GM1 ganglioside

GM1 神经节苷脂
  • 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
    Piezo1是各种生物过程所需的机械敏感离子通道,但它的监管仍然知之甚少。这里,我们使用红细胞来解决这个问题,因为它们显示Piezo1簇,强大而动态的细胞骨架和三种类型的亚微脂质结构域,分别富含胆固醇,GM1神经节苷脂/胆固醇和鞘磷脂/胆固醇。我们发现Piezo1簇存在于边缘和凹坑红细胞区域。在Yoda1对Piezo1化学活化后,Piezo1团簇的比例主要在凹坑区域增加。这种增加伴随着Ca2+流入和棘突细胞的增加,在酒窝中的GM1/胆固醇富集结构域和边缘中的胆固醇富集结构域中。相反,Piezo1激活的作用在膜胆固醇消耗时被取消。此外,在Piezo1独立的Ca2+流入时,没有观察到上述变化。在棘球细胞比例高的健康供体中,Ca2+流入,即使在静息状态下,脂质结构域和Piezo1荧光也很高,而细胞骨架膜占有率较低。因此,遗传性球形细胞增多症患者红细胞的细胞骨架膜占有率和硬度降低,Piezo1荧光增强。总之,我们表明,Piezo1受到脂质结构域和细胞骨架的不同控制,并且受到造口细胞-椎间盘细胞-棘突细胞转化的青睐。
    Piezo1 is a mechanosensitive ion channel required for various biological processes, but its regulation remains poorly understood. Here, we used erythrocytes to address this question since they display Piezo1 clusters, a strong and dynamic cytoskeleton and three types of submicrometric lipid domains, respectively enriched in cholesterol, GM1 ganglioside/cholesterol and sphingomyelin/cholesterol. We revealed that Piezo1 clusters were present in both the rim and the dimple erythrocyte regions. Upon Piezo1 chemical activation by Yoda1, the Piezo1 cluster proportion mainly increased in the dimple area. This increase was accompanied by Ca2+ influx and a rise in echinocytes, in GM1/cholesterol-enriched domains in the dimple and in cholesterol-enriched domains in the rim. Conversely, the effects of Piezo1 activation were abrogated upon membrane cholesterol depletion. Furthermore, upon Piezo1-independent Ca2+ influx, the above changes were not observed. In healthy donors with a high echinocyte proportion, Ca2+ influx, lipid domains and Piezo1 fluorescence were high even at resting state, whereas the cytoskeleton membrane occupancy was lower. Accordingly, upon decreases in cytoskeleton membrane occupancy and stiffness in erythrocytes from patients with hereditary spherocytosis, Piezo1 fluorescence was increased. Altogether, we showed that Piezo1 was differentially controlled by lipid domains and the cytoskeleton and was favored by the stomatocyte-discocyte-echinocyte transformation.
    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
    自1980年代以来,已知对培养细胞施用神经节苷脂GM1诱导或增强神经元分化。GM1的作用机制依赖于其直接相互作用和随后的膜酪氨酸激酶受体的激活,TrkA,其天然地充当NGF受体。该过程由GM1的唯一寡糖部分介导,即五糖β-Gal-(1-3)-β-GalNAc-(1-4)-[α-Neu5Ac-(2-3)]-β-Gal-(1-4)-β-Glc。在这里,我们详细介绍了GM1寡糖部分介导TrkA依赖性神经源性加工的最低结构要求。通过体外和计算机生化方法,我们证明了TrkA激活所需的GM1的最小部分是神经节苷脂寡糖β-Gal-(1-3)-β-GalNAc-(1-4)-[α-Neu5Ac-(2-3)]-β-Gal的内核。在GM1寡糖的外半乳糖的3位添加唾液酸残基,形成GD1a的寡糖,阻止了与TrkA的相互作用以及由此产生的神经生成。相反,在外部半乳糖的2位添加岩藻糖残基,形成岩藻糖-GM1寡糖,没有阻止TrkA介导的神经生成。
    Since the 1980s, it has been known that the administration of ganglioside GM1 to cultured cells induced or enhanced neuronal differentiation. GM1 mechanism of action relies on its direct interaction and subsequent activation of the membrane tyrosine kinase receptor, TrkA, which naturally serves as NGF receptor. This process is mediated by the sole oligosaccharide portion of GM1, the pentasaccharide β-Gal-(1-3)-β-GalNAc-(1-4)-[α-Neu5Ac-(2-3)]-β-Gal-(1-4)-β-Glc. Here we detailed the minimum structural requirements of the oligosaccharide portion of GM1 for mediating the TrkA dependent neuritogenic processing. By in vitro and in silico biochemical approaches, we demonstrated that the minimal portion of GM1 required for the TrkA activation is the inner core of the ganglioside\'s oligosaccharide β-Gal-(1-3)-β-GalNAc-(1-4)-[α-Neu5Ac-(2-3)]-β-Gal. The addition of a sialic acid residue at position 3 of the outer galactose of the GM1 oligosaccharide, which forms the oligosaccharide of GD1a, prevented the interaction with TrkA and the resulting neuritogenesis. On the contrary, the addition of a fucose residue at position 2 of the outer galactose, forming the Fucosyl-GM1 oligosaccharide, did not prevent the TrkA-mediated neuritogenesis.
    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
    GM1是一种主要的脑神经节苷脂,发挥神经营养作用,神经保护和抗神经炎症作用。这项研究的目的是深入了解外源性GM1在脂多糖(LPS)刺激的MG6小鼠转化的小胶质细胞系中的抗神经炎症机制。首先,我们发现GM1阻止LPS诱导的小胶质细胞转化为变形虫样形状。GM1处理抑制LPS诱导的诱导型一氧化氮合酶的表达,环氧合酶-2(COX-2),和促炎细胞因子如TNF-α,MG6细胞中的IL-1β和IL-6。在LPS处理的小鼠中,GM1还降低纹状体小胶质细胞活化并减弱COX-2表达。随后的机制研究表明,GM1抑制LPS诱导的核因子κB(NF-κB)和激活蛋白-1(AP-1)的核易位,负责产生促炎介质的两个关键转录因子。GM1通过抑制Akt/NF-κB信号传导和丝裂原活化蛋白激酶(MAPKs)的活化而表现出抗炎性,包括p38MAPK,细胞外信号调节激酶1/2(ERK1/2)和c-Jun氨基末端激酶(JNK)。此外,GM1抑制LPS诱导的转化生长因子β活化激酶1(TAK1)和NADPH氧化酶2(NOX2)的活化,IκBα/NF-κB和MAPK/AP-1信号通路的上游调节因子。GM1还抑制NOX介导的活性氧(ROS)的产生,并防止LPS诱导的MG6细胞死亡。提示GM1的抗氧化作用。总之,GM1通过抑制Akt发挥抗神经炎症和抗氧化作用,TAK1和NOX2激活。
    GM1 is a major brain ganglioside that exerts neurotrophic, neuroprotective and antineuroinflammatory effects. The aim of this study was to obtain insights into the antineuroinflammatory mechanisms of exogenous GM1 in lipopolysaccharide (LPS)-stimulated MG6 mouse transformed microglial cell line. First, we found that GM1 prevented the LPS-induced transformation of microglia into an amoeboid-like shape. GM1 treatment inhibited LPS-induced expression of inducible nitric oxide synthase, cyclooxygenase-2 (COX-2), and proinflammatory cytokines such as TNF-α, IL-1β and IL-6 in MG6 cells. In LPS-treated mice, GM1 also reduced striatal microglia activation and attenuated COX-2 expression. Subsequent mechanistic studies showed that GM1 suppressed LPS-induced nuclear translocation of nuclear factor κB (NF-κB) and activator protein-1 (AP-1), two critical transcription factors responsible for the production of proinflammatory mediators. GM1 exhibited antineuroinflammatory properties by suppressing Akt/NF-κB signaling and the activation of mitogen-activated protein kinases (MAPKs), including p38 MAPK, extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK). Furthermore, GM1 suppressed LPS-induced activation of transforming growth factor-β-activated kinase 1 (TAK1) and NADPH oxidase 2 (NOX2), upstream regulators of the IκBα/NF-κB and MAPK/AP-1 signaling pathways. GM1 also inhibited NOX-mediated reactive oxygen species (ROS) production and protected against LPS-induced MG6 cell death, suggesting an antioxidant role of GM1. In conclusion, GM1 exerts both antineuroinflammatory and antioxidative effects by inhibiting Akt, TAK1 and NOX2 activation.
    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: Video-Audio Media
    背景:细胞间的通讯对于组织的反应至关重要,适应,并在盛行的环境中茁壮成长。几种机制介导细胞间信号,包括隧道纳米管,间隙连接,和细胞外囊泡(EV)。根据当地和系统条件,电动汽车可能含有促进生存的货物,神经保护,或病理学。我们对病理性细胞间信号传导的理解已经通过使用源自人类多能干细胞(hPSC)的神经元的疾病模型得到了支持。
    方法:这里,我们使用hPSC衍生的视网膜神经节细胞(hRGC)和小鼠视觉系统来研究调节EV产生对细胞间运输和细胞存活的影响。我们通过抑制中性鞘磷脂酶(nSMase)降低鞘磷脂分解代谢成神经酰胺,探讨了EV调节对细胞存活的影响,使用GW4869。我们通过探测膜联蛋白A5,磷脂酰丝氨酸,可行的线粒体,和线粒体活性氧。在体内,我们进行了GW4869眼内注射,并测量了RGC和上丘神经元密度以及RGC顺行轴突转运。
    结果:用GW4869给药hRGC24小时后,我们发现nSMase的抑制降低了神经酰胺并增强了GM1神经节苷脂的积累。这种抑制作用也降低了小型电动汽车的密度,增加了大型电动汽车的密度,富集了促凋亡蛋白,附件A5。降低nSMase活性增加hRGC凋亡起始,由于凋亡颗粒的密度和摄取增加,由膜联蛋白A5结合磷脂鉴定,磷脂酰丝氨酸。我们通过开发GW4869处理和未处理的hRGC的共培养系统来测定线粒体的细胞间运输。在处理过的细胞中,抑制nSMase减少了活线粒体的数量,不仅在治疗中驱动线粒体活性氧,而且在共培养中添加了幼稚的hRGC。在老鼠身上,在玻璃体内单次注射GW4869后20天,我们发现上丘中RGC及其轴突受体神经元的显著损失。随后,顺行RGC轴突向丘的运输急剧减少。
    结论:总体而言,我们的数据表明,通过抑制nSMase来扰乱鞘磷脂的生理分解代谢重组质膜相关鞘脂,改变神经元生成的电动汽车的轮廓,并通过改变促存活与变性EV的平衡来促进体外和体内神经变性。视频摘要。
    Cell-to-cell communication is vital for tissues to respond, adapt, and thrive in the prevailing milieu. Several mechanisms mediate intercellular signaling, including tunneling nanotubes, gap junctions, and extracellular vesicles (EV). Depending on local and systemic conditions, EVs may contain cargoes that promote survival, neuroprotection, or pathology. Our understanding of pathologic intercellular signaling has been bolstered by disease models using neurons derived from human pluripotent stems cells (hPSC).
    Here, we used hPSC-derived retinal ganglion cells (hRGC) and the mouse visual system to investigate the influence of modulating EV generation on intercellular trafficking and cell survival. We probed the impact of EV modulation on cell survival by decreasing the catabolism of sphingomyelin into ceramide through inhibition of neutral sphingomyelinase (nSMase), using GW4869. We assayed for cell survival in vitro by probing for annexin A5, phosphatidylserine, viable mitochondria, and mitochondrial reactive oxygen species. In vivo, we performed intraocular injections of GW4869 and measured RGC and superior colliculus neuron density and RGC anterograde axon transport.
    Following twenty-four hours of dosing hRGCs with GW4869, we found that inhibition of nSMase decreased ceramide and enhanced GM1 ganglioside accumulation. This inhibition also reduced the density of small EVs, increased the density of large EVs, and enriched the pro-apoptotic protein, annexin A5. Reducing nSMase activity increased hRGC apoptosis initiation due to enhanced density and uptake of apoptotic particles, as identified by the annexin A5 binding phospholipid, phosphatidylserine. We assayed intercellular trafficking of mitochondria by developing a coculture system of GW4869-treated and naïve hRGCs. In treated cells, inhibition of nSMase reduced the number of viable mitochondria, while driving mitochondrial reactive oxygen species not only in treated, but also in naive hRGCs added in coculture. In mice, 20 days following a single intravitreal injection of GW4869, we found a significant loss of RGCs and their axonal recipient neurons in the superior colliculus. This followed a more dramatic reduction in anterograde RGC axon transport to the colliculus.
    Overall, our data suggest that perturbing the physiologic catabolism of sphingomyelin by inhibiting nSMase reorganizes plasma membrane associated sphingolipids, alters the profile of neuron-generated EVs, and promotes neurodegeneration in vitro and in vivo by shifting the balance of pro-survival versus -degenerative EVs. Video Abstract.
    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
    鞘糖脂代谢的改变与肌萎缩侧索硬化症(ALS)的病理生理机制有关。一种影响运动神经元的神经退行性疾病。因此,给药GM1,一种含唾液酸的鞘糖脂,对神经元损伤具有保护作用并支持神经元稳态,通过其生物活性成分介导的这些作用,寡糖头(GM1-OS)。这里,我们为GM1在ALS中的治疗功效增加了新的证据:其对受谷氨酸诱导的兴奋性毒性影响的WT和SOD1G93A运动神经元的施用显着增加了神经元存活和保留的神经突网络,抵抗细胞内蛋白质积累和线粒体损伤。重要的是,GM1-OS忠实地复制GM1活性,强调即使在ALS中,GM1的保护功能也严格取决于其五糖。
    Alterations in glycosphingolipid metabolism have been linked to the pathophysiological mechanisms of amyotrophic lateral sclerosis (ALS), a neurodegenerative disease affecting motor neurons. Accordingly, administration of GM1, a sialic acid-containing glycosphingolipid, is protective against neuronal damage and supports neuronal homeostasis, with these effects mediated by its bioactive component, the oligosaccharide head (GM1-OS). Here, we add new evidence to the therapeutic efficacy of GM1 in ALS: Its administration to WT and SOD1G93A motor neurons affected by glutamate-induced excitotoxicity significantly increased neuronal survival and preserved neurite networks, counteracting intracellular protein accumulation and mitochondria impairment. Importantly, the GM1-OS faithfully replicates GM1 activity, emphasizing that even in ALS the protective function of GM1 strictly depends on its pentasaccharide.
    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
    目的:本研究旨在评估药代动力学,安全,GM1在中国健康受试者和多发性骨髓瘤患者中的疗效。
    方法:本研究中使用的数据来自两项剂量递增试验:GM1-101,涉及70名健康受试者,和GM1-201,其中包括160例多发性骨髓瘤患者。使用非线性混合效应方法对90名参与者的子集进行了群体药代动力学(PopPK)分析,并对潜在的协变量进行了定量探索。观察生命体征的任何异常,体检,实验室测试,研究期间的心电图,以及任何自发报告和直接观察到的不良事件,被记录用于安全评估。此外,神经毒性量表用于评估GM1预防化疗引起的周围神经病变的疗效,并结合药代动力学参数进行暴露反应分析.
    结果:具有一级消除的单室模型最好地表征了GM1的药代动力学。分配的间隙和体积,根据最终模型估计,GM1-A分别为0.0942升/小时和3.27升/小时,GM1-B为0.0714升/小时和2.82升/小时,分别。协变量,如性别,体重,和白蛋白显著影响药代动力学参数,然而,在90%置信区间内,受试者和参考受试者之间的稳态暴露差异小于45%.与GM1相关的不良反应发生在GM1-101和GM1-201队列中的20名(28.6%)和57名(35.6%)受试者中,分别。与空白对照组相比,每个GM1剂量组在第4期和第6期结束时TNSc和FACT-Ntx评分从基线的变化较低。GM1的400mg剂量组显示出比其他剂量组更高的有效性。然而,暴露反应分析显示,随着GM1暴露的增加,疗效没有显着变化。
    结论:本研究首次对GM1进行群体药代动力学分析。GM1在健康受试者和多发性骨髓瘤患者中表现出良好的安全性。GM1被证明可有效缓解化疗引起的周围神经病变,但是这项研究观察到其疗效与暴露之间没有显着相关性。
    背景:ChiCTR2000041283和ChiCTR2000041283。
    OBJECTIVE: This study aimed to assess the pharmacokinetics, safety, and efficacy of GM1 in healthy Chinese subjects and patients with multiple myeloma.
    METHODS: The data used in this study was derived from two dose-escalation trials: GM1-101, involving 70 healthy subjects, and GM1-201, which included 160 multiple myeloma patients. Population pharmacokinetics (PopPK) analysis was conducted on a subset of 90 participants using a nonlinear mixed-effects approach, and potential covariates were explored quantitatively. Observations of any abnormalities in vital signs, physical examinations, laboratory tests, and electrocardiograms during the study period, along with any spontaneously reported and directly observed adverse events, were documented for safety evaluation. Furthermore, neurotoxicity scales were used to assess the efficacy of GM1 as a prophylaxis for chemotherapy-induced peripheral neuropathy and to perform exposure-response analyses in conjunction with pharmacokinetic parameters.
    RESULTS: A one-compartment model with first-order elimination best characterized the pharmacokinetics of GM1. The clearance and volume of distribution, as estimated by the final model, were 0.0942 L/h and 3.27 L for GM1-A, and 0.0714 L/h and 2.82 L for GM1-B, respectively. Covariates such as sex, body weight, and albumin significantly influenced pharmacokinetic parameters, yet the variation in steady-state exposure between subjects and reference subjects was less than 45% within their 90% confidence interval. Adverse reactions related to GM1 occurred in 20 (28.6%) and 57 (35.6%) subjects in the GM1-101 and GM1-201 cohorts, respectively. The changes in TNSc and FACT-Ntx scores from baseline at the end of periods 4 and 6 were lower in each GM1 dose group compared to the blank control group. The 400 mg dose group of GM1 displayed greater effectiveness than other dose groups. However, exposure-response analysis revealed no significant modification in efficacy with increasing GM1 exposure.
    CONCLUSIONS: This study provides the first population pharmacokinetic analysis of GM1. GM1 exhibits a favorable safety profile among healthy subjects and patients with multiple myeloma. GM1 proved effective in mitigating chemotherapy-induced peripheral neuropathy, but this study observed no significant correlation between its efficacy and exposure.
    BACKGROUND: ChiCTR2000041283 and ChiCTR2000041283.
    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
    阿尔茨海默病(AD)与大脑中淀粉样β(Aβ)交叉β原纤维的进行性积累有关。与GM1神经节苷脂密切相关的Aβ物种,神经元膜中丰富的鞘糖脂,促进淀粉样蛋白原纤维的形成;因此,它们可能是有吸引力的临床目标。然而,含GM1的脂质膜中Aβ的活性构象状态仍然未知。本固态核磁共振研究揭示了一种非纤维状Aβ组合,其特征是在GM1神经节苷脂簇上特异性形成的双层反平行β结构。我们的数据表明,这种独特的组合没有在含GM1的膜上转化为原纤维,但可以促进单体Aβ转化为原纤维,这表明暴露于溶剂的疏水层提供了引发Aβ原纤维形成的催化表面。我们的发现为设计针对催化活性Aβ构象物种的药物以开发抗AD治疗剂提供了结构线索。
    Alzheimer\'s disease (AD) is associated with progressive accumulation of amyloid-β (Aβ) cross-β fibrils in the brain. Aβ species tightly associated with GM1 ganglioside, a glycosphingolipid abundant in neuronal membranes, promote amyloid fibril formation; therefore, they could be attractive clinical targets. However, the active conformational state of Aβ in GM1-containing lipid membranes is still unknown. The present solid-state nuclear magnetic resonance study revealed a nonfibrillar Aβ assemblage characterized by a double-layered antiparallel β-structure specifically formed on GM1 ganglioside clusters. Our data show that this unique assemblage was not transformed into fibrils on GM1-containing membranes but could promote conversion of monomeric Aβ into fibrils, suggesting that a solvent-exposed hydrophobic layer provides a catalytic surface evoking Aβ fibril formation. Our findings offer structural clues for designing drugs targeting catalytically active Aβ conformational species for the development of anti-AD therapeutics.
    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
    这项研究试图回答St3gal5(GM3合酶)基因双等位基因和单等位基因破坏的小鼠是否可能从GM1替代疗法中受益的问题。由该唾液酸转移酶产生的GM3产生下游GD3和神经节苷脂的神经节系列。后者包括a系列(GM1+GD1a),这已被证明对神经元存活和功能最重要(尤其是GM1,GD1a为其提供了储备池)。这些双等位基因小鼠可作为患有这种相对罕见的常染色体隐性条件(ST3GAL5-/-)的儿童的模型,这些儿童患有快速的神经系统衰退,包括运动障碍,智力残疾,视觉和听力损失,未能茁壮成长,和其他严重的情况导致2-5岁的早期死亡,没有支持治疗。这里,我们研究了这两只老鼠,作为这些儿童的父母和近亲的模型,他们可能由于部分GM1缺乏而遭受长期残疾,包括帕金森病(PD)。我们发现,两种类型的小鼠表现出的运动和记忆障碍都可以通过GM1应用来解决。这表明GM1对GM1缺乏引起的疾病的潜在治疗价值。包括GM3合成酶缺乏症和PD。值得注意的是,这些研究中使用的GM1是合成的,而不是动物大脑来源的,重申前者的治疗效果。
    This study attempts to answer the question of whether mice with biallelic and monoallelic disruption of the St3gal5 (GM3 synthase) gene might benefit from GM1 replacement therapy. The GM3 produced by this sialyltransferase gives rise to downstream GD3 and the ganglio-series of gangliosides. The latter includes the a-series (GM1 + GD1a), which has proved most essential for neuron survival and function (especially GM1, for which GD1a provides a reserve pool). These biallelic mice serve as a model for children with this relatively rare autosomal recessive condition (ST3GAL5-/-) who suffer rapid neurological decline including motor loss, intellectual disability, visual and hearing loss, failure to thrive, and other severe conditions leading to an early death by 2-5 years of age without supportive care. Here, we studied both these mice, which serve as a model for the parents and close relatives of these children who are likely to suffer long-term disabilities due to partial deficiency of GM1, including Parkinson\'s disease (PD). We find that the movement and memory disorders manifested by both types of mice can be resolved with GM1 application. This suggests the potential therapeutic value of GM1 for disorders stemming from GM1 deficiency, including GM3 synthase deficiency and PD. It was noteworthy that the GM1 employed in these studies was synthetic rather than animal brain-derived, reaffirming the therapeutic efficacy of the former.
    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
    纤维聚集的α-突触核蛋白代表帕金森病的神经系统标志,被认为在该疾病中起致病作用。虽然导致α-突触核蛋白聚集的原因尚不清楚,GM1神经节苷脂相互作用被认为可以防止这一过程。GM1如何发挥这些功能还不完全清楚,尽管其可溶性寡糖(GM1-OS)的主要作用正在出现。的确,我们最近确定GM1-OS是负责GM1神经营养和神经保护特性的生物活性部分,特别是在体外和体内模型中恢复帕金森病表型。这里,我们报道了GM1-OS对α-突触核蛋白聚集的体外疗效和毒性。通过淀粉样蛋白接种聚集测定和核磁共振波谱,我们证明GM1-OS能够预防自发性和朊病毒样α-突触核蛋白聚集。此外,重组单体α-突触核蛋白的圆二色光谱显示GM1-OS没有诱导α-突触核蛋白二级结构的任何变化。重要的是,GM1-OS显着增加了受α-突触核蛋白寡聚体影响的多巴胺能神经元的存活和保留的神经突网络,以及小胶质细胞活化的减少。这些数据进一步证明神经节苷脂GM1通过其寡糖也在防止帕金森病中的α-突触核蛋白致病聚集中起作用。打开GM1-OS作为候选药物的透视窗口。
    Fibrillary aggregated α-synuclein represents the neurologic hallmark of Parkinson\'s disease and is considered to play a causative role in the disease. Although the causes leading to α-synuclein aggregation are not clear, the GM1 ganglioside interaction is recognized to prevent this process. How GM1 exerts these functions is not completely clear, although a primary role of its soluble oligosaccharide (GM1-OS) is emerging. Indeed, we recently identified GM1-OS as the bioactive moiety responsible for GM1 neurotrophic and neuroprotective properties, specifically reverting the parkinsonian phenotype both in in vitro and in vivo models. Here, we report on GM1-OS efficacy against the α-synuclein aggregation and toxicity in vitro. By amyloid seeding aggregation assay and NMR spectroscopy, we demonstrated that GM1-OS was able to prevent both the spontaneous and the prion-like α-synuclein aggregation. Additionally, circular dichroism spectroscopy of recombinant monomeric α-synuclein showed that GM1-OS did not induce any change in α-synuclein secondary structure. Importantly, GM1-OS significantly increased neuronal survival and preserved neurite networks of dopaminergic neurons affected by α-synuclein oligomers, together with a reduction of microglia activation. These data further demonstrate that the ganglioside GM1 acts through its oligosaccharide also in preventing the α-synuclein pathogenic aggregation in Parkinson\'s disease, opening a perspective window for GM1-OS as drug candidate.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号