MECP2

MECP2
  • 文章类型: Journal Article
    甲基-CpG结合蛋白2(MeCP2)的突变,例如T158M,P152R,R294X,和R306C突变,是大多数Rett综合征(RTT)病例的病因。这些突变通常会导致蛋白质表达的改变,这似乎与细胞核大小的变化有关;然而,这些观察的分子细节知之甚少。使用表达人MeCP2-E1亚型的C2C12细胞系统以及表达这些突变的小鼠模型,我们显示T158M和P152R导致MeCP2蛋白减少,而R306C的变异更温和,和R294X导致整体2.5至3倍的增加。我们还探索了MeCP2PEST结构域在蛋白酶体介导的MeCP2调节中的潜在参与。最后,我们使用R294X突变体来进一步了解在染色质背景下MeCP2和组蛋白H1之间有争议的竞争。有趣的是,在R294X中,MeCP2E1和E2亚型受到不同的影响,其中E1同工型有助于观察到的大部分整体蛋白质增加,而E2减少了一半。MeCP2调节的模式,因此,似乎在两种同工型中受到不同的调节。
    Mutations in methyl-CpG binding protein 2 (MeCP2), such as the T158M, P152R, R294X, and R306C mutations, are responsible for most Rett syndrome (RTT) cases. These mutations often result in altered protein expression that appears to correlate with changes in the nuclear size; however, the molecular details of these observations are poorly understood. Using a C2C12 cellular system expressing human MeCP2-E1 isoform as well as mouse models expressing these mutations, we show that T158M and P152R result in a decrease in MeCP2 protein, whereas R306C has a milder variation, and R294X resulted in an overall 2.5 to 3 fold increase. We also explored the potential involvement of the MeCP2 PEST domains in the proteasome-mediated regulation of MeCP2. Finally, we used the R294X mutant to gain further insight into the controversial competition between MeCP2 and histone H1 in the chromatin context. Interestingly, in R294X, MeCP2 E1 and E2 isoforms were differently affected, where the E1 isoform contributes to much of the overall protein increase observed, while E2 decreases by half. The modes of MeCP2 regulation, thus, appear to be differently regulated in the two isoforms.
    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
    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
    虽然小胶质细胞是中枢神经系统的巨噬细胞,它们的参与不仅限于免疫功能。由于对胎儿组织的访问有限,小胶质细胞在人类发育过程中的作用仍然知之甚少。为了了解小胶质细胞如何影响人类神经发育,甲基-CpG结合蛋白2(MECP2)基因在人小胶质细胞样细胞(MGLs)中被敲除.MGL中MECP2的破坏导致转录和功能扰动,包括吞噬作用受损。健康MGL与MECP2敲除(KO)神经元的共培养拯救了突触发生缺陷,提示小胶质细胞在突触形成中的作用。靶向药物筛选鉴定了CD11b激动剂ADH-503,球状体-MGL共培养物中恢复的吞噬作用和突触形成,显著改善疾病进展,和增加MeCP2无效小鼠的存活率。这些结果揭示了人类小胶质细胞吞噬作用的MECP2特异性调节,并确定了MECP2相关疾病的新型治疗方法。
    Although microglia are macrophages of the central nervous system, their involvement is not limited to immune functions. The roles of microglia during development in humans remain poorly understood due to limited access to fetal tissue. To understand how microglia can impact human neurodevelopment, the methyl-CpG binding protein 2 (MECP2) gene was knocked out in human microglia-like cells (MGLs). Disruption of the MECP2 in MGLs led to transcriptional and functional perturbations, including impaired phagocytosis. The co-culture of healthy MGLs with MECP2-knockout (KO) neurons rescued synaptogenesis defects, suggesting a microglial role in synapse formation. A targeted drug screening identified ADH-503, a CD11b agonist, restored phagocytosis and synapse formation in spheroid-MGL co-cultures, significantly improved disease progression, and increased survival in MeCP2-null mice. These results unveil a MECP2-specific regulation of human microglial phagocytosis and identify a novel therapeutic treatment for MECP2-related conditions.
    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
    表观遗传学是研究基因组和基因表达模式的可遗传变化,这些变化不是由DNA序列的直接变化引起的。这些变化的例子包括对DNA结合的组蛋白的翻译后修饰,DNA甲基化,和重建核架构。总的来说,表观遗传变化提供了一层调控,影响基因的转录活性,同时保持DNA序列不变。已经在先天性心脏病(CHD)患者中发现了影响负责修饰或感知表观遗传标记的酶的序列变异或突变。和表观遗传复合物的小分子抑制剂已显示出有望作为成人心脏病的疗法。此外,具有编码表观遗传酶的基因突变或缺失的转基因小鼠概括了人类心脏病的各个方面。一起来看,这些研究结果表明,表观遗传学领域的发展将为我们理解先天性和成人心脏病提供新的治疗机会.
    Epigenetics is the study of heritable changes to the genome and gene expression patterns that are not caused by direct changes to the DNA sequence. Examples of these changes include posttranslational modifications to DNA-bound histone proteins, DNA methylation, and remodeling of nuclear architecture. Collectively, epigenetic changes provide a layer of regulation that affects transcriptional activity of genes while leaving DNA sequences unaltered. Sequence variants or mutations affecting enzymes responsible for modifying or sensing epigenetic marks have been identified in patients with congenital heart disease (CHD), and small-molecule inhibitors of epigenetic complexes have shown promise as therapies for adult heart diseases. Additionally, transgenic mice harboring mutations or deletions of genes encoding epigenetic enzymes recapitulate aspects of human cardiac disease. Taken together, these findings suggest that the evolving field of epigenetics will inform our understanding of congenital and adult cardiac disease and offer new therapeutic opportunities.
    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
    神经发育障碍PittHopkins综合征(PTHS)引起与Rett综合征(RTT)患者相似的临床症状。然而,RTT由MECP2突变引起,而TCF4基因中的突变导致PTHS。这两种疾病背后的机械共性是未知的,但是他们的共同症状表明,趋同通路水平的破坏可能存在。我们将患者皮肤来源的成纤维细胞重编程为诱导的神经元祖细胞。有趣的是,我们发现,与健康对照组相比,PTHS患者iNPC中MeCP2水平降低,iNPC和i星形胶质细胞均以突变特异性方式显示功能和分化缺陷.当Tcf4+/-小鼠与过表达MeCP2的小鼠遗传杂交时,分子和表型缺陷显着改善,MeCP2在PTHS病理中的强调和重要作用。重要的是,出生后脑室内基因替代疗法,使用表达9型腺相关病毒载体(AAV9)的MeCP2(AAV9。P546.MeCP2)显着改善了iNPC和i星形胶质细胞的功能,并有效改善了Tcf4/-小鼠的组织学和行为缺陷。合并,我们的数据提示MeCP2在PTHS病理和常见通路中的作用之前未知,可能在多种神经发育障碍中受到影响.我们的工作突出了PTHS的潜在新治疗靶点,包括上调MeCP2表达或其下游靶标,潜在的,基于MeCP2的基因治疗。
    The neurodevelopmental disorder Pitt Hopkins syndrome (PTHS) causes clinical symptoms similar to Rett syndrome (RTT) patients. However, RTT is caused by MECP2 mutations whereas mutations in the TCF4 gene lead to PTHS. The mechanistic commonalities underling these two disorders are unknown, but their shared symptomology suggest that convergent pathway-level disruption likely exists. We reprogrammed patient skin derived fibroblasts into induced neuronal progenitor cells. Interestingly, we discovered that MeCP2 levels were decreased in PTHS patient iNPCs relative to healthy controls and that both iNPCs and iAstrocytes displayed defects in function and differentiation in a mutation-specific manner. When Tcf4+/- mice were genetically crossed with mice overexpressing MeCP2, molecular and phenotypic defects were significantly ameliorated, underlining and important role of MeCP2 in PTHS pathology. Importantly, post-natal intracerebroventricular gene replacement therapy with adeno-associated viral vector serotype 9 (AAV9)-expressing MeCP2 (AAV9.P546.MeCP2) significantly improved iNPC and iAstrocyte function and effectively ameliorated histological and behavioral defects in Tcf4+/- mice. Combined, our data suggest a previously unknown role of MeCP2 in PTHS pathology and common pathways that might be affected in multiple neurodevelopmental disorders. Our work highlights potential novel therapeutic targets for PTHS, including upregulation of MeCP2 expression or its downstream targets or, potentially, MeCP2-based gene therapy.
    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
    Nodding综合征是一种与神经炎症和tau蛋白病相关的癫痫性脑病。最初是小儿脑部疾病,与甲基-CpG结合蛋白2(MECP2)重复综合征有一些临床重叠,影响了某些贫困的东非社区,同时发生了当地的内战和国内流离失所,迫使人们依赖受污染的食物和水的条件。某些生物毒素(淡水蓝藻毒素加/减霉菌毒素)与神经炎症在Nodding综合征中的潜在作用,兴奋毒性,Tau病法,和MECP2失调特性,在这里是第一次考虑。
    Nodding syndrome is an epileptic encephalopathy associated with neuroinflammation and tauopathy. This initially pediatric brain disease, which has some clinical overlap with Methyl-CpG-binding protein 2 (MECP2) Duplication Syndrome, has impacted certain impoverished East African communities coincident with local civil conflict and internal displacement, conditions that forced dependence on contaminated food and water. A potential role in Nodding syndrome for certain biotoxins (freshwater cyanotoxins plus/minus mycotoxins) with neuroinflammatory, excitotoxic, tauopathic, and MECP2-dysregulating properties, is considered here for the first time.
    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
    目的:甲基CpG结合蛋白2(MECP2)重复综合征是一种罕见的X连锁基因组疾病,主要影响男性,通常表现为癫痫和自闭症谱系障碍(ASD)合并症。转基因系MeCP2Tg1用于模拟MECP2重复综合征,并显示自闭症-癫痫并存。先前的工作表明,兴奋性/抑制性(E/I)失衡是癫痫和ASD的潜在共同机制。投射神经元和小白蛋白(PV)中间神经元占海马中E/I平衡的大部分。因此,我们探讨了MeCP2Tg1小鼠海马中投射和PV+神经元的结构变化是如何发生的,以及这些形态学变化是否有助于癫痫易感性.
    方法:我们使用设计药物小鼠模型专门激活的中间神经元Designer受体来抑制海马中的抑制性神经元,以验证MeCP2Tg1的癫痫易感性(FVB,一种近交系,称为对Friend白血病病毒敏感)小鼠。记录脑电图以定义癫痫发作。我们在MeCP2Tg1(FVB):CaMKIIα-Cre(C57BL/6)小鼠或MeCP2Tg1:PV-Cre(C57BL/6)小鼠及其同窝对照中进行了病毒的眼眶后注射,以特异性标记投射和PV神经元进行结构分析。
    结果:MeCP2Tg1小鼠癫痫易感性增加。MeCP2Tg1小鼠海马中PV神经元数量减少,树突复杂性降低。与野生型小鼠相比,MeCP2Tg1小鼠的树突复杂性增加,MeCP2Tg1小鼠齿状回总树突棘密度也增加。MeCP2Tg1小鼠的CA1中总树突棘密度增加。
    结论:MeCP2的过表达可能会破坏关键的信号通路,导致PV中间神经元的树突复杂性降低,投射神经元的树突脊柱密度增加。与MeCP2相关的兴奋性和抑制性神经元结构的这种相互调节暗示了其作为癫痫发展中的潜在靶标的重要性,并为自闭症和癫痫的共同发生提供了新的视角。
    OBJECTIVE: Methyl CpG-binding protein 2 (MECP2) duplication syndrome is a rare X-linked genomic disorder affecting predominantly males, which is usually manifested as epilepsy and autism spectrum disorder (ASD) comorbidity. The transgenic line MeCP2Tg1 was used for mimicking MECP2 duplication syndrome and showed autism-epilepsy co-occurrence. Previous works suggested that the excitatory/inhibitory (E/I) imbalance is a potential common mechanism for both epilepsy and ASD. The projection neurons and parvalbumin (PV) interneurons account for the majority of E/I balance in the hippocampus. Therefore, we explored how structural changes of projection and PV+ neurons occur in the hippocampus of MeCP2Tg1 mice and whether these morphological changes contribute to epilepsy susceptibility.
    METHODS: We used the interneuron Designer receptors exclusively activated by designer drugs mouse model to inhibit inhibitory neurons in the hippocampus to verify the epilepsy susceptibility of MeCP2Tg1 (FVB, an inbred strain named as sensitivity to Friend leukemia virus) mice. Electroencephalograms were recorded for the definition of seizure. We performed retro-orbital injection of virus in MeCP2Tg1 (FVB):CaMKIIα-Cre (C57BL/6) mice or MeCP2Tg1:PV-Cre (C57BL/6) mice and their littermate controls to specifically label projection and PV+ neurons for structural analysis.
    RESULTS: Epilepsy susceptibility was increased in MeCP2Tg1 mice. There was a reduced number of PV neurons and reduced dendritic complexity in the hippocampus of MeCP2Tg1 mice. The dendritic complexity in MeCP2Tg1 mice was increased compared to wild-type mice, and total dendritic spine density in dentate gyrus of MeCP2Tg1 mice was also increased. Total dendritic spine density was increased in CA1 of MeCP2Tg1 mice.
    CONCLUSIONS: Overexpression of MeCP2 may disrupt crucial signaling pathways, resulting in decreased dendritic complexity of PV interneurons and increased dendritic spine density of projection neurons. This reciprocal modulation of excitatory and inhibitory neuronal structures associated with MeCP2 implies its significance as a potential target in the development of epilepsy and offers a novel perspective on the co-occurrence of autism and epilepsy.
    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
    迄今为止,Rett综合征(RTT),主要由X连锁MECP2基因突变引起的遗传性疾病,越来越被认为是一种广谱病理学,而不仅仅是神经发育疾病,由于大量的外周共病和受损的代谢途径,影响患者。改变的分子过程包括受损的线粒体功能,受干扰的氧化还原稳态,慢性亚临床炎症和不适当的胆固醇代谢。10年前首次定义了持续性亚临床炎症,作为以前无法识别的RTT功能,在病理进展和表型严重程度的调节中起作用。鉴于此,本工作旨在回顾目前有关RTT中慢性炎症状态和免疫/炎症功能改变的知识,以及调查这种疾病背后的新机制,特别关注关于炎症小体系统的最新发现,自身免疫反应和肠道微生物和真菌群。在这些基础上,虽然还需要进一步的研究,能够重新建立足够免疫/炎症反应的未来治疗策略可能是RTT患者的潜在方法.
    To date, Rett syndrome (RTT), a genetic disorder mainly caused by mutations in the X-linked MECP2 gene, is increasingly considered a broad-spectrum pathology, instead of just a neurodevelopmental disease, due to the multitude of peripheral co-morbidities and the compromised metabolic pathways, affecting the patients. The altered molecular processes include an impaired mitochondrial function, a perturbed redox homeostasis, a chronic subclinical inflammation and an improper cholesterol metabolism. The persistent subclinical inflammatory condition was first defined ten years ago, as a previously unrecognized feature of RTT, playing a role in the pathology progress and modulation of phenotypical severity. In light of this, the present work aims at reviewing the current knowledge on the chronic inflammatory status and the altered immune/inflammatory functions in RTT, as well as investigating the emerging mechanisms underlying this condition with a special focus on the latest findings about inflammasome system, autoimmunity responses and intestinal micro- and mycobiota. On these bases, although further research is needed, future therapeutic strategies able to re-establish an adequate immune/inflammatory response could represent potential approaches for RTT patients.
    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
    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)

公众号