fragile X mental retardation protein

脆性 X 智力迟钝蛋白
  • 文章类型: Journal Article
    脆性X综合征(FXS)的症状,由Fmr1的单个基因突变引起的,越来越多地与大脑皮层内星形胶质细胞信号的紊乱有关。我们最近证明了嘌呤能信号通路,利用核苷三磷酸及其代谢产物促进双向神经胶质和神经胶质-神经元相互作用,在源自FXS的Fmr1敲除(KO)小鼠模型的皮质星形胶质细胞中上调。提高Fmr1KOP2Y嘌呤能受体水平与细胞内钙释放延长相关,突触蛋白分泌升高,和发育电路的过度活跃。然而,由于相对缺乏可用于测量嘌呤和嘧啶的灵敏和可重现的定量方法,确定Fmr1KO星形胶质细胞中这些因子的丰度是有限的.因此,我们开发了一种与质谱联用的亲水相互作用液相色谱方案,以比较野生型和Fmr1KO小鼠星形胶质细胞之间细胞内和细胞外嘌呤能分子的丰度。UDP浓度的显著差异,ATP,AMP,相对于WT,在Fmr1KO星形胶质细胞内发现了腺苷细胞内储存。与从WT星形胶质细胞收集的培养基相比,Fmr1KO星形胶质细胞条件培养基中腺苷的细胞外水平也显着升高。星形细胞膜结合的CD39外核苷酸酶的糖基化,促进突触释放后的配体分解,在Fmr1KO星形胶质细胞培养物中也升高。一起,这些差异表明Fmr1KO皮质星形胶质细胞内嘌呤能信号系统的进一步失调,可能导致FXS嘌呤能受体激活和细胞病理学的显着改变。
    The symptoms of fragile X syndrome (FXS), caused by a single gene mutation to Fmr1, have been increasingly linked to disordered astrocyte signalling within the cerebral cortex. We have recently demonstrated that the purinergic signalling pathway, which utilizes nucleoside triphosphates and their metabolites to facilitate bidirectional glial and glial-neuronal interactions, is upregulated in cortical astrocytes derived from the Fmr1 knockout (KO) mouse model of FXS. Heightened Fmr1 KO P2Y purinergic receptor levels were correlated with prolonged intracellular calcium release, elevated synaptogenic protein secretion, and hyperactivity of developing circuits. However, due to the relative lack of sensitive and reproducible quantification methods available for measuring purines and pyrimidines, determining the abundance of these factors in Fmr1 KO astrocytes was limited. We therefore developed a hydrophilic interaction liquid chromatography protocol coupled with mass spectrometry to compare the abundance of intracellular and extracellular purinergic molecules between wildtype and Fmr1 KO mouse astrocytes. Significant differences in the concentrations of UDP, ATP, AMP, and adenosine intracellular stores were found within Fmr1 KO astrocytes relative to WT. The extracellular level of adenosine was also significantly elevated in Fmr1 KO astrocyte-conditioned media in comparison to media collected from WT astrocytes. Glycosylation of the astrocyte membrane-bound CD39 ectonucleotidase, which facilitates ligand breakdown following synaptic release, was also elevated in Fmr1 KO astrocyte cultures. Together, these differences demonstrated further dysregulation of the purinergic signalling system within Fmr1 KO cortical astrocytes, potentially leading to significant alterations in FXS purinergic receptor activation and cellular pathology.
    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
    脆性X相关震颤/共济失调综合征(FXTAS)是一种年龄相关的神经退行性疾病,由X染色体上FMR1基因的预突变引起。尽管FXTAS对身体和认知的影响非常普遍,没有研究检查有症状的男性和女性的语言,限制效用作为FXTAS临床试验中的结果指标。这项工作的目的是确定(a)具有FXTAS症状的男性和女性FMR1前突变携带者在语言使用上的差异程度,以及(b)语言产生是否可以预测FXTAS症状。31个具有FMR1前突变的个体(21M,10F),年龄58-85岁,有FXTAS的一些症状,是从一项更大的横断面研究中招募的。参与者完成了五分钟的单一语言样本。评估语言成绩单的流失率,词汇语义,语法,和语速。多变量线性和序数回归用于预测FXTAS相关症状,认知功能,和执行功能。男性和女性在语言使用上没有差异。语言产生预测FXTAS症状严重程度,认知功能,和执行功能。语言产生困难可能与FXTAS相关症状同时发生,并且可能是未来临床试验中可行的结果指标。需要未来的研究。
    Fragile X-associated tremor/ataxia syndrome (FXTAS) is an age-related neurodegenerative disorder caused by a premutation of the FMR1 gene on the X chromosome. Despite the pervasive physical and cognitive effects of FXTAS, no studies have examined language in symptomatic males and females, limiting utility as an outcome measure in clinical trials of FXTAS. The goal of this work is to determine (a) the extent to which male and female FMR1 premutation carriers with FXTAS symptoms differ in their language use and (b) whether language production predicts FXTAS symptoms. Thirty-one individuals with the FMR1 premutation (21M, 10F), ages 58-85 years with some symptoms of FXTAS, were recruited from a larger cross-sectional study. Participants completed a five-minute monologic language sample. Language transcripts were assessed for rate of dysfluencies, lexical-semantics, syntax, and speech rate. Multivariable linear and ordinal regressions were used to predict FXTAS-associated symptoms, cognitive functioning, and executive functioning. Males and females did not differ in their language use. Language production predicted FXTAS symptom severity, cognitive functioning, and executive functioning. Language production difficulties may co-occur with FXTAS-associated symptoms and may be a viable outcome measure in future clinical trials, with future research needed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: News
    暂无摘要。
    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
    早期卵巢功能不全(FXPOI)与FMR1基因前突变之间的关系已得到充分证实。近年来,虽然,有人认为后者与低卵巢储备之间存在潜在的关系.为了探索它,我们在巴塞罗那(西班牙)一所大学专科学校转诊中心的一项IVF项目中进行了一项回顾性研究.数据来自2018年1月至2021年12月在我们机构接受FMR1基因检测的385名女性。我们比较了其中93例FMR1基因前突变的患病率,小于35岁,卵巢储备(DOR)减少,特征在于抗苗勒管激素的水平<1.1ng/mL和窦卵泡计数<5;和132个卵子供体通过用作对照的方案筛选。我们发现DOR组(7例(7.69%))的FMR1前突变患病率高于对照组(1例(1.32%))。Fisher精确检验p值=0.012)。我们得出的结论是,与年轻卵子捐赠者代表的普通人群相比,在卵巢储备功能减退的年轻患者中,FMR1基因前突变的发生率较高.尽管这些发现需要在DOR中更大的患者队列中进行进一步的前瞻性验证,他们建议,在临床实践中,应在患有DOR的不育年轻患者中确定FMR1预突变,以便为他们提供足够的遗传咨询。
    The relationship between premature ovarian insufficiency (FXPOI) and premutation in the FMR1 gene is well established. In recent years, though, a potential relationship between the latter and a low ovarian reserve has been suggested. To explore it, we conducted a retrospective study in an IVF program at a university tertiary referral center in Barcelona (Spain). Data were obtained retrospectively from a total of 385 women referred for FMR1 gene testing at our institution from January 2018 to December 2021. We compared the prevalence of FMR1 gene premutation between 93 of them, younger than 35 years, with a diminished ovarian reserve (DOR), characterized by levels of anti-Mullerian hormone < 1.1 ng/mL and antral follicle count < 5; and 132 egg donors screened by protocol that served as the controls. We found a higher prevalence of FMR1 premutation in the DOR group (seven patients (7.69%)) than in the control group (one patient (1.32%)), Fisher-exact test p-value = 0.012). We concluded that compared with the general population represented by young egg donors, the prevalence of FMR1 gene premutation is higher in young patients with a diminished ovarian reserve. Although these findings warrant further prospective validation in a larger cohort of patients within DOR, they suggest that, in clinical practice, FMR1 premutation should be determined in infertile young patients with DOR in order to give them adequate genetic counselling.
    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
    脆性X综合征(FXS)是智力障碍的最常见遗传形式,是由编码脆性X信使核糖核蛋白(FMRP)的基因突变引起的。FMRP是一种进化上保守且富含神经的RNA结合蛋白(RBP),具有RNA编辑功能,RNA转运,蛋白质翻译特异性靶RNA在神经发育中发挥关键作用,包括神经突形态发生的调节,突触可塑性,和认知功能。FMRP的不同生物学功能通过其与不同组的神经元RNA和蛋白质结合配偶体的协同相互作用来调节。这里,我们关注FMRP与microRNA(miRNA)通路组分之间的相互作用。使用果蝇S2细胞模型系统,我们表明,FMRP的果蝇直系同源物(dFMRP)在直接连接到报告mRNA时可以抑制翻译。这种镇压需要AGO1,GW182和MOV10/Armitage的活性,与含miRNA的RNA诱导沉默复合物(miRISC)相关的保守蛋白。此外,我们发现未标记的dFMRP可以与翻译报告分子中的短茎环序列相互作用,外源性miR-958抑制的先决条件。最后,我们证明了dFmr1与GW182在遗传上相互作用以控制神经突形态发生。这些数据表明,dFMRP可能会将miRISC招募到附近的miRNA结合位点,并通过其与miRNA途径的进化保守成分的协同相互作用来抑制翻译。
    Fragile X Syndrome (FXS) is the most common inherited form of intellectual disability and is caused by mutations in the gene encoding the Fragile X messenger ribonucleoprotein (FMRP). FMRP is an evolutionarily conserved and neuronally enriched RNA-binding protein (RBP) with functions in RNA editing, RNA transport, and protein translation. Specific target RNAs play critical roles in neurodevelopment, including the regulation of neurite morphogenesis, synaptic plasticity, and cognitive function. The different biological functions of FMRP are modulated by its cooperative interaction with distinct sets of neuronal RNA and protein-binding partners. Here, we focus on interactions between FMRP and components of the microRNA (miRNA) pathway. Using the Drosophila S2 cell model system, we show that the Drosophila ortholog of FMRP (dFMRP) can repress translation when directly tethered to a reporter mRNA. This repression requires the activity of AGO1, GW182, and MOV10/Armitage, conserved proteins associated with the miRNA-containing RNA-induced silencing complex (miRISC). Additionally, we find that untagged dFMRP can interact with a short stem-loop sequence in the translational reporter, a prerequisite for repression by exogenous miR-958. Finally, we demonstrate that dFmr1 interacts genetically with GW182 to control neurite morphogenesis. These data suggest that dFMRP may recruit the miRISC to nearby miRNA binding sites and repress translation via its cooperative interactions with evolutionarily conserved components of the miRNA pathway.
    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
    脆性X智力低下蛋白(FMRP),RNA结合蛋白(RBP),在人类肿瘤中异常高表达,在肿瘤侵袭中起重要作用,转移和免疫逃避。然而,目前尚无FMRP的小分子抑制剂。在这项研究中,我们开发了第一个FMRP靶向降解剂,该降解剂基于PROteasolsisTArgeting嵌合体(PROTAC)技术,并通过将FMRP靶向G-四链体RNA(sc1)连接到vonHippel-Lindau(VHL)靶向配体肽(命名为sc1-VHLL),构建了异双功能PROTAC。Sc1-VHLL在小鼠和人类癌细胞中通过泛素化途径特异性降解内源性FMRP。FMRP降解显著改变了癌细胞的分泌模式,导致促炎细胞因子的较高表达和较少量的免疫调节内容物。此外,sc1-VHLL,当包封到可电离的脂质体纳米颗粒(LNP)中时,可以有效地靶向肿瘤部位并降解癌细胞中的FMRP。在CT26荷瘤小鼠模型中,FMRP在肿瘤内的降解显著促进了淋巴细胞和CD8T细胞的浸润,降低了Treg细胞的比例,重塑促炎肿瘤微环境,从而将冷肿瘤转化为热肿瘤。当联合免疫检查点阻断(ICB)治疗时,基于sc1-VHLL的治疗显著抑制肿瘤生长。
    Fragile X mental retardation protein (FMRP), an RNA binding protein (RBP), is aberrantly hyper-expressed in human tumors and plays an essential role in tumor invasion, metastasis and immune evasion. However, there is no small-molecule inhibitor for FMRP so far. In this study, we developed the first FMRP-targeting degrader based on PROteolysis TArgeting Chimera (PROTAC) technology and constructed a heterobifunctional PROTAC through linking a FMRP-targeting G-quadruplex RNA (sc1) to a von Hippel-Lindau (VHL)-targeting ligand peptide (named as sc1-VHLL). Sc1-VHLL specifically degraded endogenous FMRP via ubiquitination pathway in both mouse and human cancer cells. The FMRP degradation significantly changed the secretion pattern of cancer cells, resulting in higher expression of pro-inflammatory cytokine and smaller amounts of immunomodulatory contents. Furthermore, sc1-VHLL, when encapsulated into ionizable liposome nanoparticles (LNP) efficiently targeted tumor site and degraded FMRP in cancer cells. In CT26 tumor-bearing mouse model, FMRP degradation within tumors substantially promoted the infiltration of lymphocytes and CD8 T cells and reduced the proportion of Treg cells, reshaping the proinflammatory tumor microenvironment and accordingly transforming cold tumor into hot tumor. When combined with immune checkpoint blockade (ICB) therapy, sc1-VHLL based treatment remarkably inhibited the tumor growth.
    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
    目前尚不清楚mRNA是否在细胞质中发挥结构作用。这里,我们报道了脆性X相关蛋白1(FXR1)网络,存在于整个细胞质中的mRNA蛋白(mRNP)网络,由FXR1介导的非常长的mRNA的包装形成。这些mRNA充当基础缩合物支架并浓缩FXR1分子。FXR1网络包含多个蛋白质结合位点,并充当相互作用蛋白质的信号支架。我们表明,RhoA信号诱导的肌动球蛋白重组必须在激酶及其底物之间提供空间接近性。FXR1的点突变,在其同源FMR1中发现,它们会导致脆性X综合征,扰乱网络。FXR1网络破坏可防止肌动球蛋白重塑-这是调节细胞形状的重要且普遍存在的过程,迁移,和突触功能。我们的发现揭示了细胞质mRNA的结构作用,并显示了FXR1RNA结合蛋白作为FXR1网络的一部分如何充当信号传导反应的组织者。
    It is currently not known whether mRNAs fulfill structural roles in the cytoplasm. Here, we report the fragile X-related protein 1 (FXR1) network, an mRNA-protein (mRNP) network present throughout the cytoplasm, formed by FXR1-mediated packaging of exceptionally long mRNAs. These mRNAs serve as an underlying condensate scaffold and concentrate FXR1 molecules. The FXR1 network contains multiple protein binding sites and functions as a signaling scaffold for interacting proteins. We show that it is necessary for RhoA signaling-induced actomyosin reorganization to provide spatial proximity between kinases and their substrates. Point mutations in FXR1, found in its homolog FMR1, where they cause fragile X syndrome, disrupt the network. FXR1 network disruption prevents actomyosin remodeling-an essential and ubiquitous process for the regulation of cell shape, migration, and synaptic function. Our findings uncover a structural role for cytoplasmic mRNA and show how the FXR1 RNA-binding protein as part of the FXR1 network acts as an organizer of signaling reactions.
    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
    脆性X综合征(FXS),人类智力残疾最常见的遗传形式,是由FMR1基因的功能丧失突变引起的单基因神经发育障碍。FMR1编码脆性X信使核糖核酸蛋白(FMRP),这是一种调节突触蛋白翻译的RNA结合蛋白。FMRP表达的缺失对突触可塑性和功能有许多重要影响,导致FXS临床表型。在过去的十年里,在FXS患者以及小鼠模型(Fmr1-/ymice)中已经描述了视觉神经感觉表型,以与视网膜感知改变相关的视网膜缺陷为特征。然而,尽管已经在中枢神经系统(CNS)的大脑部分研究了不存在FMRP的转录组学谱,没有视网膜的实际数据,视网膜是中枢神经系统的延伸。在这里,我们研究了Fmr1-/ymice整个视网膜mRNA的转录组学特征。有趣的是,与其他脑部研究相比,我们在视网膜mRNA表达上发现了Fmrp缺失的特异性特征,且常见基因很少.这些视网膜特异性基因的基因本体论证明了视网膜发育基因以及突触基因的富集。这些改变可能与所报道的FXS病症的视网膜表型有关。总之,我们第一次描述,在没有FMRP的情况下,视网膜特异性转录组的变化。
    Fragile X Syndrome (FXS), the most common inherited form of human intellectual disability, is a monogenic neurodevelopmental disorder caused by a loss-of-function mutation of the FMR1 gene. FMR1 is encoding the Fragile X Messenger Ribonucleo Protein (FMRP) an RNA-binding protein that regulates the translation of synaptic proteins. The absence of FMRP expression has many important consequences on synaptic plasticity and function, leading to the FXS clinical phenotype. Over the last decade, a visual neurosensorial phenotype had been described in the FXS patients as well as in the murine model (Fmr1-/ymice), characterized by retinal deficits associated to retinal perception alterations. However, although the transcriptomic profile in the absence of FMRP has been studied in the cerebral part of the central nervous system (CNS), there are no actual data for the retina which is an extension of the CNS. Herein, we investigate the transcriptomic profile of mRNA from whole retinas of Fmr1-/ymice. Interestingly, we found a specific signature of Fmrp absence on retinal mRNA expression with few common genes compared to other brain studies. Gene Ontology on these retinal specific genes demonstrated an enrichment in retinal development genes as well as in synaptic genes. These alterations could be linked to the reported retinal phenotype of the FXS condition. In conclusion, we describe for the first time, retinal-specific transcriptomic changes in the absence of FMRP.
    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
    脆性X综合征(FXS)是由Xq27.3染色体区域上FMR1基因的完全突变引起的。它是自闭症谱系障碍(ASD)和遗传性智力障碍(ID)的最常见的单基因原因。除了ASD和ID和其他症状,FXS患者可能出现睡眠问题和昼夜节律(CR)受损.FXS的果蝇模型,例如DFMR1B55,代表了FXS领域研究的优秀模型。在这项研究中,分析了dFMR1B55突变体的睡眠模式和CR,使用基于连续高分辨率视频的新平台,该平台与高度定制的开源软件版本集成在一起。这种方法提供了更敏感的结果,这对于这个果蝇模型的所有进一步研究都是至关重要的。该研究表明,dFMR1B55雄性突变体睡眠更多,可以被认为是弱节律性苍蝇,而不是完全心律失常,并提供了一个很好的遗传障碍替代动物模型,其中包括CR受损和睡眠行为。当前研究中使用的负担得起的摄像和软件的结合是对以前方法的重大改进,并且将使这种高分辨率行为监测方法能够更广泛地适应。
    Fragile X syndrome (FXS) is caused by the full mutation in the FMR1 gene on the Xq27.3 chromosome region. It is the most common monogenic cause of autism spectrum disorder (ASD) and inherited intellectual disability (ID). Besides ASD and ID and other symptoms, individuals with FXS may exhibit sleep problems and impairment of circadian rhythm (CR). The Drosophila melanogaster models of FXS, such as dFMR1B55, represent excellent models for research in the FXS field. During this study, sleep patterns and CR in dFMR1B55 mutants were analyzed, using a new platform based on continuous high-resolution videography integrated with a highly-customized version of an open-source software. This methodology provides more sensitive results, which could be crucial for all further research in this model of fruit flies. The study revealed that dFMR1B55 male mutants sleep more and can be considered weak rhythmic flies rather than totally arrhythmic and present a good alternative animal model of genetic disorder, which includes impairment of CR and sleep behavior. The combination of affordable videography and software used in the current study is a significant improvement over previous methods and will enable broader adaptation of such high-resolution behavior monitoring methods.
    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
    脆性X综合征(FXS)是由脆性X信使核糖核蛋白1(FMR1)基因的转录沉默引起的自闭症谱系障碍的最常见遗传原因。鉴于行为和分子变化的早期发作,必须知道治疗干预的最佳时机。病例报告记录了2至14岁的FXS儿童中二甲双胍治疗的益处。在这项研究中,我们从出生时开始对Fmr1-/y小鼠给予二甲双胍,这些小鼠纠正了丝裂原-2激活的蛋白激酶/细胞外信号调节激酶和雷帕霉素复合物1的哺乳动物/机制靶标信号通路以及FMRP的特异性突触mRNA结合靶标。二甲双胍挽救了Fmr1-/y小鼠的超声发声和重复行为中的呼叫数量增加。我们的发现表明,在小鼠中,生命早期二甲双胍干预在治疗FXS病理生理学方面是有效的.
    Fragile X syndrome (FXS) is the most common genetic cause of autism spectrum disorder engendered by transcriptional silencing of the fragile X messenger ribonucleoprotein 1 (FMR1) gene. Given the early onset of behavioral and molecular changes, it is imperative to know the optimal timing for therapeutic intervention. Case reports documented benefits of metformin treatment in FXS children between 2 and 14 y old. In this study, we administered metformin from birth to Fmr1-/y mice which corrected up-regulated mitogen-2 activated protein kinase/extracellular signal-regulated kinase and mammalian/mechanistic target of rapamycin complex 1 signaling pathways and specific synaptic mRNA-binding targets of FMRP. Metformin rescued increased number of calls in ultrasonic vocalization and repetitive behavior in Fmr1-/y mice. Our findings demonstrate that in mice, early-in-life metformin intervention is effective in treating FXS pathophysiology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号