FXS

FXS
  • 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综合征(FXS),最常见的单基因形式的智力残疾,是由FMR1基因的转录沉默引起的,这可能使神经元过度兴奋。在这里,我们显示海马背侧CA1区域的锥体细胞(PC)在幼年Fmr1敲除(KO)小鼠中响应于阈值刺激而引起的动作电位(AP)数量比野生型(WT)小鼠更大。因为Kv7/M通道调节大鼠CA1PC兴奋性,我们调查了它们的功能障碍是否在Fmr1KO小鼠中产生神经元过度兴奋。免疫组织化学和蛋白质印迹分析显示,基因型之间Kv7.2和Kv7.3通道亚基的表达没有差异;然而,Kv7/M通道介导的电流在Fmr1KO小鼠中降低。在两种基因型中,XE991(10μM)的浴应用,Kv7/M通道的阻断剂:产生了增加的AP数,产生了增加的输入电阻,通过增加平均速度,产生了降低的AP电压阈值,并形成了AP介质后超极化。雷替加宾(10μM),Kv7/M频道的开瓶器,产生与XE991相反的效果。XE991和瑞替加滨都消除了在基因型之间的对照条件中发现的所有这些参数的差异。此外,低浓度的瑞替加滨(2.5μM)使Fmr1KO小鼠的CA1PC兴奋性正常化。最后,在Fmr1KO小鼠中,背侧CA1区4-氨基吡啶(200μM)诱发的离体癫痫样事件更为频繁,并被瑞替加滨(5-10μM)废除。我们得出结论,Fmr1KO小鼠的CA1PCs表现出过度兴奋,由Kv7/M通道功能障碍引起,癫痫样活动增加,被瑞替加宾废除了。关键点:Fmr1基因敲除小鼠海马CA1区的背侧锥体细胞表现出过度兴奋。Kv7/M通道活动,但不是表达,在Fmr1敲除小鼠的海马CA1区的锥体细胞中减少。Kv7/M通道功能障碍通过增加输入阻力导致Fmr1敲除小鼠海马CA1区锥体细胞过度兴奋,降低AP电压阈值和超极化后整形介质。Kv7/M通道开放剂使Fmr1敲除小鼠海马CA1区锥体细胞的神经元兴奋性正常化。在Fmr1KO小鼠中,背侧CA1区诱发的离体癫痫样事件更为频繁,根据药物浓度,Kv7/M通道开放剂消除了这种癫痫样活性。Kv7/M通道可以代表用于治疗脆性X综合征中与海马改变相关的症状的治疗靶标。
    Fragile X syndrome (FXS), the most frequent monogenic form of intellectual disability, is caused by transcriptional silencing of the FMR1 gene that could render neuronal hyperexcitability. Here we show that pyramidal cells (PCs) in the dorsal CA1 region of the hippocampus elicited a larger action potential (AP) number in response to suprathreshold stimulation in juvenile Fmr1 knockout (KO) than wild-type (WT) mice. Because Kv7/M channels modulate CA1 PC excitability in rats, we investigated if their dysfunction produces neuronal hyperexcitability in Fmr1 KO mice. Immunohistochemical and western blot analyses showed no differences in the expression of Kv7.2 and Kv7.3 channel subunits between genotypes; however, the current mediated by Kv7/M channels was reduced in Fmr1 KO mice. In both genotypes, bath application of XE991 (10 μM), a blocker of Kv7/M channels: produced an increased AP number, produced an increased input resistance, produced a decreased AP voltage threshold and shaped AP medium afterhyperpolarization by increasing mean velocities. Retigabine (10 μM), an opener of Kv7/M channels, produced opposite effects to XE991. Both XE991 and retigabine abolished differences in all these parameters found in control conditions between genotypes. Furthermore, a low concentration of retigabine (2.5 μM) normalized CA1 PC excitability of Fmr1 KO mice. Finally, ex vivo seizure-like events evoked by 4-aminopyiridine (200 μM) in the dorsal CA1 region were more frequent in Fmr1 KO mice, and were abolished by retigabine (5-10 μM). We conclude that CA1 PCs of Fmr1 KO mice exhibit hyperexcitability, caused by Kv7/M channel dysfunction, and increased epileptiform activity, which were abolished by retigabine. KEY POINTS: Dorsal pyramidal cells of the hippocampal CA1 region of Fmr1 knockout mice exhibit hyperexcitability. Kv7/M channel activity, but not expression, is reduced in pyramidal cells of the hippocampal CA1 region of Fmr1 knockout mice. Kv7/M channel dysfunction causes hyperexcitability in pyramidal cells of the hippocampal CA1 region of Fmr1 knockout mice by increasing input resistance, decreasing AP voltage threshold and shaping medium afterhyperpolarization. A Kv7/M channel opener normalizes neuronal excitability in pyramidal cells of the hippocampal CA1 region of Fmr1 knockout mice. Ex vivo seizure-like events evoked in the dorsal CA1 region were more frequent in Fmr1 KO mice, and such an epileptiform activity was abolished by a Kv7/M channel opener depending on drug concentration. Kv7/M channels may represent a therapeutic target for treating symptoms associated with hippocampal alterations in fragile X syndrome.
    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信使核糖核蛋白1(FMRP)是一种广泛表达的RNA结合蛋白,参与mRNA代谢的几个步骤。编码FMRP的FMR1基因突变是脆性X综合征(FXS)的原因,智力障碍和自闭症谱系障碍的主要遗传原因,和脆性X相关震颤共济失调综合征(FXTAS),老年男性的神经退行性疾病。尽管FMRP主要在神经元中表达,它也存在于神经胶质细胞中,其缺乏或表达改变会影响神经胶质细胞的功能,对脑部疾病的病理生理学有影响。本综述侧重于神经胶质亚型的作用的最新进展,星形胶质细胞,少突胶质细胞和小胶质细胞,在FXS和FXTAS的病理生理学中,并描述了这些细胞中FMRP的缺失或表达降低如何影响神经胶质和神经元功能。我们还将简要讨论FMRP在放射状神经胶质细胞中的作用及其对神经发育和神经胶质瘤的影响,并推测神经胶质FMRP在其他脑部疾病中的作用。
    Fragile X messenger ribonucleoprotein 1 (FMRP) is a widely expressed RNA binding protein involved in several steps of mRNA metabolism. Mutations in the FMR1 gene encoding FMRP are responsible for fragile X syndrome (FXS), a leading genetic cause of intellectual disability and autism spectrum disorder, and fragile X-associated tremor-ataxia syndrome (FXTAS), a neurodegenerative disorder in aging men. Although FMRP is mainly expressed in neurons, it is also present in glial cells and its deficiency or altered expression can affect functions of glial cells with implications for the pathophysiology of brain disorders. The present review focuses on recent advances on the role of glial subtypes, astrocytes, oligodendrocytes and microglia, in the pathophysiology of FXS and FXTAS, and describes how the absence or reduced expression of FMRP in these cells can impact on glial and neuronal functions. We will also briefly address the role of FMRP in radial glial cells and its effects on neural development, and gliomas and will speculate on the role of glial FMRP in other brain disorders.
    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FMR1基因的表观遗传沉默引起的神经系统疾病。FMR1的再激活是FXS的潜在治疗方法,可以纠正疾病的根本原因。这里,使用基于候选的shRNA筛选,我们确定了9种促进FXS细胞中FMR1沉默的表观遗传抑制因子(称为FMR1沉默因子,或FMR1-SF)。用shRNA或小分子抑制FMR1-SF在培养的未分化诱导多能干细胞中重新激活FMR1,来自FXS患者的神经祖细胞(NPC)和有丝分裂后神经元。FMR1-SF之一是组蛋白甲基转移酶EZH2,这是FDA批准的小分子抑制剂,EPZ6438(也称为tazemetostat),是可用的。我们表明EPZ6438基本上纠正了培养的FXS神经元的特征性分子和电生理异常。不幸的是,EZH2抑制剂不能有效地穿过血脑屏障,限制其对FXS的治疗用途。最近,基于反义寡核苷酸(ASO)的方法已被开发为某些中枢神经系统疾病的有效治疗选择。因此,我们获得了靶向EZH2的有效ASO,并证明它们重新激活FMR1表达并纠正培养的FXS神经元的分子和电生理异常,并重新激活移植在小鼠脑内的人FXSNPC中的FMR1表达。总的来说,我们的结果建立了EZH2抑制一般,特别是EZH2ASO,作为FXS的治疗方法。
    Fragile X Syndrome (FXS) is a neurological disorder caused by epigenetic silencing of the FMR1 gene. Reactivation of FMR1 is a potential therapeutic approach for FXS that would correct the root cause of the disease. Here, using a candidate-based shRNA screen, we identify nine epigenetic repressors that promote silencing of FMR1 in FXS cells (called FMR1 Silencing Factors, or FMR1- SFs). Inhibition of FMR1-SFs with shRNAs or small molecules reactivates FMR1 in cultured undifferentiated induced pluripotent stem cells, neural progenitor cells (NPCs) and post-mitotic neurons derived from FXS patients. One of the FMR1-SFs is the histone methyltransferase EZH2, for which an FDA-approved small molecule inhibitor, EPZ6438 (also known as tazemetostat), is available. We show that EPZ6438 substantially corrects the characteristic molecular and electrophysiological abnormalities of cultured FXS neurons. Unfortunately, EZH2 inhibitors do not efficiently cross the blood-brain barrier, limiting their therapeutic use for FXS. Recently, antisense oligonucleotide (ASO)-based approaches have been developed as effective treatment options for certain central nervous system disorders. We therefore derived efficacious ASOs targeting EZH2 and demonstrate that they reactivate FMR1 expression and correct molecular and electrophysiological abnormalities in cultured FXS neurons, and reactivate FMR1 expression in human FXS NPCs engrafted within the brains of mice. Collectively, our results establish EZH2 inhibition in general, and EZH2 ASOs in particular, as a therapeutic approach for FXS.
    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)是遗传性自闭症和智力障碍的主要原因。由于脆性X信使核糖核蛋白(FMRP)丢失导致的蛋白质合成异常是FXS的主要缺陷,导致过多的细胞和行为异常。然而,迄今为止没有治疗方法。在这项研究中,我们发现,使用名为AMN082的正变构调节剂激活代谢型谷氨酸受体7(mGluR7),通过ERK1/2和eIF4E信号传导,以不依赖FMRP的方式抑制蛋白质合成.我们进一步证明,AMN082的治疗导致神经元兴奋性的降低,这反过来又改善了Fmr1KO小鼠的听原癫痫发作易感性,FXS小鼠模型。在评估动物的行为时,我们发现AMN082治疗减少了Fmr1KO小鼠的重复行为并改善了学习和记忆。这项研究揭示了mGluR7和AMN082的新功能,并表明mGluR7的激活是治疗FXS的潜在治疗方法。
    Fragile X syndrome (FXS) is the leading cause of inherited autism and intellectual disabilities. Aberrant protein synthesis due to the loss of fragile X messenger ribonucleoprotein (FMRP) is the major defect in FXS, leading to a plethora of cellular and behavioral abnormalities. However, no treatments are available to date. In this study, we found that activation of metabotropic glutamate receptor 7 (mGluR7) using a positive allosteric modulator named AMN082 represses protein synthesis through ERK1/2 and eIF4E signaling in an FMRP-independent manner. We further demonstrated that treatment of AMN082 leads to a reduction in neuronal excitability, which in turn ameliorates audiogenic seizure susceptibility in Fmr1 KO mice, the FXS mouse model. When evaluating the animals\' behavior, we showed that treatment of AMN082 reduces repetitive behavior and improves learning and memory in Fmr1 KO mice. This study uncovers novel functions of mGluR7 and AMN082 and suggests the activation of mGluR7 as a potential therapeutic approach for treating FXS.
    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)是遗传性智力障碍的主要原因。在FXS个体的神经行为功能障碍中,语言发展和识字受到损害。最近的证据假设,兴奋性谷氨酸能和GABA能抑制性神经传递平衡的破坏可能是认知功能受损的原因。在这项研究中,我们第一次评估,安全,耐受性,经颅经颅直流刺激(tDCS)联合标准言语治疗增强FXS患者语言功能的疗效。
    总共,16名成人FXS患者入组。参与者接受45分钟的阳极tDCS联合言语治疗5周(每周3次)。使用语法接受测试2(TROG-2)和意大利语语言考试的子测试(EsamedelLinguaggio-II,EDL-II)。在基线和治疗后收集左右背外侧前额叶皮质经颅磁刺激和并发脑电图(TMS-EEG)记录,以评估皮质反应性和连通性变化。
    联合治疗5周后,我们观察到写作有显著改善(7.5%),读数(20.3%),重复(13.3%),和TROG-2(10.2%)测试。与临床改变平行,TMS-EEG结果显示,与基线(0.18±0.84μV)相比,治疗后左额叶皮层的TMS诱发电位振幅(-0.73±0.87μV)存在显着差异。
    我们的研究提供了新的证据,表明左阳极前额叶tDCS结合标准言语治疗可以有效增强FXS患者的语言功能,主要通过诱导功能失调的前额叶皮质兴奋性的再平衡。
    UNASSIGNED: Fragile X syndrome (FXS) is the leading cause of genetic intellectual disability. Among the neurobehavioral dysfunctions in FXS individuals, language development and literacy are compromised. Recent evidence hypothesized that the disruption of excitatory glutamatergic and GABAergic inhibitory neurotransmission balance might be responsible for impairment in cognitive function. In this study, we evaluated for the first time, the safety, tolerability, and efficacy of anodal prefrontal transcranial direct current stimulation (tDCS) combined with standard speech therapy to enhance language function in FXS patients.
    UNASSIGNED: In total, 16 adult FXS patients were enrolled. Participants underwent 45 min of anodic tDCS combined with speech therapy for 5 weeks (3 times per week). Language function was evaluated using the Test for Reception of Grammar-Version 2 (TROG-2) and subtests of the Italian Language Examination (Esame del Linguaggio - II, EDL-II). Right and left dorsolateral prefrontal cortex transcranial magnetic stimulation and concurrent electroencephalography (TMS-EEG) recordings were collected at baseline and after the treatment to evaluate cortical reactivity and connectivity changes.
    UNASSIGNED: After 5 weeks of combined therapy, we observed a significant improvement in the writing (7.5%), reading (20.3%), repetition (13.3%), and TROG-2 (10.2%) tests. Parallelly with clinical change, TMS-EEG results showed a significant difference in TMS-evoked potential amplitude over the left frontal cortex after treatment (-0.73 ± 0.87 μV) compared to baseline (0.18 ± 0.84 μV).
    UNASSIGNED: Our study provides novel evidence that left anodal prefrontal tDCS combined with standard speech therapy could be effective in enhancing language function in FXS patients, mainly by inducing a rebalance of the dysfunctional prefrontal cortical excitability.
    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),遗传性智力残疾和自闭症的主要原因,是由FMR1基因的转录沉默引起的,其编码脆性X信使核糖核蛋白(FMRP)。FMRP与许多参与突触可塑性并参与自闭症谱系障碍的大脑mRNA相互作用。我们发表的研究表明,单一来源,以大豆为基础的饮食与癫痫发作和自闭症增加有关。因此,对于响应大豆消费的FXS中无偏倚的蛋白质标记鉴定存在迫切需求.在这里,我们提出了一种在FXS小鼠模型中整合质谱成像和无标记蛋白质组学的空间蛋白质组学方法,以绘制海马和下丘脑脑区蛋白质的空间分布和定量水平.总的来说,1250种独特的肽是空间分辨的,证明了组织切片中存在的各种肽和策略的广泛覆盖范围。一组已知参与糖酵解的蛋白质,突触传递,和共表达网络分析表明,大豆蛋白与Fmr1KO大脑中的代谢和突触过程之间存在显着关联。最终,这项空间蛋白质组学工作代表了确定FXS潜在候选蛋白标记和新治疗靶点的关键一步.
    Fragile X syndrome (FXS), the leading cause of inherited intellectual disability and autism, is caused by the transcriptional silencing of the FMR1 gene, which encodes the fragile X messenger ribonucleoprotein (FMRP). FMRP interacts with numerous brain mRNAs that are involved in synaptic plasticity and implicated in autism spectrum disorders. Our published studies indicate that single-source, soy-based diets are associated with increased seizures and autism. Thus, there is an acute need for an unbiased protein marker identification in FXS in response to soy consumption. Herein, we present a spatial proteomics approach integrating mass spectrometry imaging with label-free proteomics in the FXS mouse model to map the spatial distribution and quantify levels of proteins in the hippocampus and hypothalamus brain regions. In total, 1250 unique peptides were spatially resolved, demonstrating the diverse array of peptidomes present in the tissue slices and the broad coverage of the strategy. A group of proteins that are known to be involved in glycolysis, synaptic transmission, and coexpression network analysis suggest a significant association between soy proteins and metabolic and synaptic processes in the Fmr1KO brain. Ultimately, this spatial proteomics work represents a crucial step toward identifying potential candidate protein markers and novel therapeutic targets for FXS.
    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)是人类遗传性智力障碍和自闭症的最常见单基因病因。Fmr1KO小鼠的特征良好的分子表型之一,FXS的模型,是突触蛋白的翻译增加。尽管这种上调在成年期稳定下来,可塑性关键时期的异常对回路形成和突触特性有长期影响。使用从成人中分离的突触神经素的高分辨率定量蛋白质组学,Fmr1KO小鼠的大脑发育,我们显示了调节谷氨酸能突触的突触后受体活性的蛋白质的不同丰度。我们研究了AMPA受体的组成和穿梭在成年Fmr1KO和WT小鼠使用各种互补的实验策略,如表面蛋白交联,表面受体的免疫染色,和电生理学。我们发现AMPAR的活性依赖性突触传递在成年Fmr1KO小鼠中受损。此外,我们显示Fmr1KO突触AMPAR含有更多GluA2亚基,这可以解释为突触AMPAR亚型向成年Fmr1KO突触中Ca2+不可渗透受体数量增加的转换.
    Fragile X syndrome (FXS) is the most common monogenetic cause of inherited intellectual disability and autism in humans. One of the well-characterized molecular phenotypes of Fmr1 KO mice, a model of FXS, is increased translation of synaptic proteins. Although this upregulation stabilizes in adulthood, abnormalities during the critical period of plasticity have long-term effects on circuit formation and synaptic properties. Using high-resolution quantitative proteomics of synaptoneurosomes isolated from the adult, developed brains of Fmr1 KO mice, we show a differential abundance of proteins regulating the postsynaptic receptor activity of glutamatergic synapses. We investigated the AMPA receptor composition and shuttling in adult Fmr1 KO and WT mice using a variety of complementary experimental strategies such as surface protein crosslinking, immunostaining of surface receptors, and electrophysiology. We discovered that the activity-dependent synaptic delivery of AMPARs is impaired in adult Fmr1 KO mice. Furthermore, we show that Fmr1 KO synaptic AMPARs contain more GluA2 subunits that can be interpreted as a switch in the synaptic AMPAR subtype toward an increased number of Ca2+-impermeable receptors in adult Fmr1 KO synapses.
    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
    FMRP是由脆性X信使核糖核蛋白1基因(FMR1)编码的多功能蛋白。FMR1基因的失活导致脆性X综合征(FXS),严重的神经发育障碍.FMRP缺乏导致异常的神经突生长,这可能会导致学习和记忆能力异常。然而,FMRP调节神经元发育的机制尚不清楚。我们发现FMRP增强4EBP2的翻译,4EBP2是4EBPs的神经元特异性形式,通过抑制eIF4E和eIF4G之间的相互作用使eIF4E失活。4EBP2的耗尽导致异常的神经突生长。此外,通过4EBP2的异位表达克服了FMRP耗竭时神经突生长的损害。这些结果表明FMRP通过在翻译水平上增强4EBP2表达来控制神经元发育。此外,用4EGI-1治疗,这是一种阻断eIF4E活性的化学物质,在FMRP耗尽和4EBP2耗尽的细胞中恢复了神经突长度。总之,我们发现4EBP2在神经元发育中作为FMRP活性的关键下游调节因子,FMRP通过增强4EBP2翻译抑制eIF4E活性。
    FMRP is a multifunctional protein encoded by the Fragile X Messenger Ribonucleoprotein 1 gene (FMR1). The inactivation of the FMR1 gene results in fragile X syndrome (FXS), a serious neurodevelopmental disorder. FMRP deficiency causes abnormal neurite outgrowth, which is likely to lead to abnormal learning and memory capabilities. However, the mechanism of FMRP in modulating neuronal development remains unknown. We found that FMRP enhances the translation of 4EBP2, a neuron-specific form of 4EBPs that inactivates eIF4E by inhibiting the interaction between eIF4E and eIF4G. Depletion of 4EBP2 results in abnormal neurite outgrowth. Moreover, the impairment of neurite outgrowth upon FMRP depletion was overcome by the ectopic expression of 4EBP2. These results suggest that FMRP controls neuronal development by enhancing 4EBP2 expression at the translational level. In addition, treatment with 4EGI-1, a chemical that blocks eIF4E activity, restored neurite length in FMRP-depleted and 4EBP2-depleted cells. In conclusion, we discovered that 4EBP2 functions as a key downstream regulator of FMRP activity in neuronal development and that FMRP represses eIF4E activity by enhancing 4EBP2 translation.
    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
    补体系统是先天免疫系统的一部分,已被证明在自闭症谱系障碍(ASD)中发生改变。脆性X综合征(FXS)是ASD的主要遗传原因,研究表明该疾病患者的免疫系统失调。为了评估FXS的动物模型是否存在补体信号改变,我们用脂多糖(LPS)处理雄性Fmr1基因敲除(KO)小鼠,并在24小时后收集海马。评估补体基因C1q的表达,C3和C4在野生型(WT)和敲除小鼠两者中鉴定C3的上调。两种基因型的C3水平也增加。C3表达与细胞因子IL-6、IL-1β、和TNF-α鉴定了与WT小鼠相比,Fmr1KO中基因表达之间的不同关系。我们的发现并不支持我们最初的假设,即缺乏FMR1基因会改变补体系统信号。并且在Fmr1KO小鼠中响应LPS的补体系统的诱导与野生型特异性不同。
    The complement system is part of the innate immune system and has been shown to be altered in autism spectrum disorder (ASD). Fragile-X syndrome (FXS) is the main genetic cause of ASD and studies suggest a dysregulation in the immune system in patients with the disorder. To assess if an animal model of FXS presents with altered complement signaling, we treated male Fmr1 knockout (KO) mice with lipopolysaccharide (LPS) and collected the hippocampus 24 h later. Assessment of the expression of the complement genes C1q, C3, and C4 identified the upregulation of C3 in both wild-type (WT) and knockout mice. Levels of C3 also increased in both genotypes. Analysis of the correlation between the expression of C3 and the cytokines IL-6, IL-1β, and TNF-α identified a different relationship between the expression of the genes in Fmr1 KO when compared to WT mice. Our findings did not support our initial hypotheses that the lack of the FMR1 gene would alter complement system signaling, and that the induction of the complement system in response to LPS in Fmr1 KO mice differed from wild-type conspecifics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号