cryoelectron microscopy

冷冻电子显微镜
  • 文章类型: Journal Article
    Kisspeptin受体(KISS1R),属于A类肽-GPCR家族,在kisspeptin刺激后对生殖生理的调节中起关键作用,被认为是生殖疾病的有吸引力的药物靶标。这里,我们证明,除了众所周知的Gq/11途径外,KISS1R还可以与Gi/o途径偶联。我们进一步解析了与合成激动剂TAK448结合的KISS1R-Gq和KISS1R-Gi复合物的低温电子显微镜(cryo-EM)结构以及与内源性激动剂KP54结合的KISS1R-Gq复合物的结构。高分辨率结构为其配体识别KISS1R的机制提供了清晰的见解,并且可以促进具有高亲和力的靶向药物的设计以提高治疗效果。此外,结构和功能分析表明胞外环(ECLs)的构象差异,受体的胞内环(ICL),Gα亚基的“波浪形钩子”可能解释了G蛋白偶联对KISS1R信号传导的特异性。
    Kisspeptin receptor (KISS1R), belonging to the class A peptide-GPCR family, plays a key role in the regulation of reproductive physiology after stimulation by kisspeptin and is regarded as an attractive drug target for reproductive diseases. Here, we demonstrated that KISS1R can couple to the Gi/o pathway besides the well-known Gq/11 pathway. We further resolved the cryo-electron microscopy (cryo-EM) structure of KISS1R-Gq and KISS1R-Gi complexes bound to the synthetic agonist TAK448 and structure of KISS1R-Gq complex bound to the endogenous agonist KP54. The high-resolution structures provided clear insights into mechanism of KISS1R recognition by its ligand and can facilitate the design of targeted drugs with high affinity to improve treatment effects. Moreover, the structural and functional analyses indicated that conformational differences in the extracellular loops (ECLs), intracellular loops (ICLs) of the receptor, and the \"wavy hook\" of the Gα subunit may account for the specificity of G protein coupling for KISS1R signaling.
    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的北极(E22G)突变(Aβ增强了阿尔茨海默病(AD)中Aβ40原纤维的积累。与零星的AD不同,具有突变的家族性AD(FAD)患者在斑块核心中表现出更多的Aβ40。然而,E22GAβ40原纤维的结构细节仍然难以捉摸,阻碍治疗进展。这里,我们通过冷冻电子显微镜(cryoEM)和固态核磁共振(SSNMR)对体外制备的E22GAβ40原纤维进行共分析,确定了独特的W形平行β-折叠结构。E22GAβ40原纤维在FAD的棉绒斑块中表现出典型的淀粉样蛋白特征,如低硫黄素-T荧光和不太紧凑的未捆绑形态。此外,动力学和MD研究揭示了先前未确定的体外证据,即E22GAβ40而不是Aβ42可能引发FAD中的Aβ错误折叠,并通过交叉接种促进野生型(WT)Aβ40/Aβ42的后续错误折叠。结果提供了对北极突变如何通过Aβ40积累和交叉传播促进AD的见解。
    Arctic (E22G) mutation in amyloid-β (Aβ enhances Aβ40 fibril accumulation in Alzheimer\'s disease (AD). Unlike sporadic AD, familial AD (FAD) patients with the mutation exhibit more Aβ40 in the plaque core. However, structural details of E22G Aβ40 fibrils remain elusive, hindering therapeutic progress. Here, we determine a distinctive W-shaped parallel β-sheet structure through co-analysis by cryo-electron microscopy (cryoEM) and solid-state nuclear magnetic resonance (SSNMR) of in-vitro-prepared E22G Aβ40 fibrils. The E22G Aβ40 fibrils displays typical amyloid features in cotton-wool plaques in the FAD, such as low thioflavin-T fluorescence and a less compact unbundled morphology. Furthermore, kinetic and MD studies reveal previously unidentified in-vitro evidence that E22G Aβ40, rather than Aβ42, may trigger Aβ misfolding in the FAD, and prompt subsequent misfolding of wild-type (WT) Aβ40/Aβ42 via cross-seeding. The results provide insight into how the Arctic mutation promotes AD via Aβ40 accumulation and cross-propagation.
    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
    严重急性呼吸综合征冠状病毒2(SARS-CoV-2)刺突蛋白结合受体血管紧张素转换酶2(ACE2),并通过其S2结构域的重折叠驱动病毒-宿主膜融合。而S1结构域包含高序列变异性,S2结构域是保守的,是一个有前景的泛β-冠状病毒疫苗靶标.我们应用低温电子断层摄影术来捕获S2重折叠的中间体,并了解抗体对S2茎螺旋的抑制作用。亚放射图平均显示ACE2二聚体在过渡到S2中间体之前交联尖峰,在重新折叠的各个阶段被捕获。靶向S2茎螺旋的泛β冠状病毒中和抗体结合并抑制穗前发夹中间体的重折叠。结合分子动力学模拟,这些结构阐明了SARS-CoV-2进入的过程,并揭示了泛β冠状病毒S2靶向抗体如何通过阻止发夹前中间体来中和感染性。
    The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein binds the receptor angiotensin converting enzyme 2 (ACE2) and drives virus-host membrane fusion through refolding of its S2 domain. Whereas the S1 domain contains high sequence variability, the S2 domain is conserved and is a promising pan-betacoronavirus vaccine target. We applied cryo-electron tomography to capture intermediates of S2 refolding and understand inhibition by antibodies to the S2 stem-helix. Subtomogram averaging revealed ACE2 dimers cross-linking spikes before transitioning into S2 intermediates, which were captured at various stages of refolding. Pan-betacoronavirus neutralizing antibodies targeting the S2 stem-helix bound to and inhibited refolding of spike prehairpin intermediates. Combined with molecular dynamics simulations, these structures elucidate the process of SARS-CoV-2 entry and reveal how pan-betacoronavirus S2-targeting antibodies neutralize infectivity by arresting prehairpin intermediates.
    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
    DNA聚合酶theta(Polθ)是一种DNA解旋酶-聚合酶蛋白,可促进DNA修复,并且对同源定向修复(HDR)因子具有合成致死性。因此,Pole是HDR缺陷癌症中一种有前途的精准肿瘤药物靶标。这里,我们使用cryo-EM表征了Polθ解旋酶(Polθ-hel)小分子抑制剂(AB25583)的结合和作用机制。AB25583对Pole-hel,选择性杀死BRCA1/2缺陷细胞,并在携带致病性BRCA1/2突变的癌细胞中与奥拉帕尼协同作用。Cryo-EM在3.0-3.2µ上主要发现二聚体Polθ-hel:AB25583复杂结构。这些结构揭示了解旋酶中央通道深处的结合袋,这强调了AB25583的高特异性和效力。低温EM结构与生化数据的结合表明,AB25583通过变构机制抑制了Polθ-hel解旋酶的ATPase活性。这些关于AB25583抑制的详细结构数据和见解为加速HDR缺陷型癌症中靶向Pole-hel的药物开发铺平了道路。
    DNA polymerase theta (Polθ) is a DNA helicase-polymerase protein that facilitates DNA repair and is synthetic lethal with homology-directed repair (HDR) factors. Thus, Polθ is a promising precision oncology drug-target in HDR-deficient cancers. Here, we characterize the binding and mechanism of action of a Polθ helicase (Polθ-hel) small-molecule inhibitor (AB25583) using cryo-EM. AB25583 exhibits 6 nM IC50 against Polθ-hel, selectively kills BRCA1/2-deficient cells, and acts synergistically with olaparib in cancer cells harboring pathogenic BRCA1/2 mutations. Cryo-EM uncovers predominantly dimeric Polθ-hel:AB25583 complex structures at 3.0-3.2 Å. The structures reveal a binding-pocket deep inside the helicase central-channel, which underscores the high specificity and potency of AB25583. The cryo-EM structures in conjunction with biochemical data indicate that AB25583 inhibits the ATPase activity of Polθ-hel helicase via an allosteric mechanism. These detailed structural data and insights about AB25583 inhibition pave the way for accelerating drug development targeting Polθ-hel in HDR-deficient cancers.
    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
    胆碱-甘氨酸甜菜碱途径在高渗环境中的细菌生存中起重要作用。胆碱转运蛋白BetT的渗透激活促进了外部胆碱的摄取,以合成渗透保护性甘氨酸甜菜碱。这里,我们报道了apo和胆碱结合状态的丁香假单胞菌BetT的低温电子显微镜结构。我们的结构表明,BetT与一个原聚体的C端结构域(CTD)形成一个结构域交换的三聚体,该结构域与相邻原聚体的跨膜结构域(TMD)相互作用。底物胆碱结合在TMD中心部分的色氨酸棱镜内。加上功能表征,我们的结果表明,在假单胞菌中,包括植物病原体丁香假单胞菌和人类病原体铜绿假单胞菌,在没有渗透胁迫的情况下,BetT通过CTD介导的自动抑制被锁定在低活性状态,它的高渗激活涉及这种自身抑制作用的释放。
    The choline-glycine betaine pathway plays an important role in bacterial survival in hyperosmotic environments. Osmotic activation of the choline transporter BetT promotes the uptake of external choline for synthesizing the osmoprotective glycine betaine. Here, we report the cryo-electron microscopy structures of Pseudomonas syringae BetT in the apo and choline-bound states. Our structure shows that BetT forms a domain-swapped trimer with the C-terminal domain (CTD) of one protomer interacting with the transmembrane domain (TMD) of a neighboring protomer. The substrate choline is bound within a tryptophan prism at the central part of TMD. Together with functional characterization, our results suggest that in Pseudomonas species, including the plant pathogen P. syringae and the human pathogen Pseudomonas aeruginosa, BetT is locked at a low-activity state through CTD-mediated autoinhibition in the absence of osmotic stress, and its hyperosmotic activation involves the release of this autoinhibition.
    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
    破译大分子复合物中的蛋白质-配体相互作用对于理解分子机制至关重要,潜在的生物过程,和药物开发。近年来,低温样品电子显微镜(cryoEM)已成为一种强大的技术,可以确定大分子的结构并研究以近原子分辨率结合配体的方式。由于跨感兴趣分子的各向异性分辨率和数据中的固有噪声,在冷冻EM图中识别和建模非蛋白质分子通常是具有挑战性的。在这篇文章中,向读者介绍了目前用于配体鉴定的各种软件和方法,模型建筑,和使用选定的大分子细化原子坐标。识别配体存在的最简单方法之一,正如烯醇酶所说明的,是减去有和没有配体获得的两个图谱。即使在较高的阈值下,配体的额外密度也可能在差异图中突出。有实例,如在代谢型谷氨酸受体mGlu5的情况下所示,当不能生成这样的简单差异图。最近引入的衍生Fo-Fc省略图的方法可以用作验证和证明配体存在的工具。最后,以研究良好的β-半乳糖苷酶为例,分析了分辨率对冷冻EM图中配体和溶剂分子建模的影响,并对冷冻EM如何用于药物发现进行了展望。
    Deciphering the protein-ligand interactions in a macromolecular complex is crucial for understanding the molecular mechanism, underlying biological processes, and drug development. In recent years, cryogenic sample electron microscopy (cryoEM) has emerged as a powerful technique to determine the structures of macromolecules and to investigate the mode of ligand binding at near-atomic resolution. Identifying and modeling non-protein molecules in cryoEM maps is often challenging due to anisotropic resolution across the molecule of interest and inherent noise in the data. In this article, the readers are introduced to various software and methods currently used for ligand identification, model building, and refinement of atomic coordinates using selected macromolecules. One of the simplest ways to identify the presence of a ligand, as illustrated with the enolase enzyme, is to subtract the two maps obtained with and without the ligand. The extra density of the ligand is likely to stand out in the difference map even at a higher threshold. There are instances, as shown in the case of metabotropic Glutamate receptor mGlu5, when such simple difference maps cannot be generated. The recently introduced method of deriving the Fo-Fc omit map can serve as a tool for validating and demonstrating the presence of the ligand. Finally, using the well-studied β-galactosidase as an example, the effect of resolution on modeling the ligands and solvent molecules in cryoEM maps is analyzed, and an outlook on how cryoEM can be used in drug discovery is presented.
    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
    微生物离子抽运视紫红质(MR)是广泛研究的视网膜结合膜蛋白。然而,它们的生物发生,包括寡聚化和视网膜掺入,仍然知之甚少。细菌吸收绿光的质子泵蛋白视紫红质(GPR)已成为MR的模型蛋白,并在此使用低温电子显微镜(cryo-EM)和分子动力学(MD)模拟来解决这些悬而未决的问题。具体来说,关于GPR化学计量的相互矛盾的研究报告了五聚体和六聚体混合物,但没有提供可能的组装机制。我们报告了GPR突变体的五聚体和六聚体冷冻-EM结构,揭示未加工的N端信号肽在六聚体GPR组装中的作用。此外,某些表达蛋白视紫红质的细菌缺乏视网膜生物合成途径,表明他们从环境中清除了辅因子。我们通过解决无视网膜蛋白聚糖的低温EM结构来阐明这一假设,与质谱和MD模拟一起表明癸酸酯在发色团结合袋中充当视网膜的临时占位符。进一步的MD模拟阐明了癸酸酯和视黄醛交换的可能途径,提供清除视网膜的机制.总的来说,我们的发现提供了对MR的生物发生的见解,包括它们的寡聚组装,通过潜在的辅因子清除机制,在原药化学计量和视网膜掺入中的变化。
    Microbial ion-pumping rhodopsins (MRs) are extensively studied retinal-binding membrane proteins. However, their biogenesis, including oligomerisation and retinal incorporation, remains poorly understood. The bacterial green-light absorbing proton pump proteorhodopsin (GPR) has emerged as a model protein for MRs and is used here to address these open questions using cryo-electron microscopy (cryo-EM) and molecular dynamics (MD) simulations. Specifically, conflicting studies regarding GPR stoichiometry reported pentamer and hexamer mixtures without providing possible assembly mechanisms. We report the pentameric and hexameric cryo-EM structures of a GPR mutant, uncovering the role of the unprocessed N-terminal signal peptide in the assembly of hexameric GPR. Furthermore, certain proteorhodopsin-expressing bacteria lack retinal biosynthesis pathways, suggesting that they scavenge the cofactor from their environment. We shed light on this hypothesis by solving the cryo-EM structure of retinal-free proteoopsin, which together with mass spectrometry and MD simulations suggests that decanoate serves as a temporary placeholder for retinal in the chromophore binding pocket. Further MD simulations elucidate possible pathways for the exchange of decanoate and retinal, offering a mechanism for retinal scavenging. Collectively, our findings provide insights into the biogenesis of MRs, including their oligomeric assembly, variations in protomer stoichiometry and retinal incorporation through a potential cofactor scavenging mechanism.
    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
    转谷氨酰胺酶2(TG2)是一种GTP结合酶,蛋白质交联酶已被研究为乳糜泻的治疗靶标,神经系统疾病,和侵袭性癌症。TG2已被建议采用调节其功能的两种构象状态:GTP结合,闭合构象,和钙结合,交联活性开放构象。组成型采用开放构象的TG2突变体对癌细胞具有细胞毒性。因此,结合和稳定TG2开放构象的小分子可以提供一种新的治疗策略。这里,我们研究了TG2,使用静态和时间分辨小角度X射线散射(SAXS)和单粒子冷冻电子显微镜(cryo-EM),以确定负责赋予其生物学效应的构象状态。我们还描述了一种新开发的TG2抑制剂,LM11可有效杀死胶质母细胞瘤细胞,并使用SAXS研究LM11如何影响TG2的构象状态。使用SAXS和低温EM,我们显示鸟嘌呤核苷酸结合并稳定单体闭合构象,而钙结合到可以形成更高阶寡聚体的开放状态。SAXS分析提示组成型采用开放状态的TG2突变体如何通过与野生型TG2的替代机制结合核苷酸。此外,我们使用时间分辨SAXS来显示LM11增加钙结合和稳定开放构象的能力,鸟嘌呤核苷酸是不可逆的,对癌细胞有细胞毒性。一起来看,我们的发现表明,TG2的构象动力学比以前提出的更复杂,并强调了LM11对TG2的构象稳定如何维持TG2处于细胞毒性构象状态。
    Transglutaminase 2 (TG2) is a GTP-binding, protein-crosslinking enzyme that has been investigated as a therapeutic target for Celiac disease, neurological disorders, and aggressive cancers. TG2 has been suggested to adopt two conformational states that regulate its functions: a GTP-bound, closed conformation, and a calcium-bound, crosslinking-active open conformation. TG2 mutants that constitutively adopt an open conformation are cytotoxic to cancer cells. Thus, small molecules that bind and stabilize the open conformation of TG2 could offer a new therapeutic strategy. Here, we investigate TG2, using static and time-resolved small-angle X-ray scattering (SAXS) and single-particle cryoelectron microscopy (cryo-EM), to determine the conformational states responsible for conferring its biological effects. We also describe a newly developed TG2 inhibitor, LM11, that potently kills glioblastoma cells and use SAXS to investigate how LM11 affects the conformational states of TG2. Using SAXS and cryo-EM, we show that guanine nucleotides bind and stabilize a monomeric closed conformation while calcium binds to an open state that can form higher order oligomers. SAXS analysis suggests how a TG2 mutant that constitutively adopts the open state binds nucleotides through an alternative mechanism to wildtype TG2. Furthermore, we use time resolved SAXS to show that LM11 increases the ability of calcium to bind and stabilize an open conformation, which is not reversible by guanine nucleotides and is cytotoxic to cancer cells. Taken together, our findings demonstrate that the conformational dynamics of TG2 are more complex than previously suggested and highlight how conformational stabilization of TG2 by LM11 maintains TG2 in a cytotoxic conformational state.
    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
    尼帕病毒感染,世界卫生组织承认的最优先疾病之一,强调迫切需要针对潜在的流行病和大流行病制定有效的对策。这里,我们确定了一种完全人单结构域抗体,该抗体靶向位于尼帕病毒G蛋白(受体结合蛋白,RBP),通过高分辨率低温电子显微镜(cryo-EM)的结构阐明。这种独特的结合模式破坏了G蛋白的四聚化,因此阻碍了F蛋白的激活并抑制了病毒膜融合。此外,我们的研究表明,这种紧凑型抗体在血脑屏障(BBB)中显示出增强的通透性,并在尼帕病毒感染的鼠模型中在消除脑内假病毒方面表现出卓越的功效,特别是与IgG1形式的充分表征的抗体m102.4相比。因此,这种单结构域抗体有望作为预防尼帕病毒感染的治疗候选药物,并对疫苗开发具有潜在意义.
    Nipah virus infection, one of the top priority diseases recognized by the World Health Organization, underscores the urgent need to develop effective countermeasures against potential epidemics and pandemics. Here, we identify a fully human single-domain antibody that targets a highly conserved cryptic epitope situated at the dimeric interface of the Nipah virus G protein (receptor binding protein, RBP), as elucidated through structures by high-resolution cryo-electron microscopy (cryo-EM). This unique binding mode disrupts the tetramerization of the G protein, consequently obstructing the activation of the F protein and inhibiting viral membrane fusion. Furthermore, our investigations reveal that this compact antibody displays enhanced permeability across the blood-brain barrier (BBB) and demonstrates superior efficacy in eliminating pseudovirus within the brain in a murine model of Nipah virus infection, particularly compared to the well-characterized antibody m102.4 in an IgG1 format. Consequently, this single-domain antibody holds promise as a therapeutic candidate to prevent Nipah virus infections and has potential implications for vaccine development.
    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
    ABCA4是一种ATP结合盒(ABC)转运蛋白,可通过促进N-视黄亚烷基磷脂酰乙醇胺跨视杆和视锥感光细胞膜的转运来防止有毒的类视黄醇化合物的积聚。ABCA4中超过1500个错义突变,许多在核苷酸结合域(NBD)中,与Stargardt病(STGD1)有遗传联系。这里,我们通过低温电子显微镜显示,ABCA4在AMP-PNP结合后从开放的向外构象转化为封闭构象。结构信息和生化研究用于进一步定义NBD在ABCA4功能特性中的作用以及突变导致活性丧失的机制。我们表明,ABCA4的功能活性需要两个NBD中的ATPase活性。WalkerA天冬酰胺残基的突变导致底物激活的ATPase活性严重降低,这是由于与相对NBD的D环内残基的极性相互作用丧失。包括R1108C在内的其他NBD残基中的疾病突变的结构基础,R2077W,R2107H和L2027F对ABCA4的构造和功效影响停止了描写。总的来说,我们的研究为ABCA4的结构和功能以及STGD1的潜在机制提供了见解.
    ABCA4 is an ATP-binding cassette (ABC) transporter that prevents the buildup of toxic retinoid compounds by facilitating the transport of N-retinylidene-phosphatidylethanolamine across membranes of rod and cone photoreceptor cells. Over 1500 missense mutations in ABCA4, many in the nucleotide binding domains (NBDs), have been genetically linked to Stargardt disease (STGD1). Here, we show by Cryo-electron microscopy that ABCA4 is converted from an open outward conformation to a closed conformation upon the binding of AMP-PNP. Structural information and biochemical studies were used to further define the role of the NBDs in the functional properties of ABCA4 and the mechanisms by which mutations lead to the loss in activity. We show that ATPase activity in both NBDs is required for the functional activity of ABCA4. Mutations in Walker A asparagine residues cause a severe reduction in substrate-activated ATPase activity due to the loss in polar interactions with residues within the D-loops of the opposing NBD. The structural basis for how disease mutations in other NBD residues including the R1108C, R2077W, R2107H and L2027F affect the structure and function of ABCA4 is described. Collectively, our studies provide insight into the structure and function of ABCA4 and mechanisms underlying STGD1.
    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