G(M1) Ganglioside

G (M1) 神经节苷脂
  • 文章类型: Journal Article
    通过贝叶斯优化确定了GM1模拟物的合成糖共聚物的最佳结构。通过酶联免疫吸附测定(ELISA)评估了不同组合物中携带半乳糖和神经氨酸单元的糖共聚物与霍乱毒素B亚基(CTB)的相互作用。高斯过程回归,使用ELISA结果,预测了将表现出与CTB更强相互作用的糖共聚物的组成。经过五个周期的优化,含有60摩尔%半乳糖和25摩尔%神经氨酸的糖共聚物对CTB的IC50值为75μM,代表合成的糖共聚物中的最低值。
    The optimal structure of synthetic glycopolymers for GM1 mimetics was determined through Bayesian optimization. The interactions of glycopolymers carrying galactose and neuraminic acid units in different compositions with cholera toxin B subunit (CTB) were assessed by an enzyme-linked immunosorbent assay (ELISA). Gaussian process regression, using the ELISA results, predicted the composition of glycopolymers that would exhibit stronger interactions with CTB. Following five cycles of optimization, the glycopolymers carrying 60 mol% galactose and 25 mol% neuraminic acid demonstrated an IC50 value of 75 μM for CTB, representing the lowest value among the synthesized glycopolymers.
    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
    单唾液酸神经节苷脂GM1(GM1)在缺血性卒中的临床治疗中一直被用作神经系统疾病的治疗剂。然而,GM1神经保护功能的潜在机制至今仍不清楚。在这项研究中,我们研究了GM1在缺血再灌注(I/R)脑损伤模型中的作用.大脑中动脉闭塞再灌注(MCAO/R)大鼠接受GM1(60mg·kg-1·d-1,尾静脉注射)治疗2周。结果表明,GM1可以明显减轻MCAO/R诱导的缺血顶叶皮质的神经功能障碍,抑制炎症反应和细胞凋亡。我们进一步揭示了GM1抑制MCAO/R损伤诱导的NFκB/MAPK信号通路的激活。探讨其神经保护作用的潜在机制,引入转录组测序来筛选差异表达基因(DEGs)。通过功能丰富和PPI网络分析,Sptbn1被鉴定为由GM1处理调节的网络中的节点基因。在MCAO/R大鼠模型和氧糖剥夺再灌注(OGD/R)模型中原代培养大鼠皮层神经元,我们首先发现SPTBN1参与减轻GM1给药后I/R诱导的神经元损伤。在SPTBN1敲低SH-SY5Y细胞中,用GM1(20μM)处理显著增加SPTBN1水平。此外,OGD/R降低SPTBN1过表达SH-SY5Y细胞中SPTBN1的水平。这些结果表明,GM1可能通过调节炎症反应来实现其有效的神经保护作用。细胞凋亡,以及通过SPTBN1的细胞膜和细胞骨架信号。因此,SPTBN1可能是缺血性卒中治疗的潜在靶点。
    Monosialoganglioside GM1 (GM1) has long been used as a therapeutic agent for neurological diseases in the clinical treatment of ischemic stroke. However, the mechanism underlying the neuroprotective function of GM1 is still obscure until now. In this study, we investigated the effects of GM1 in ischemia and reperfusion (I/R) brain injury models. Middle cerebral artery occlusion and reperfusion (MCAO/R) rats were treated with GM1 (60 mg·kg-1·d-1, tail vein injection) for 2 weeks. The results showed that GM1 substantially attenuated the MCAO/R-induced neurological dysfunction and inhibited the inflammatory responses and cell apoptosis in ischemic parietal cortex. We further revealed that GM1 inhibited the activation of NFκB/MAPK signaling pathway induced by MCAO/R injury. To explore its underlying mechanism of the neuroprotective effect, transcriptome sequencing was introduced to screen the differentially expressed genes (DEGs). By function enrichment and PPI network analyses, Sptbn1 was identified as a node gene in the network regulated by GM1 treatment. In the MCAO/R model of rats and oxygen-glucose deprivation and reperfusion (OGD/R) model of primary culture of rat cortical neurons, we first found that SPTBN1 was involved in the attenuation of I/R induced neuronal injury after GM1 administration. In SPTBN1-knockdown SH-SY5Y cells, the treatment with GM1 (20 μM) significantly increased SPTBN1 level. Moreover, OGD/R decreased SPTBN1 level in SPTBN1-overexpressed SH-SY5Y cells. These results indicated that GM1 might achieve its potent neuroprotective effects by regulating inflammatory response, cell apoptosis, and cytomembrane and cytoskeleton signals through SPTBN1. Therefore, SPTBN1 may be a potential target for the treatment of ischemic stroke.
    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
    淀粉样蛋白β(Aβ)是阿尔茨海默病(AD)中神经元损伤和认知障碍的主要原因。Aβ破坏AMPA受体介导的突触可塑性,早期AD进展的关键因素。大量研究认为Aβ寡聚体阻碍突触可塑性,特别是长期增强(LTP),通过破坏GluA1(由GRIA1编码)功能,尽管确切的机制尚不清楚。在这项研究中,我们证明Aβ介导GM1神经节苷脂在培养细胞的脂筏结构域中的积累,和GluA1通过与GM1直接结合而在脂筏中表现出优先定位。Aβ通过增加这些区域中的GM1来增强GluA1的移植物定位。此外,化学LTP刺激在Aβ处理的神经元中诱导脂质筏依赖性GluA1内化,导致细胞表面和突触后GluA1表达减少。与此一致,破坏脂筏和移植物中GluA1的定位可挽救Aβ介导的海马LTP抑制。这些发现揭示了由Aβ诱导的GluA1运输中的一种新的功能缺陷,为AD相关认知功能障碍的潜在机制提供新的见解。
    Amyloid β (Aβ) is a central contributor to neuronal damage and cognitive impairment in Alzheimer\'s disease (AD). Aβ disrupts AMPA receptor-mediated synaptic plasticity, a key factor in early AD progression. Numerous studies propose that Aβ oligomers hinder synaptic plasticity, particularly long-term potentiation (LTP), by disrupting GluA1 (encoded by GRIA1) function, although the precise mechanism remains unclear. In this study, we demonstrate that Aβ mediates the accumulation of GM1 ganglioside in lipid raft domains of cultured cells, and GluA1 exhibits preferential localization in lipid rafts via direct binding to GM1. Aβ enhances the raft localization of GluA1 by increasing GM1 in these areas. Additionally, chemical LTP stimulation induces lipid raft-dependent GluA1 internalization in Aβ-treated neurons, resulting in reduced cell surface and postsynaptic expression of GluA1. Consistent with this, disrupting lipid rafts and GluA1 localization in rafts rescues Aβ-mediated suppression of hippocampal LTP. These findings unveil a novel functional deficit in GluA1 trafficking induced by Aβ, providing new insights into the mechanism underlying AD-associated cognitive dysfunction.
    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
    背景:多灶性运动神经病(MMN)是一种罕见的,以不对称远端肢体无力为特征的慢性免疫介导的多发性神经病。MMN的一个重要特征是存在抗神经节苷脂的IgM抗体,特别是GM1和较少的GM2。针对GM1的抗体与运动神经元(MN)结合并通过补体激活引起损伤。施万细胞(SCs)的参与,在MMN的发病机制中表达GM1和GM2是未知的。
    方法:结合我们2007年和2015年在荷兰MMN患者中进行的横断面和随访研究的数据,我们评估了124例MMN患者血清中GM1和GM2IgM抗体的存在,并研究了它们与SC的结合和补体激活特性.我们还评估了IgM结合和补体沉积与临床特征的关系。
    结果:124例患者中有13例(10%)的IgM抗GM2ELISA滴度呈阳性。使用抗GM2IgM的MMN患者的症状发作年龄显着降低。IgM与SC的结合与IgM抗GM2滴度相关。我们发现IgM抗GM2滴度与MN结合或与IgM抗GM1滴度之间没有相关性。在血清与可溶性GM2预孵育后,IgM与SC的结合减少,但与可溶性GM1无关。IgM抗GM2与SC的结合与补体激活相关,如SCs上C3固定增加和上清液中C5a形成所反映的。
    结论:循环IgM抗GM2抗体定义了具有较早发病的MMN患者亚组。这些抗体可能特异性靶向SCs并激活补体,与MNs上的IgM抗GM1相似。我们的数据表明,IgM抗体与SC和MN结合的补体激活是MMN病理学的基础。
    BACKGROUND: Multifocal motor neuropathy (MMN) is a rare, chronic immune-mediated polyneuropathy characterized by asymmetric distal limb weakness. An important feature of MMN is the presence of IgM antibodies against gangliosides, in particular GM1 and less often GM2. Antibodies against GM1 bind to motor neurons (MNs) and cause damage through complement activation. The involvement of Schwann cells (SCs), expressing GM1 and GM2, in the pathogenesis of MMN is unknown.
    METHODS: Combining the data of our 2007 and 2015 combined cross-sectional and follow-up studies in Dutch patients with MMN, we evaluated the presence of IgM antibodies against GM1 and GM2 in serum from 124 patients with MMN and investigated their binding to SCs and complement-activating properties. We also assessed the relation of IgM binding and complement deposition with clinical characteristics.
    RESULTS: Thirteen out of 124 patients (10%) had a positive ELISA titer for IgM anti-GM2. Age at onset of symptoms was significantly lower in MMN patients with anti-GM2 IgM. IgM binding to SCs correlated with IgM anti-GM2 titers. We found no correlation between IgM anti-GM2 titers and MN binding or with IgM anti-GM1 titers. IgM binding to SCs decreased upon pre-incubation of serum with soluble GM2, but not with soluble GM1. IgM anti-GM2 binding to SCs correlated with complement activation, as reflected by increased C3 fixation on SCs and C5a formation in the supernatant.
    CONCLUSIONS: Circulating IgM anti-GM2 antibodies define a subgroup of patients with MMN that has an earlier onset of disease. These antibodies probably target SCs specifically and activate complement, similarly as IgM anti-GM1 on MNs. Our data indicate that complement activation by IgM antibodies bound to SCs and MNs underlies MMN pathology.
    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
    内质网-质膜(ER-PM)连接介导穿过神经元膜的Ca2+通量。这些膜接触位点的性质由它们的脂质含量定义,但很少有人关注鞘糖脂(GSL)。这里,我们发现GM1-神经节苷脂,神经元膜中丰富的GSL,是ER-PM连接的组成部分;它与突触蛋白/受体相互作用并调节Ca2信号传导。在神经退行性溶酶体贮积病模型中,GM1-神经节苷脂增多症,由于β-半乳糖苷酶缺乏,GM1在ER-PM连接处的致病性积累极大地改变了神经元Ca2稳态。机械上,我们显示GM1与磷酸化的N-甲基D-天冬氨酸受体(NMDAR)Ca2通道相互作用,从而增加Ca2+通量,激活细胞外信号调节激酶(ERK)信号,并在不增加突触连通性的情况下增加突触棘的数量。因此,GM1在ER-PM连接处的聚集会改变突触可塑性,并使GM1神经节苷异位症的广义神经元细胞死亡特征恶化。
    Endoplasmic reticulum-plasma membrane (ER-PM) junctions mediate Ca2+ flux across neuronal membranes. The properties of these membrane contact sites are defined by their lipid content, but little attention has been given to glycosphingolipids (GSLs). Here, we show that GM1-ganglioside, an abundant GSL in neuronal membranes, is integral to ER-PM junctions; it interacts with synaptic proteins/receptors and regulates Ca2+ signaling. In a model of the neurodegenerative lysosomal storage disease, GM1-gangliosidosis, pathogenic accumulation of GM1 at ER-PM junctions due to β-galactosidase deficiency drastically alters neuronal Ca2+ homeostasis. Mechanistically, we show that GM1 interacts with the phosphorylated N-methyl D-aspartate receptor (NMDAR) Ca2+ channel, thereby increasing Ca2+ flux, activating extracellular signal-regulated kinase (ERK) signaling, and increasing the number of synaptic spines without increasing synaptic connectivity. Thus, GM1 clustering at ER-PM junctions alters synaptic plasticity and worsens the generalized neuronal cell death characteristic of GM1-gangliosidosis.
    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
    神经节苷脂GM1是一类主要位于神经系统的糖脂。包含神经酰胺锚和含有唾液酸的寡糖链,GM1在各种细胞过程中起着关键作用,包括信号转导,细胞粘附,和膜组织。此外,GM1与几种神经系统疾病的发病机理有关,如帕金森病,老年痴呆症,和中风。在这项研究中,通过创建神经细胞模型膜模拟系统并采用严格的分子模型,我们利用粗粒分子动力学方法来探索不同GM1神经节苷脂浓度下多组分神经元质膜的结构和动态特征。模拟结果表明,随着GM1浓度的增加,GM1分子之间形成更多的氢键,导致形成更大的集群,这导致膜流动性降低,脂质排序增加,降低膜厚度和表面积以及更高水平的GM1解离。经过细致的分析,在考虑GM1的结构属性时,我们为细胞膜的结构和动态特性提供了有价值的见解。这项研究为探索膜特性提供了一种可靠的方法,并增强了我们对GM1分子的理解,作为该领域实验和计算研究人员的资源。
    Ganglioside GM1 is a class of glycolipids predominantly located in the nervous system. Comprising a ceramide anchor and an oligosaccharide chain containing sialic acid, GM1 plays a pivotal role in various cellular processes, including signal transduction, cell adhesion, and membrane organization. Moreover, GM1 has been implicated in the pathogenesis of several neurological disorders, such as Parkinson\'s disease, Alzheimer\'s disease, and stroke. In this study, by creating a neural cell model membrane simulation system and employing rigorous molecular models, we utilize a coarse-grained molecular dynamics approach to explore the structural and dynamic characteristics of multi-component neuronal plasma membranes at varying GM1 ganglioside concentrations. The simulation results reveal that as GM1 concentration increases, a greater number of hydrogen bonds form between GM1 molecules, resulting in the formation of larger clusters, which leads to reduced membrane fluidity, increased lipid ordering, decreased membrane thickness and surface area and higher levels of GM1 dissociation. Through a meticulous analysis, while considering GM1\'s structural attributes, we offer valuable insights into the structural and dynamic traits of the cell membrane. This study provides a robust methodology for exploring membrane characteristics and enhances our comprehension of GM1 molecules, serving as a resource for both experimental and computational researchers in this field.
    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
    神经节苷脂GM1及其在帕金森病(PD)中的神经保护作用研究,特别是在减轻α-突触核蛋白(aSyn)的聚集方面,在各种模式生物中都很成熟。这个重要的分子,GM1与防止aSyn聚集密切相关,它的缺乏被认为在PD的启动中起着关键作用。在我们目前的研究中,我们试图阐明GM1和aSyn之间的细胞溶质相互作用,基于以前的报道表明神经节苷脂和单体aSyn存在于神经元细胞溶质中。来自小鼠大脑的神经元细胞溶胶的Native-PAGE和Western印迹分析证明了正常小鼠大脑的神经元细胞溶胶中GM1和单体aSyn的存在。为了证明GM1的足够水平可以防止aSyn的聚集,我们使用NG108-15和SH-SY5Y细胞,有或没有1-苯基-2-棕榈酰-3-吗啉代-1-丙醇(PPMP)处理,抑制GM1的合成/表达。与未处理的细胞相比,用PPMP处理以减少GM1表达的细胞显示聚集的aSyn的形成显著增加。因此,我们证明了足够的GM1可以防止aSyn的聚集。为了发生这种情况,Syn和GM1必须显示接近神经元内。本研究为神经元胞质溶胶中的这种共定位提供了证据,这也促进了与上述两种细胞类型的研究中揭示的反向相互作用。这增加了对GM1如何防止aSyn聚集和帕金森病发作的解释。
    Research on GM1 ganglioside and its neuroprotective role in Parkinson\'s disease (PD), particularly in mitigating the aggregation of α-Synuclein (aSyn), is well established across various model organisms. This essential molecule, GM1, is intimately linked to preventing aSyn aggregation, and its deficiency is believed to play a key role in the initiation of PD. In our current study, we attempted to shed light on the cytosolic interactions between GM1 and aSyn based on previous reports demonstrating gangliosides and monomeric aSyn to be present in neuronal cytosol. Native-PAGE and Western blot analysis of neuronal cytosol from mouse brains demonstrated the presence of both GM1 and monomeric aSyn in the neuronal cytosol of normal mouse brain. To demonstrate that an adequate level of GM1 prevents the aggregation of aSyn, we used NG108-15 and SH-SY5Y cells with and without treatment of 1-phenyl-2-palmitoyl-3-morpholino-1-propanol (PPMP), which inhibits the synthesis/expression of GM1. Cells treated with PPMP to reduce GM1 expression showed a significant increase in the formation of aggregated aSyn compared to untreated cells. We thus demonstrated that sufficient GM1 prevents the aggregation of aSyn. For this to occur, aSyn and GM1 must show proximity within the neuron. The present study provides evidence for such co-localization in neuronal cytosol, which also facilitates the inverse interaction revealed in studies with the two cell types above. This adds to the explanation of how GM1 prevents the aggregation of aSyn and onset of Parkinson\'s disease.
    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
    掌握跨人类细胞膜的选择性分子运输对医疗保健生物技术提出了巨大的挑战,同时为药物输送提供了突破的前景。基因治疗,和诊断成像。霍乱毒素B亚基(CTB)具有成为这些应用有用的货物转运蛋白的潜力。CTB是一种强大的蛋白质,适合于多种应用的重新设计;然而,蛋白质重新设计主要集中在蛋白质的N和C末端的修饰上。充分利用合理的重新设计需要详细了解表面残基对蛋白质稳定性和结合活性的贡献。这里,我们对CTB的58个表面残基进行了基于Rosetta的计算饱和扫描,包括GM1结合位点,分析配体结合和无配体的结构,以破译对蛋白质稳定性和GM1亲和力的突变影响。差示扫描荧光法和等温滴定量热法的补充实验结果为这些位置之间的40个丙氨酸突变体提供了解链温度和GM1结合亲和力。结果表明,CTB可以适应不同的突变,同时保持其稳定性和配体结合亲和力。这些突变可能允许寡糖结合特异性的修饰,以改变其细胞靶向,改变B亚基的细胞内路由,或通过改变蛋白质稳定性影响其保质期和体内半衰期。我们预计这里呈现的突变空间图将作为未来CTB重新设计的基石,为创新生物技术工具的发展铺平道路。
    Mastering selective molecule trafficking across human cell membranes poses a formidable challenge in healthcare biotechnology while offering the prospect of breakthroughs in drug delivery, gene therapy, and diagnostic imaging. The cholera toxin B-subunit (CTB) has the potential to be a useful cargo transporter for these applications. CTB is a robust protein that is amenable to reengineering for diverse applications; however, protein redesign has mostly focused on modifications of the N- and C-termini of the protein. Exploiting the full power of rational redesign requires a detailed understanding of the contributions of the surface residues to protein stability and binding activity. Here, we employed Rosetta-based computational saturation scans on 58 surface residues of CTB, including the GM1 binding site, to analyze both ligand-bound and ligand-free structures to decipher mutational effects on protein stability and GM1 affinity. Complimentary experimental results from differential scanning fluorimetry and isothermal titration calorimetry provided melting temperatures and GM1 binding affinities for 40 alanine mutants among these positions. The results showed that CTB can accommodate diverse mutations while maintaining its stability and ligand binding affinity. These mutations could potentially allow modification of the oligosaccharide binding specificity to change its cellular targeting, alter the B-subunit intracellular routing, or impact its shelf-life and in vivo half-life through changes to protein stability. We anticipate that the mutational space maps presented here will serve as a cornerstone for future CTB redesigns, paving the way for the development of innovative biotechnological tools.
    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
    单唾液酸神经节苷脂(GM1),一种普遍存在的脂筏成分,和血红素,血红素蛋白如血红蛋白的组成部分,对于调节细胞通讯和氧气运输的大脑神经元和红细胞红细胞的细胞膜至关重要。原卟啉IX(PPIX)及其衍生物血红素,相反,当过量引起血液系统疾病时显示出显著的细胞毒性作用,比如地中海贫血,贫血,疟疾,和神经变性。然而,到目前为止,人们对它们与细胞膜相互作用的深入分子病因知之甚少。在这里,已经研究了在PPIX及其衍生物血红素存在下磷脂和GM1二元混合物的聚合物垫支持的脂质双层(SLB)的结构,以预测模型磷脂膜中的分子相互作用。已采用基于高分辨率同步加速器的X射线散射技术来探索不同成分和浓度的组件的平面外结构。结构变化已与从Langmuir单层等温线获得的平均分子面积的等压变化相结合,以预测添加剂引起的膜冷凝和流化。PPIX诱导的无GM1磷脂SLB流态化,在存在GM1的情况下,结构变化提高了2倍,从而逆转为缩合。在原始SLB中观察到血红素浓度依赖性线性缩合效应。效果明显下降,并且观察到在含有GM1的混合SLB中失去了线性。我们的研究表明,GM1改变了血红素和PPIX与膜的相互作用,这可以用疏水和静电相互作用来解释。我们的研究表明GM1与PPIX和血红素的有利和不利的相互作用,分别,在膜中。在SLB和底层聚合物垫层中观察到的结构变化导致了分子特异性相互作用模型的提出,该模型可以使专门用于药物设计的制药行业受益。我们的研究可能丰富了我们对神经退行性疾病和药物-膜相互作用的基本生物物理理解。
    Monosialoganglioside (GM1), a ubiquitous component of lipid rafts, and hemin, an integral part of heme proteins such as hemoglobin, are essential to the cell membranes of brain neurons and erythrocyte red blood cells for regulating cellular communication and oxygen transport. Protoporphyrin IX (PPIX) and its derivative hemin, on the contrary, show significant cytotoxic effects when in excess causing hematological diseases, such as thalassemia, anemia, malaria, and neurodegeneration. However, the in-depth molecular etiology of their interactions with the cell membrane has so far been poorly understood. Herein, the structure of the polymer cushion-supported lipid bilayer (SLB) of the binary mixture of phospholipid and GM1 in the presence of PPIX and its derivative hemin has been investigated to predict the molecular interactions in model phospholipid membranes. A high-resolution synchrotron-based X-ray scattering technique has been employed to explore the out-of-plane structure of the assembly at different compositions and concentrations. The structural changes have been complemented with the isobaric changes in the mean molecular area obtained from the Langmuir monolayer isotherm to predict the additive-induced membrane condensation and fluidization. PPIX-induced fluidization of phospholipid SLB without GM1 was witnessed, which was reversed to condensation with 2-fold higher structural changes in the presence of GM1. A hemin concentration-dependent linear condensing effect was observed in the pristine SLB. The effect was significantly reduced, and the linearity was observed to be lost in the mixed SLB containing GM1. Our study shows that GM1 alters the interaction of hemin and PPIX with the membrane, which could be explained with the aid of hydrophobic and electrostatic interactions. Our study indicates favorable and unfavorable interactions of GM1 with PPIX and hemin, respectively, in the membrane. The observed structural changes in both SLB and the underlying polymer cushion layer lead to the proposal of a molecule-specific interaction model that can benefit the pharmaceutical industries specialized for drug designing. Our study potentially enriches our fundamental biophysical understanding of neurodegenerative diseases and drug-membrane interactions.
    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
    力快速转化为生物信号使活细胞能够响应其环境中的机械力。该力被认为最初影响质膜,然后改变膜蛋白的行为。磷脂酶D2(PLD2)是一种机械敏感性酶,由胆固醇和饱和神经节苷脂(GM1)组成的结构化膜-脂质位点调节。在这里,我们显示TWIK相关的K通道(TREK-1)的拉伸激活是由PLD2和涉及哺乳动物细胞中有序GM1和4,5-二磷酸(PIP2)簇的空间模式机械诱发的。首先,机械力使有序的脂质变形,它破坏了PLD2与GM1脂质的相互作用,并允许TREK-1和PLD2的复合物与PIP2簇结合。与PIP2的结合激活了酶,产生第二信使磷脂酸(PA)的通道。催化非活性PLD2的共表达抑制生物膜中的TREK-1拉伸电流。胆固醇的细胞摄取抑制培养物中的TREK-1电流,星形胶质细胞中胆固醇的消耗从小鼠脑中的GM1脂质释放TREK-1。果蝇中PLD2直系同源物的耗尽导致对机械力的超敏反应。我们得出的结论是,PLD2机械敏感性与TREK-1离子渗透性结合会引起机械诱发反应。
    “哎呀!”:你刚刚在咖啡桌的尖角上刺伤了你的小脚趾。这种痛苦的感觉源于神经细胞将有关外力的信息转化为大脑可以解释的电信号。越来越多,新的证据表明,这一过程可能始于这些细胞膜内的脂肪结构。细胞膜是由两个相互连接的,其中嵌入结构或分子自由移动的柔性脂质薄片。这种组织允许膜对外力做出物理响应,反过来,设置分子事件的运动链,帮助微调细胞如何将这些信息传递到大脑。例如,一种被称为PLD2的酶与脂筏结合-精确排列,膜中的刚性脂肪“团块”,部分由胆固醇形成。PLD2还被证明与离子通道TREK-1物理相互作用,然后激活离子通道TREK-1,这是一种基于膜的蛋白质,有助于防止神经细胞传递疼痛信号。然而,由于所涉及分子的性质和大小,难以研究这些相互作用的确切机制。为了解决这个问题,彼得森等人。将一种称为超分辨率成像的技术与一种新方法相结合,使他们能够观察膜脂对压力和流体剪切的反应。实验表明,机械力破坏了脂筏的精心排列,导致PLD2和TREK-1释放。然后,它们可以穿过周围的膜,到达打开TREK-1的开关。进一步的工作表明,小鼠细胞的胆固醇水平直接影响了团块如何形成和与PLD2结合,上下拨TREK-1介导的保护信号。总的来说,Petersen等人的研究。显示神经细胞膜可以包含基于胆固醇的“脂肪传感器”,有助于检测外力并参与疼痛调节。通过解剖这些过程,有可能更好地了解和治疗糖尿病和狼疮等疾病,这与疼痛敏感性和组织中胆固醇水平升高有关。
    Rapid conversion of force into a biological signal enables living cells to respond to mechanical forces in their environment. The force is believed to initially affect the plasma membrane and then alter the behavior of membrane proteins. Phospholipase D2 (PLD2) is a mechanosensitive enzyme that is regulated by a structured membrane-lipid site comprised of cholesterol and saturated ganglioside (GM1). Here we show stretch activation of TWIK-related K+ channel (TREK-1) is mechanically evoked by PLD2 and spatial patterning involving ordered GM1 and 4,5-bisphosphate (PIP2) clusters in mammalian cells. First, mechanical force deforms the ordered lipids, which disrupts the interaction of PLD2 with the GM1 lipids and allows a complex of TREK-1 and PLD2 to associate with PIP2 clusters. The association with PIP2 activates the enzyme, which produces the second messenger phosphatidic acid (PA) that gates the channel. Co-expression of catalytically inactive PLD2 inhibits TREK-1 stretch currents in a biological membrane. Cellular uptake of cholesterol inhibits TREK-1 currents in culture and depletion of cholesterol from astrocytes releases TREK-1 from GM1 lipids in mouse brain. Depletion of the PLD2 ortholog in flies results in hypersensitivity to mechanical force. We conclude PLD2 mechanosensitivity combines with TREK-1 ion permeability to elicit a mechanically evoked response.
    “Ouch!”: you have just stabbed your little toe on the sharp corner of a coffee table. That painful sensation stems from nerve cells converting information about external forces into electric signals the brain can interpret. Increasingly, new evidence is suggesting that this process may be starting at fat-based structures within the membrane of these cells. The cell membrane is formed of two interconnected, flexible sheets of lipids in which embedded structures or molecules are free to move. This organisation allows the membrane to physically respond to external forces and, in turn, to set in motion chains of molecular events that help fine-tune how cells relay such information to the brain. For instance, an enzyme known as PLD2 is bound to lipid rafts – precisely arranged, rigid fatty ‘clumps’ in the membrane that are partly formed of cholesterol. PLD2 has also been shown to physically interact with and then activate the ion channel TREK-1, a membrane-based protein that helps to prevent nerve cells from relaying pain signals. However, the exact mechanism underpinning these interactions is difficult to study due to the nature and size of the molecules involved. To address this question, Petersen et al. combined a technology called super-resolution imaging with a new approach that allowed them to observe how membrane lipids respond to pressure and fluid shear. The experiments showed that mechanical forces disrupt the careful arrangement of lipid rafts, causing PLD2 and TREK-1 to be released. They can then move through the surrounding membrane where they reach a switch that turns on TREK-1. Further work revealed that the levels of cholesterol available to mouse cells directly influenced how the clumps could form and bind to PLD2, and in turn, dialled up and down the protective signal mediated by TREK-1. Overall, the study by Petersen et al. shows that the membrane of nerve cells can contain cholesterol-based ‘fat sensors’ that help to detect external forces and participate in pain regulation. By dissecting these processes, it may be possible to better understand and treat conditions such as diabetes and lupus, which are associated with both pain sensitivity and elevated levels of cholesterol in tissues.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号