Mecp2

MECP2
  • 文章类型: Journal Article
    增生性玻璃体视网膜病变(PVR)是一种复杂的疾病,显着有助于复发性视网膜脱离。它的发育受到上皮-间质转化(EMT)的显著影响,其中凋亡作为EMT的调节剂起着至关重要的作用。然而,MeCP2在调节视网膜色素上皮(RPE)细胞凋亡和EMT中的功能及其对PVR发展的影响仍未得到充分了解。因此,我们研究了MeCP2对增殖的影响,迁移,ARPE-19细胞的凋亡和EMT为PVR的病因提供了新的视角。观察重组人MeCP2蛋白和MeCP2敲低诱导ARPE-19细胞的形态学变化。进行伤口愈合测定以验证重组人MeCP2蛋白和MeCP2敲低对ARPE-19细胞迁移的影响。此外,使用CCK-8测定和流式细胞术评估细胞增殖。蛋白质印迹分析,定量逆转录酶-聚合酶链反应(qRT-PCR),和免疫荧光分析进行测量与细胞凋亡相关的蛋白质水平,细胞周期和EMT。Western印迹分析和免疫荧光测定证实MeCP2促进ARPE-19细胞中的EMT形成。CCK-8检测显示MeCP2处理增强了ARPE-19细胞的增殖,而MeCP2敲低抑制ARPE-19细胞增殖。用重组人MeCP2蛋白和MeCP2敲低处理改变了ARPE-19细胞的形态。伤口愈合实验证明MeCP2敲低抑制ARPE-19细胞迁移,和MeCP2处理促进ARPE-19细胞迁移。MeCP2敲低诱导G0/G1期阻滞,抑制细胞生长,和qRT-PCR数据表明细胞周期相关基因的表达降低。在ARPE-19细胞中MeCP2敲低后观察到细胞凋亡增加。总的来说,MeCP2处理刺激细胞增殖,迁移和EMT形成;相反,MeCP2敲低抑制EMT,细胞增殖,迁移和细胞周期G1/S相变,并诱导细胞凋亡。
    Proliferative vitreoretinopathy (PVR) is a complex disease that significantly contributes to recurrent retinal detachment. Its development is notably affected by epithelial-mesenchymal transition (EMT), where apoptosis plays a crucial role as a regulator of EMT. However, the function of MeCP2 in governing apoptosis and EMT in retinal pigment epithelial (RPE) cells and its implications for PVR development have remained inadequately understood. Thus, we investigated the impact of MeCP2 on proliferation, migration, apoptosis and EMT in ARPE-19 cells to provide a fresh perspective on the etiology of PVR. The morphological changes in ARPE-19 cells induced by recombinant human MeCP2 protein and MeCP2 knockdown were observed. Wound healing assay were performed to verify the effects of recombinant human MeCP2 protein and MeCP2 knockdown on ARPE-19 cell migration. Furthermore, cell proliferation was assessed using the CCK-8 assay and flow cytometry. Western blot analysis, quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), and immunofluorescence analysis were conducted to measure the protein levels associated with apoptosis, cell cycle and EMT. Western blot analysis and immunofluorescence assays confirmed that MeCP2 promoted EMT formation in ARPE-19 cells. The CCK-8 assay revealed that MeCP2 treatment enhanced the proliferation of ARPE-19 cells, whereas MeCP2 knockdown inhibited ARPE-19 cell proliferation. Treatment with recombinant human MeCP2 protein and MeCP2 knockdown altered the morphology of ARPE-19 cells. Wound healing assay demonstrated that MeCP2 knockdown inhibited ARPE-19 cell migration, and MeCP2 treatment promoted ARPE-19 cell migration. MeCP2 knockdown induced a G0/G1 phase block, inhibiting cell growth, and qRT-PCR data indicated reduced expression of cell cycle-related genes. Increased apoptosis was observed after MeCP2 knockdown in ARPE-19 cells. Overall, MeCP2 treatment stimulates cell proliferation, migration and EMT formation; conversely, MeCP2 knockdown inhibits EMT, cell proliferation, migration and cell cycle G1/S phase transition, and induces apoptosis.
    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
    关于MECP2功能及其在Rett综合征(RTT)中的意义的研究传统上集中在神经元上。这里,使用人类胚胎干细胞(hESC)系,我们对MECP2功能丧失进行建模,以探讨其对星形胶质细胞(AST)发育和脑功能障碍的影响.RTThESC衍生的大脑类器官的超微结构分析显示,与对照组(CTRs)相比,线粒体明显较小,在神经胶质和神经元中尤其明显。采用多元组学方法,我们观察到,与神经元相比,ASTs中MECP2突变后,核编码线粒体基因亚组的基因表达和可及性增加.对hESC衍生的AST的分析显示,线粒体呼吸减少,RTT与CTR中的三羧酸循环和电子传递链中的关键蛋白发生了变化。此外,RTTAST在基础条件下表现出增加的细胞溶质氨基酸,随着能源需求的增加而耗尽。值得注意的是,从RTTAST中分离出的线粒体在转移到皮质神经元时表现出增加的活性氧并影响神经元活性。这些发现强调了MECP2突变对ASTs和神经元中线粒体和代谢途径的不同影响,提示AST线粒体功能失调可能通过影响神经元健康而导致RTT病理生理学。
    Studies on MECP2 function and its implications in Rett Syndrome (RTT) have traditionally centered on neurons. Here, using human embryonic stem cell (hESC) lines, we modeled MECP2 loss-of-function to explore its effects on astrocyte (AST) development and dysfunction in the brain. Ultrastructural analysis of RTT hESC-derived cerebral organoids revealed significantly smaller mitochondria compared to controls (CTRs), particularly pronounced in glia versus neurons. Employing a multiomics approach, we observed increased gene expression and accessibility of a subset of nuclear-encoded mitochondrial genes upon mutation of MECP2 in ASTs compared to neurons. Analysis of hESC-derived ASTs showed reduced mitochondrial respiration and altered key proteins in the tricarboxylic acid cycle and electron transport chain in RTT versus CTRs. Additionally, RTT ASTs exhibited increased cytosolic amino acids under basal conditions, which were depleted upon increased energy demands. Notably, mitochondria isolated from RTT ASTs exhibited increased reactive oxygen species and influenced neuronal activity when transferred to cortical neurons. These findings underscore MECP2 mutation\'s differential impact on mitochondrial and metabolic pathways in ASTs versus neurons, suggesting that dysfunctional AST mitochondria may contribute to RTT pathophysiology by affecting neuronal health.
    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
    尽管已观察到Rett综合征(RTT)的长期生存率,关于RTT老年人的信息有限。我们假设延长RTT的寿命与MECP2中与轻度严重程度相关的遗传变异有关,在老年人中,临床特征不会是静态的。为了解决这些假设,我们比较了MECP2变异体的分布和临床严重程度在经典RTT的年轻个体(30岁以下)和老年个体(30岁以上)之间的差异.与预期相反,在较老的队列中观察到重度MECP2变体(R106W)的富集。总体严重程度在队列之间没有差异,但不同队列的具体临床特征不同.从第一次到最后一次访问的总体严重程度在年轻队列中增加,但在老年队列中没有增加。虽然从第一次到最后一次就诊,老年队列中的一些特定临床特征是稳定的,其他人则表现出改善或恶化。这些数据不支持轻度MECP2变异或总体严重程度较低导致RTT寿命延长的假设,但证明了RTT成人的临床特征随年龄增加而变化。需要额外的工作来了解患有RTT的成年人的疾病进展。
    Although long-term survival in Rett syndrome (RTT) has been observed, limited information on older people with RTT exists. We hypothesized that increased longevity in RTT would be associated with genetic variants in MECP2 associated with milder severity, and that clinical features would not be static in older individuals. To address these hypotheses, we compared the distribution of MECP2 variants and clinical severity between younger individuals with Classic RTT (under 30 years old) and older individuals (over 30 years old). Contrary to expectation, enrichment of a severe MECP2 variant (R106W) was observed in the older cohort. Overall severity was not different between the cohorts, but specific clinical features varied between the cohorts. Overall severity from first to last visit increased in the younger cohort but not in the older cohort. While some specific clinical features in the older cohort were stable from the first to the last visit, others showed improvement or worsening. These data do not support the hypothesis that mild MECP2 variants or less overall severity leads to increased longevity in RTT but demonstrate that clinical features change with increasing age in adults with RTT. Additional work is needed to understand disease progression in adults with RTT.
    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
    “AT-hook”是一种特殊的DNA结合域,与富含AT的序列对应的小沟中的DNA相互作用。该结构域首先在结构因子的HMGA蛋白家族中被描述,后来在各种转录因子和染色质蛋白中被描述,通常与大沟DNA结合域相关。在这次审查中,使用文献检索,我们鉴定了大约一百种含有AT-hook的蛋白质,主要是染色质蛋白和转录因子。在考虑了含AT-hook蛋白的原型之后,HMGA家族,我们回顾了那些已经更详细研究过的,并且涉及各种病理,特别关注癌症。这篇综述表明,AT-hook是一个结构域,它不仅赋予蛋白质与DNA相互作用的能力,而且还赋予蛋白质与RNA和蛋白质相互作用的能力。该结构域可以具有酶活性,并且可以影响大沟DNA结合结构域和染色质对接模块(当存在时)的活性。其活性可以通过翻译后修饰来调节。对含AT钩蛋白功能的未来研究将使我们能够更好地破译它们的功能和对不同病理的贡献,并最终揭示它们的相互影响。
    The \"AT-hook\" is a peculiar DNA-binding domain that interacts with DNA in the minor groove in correspondence to AT-rich sequences. This domain has been first described in the HMGA protein family of architectural factors and later in various transcription factors and chromatin proteins, often in association with major groove DNA-binding domains. In this review, using a literature search, we identified about one hundred AT-hook-containing proteins, mainly chromatin proteins and transcription factors. After considering the prototypes of AT-hook-containing proteins, the HMGA family, we review those that have been studied in more detail and that have been involved in various pathologies with a particular focus on cancer. This review shows that the AT-hook is a domain that gives proteins not only the ability to interact with DNA but also with RNA and proteins. This domain can have enzymatic activity and can influence the activity of the major groove DNA-binding domain and chromatin docking modules when present, and its activity can be modulated by post-translational modifications. Future research on the function of AT-hook-containing proteins will allow us to better decipher their function and contribution to the different pathologies and to eventually uncover their mutual influences.
    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
    Mecp2敲除(KO)小鼠的社会记忆障碍是由于从腹侧海马(vHIP)到内侧前额叶皮层(mPFC)的单突触投射中的神经元活动改变所致。在这个Rett综合征模型中海马网络是过度活跃的,这种非典型的神经元活动通过这种单突触投射传播到mPFC,导致mPFC网络活动和社交记忆缺陷的改变。然而,在Mecp2KO小鼠中,vHIP锥体神经元(PYR)和mPFCPYR与小白蛋白中间神经元(PV-IN)之间的投射内细胞功能障碍导致社会记忆障碍的潜在机制尚未阐明.我们使用新的4室社交记忆竞技场证实了Mecp2KO小鼠的社交记忆(而不是社交能力)缺陷,旨在最大程度地减少在社交互动过程中同时对Ca2传感器信号进行体内光纤测光所需的束缚对光纤的影响。野生型(WT)小鼠的mPFCPYR在探索新型玩具小鼠以及与熟悉和新型小鼠的相互作用期间显示Ca2信号幅度增加,而Mecp2KO小鼠的PYRs在仅与活小鼠相互作用时显示较小的Ca2+信号。另一方面,与PYR中的信号相比,Mecp2KO小鼠的mPFCPV-INs在与熟悉的笼伴侣相互作用期间显示出更大的Ca2信号,在WT小鼠中不存在差异。这些观察结果表明,在社会互动过程中,Mecp2KO小鼠的mPFC网络中的抑制和兴奋异常增强,可能导致他们在社会记忆中的赤字。
    Social memory impairments in Mecp2 knockout (KO) mice result from altered neuronal activity in the monosynaptic projection from the ventral hippocampus (vHIP) to the medial prefrontal cortex (mPFC). The hippocampal network is hyperactive in this model for Rett syndrome, and such atypically heightened neuronal activity propagates to the mPFC through this monosynaptic projection, resulting in altered mPFC network activity and social memory deficits. However, the underlying mechanism of cellular dysfunction within this projection between vHIP pyramidal neurons (PYR) and mPFC PYRs and parvalbumin interneurons (PV-IN) resulting in social memory impairments in Mecp2 KO mice has yet to be elucidated. We confirmed social memory (but not sociability) deficits in Mecp2 KO mice using a new 4-chamber social memory arena, designed to minimize the impact of the tethering to optical fibers required for simultaneous in vivo fiber photometry of Ca2+-sensor signals during social interactions. mPFC PYRs of wildtype (WT) mice showed increases in Ca2+ signal amplitude during explorations of a novel toy mouse and interactions with both familiar and novel mice, while PYRs of Mecp2 KO mice showed smaller Ca2+ signals during interactions only with live mice. On the other hand, mPFC PV-INs of Mecp2 KO mice showed larger Ca2+ signals during interactions with a familiar cage-mate compared to those signals in PYRs, a difference absent in the WT mice. These observations suggest atypically heightened inhibition and impaired excitation in the mPFC network of Mecp2 KO mice during social interactions, potentially driving their deficit in social memory.
    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
    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

       PDF(Pubmed)

  • 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

公众号