G proteins

G 蛋白
  • 文章类型: Journal Article
    在植物异源三聚体G蛋白介导的信号转导研究中,它们在低钾刺激透射率中的作用仍有待阐明。这里,我们发现,随着外界钾浓度的降低,野生型拟南芥的主根生长逐渐受到抑制,而G蛋白β亚基AGB1和γ亚基AGG1,AGG2和AGG3突变体的主根在低钾条件(LK)下仍可以生长。外源NAA的施用减弱了agb1和agg1/2/3的初根伸长,但促进了LK胁迫下野生型幼苗的生长。使用ProDR5:GFP,ProPIN1:PIN1-GFP和ProPIN2:PIN2-GFP报告系,在LK下,在野生型根中观察到胚根顶端的生长素浓度降低,PIN1和PIN2外排载体丰度降低,在agb1和agg1/2/3中没有记录的现象。进一步的蛋白水解和转录评估显示,在LK下,野生型背景下PIN1的降解增强和PIN2的表达抑制,与在agb1和agg1/2/3突变体中观察到的稳定性相反。我们的结果表明,G蛋白β和γ亚基通过改变PIN1降解和PIN2生物合成来调节生长素的再分布,在LK下抑制拟南芥根生长中起关键作用。
    In the investigation of heterotrimeric G protein-mediated signal transduction in planta, their roles in the transmittance of low K+ stimuli remain to be elucidated. Here, we found that the primary root growth of wild-type Arabidopsis was gradually inhibited with the decrease of external K+ concentrations, while the primary root of the mutants for G protein β subunit AGB1 and γ subunits AGG1, AGG2 and AGG3 could still grow under low K+ conditions (LK). Exogenous NAA application attenuated primary root elongation in agb1 and agg1/2/3 but promoted the growth in wild-type seedlings under LK stress. Using ProDR5:GFP, ProPIN1:PIN1-GFP and ProPIN2:PIN2-GFP reporter lines, a diminishment in auxin concentration at the radicle apex and a reduction in PIN1and PIN2 efflux carrier abundance were observed in wild-type roots under LK, a phenomenon not recorded in the agb1 and agg1/2/3. Further proteolytic and transcriptional assessments revealed an enhanced degradation of PIN1 and a suppressed expression of PIN2 in the wild-type background under LK, contrasting with the stability observed in the agb1 and agg1/2/3 mutants. Our results indicate that the G protein β and γ subunits play pivotal roles in suppressing of Arabidopsis root growth under LK by modulating auxin redistribution via alterations in PIN1 degradation and PIN2 biosynthesis.
    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
    人类G蛋白偶联受体(GPCRs)家族由大约800个不同的成员组成,目前约有35%的药物靶向GPCRs。然而,GPCR结构生物学,结构引导药物设计所必需的,落后于其他膜蛋白,直到2000年才解决了GPCR(视紫红质)的第一个晶体结构。从2007年开始,蛋白质工程促进了其他GPCR结构的确定,新的结晶技术,与抗体片段复合,和其他策略。最近,使用骆驼科动物仅重链抗体片段(纳米抗体)作为晶体学伴侣已经彻底改变了GPCR结构生物学领域,帮助确定超过340个GPCR结构。在大多数情况下,作为与纳米抗体(Nbs)的复合物解决的GPCR结构揭示了同源或非天然配体的结合模式;在少数情况下,相同的Nb已经充当GPCR信号传导的正构或变构调节剂。在这篇综述中,我们总结了过去十年中构思和实施的多种巧妙策略,以利用纳米体的发现从结构角度研究GPCRs。意义陈述G蛋白偶联受体(GPCRs)是主要的药理学靶标,并且以高分辨率确定其结构对于结构指导药物设计和了解其功能至关重要。单结构域抗体(纳米抗体)极大地促进了GPCRs的结构确定,通过直接与受体或间接通过蛋白质伴侣形成复合物。
    The family of human G protein-coupled receptors (GPCRs) is comprised of about 800 different members, with about 35% of current pharmaceutical drugs targeting GPCRs. However, GPCR structural biology, necessary for structure-guided drug design, has lagged behind that of other membrane proteins, and it was not until the year 2000 when the first crystal structure of a GPCR (rhodopsin) was solved. Starting in 2007, the determination of additional GPCR structures was facilitated by protein engineering, new crystallization techniques, complexation with antibody fragments, and other strategies. More recently, the use of camelid heavy-chain-only antibody fragments (nanobodies) as crystallographic chaperones has revolutionized the field of GPCR structural biology, aiding in the determination of more than 340 GPCR structures to date. In most cases, the GPCR structures solved as complexes with nanobodies (Nbs) have revealed the binding mode of cognate or non-natural ligands; in a few cases, the same Nb has acted as an orthosteric or an allosteric modulator of GPCR signaling. In this review we summarize the multiple ingenious strategies that have been conceived and implemented in the last decade to capitalize on the discovery of nanobodies to study GPCRs from a structural perspective. Significance Statement G protein-coupled receptors (GPCRs) are major pharmacological targets, and the determination of their structures at high resolution has been essential for structure-guided drug design and for insights about their functions. Single domain antibodies (nanobodies) have greatly facilitated the structural determination of GPCRs, by forming complexes directly with the receptors or indirectly through protein partners.
    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
    G蛋白偶联受体(GPCRs)是人类基因组中编码的最大的跨膜受体。它们启动由大量细胞外刺激触发的细胞反应,从神经递质或激素到光子。在刺激时,GPCRs激活细胞质中的异源三聚体G蛋白(Gαβγ),然后将信号传递给它们的效应子,以引起细胞反应。鉴于GPCRs和G蛋白在生理和疾病中的广泛生物学和生物医学相关性,人们非常有兴趣开发和优化方法,以高精度和跨实验系统测量它们在细胞通信中的功能。这篇综述提供了测量GPCR-G蛋白信号的方法的历史观点,从第二信使的量化和其他活动的间接读数,直接检测G蛋白活性的生物传感器。后者是对具有不同实验能力的G蛋白活性的各种光学生物传感器的设计原理的更详细概述的重点。我们将强调检测不同G蛋白激活标志的生物传感器的优点和局限性,如Gα和Gβγ的解离或Gα上的核苷酸交换,以及它们对检测内源性和外源性信号传导成分介导的信号传导的适用性,或在生理相关的系统,如原代细胞。总的来说,本综述旨在对直接测量G蛋白活性的生物传感器的最新技术进行评估,以使读者能够就当前可用工具的选择和实施做出明智的决定.重要性陈述G蛋白活性生物传感器已成为评估GPCR信号传导和药理学的基本和广泛的工具。然而,研究人员面临着从越来越多的G蛋白活性生物传感器中进行选择的挑战.这篇综述概述了用于直接检测细胞中G蛋白活性的不同光学生物传感器设计的特征和功能,目的是促进合理选择符合研究人员具体科学问题和需求的系统。
    G protein-coupled receptors (GPCRs) are the largest class of transmembrane receptors encoded in the human genome, and they initiate cellular responses triggered by a plethora of extracellular stimuli ranging from neurotransmitters and hormones to photons. Upon stimulation, GPCRs activate heterotrimeric G proteins (Gαβγ) in the cytoplasm, which then convey signals to their effectors to elicit cellular responses. Given the broad biological and biomedical relevance of GPCRs and G proteins in physiology and disease, there is great interest in developing and optimizing approaches to measure their signaling activity with high accuracy and across experimental systems pertinent to their functions in cellular communication. This review provides a historical perspective on approaches to measure GPCR-G protein signaling, from quantification of second messengers and other indirect readouts of activity to biosensors that directly detect the activity of G proteins. The latter is the focus of a more detailed overview of the evolution of design principles for various optical biosensors of G protein activity with different experimental capabilities. We will highlight advantages and limitations of biosensors that detect different G protein activation hallmarks, like dissociation of Gα and Gβγ or nucleotide exchange on Gα, as well as their suitability to detect signaling mediated by endogenous versus exogenous signaling components or in physiologically relevant systems like primary cells. Overall, this review intends to provide an assessment of the state-of-the-art for biosensors that directly measure G protein activity to allow readers to make informed decisions on the selection and implementation of currently available tools. SIGNIFICANCE STATEMENT: G protein activity biosensors have become essential and widespread tools to assess GPCR signaling and pharmacology. Yet, investigators face the challenge of choosing from a growing list of G protein activity biosensors. This review provides an overview of the features and capabilities of different optical biosensor designs for the direct detection of G protein activity in cells, with the aim of facilitating the rational selection of systems that align with the specific scientific questions and needs of investigators.
    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
    G蛋白偶联受体(GPCR)的F类由10个卷曲蛋白(FZD1-10)和平滑蛋白(SMO)组成。FZD结合无翼/Int-1(WNT)家族的分泌的脂糖蛋白并被其激活,而SMO则被Hedgehog(Hh)家族的形态发生素作用于跨膜蛋白Patched(PTCH)间接激活。我们对FZD和SMO作为动态跨膜受体和分子机器的理解的进展,自第一份F类GPCRIUPHAR命名报告以来的过去14年中出现的,对更新进行校正。本文重点介绍了分子药理学和结构生物学的进展,为配体识别提供了新的机制见解,受体激活机制,信号启动和信号规范。此外,F类GPCRs继续发展为药物靶标,基因编码的生物传感器和CRISP/Cas9编辑的细胞系统等新技术和工具为这些受体的精细功能分析做出了贡献。此外,晶体结构分析和低温电子显微镜的进步有助于我们对结构-功能关系的知识的快速发展,为药物开发提供了一个很好的起点。尽管取得了进展,但在充分理解WNT/FZD和Hh/SMO信号系统的复杂性方面仍然存在问题和挑战。重要性陈述近年来的研究带来了对卷曲和平滑的激活机制的实质性功能和结构见解。虽然这一进展进一步加深了我们对配体识别的机械理解,受体激活,信号规格和启动,出现了更广泛的机会,允许靶向F类GPCRs用于使用生物制剂和小分子化合物的治疗和再生医学。
    The class F of G protein-coupled receptors (GPCRs) consists of ten Frizzleds (FZD1-10) and Smoothened (SMO). FZDs bind and are activated by secreted lipoglycoproteins of the Wingless/Int-1 (WNT) family and SMO is indirectly activated by the Hedgehog (Hh) family of morphogens acting on the transmembrane protein Patched (PTCH). The advance of our understanding of FZDs and SMO as dynamic transmembrane receptors and molecular machines, which emerged during the past 14 years since the first class F GPCR IUPHAR nomenclature report, justifies an update. This article focuses on the advances in molecular pharmacology and structural biology providing new mechanistic insight into ligand recognition, receptor activation mechanisms, signal initiation and signal specification. Furthermore, class F GPCRs continue to develop as drug targets, and novel technologies and tools such as genetically encoded biosensors and CRISP/Cas9 edited cell systems have contributed to refined functional analysis of these receptors. Also, advances in crystal structure analysis and cryogenic electron microscopy contribute to a rapid development of our knowledge about structure-function relationships providing a great starting point for drug development. Despite the progress questions and challenges remain to fully understand the complexity of the WNT/FZD and Hh/SMO signaling systems. Significance Statement The recent years of research have brought about substantial functional and structural insight into mechanisms of activation of Frizzleds and Smoothened. While the advance furthers our mechanistic understanding of ligand recognition, receptor activation, signal specification and initiation, broader opportunities emerge that allow targeting class F GPCRs for therapy and regenerative medicine employing both biologics and small molecule compounds.
    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
    “分子胶”定义为可以是内源性或合成的小分子,可促进蛋白质之间在其界面处的相互作用。变构调节剂,特别是GPCR变构调节剂,可以促进给定受体换能器的缔合和解离,但可以在与界面相距一定距离的地方实现这一目标。然而,在存在变构调节剂的情况下,GPCRG蛋白复合物的最新结构表明,某些GPCR变构调节剂可以充当“分子胶”,在界面处与受体和换能器相互作用,从而以正负方式偏置换能器信号传导取决于换能器。鉴于这些现象,我们讨论了这类变构调节剂用作分子工具和未来药物开发的含义。
    \"Molecular Glues\" are defined as small molecules that can either be endogenous or synthetic which promote interactions between proteins at their interface. Allosteric modulators, specifically GPCR allosteric modulators, can promote both the association and the dissociation of a given receptor\'s transducer but accomplishes this \"at a distance\" from the interface. However, recent structures of GPCR G protein complexes in the presence of allosteric modulators indicate that some GPCR allosteric modulators can act as \"molecular glues\" interacting with both the receptor and the transducer at the interface biasing transducer signaling in both a positive and negative manner depending on the transducer. Given these phenomena we discuss the implications for this class of allosteric modulators to be used as molecular tools and for future drug development.
    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
    AB型细菌蛋白毒素的细胞毒性坏死因子(CNF)家族在其RhoGTP酶底物上催化两种类型的修饰:脱酰胺和转谷氨酰胺。已经确定,大肠杆菌CNF1及其紧密同源蛋白主要催化脱酰胺作用,而博德特氏菌属皮肤坏死毒素(DNT)主要催化转谷氨酰胺作用。快速扩增的微生物基因组测序数据已经揭示存在至少13种CNF1同源物的全长变体。来自大肠杆菌菌株GN02091的CNFx与CNF家族的所有其他成员相距最远,在蛋白质水平上具有50%-55%的序列同一性,在DNA水平上每个位点具有0.45-0.52个核苷酸取代。CNFx修饰RhoA,Rac1和Cdc42和CNF1一样,激活人HEK293T细胞中的下游SRE依赖性有丝分裂信号通路,但EC50值高出1000倍。与其他先前表征的CNF毒素不同,CNFx主要通过转谷氨酰胺修饰Rho蛋白,如凝胶移位测定所证明的,并通过MALDI质谱分析证实,当与Rho蛋白底物在大肠杆菌BL21细胞中共表达或通过直接处理HEK293T细胞时。CNF1和CNFx序列的比较鉴定了对应于CNF1中832和862位的两个关键活性位点残基。每个毒素中这些残基处的相互位点特异性突变揭示了定义CNF中脱酰胺酶与转谷氨酰胺酶活性的偏好的分层规则。还发现在CNFx的C-末端的另外的独特Cys残基对于延迟货物递送是关键的。重要毒性坏死因子(CNF)毒素不仅在致病性大肠杆菌和其他细菌病原体中发挥重要的毒力作用,但是在越来越多的临床分离株的基因组中也发现了类似CNF的基因。利用CNF毒素之间的进化关系的力量,可以破译分级活性位点决定子,这些决定子定义它们是否通过脱酰胺或转凝集来修饰其RhoGTP酶底物。随着我们的发现,与其他已知的CNF不同,远处的CNF变体(CNFx)主要使其RhoGTP酶底物转谷氨酰胺,“CNFs脱酰胺酯和DNT转谷氨酸酯”的范例最终可以归因于活性位点内的两个关键氨基酸残基,而不是先前鉴定的催化性Cys-His二位残基。我们的方法和研究结果的意义在于,它们可以应用于破译其他细菌蛋白质的酶反应决定簇和底物特异性,以开发精确治疗策略。
    The cytotoxic necrotizing factor (CNF) family of AB-type bacterial protein toxins catalyze two types of modification on their Rho GTPase substrates: deamidation and transglutamination. It has been established that E. coli CNF1 and its close homolog proteins catalyze primarily deamidation and Bordetella dermonecrotic toxin (DNT) catalyzes primarily transglutamination. The rapidly expanding microbial genome sequencing data have revealed that there are at least 13 full-length variants of CNF1 homologs. CNFx from E. coli strain GN02091 is the most distant from all other members of the CNF family with 50%-55% sequence identity at the protein level and 0.45-0.52 nucleotide substitutions per site at the DNA level. CNFx modifies RhoA, Rac1, and Cdc42, and like CNF1, activates downstream SRE-dependent mitogenic signaling pathways in human HEK293T cells, but at a 1,000-fold higher EC50 value. Unlike other previously characterized CNF toxins, CNFx modifies Rho proteins primarily through transglutamination, as evidenced by gel-shift assay and confirmed by MALDI mass spectral analysis, when coexpressed with Rho-protein substrates in E. coli BL21 cells or through direct treatment of HEK293T cells. A comparison of CNF1 and CNFx sequences identified two critical active-site residues corresponding to positions 832 and 862 in CNF1. Reciprocal site-specific mutations at these residues in each toxin revealed hierarchical rules that define the preference for deamidase versus a transglutaminase activity in CNFs. An additional unique Cys residue at the C-terminus of CNFx was also discovered to be critical for retarding cargo delivery.IMPORTANCECytotoxic necrotizing factor (CNF) toxins not only play important virulence roles in pathogenic E. coli and other bacterial pathogens, but CNF-like genes have also been found in an expanding number of genomes from clinical isolates. Harnessing the power of evolutionary relationships among the CNF toxins enabled the deciphering of the hierarchical active-site determinants that define whether they modify their Rho GTPase substrates through deamidation or transglutamination. With our finding that a distant CNF variant (CNFx) unlike other known CNFs predominantly transglutaminates its Rho GTPase substrates, the paradigm of \"CNFs deamidate and DNTs transglutaminate\" could finally be attributed to two critical amino acid residues within the active site other than the previously identified catalytic Cys-His dyad residues. The significance of our approach and research findings is that they can be applied to deciphering enzyme reaction determinants and substrate specificities for other bacterial proteins in the development of precision therapeutic strategies.
    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
    G蛋白偶联受体(GPCRs)是有效的鸟嘌呤核苷酸交换因子(GEF),可响应各种细胞外刺激,在G蛋白异源三聚体的Gα亚基上将GDP交换为GTP,包括神经递质和光。GPCRs主要通过激活的G蛋白传播信号,GαGTP,和游离Gβγ是主要的疾病驱动因素。证据表明,细胞所需的环境低阈值信号传导可能由诸如非GPCRGEF和鸟嘌呤核苷酸解离抑制剂(GDI)的信号传导调节剂补充。G蛋白信号激活因子3(AGS3)被认为是参与多种健康和疾病相关过程的GDI。然而,对AGS3的理解有限,并且在活细胞中没有关于其结构-功能关系或信号调节的重要信息。这里,我们采用了一种新型光遗传学GDI的硅结构引导工程,基于AGS3的G蛋白调节(GPR)基序,了解其GDI活性,并在光命令下诱导活细胞中独立的Gβγ信号传导。我们的结果表明,OptoGDI的质膜募集有效地释放Gβγ,其亚细胞靶向作用产生局部PIP3并触发巨噬细胞迁移。因此,我们提出OptoGDI作为光学解剖GDI介导的信号通路和在细胞和体内触发GPCR非依赖性Gβγ信号的强大工具。
    G protein-coupled receptors (GPCRs) are efficient Guanine nucleotide exchange factors (GEFs) and exchange GDP to GTP on the Gα subunit of G protein heterotrimers in response to various extracellular stimuli, including neurotransmitters and light. GPCRs primarily broadcast signals through activated G proteins, GαGTP, and free Gβγ, and are major disease drivers. Evidence shows that the ambient low threshold signaling required for cells is likely supplemented by signaling regulators such as non-GPCR GEFs and Guanine nucleotide Dissociation Inhibitors (GDIs). Activators of G protein Signaling 3 (AGS3) are recognized as a GDI involved in multiple health and disease-related processes. Nevertheless, understanding of AGS3 is limited, and no significant information is available on its structure-function relationship or signaling regulation in living cells. Here, we employed in silico structure-guided engineering of a novel optogenetic GDI, based on the AGS3\'s G protein regulatory (GPR) motif, to understand its GDI activity and induce standalone Gβγ signaling in living cells on optical command. Our results demonstrate that plasma membrane recruitment of OptoGDI efficiently releases Gβγ, and its subcellular targeting generated localized PIP3 and triggered macrophage migration. Therefore, we propose OptoGDI as a powerful tool for optically dissecting GDI-mediated signaling pathways and triggering GPCR-independent Gβγ signaling in cells and in vivo.
    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
    在小儿脑病中突变的GNAO1编码主要的神经元G蛋白Gαo。在>80个致病突变中,大多数是跨Gαo序列的单个氨基酸取代。我们对Gαo突变体进行了广泛的表征,显示出异常的GTP吸收和水解,以及结合Gβγ和RGS19的缺陷。对于导致癫痫的突变子集,Gαo的质膜定位降低;对于更严重的突变体,与GPCRs的显性相互作用也会出现。致病性突变体大量获得与Ric8A的相互作用,令人惊讶的是,Ric8B蛋白,将它们从细胞质离域到高尔基。在这两个强制性Gα亚基伴侣中,Ric8A通常负责Gαi/o,Gαq,和Gα12/13亚家族,和Ric8B仅用于Gαs/olf。Ric8A/B通过使神经元G蛋白信号网络失衡而参与与致病性Gαo的新形态相互作用时,介导了疾病的优势。由于Gαo-Ric8B相互作用的强度与疾病严重程度相关,我们的研究进一步确定了GNAO1脑病临床表现的有效生物标志物和预测因子.我们的工作发现了小儿脑病潜在突变的新形态分子机制,并为G蛋白功能失调和其他遗传疾病引起的其他疾病提供了见解。
    GNAO1 mutated in pediatric encephalopathies encodes the major neuronal G protein Gαo. Of the more than 80 pathogenic mutations, most are single amino acid substitutions spreading across the Gαo sequence. We performed extensive characterization of Gαo mutants, showing abnormal GTP uptake and hydrolysis and deficiencies in binding Gβγ and RGS19. Plasma membrane localization of Gαo was decreased for a subset of mutations that leads to epilepsy; dominant interactions with GPCRs also emerged for the more severe mutants. Pathogenic mutants massively gained interaction with Ric8A and, surprisingly, Ric8B proteins, relocalizing them from cytoplasm to Golgi. Of these 2 mandatory Gα-subunit chaperones, Ric8A is normally responsible for the Gαi/Gαo, Gαq, and Gα12/Gα13 subfamilies, and Ric8B solely responsible for Gαs/Gαolf. Ric8 mediates the disease dominance when engaging in neomorphic interactions with pathogenic Gαo through imbalance of the neuronal G protein signaling networks. As the strength of Gαo-Ric8B interactions correlates with disease severity, our study further identifies an efficient biomarker and predictor for clinical manifestations in GNAO1 encephalopathies. Our work uncovers the neomorphic molecular mechanism of mutations underlying pediatric encephalopathies and offers insights into other maladies caused by G protein malfunctioning and further genetic diseases.
    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
    GNAO1基因突变,编码丰富的大脑G蛋白Gαo,导致以发育迟缓为特征的神经系统疾病,癫痫,和运动异常。有超过50个突变等位基因与GNAO1疾病相关;R209H突变导致肌张力障碍,卵巢狭窄症,和没有癫痫发作的发育迟缓。人类突变等位基因(Gnao1/R209H)的杂合小鼠在野外测试中表现出过度活跃,但没有癫痫发作。我们开发了表达人类GNAO1Gαo亚型1的两个剪接变体的自我互补腺相关病毒载体(scAAV9)(GoA,GNAO1.1)和2(GoB,GNAO1.2).双侧纹状体内注射scAAV9-GNAO1.1或scAAV9-GNAO1.2可显着逆转开放场测试中与突变相关的多动症。GNAO1过表达并没有增加癫痫的易感性,GNAO1载体治疗的潜在副作用。这代表了在体内应用GNAO1脑病的成功临床前基因治疗的第一份报告。需要进一步的研究来揭示scAAV9介导的Gαo表达后导致行为改善的分子机制,并完善载体设计。GNAO1突变引起一系列发育,癫痫,和运动障碍。这里,我们显示表达野生型Gαo蛋白的scAAV9-GNAO1的纹状体内递送降低了Gnao1+/R209H小鼠模型的过度活性,携带最常见的运动障碍相关突变之一。这是针对GNAO1脑病的基因治疗在体内应用于患者等位基因模型的首次报道。
    Mutations in the GNAO1 gene, which encodes the abundant brain G-protein Gα o, result in neurologic disorders characterized by developmental delay, epilepsy, and movement abnormalities. There are over 50 mutant alleles associated with GNAO1 disorders; the R209H mutation results in dystonia, choreoathetosis, and developmental delay without seizures. Mice heterozygous for the human mutant allele (Gnao1 +/R209H) exhibit hyperactivity in open field tests but no seizures. We developed self-complementary adeno-associated virus serotype 9 (scAAV9) vectors expressing two splice variants of human GNAO1 Gα o isoforms 1 (GoA, GNAO1.1) and 2 (GoB, GNAO1.2). Bilateral intrastriatal injections of either scAAV9-GNAO1.1 or scAAV9-GNAO1.2 significantly reversed mutation-associated hyperactivity in open field tests. GNAO1 overexpression did not increase seizure susceptibility, a potential side effect of GNAO1 vector treatment. This represents the first report of successful preclinical gene therapy for GNAO1 encephalopathy applied in vivo. Further studies are needed to uncover the molecular mechanism that results in behavior improvements after scAAV9-mediated Gα o expression and to refine the vector design. SIGNIFICANCE STATEMENT: GNAO1 mutations cause a spectrum of developmental, epilepsy, and movement disorders. Here we show that intrastriatal delivery of scAAV9-GNAO1 to express the wild-type Gα o protein reduces the hyperactivity of the Gnao1 +/R209H mouse model, which carries one of the most common movement disorder-associated mutations. This is the first report of a gene therapy for GNAO1 encephalopathy applied in vivo on a patient-allele model.
    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
    人和非人灵长类动物的背纹状体由尾状核和壳核组成。这两个区域接收不同的皮质投影并且在功能上是不同的。尾状参与目标导向行为的控制,而壳核与习惯的学习和形成有关。以前的报道表明,乙醇对这两个区域的神经传递有不同的影响。因为神经递质主要通过G蛋白偶联受体(GPCRs)发出信号来调节神经元活动,本研究旨在确定乙醇是否对参与GPCRs运输和功能的蛋白质的表达具有区域依赖性影响,包括G蛋白亚基及其效应子,蛋白激酶,和细胞骨架的元素。进行蛋白质印迹以检查在自由进入条件下自我施用乙醇1年的雄性食蟹猴的尾状核和壳核中的蛋白质水平,以及在相同操作条件下自行施用等热量甜味溶液的对照动物。在研究的18种蛋白质中,我们发现一种蛋白质(PKCβ)的水平升高,和13种蛋白质(Gαi1/3,Gαi2,Gαo,Gβ1γ,PKCα,PKCε,CaMKII,GSK3β,β-肌动蛋白,cofilin,α-微管蛋白,和微管蛋白聚合促进蛋白)在饮酒猕猴的尾状中减少。然而,乙醇不会改变壳核中任何蛋白质的表达。这些观察结果强调了尾状核对慢性乙醇暴露引起的蛋白质表达变化的独特脆弱性。这些改变是否与乙醇诱导的GPCR功能和神经传递失调有关,值得进一步研究。
    The dorsal striatum is composed of the caudate nucleus and the putamen in human and non-human primates. These two regions receive different cortical projections and are functionally distinct. The caudate is involved in the control of goal-directed behaviors, while the putamen is implicated in habit learning and formation. Previous reports indicate that ethanol differentially influences neurotransmission in these two regions. Because neurotransmitters primarily signal through G protein-coupled receptors (GPCRs) to modulate neuronal activity, the present study aimed to determine whether ethanol had a region-dependent impact on the expression of proteins that are involved in the trafficking and function of GPCRs, including G protein subunits and their effectors, protein kinases, and elements of the cytoskeleton. Western blotting was performed to examine protein levels in the caudate and the putamen of male cynomolgus macaques that self-administered ethanol for 1 year under free access conditions, along with control animals that self-administered an isocaloric sweetened solution under identical operant conditions. Among the 18 proteins studied, we found that the levels of one protein (PKCβ) were increased, and 13 proteins (Gαi1/3, Gαi2, Gαo, Gβ1γ, PKCα, PKCε, CaMKII, GSK3β, β-actin, cofilin, α-tubulin, and tubulin polymerization promoting protein) were reduced in the caudate of alcohol-drinking macaques. However, ethanol did not alter the expression of any proteins examined in the putamen. These observations underscore the unique vulnerability of the caudate nucleus to changes in protein expression induced by chronic ethanol exposure. Whether these alterations are associated with ethanol-induced dysregulation of GPCR function and neurotransmission warrants future investigation.
    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