1(DNMT1)和MeCP2(甲基-CpG结合蛋白2),我们研究了该复合物在卒中后Ncx1转录调控中的作用。
    结果:在SH-SY5Y细胞中进行的荧光素酶实验表明,Br活性被REST选择性地降低,而Ht活性被DNMT1、MeCP2和REST降低。值得注意的是,Ht-RE1的定点突变阻止了Ncx1的REST依赖性下调。此外,在短暂大脑中动脉闭塞的8周龄雄性野生型小鼠(C57BL/6)的颞顶皮层中,DNMT1、MeCP2和REST与Ht启动子的结合增加,随之而来的DNA启动子超甲基化。侧脑室注射siREST可防止DNMT1/MeCP2结合Ht和Ncx1下调,从而减少中风引起的损伤。始终如一,在接受氧和葡萄糖剥夺加复氧的皮质神经元中,Ncx1敲低抵消了去甲基化剂5-氮杂胞苷诱导的神经元保护作用。对于两个实验组之间的比较,使用学生的t检验,而对于两个以上的实验组,使用了一元方差分析,其次是Tukey或NewmanKeuls.P<0.05时具有统计学意义。
    结论:如果这项研究的结果在人类中得到证实,可以断言DNMT1/MeCP2/REST复合物破坏可能是减少大脑中HtDNA甲基化的新药理学策略,改善中风损伤。
    BACKGROUND: REST (Repressor-Element 1 [RE1]-silencing transcription factor) inhibits Na+/Ca2+exchanger-1 (Ncx1) transcription in neurons through the binding of RE1 site on brain promoter (Br) after stroke. We identified a new putative RE1 site in Ncx1 heart promoter (Ht) sequence (Ht-RE1) that participates in neuronal Ncx1 transcription. Because REST recruits DNA-methyltransferase-1 (DNMT1) and MeCP2 (methyl-CpG binding protein 2) on different neuronal genes, we investigated the role of this complex in Ncx1 transcriptional regulation after stroke.
    RESULTS: Luciferase experiments performed in SH-SY5Y cells demonstrated that Br activity was selectively decreased by REST, whereas Ht activity was reduced by DNMT1, MeCP2, and REST. Notably, site-direct mutagenesis of Ht-RE1 prevented REST-dependent downregulation of Ncx1. Furthermore, in temporoparietal cortex of 8-week-old male wild-type mice (C57BL/6) subjected to transient middle cerebral artery occlusion, DNMT1, MeCP2, and REST binding to Ht promoter was increased, with a consequent DNA promoter hypermethylation. Intracerebroventricular injection of siREST prevented DNMT1/MeCP2 binding to Ht and Ncx1 downregulation, thus causing a reduction in stroke-induced damage. Consistently, in cortical neurons subjected to oxygen and glucose deprivation plus reoxygenation Ncx1 knockdown counteracted neuronal protection induced by the demethylating agent 5-azacytidine. For comparisons between 2 experimental groups, Student\'s t test was used, whereas for more than 2 experimental groups, 1-way ANOVA was used, followed by Tukey or Newman Keuls. Statistical significance was set at P<0.05.
    CONCLUSIONS: If the results of this study are confirmed in humans, it could be asserted that DNMT1/MeCP2/REST complex disruption could be a new pharmacological strategy to reduce DNA methylation of Ht in the brain, ameliorating stroke damage.
    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
    新的遗传工具的出现导致了许多智力和发育障碍的遗传基础的发现。这为研究和治疗发展创造了令人兴奋的机会,和一些遗传性疾病(例如,脊髓性肌萎缩症)最近已使用基于基因的疗法进行治疗。MECP2在X染色体上发现,并调节数千个基因的转录。MECP2基因产物的缺失导致Rett综合征,一种主要在女性身上发现的疾病,以发育回归为特征,运动功能障碍,中线手的刻板印象,自主神经系统功能障碍,癫痫,脊柱侧弯,和自闭症样的行为。MECP2的重复导致MECP2重复综合征(MDS)。MDS主要在男性中发现,并表现为发育迟缓,低张力,自闭症特征,难治性癫痫,和反复呼吸道感染。虽然这两种疾病有几个共同的特点,他们的差异(例如,受影响的性别,发病年龄,基因型/表型相关性)对于区分基于基因的治疗很重要,因为它们需要相反的解决方案。这篇综述探讨了这两种疾病的临床特征,并强调了这些重要的临床差异。
    The emergence of new genetic tools has led to the discovery of the genetic bases of many intellectual and developmental disabilities. This creates exciting opportunities for research and treatment development, and a few genetic disorders (e.g., spinal muscular atrophy) have recently been treated with gene-based therapies. MECP2 is found on the X chromosome and regulates the transcription of thousands of genes. Loss of MECP2 gene product leads to Rett Syndrome, a disease found primarily in females, and is characterized by developmental regression, motor dysfunction, midline hand stereotypies, autonomic nervous system dysfunction, epilepsy, scoliosis, and autistic-like behavior. Duplication of MECP2 causes MECP2 Duplication Syndrome (MDS). MDS is found mostly in males and presents with developmental delay, hypotonia, autistic features, refractory epilepsy, and recurrent respiratory infections. While these two disorders share several characteristics, their differences (e.g., affected sex, age of onset, genotype/phenotype correlations) are important to distinguish in the light of gene-based therapy because they require opposite solutions. This review explores the clinical features of both disorders and highlights these important clinical differences.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号