cryoelectron microscopy

冷冻电子显微镜
  • 文章类型: Journal Article
    5'-3'外核糖核酸酶Rat1/Xrn2负责终止RNAPII的真核mRNA转录。Rat1与其伴侣蛋白形成复合物,Rai1和Rtt103,并充当“鱼雷”以结合转录RNAPII并从中解离DNA/RNA。在这里,我们报告了Rat1-Rai1-Rtt103复合物和三个Rat1-Rai1相关的RNAPII复合物(1型,1b型,和2型)来自酵母,Komagataellaphaffii.Rat1-Rai1-Rtt103结构表明Rat1和Rai1形成异四聚体,单个Rtt103结合在两个Rai1分子之间。在1型复合体中,Rat1-Rai1形成异二聚体并与RNAPII的RNA退出位点结合以将RNA提取到Rat1外切核酸酶活性位点中。这种相互作用改变了RNA路径,有利于终止(“预终止”状态)。1b型和2型复合物没有结合的DNA/RNA,可能代表“终止后”状态。这些结构说明了真核mRNA转录的终止机制。
    The 5´-3´ exoribonuclease Rat1/Xrn2 is responsible for the termination of eukaryotic mRNA transcription by RNAPII. Rat1 forms a complex with its partner proteins, Rai1 and Rtt103, and acts as a \"torpedo\" to bind transcribing RNAPII and dissociate DNA/RNA from it. Here we report the cryo-electron microscopy structures of the Rat1-Rai1-Rtt103 complex and three Rat1-Rai1-associated RNAPII complexes (type-1, type-1b, and type-2) from the yeast, Komagataella phaffii. The Rat1-Rai1-Rtt103 structure revealed that Rat1 and Rai1 form a heterotetramer with a single Rtt103 bound between two Rai1 molecules. In the type-1 complex, Rat1-Rai1 forms a heterodimer and binds to the RNA exit site of RNAPII to extract RNA into the Rat1 exonuclease active site. This interaction changes the RNA path in favor of termination (the \"pre-termination\" state). The type-1b and type-2 complexes have no bound DNA/RNA, likely representing the \"post-termination\" states. These structures illustrate the termination mechanism of eukaryotic mRNA transcription.
    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
    胶原蛋白的翻译后加工对于其正确的组装和功能至关重要。胶原蛋白加工的破坏导致组织发育和结构紊乱,例如成骨不全症(OI)。与OI相关的胶原蛋白加工机器包括3-羟化酶1(P3H1),肽基氨酰顺反异构酶B(PPIB),和软骨相关蛋白(CRTAP),其结构组织和机制不明确。我们确定了P3H1/CRTAP/PPIB复合物的低温-EM结构。P3H1和PPIB的活性位点形成面对面的双功能反应中心,指示耦合的修改机制。P3H1/CRTAP/PPIB/胶原肽复合物的结构揭示了多个结合位点,暗示了一个底物相互作用区。出乎意料的是,观察到二元三元络合物,三元和二元三元状态之间的平衡可以通过P3H1/PPIB活性位点的突变和PPIB抑制剂的添加而改变。这些发现为P3H1/CRTAP/PPIB加工胶原蛋白的结构基础和胶原蛋白相关疾病的分子病理学提供了见解。
    Collagen posttranslational processing is crucial for its proper assembly and function. Disruption of collagen processing leads to tissue development and structure disorders like osteogenesis imperfecta (OI). OI-related collagen processing machinery includes prolyl 3-hydroxylase 1 (P3H1), peptidyl-prolyl cis-trans isomerase B (PPIB), and cartilage-associated protein (CRTAP), with their structural organization and mechanism unclear. We determine cryo-EM structures of the P3H1/CRTAP/PPIB complex. The active sites of P3H1 and PPIB form a face-to-face bifunctional reaction center, indicating a coupled modification mechanism. The structure of the P3H1/CRTAP/PPIB/collagen peptide complex reveals multiple binding sites, suggesting a substrate interacting zone. Unexpectedly, a dual-ternary complex is observed, and the balance between ternary and dual-ternary states can be altered by mutations in the P3H1/PPIB active site and the addition of PPIB inhibitors. These findings provide insights into the structural basis of collagen processing by P3H1/CRTAP/PPIB and the molecular pathology of collagen-related disorders.
    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
    在真核生物中,前导链DNA由Polε合成,滞后链由Polδ合成。当与DNA夹PCNA配对时,这些复制聚合酶具有更高的持续合成能力。虽然已经确定了酵母Polε催化结构域的结构,Polε如何与PCNA相互作用在任何真核生物中都是未知的,人类或酵母。在这里,我们报告了人类Polε-PCNA-DNA复合物的两种低温EM结构,一个处于进入的核苷酸结合状态,另一个处于核苷酸交换状态。结构揭示了Pole催化结构域与PCNA之间意想不到的三点界面,具有保守的PIP(PCNA相互作用肽)基序,唯一的P域,和拇指结构域各自与PCNA三聚体的不同原聚体相互作用。我们建议,当PCNA与Pole一起工作时,多点接口可防止其他包含PIP的因素招募到PCNA。两种状态的比较表明,指状结构域围绕P结构域的含[4Fe-4S]簇的尖端旋转,以调节核苷酸交换和传入的核苷酸结合。
    In eukaryotes, the leading strand DNA is synthesized by Polε and the lagging strand by Polδ. These replicative polymerases have higher processivity when paired with the DNA clamp PCNA. While the structure of the yeast Polε catalytic domain has been determined, how Polε interacts with PCNA is unknown in any eukaryote, human or yeast. Here we report two cryo-EM structures of human Polε-PCNA-DNA complex, one in an incoming nucleotide bound state and the other in a nucleotide exchange state. The structures reveal an unexpected three-point interface between the Polε catalytic domain and PCNA, with the conserved PIP (PCNA interacting peptide)-motif, the unique P-domain, and the thumb domain each interacting with a different protomer of the PCNA trimer. We propose that the multi-point interface prevents other PIP-containing factors from recruiting to PCNA while PCNA functions with Polε. Comparison of the two states reveals that the finger domain pivots around the [4Fe-4S] cluster-containing tip of the P-domain to regulate nucleotide exchange and incoming nucleotide binding.
    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
    在白色念珠菌中,Cdr1将唑类药物从细胞中泵出,以减少有害浓度的细胞内积累,导致唑类药物耐药性。米伯霉素肟,一种兽用抗寄生虫药,强烈且特异性地抑制Cdr1。然而,Cdr1如何识别和出口唑类药物,以及米尔贝霉素肟如何抑制Cdr1仍不清楚。这里,我们报道了Cdr1在不同状态下的三种低温EM结构:apo态(Cdr1Apo),氟康唑结合态(Cdr1Flu),和米尔贝霉素肟抑制状态(Cdr1Mil)。氟康唑底物和米尔贝霉素肟抑制剂都主要通过疏水相互作用在Cdr1的中心腔内被识别。建议氟康唑通过TM2,TM5,TM8和TM11驱动的横向途径从结合位点输出到环境中。我们的发现揭示了米尔贝霉素肟的抑制机制,通过竞争抑制了Cdr1,阻碍出口,并阻碍基材进入。这些发现促进了我们对白色念珠菌中Cdr1介导的唑类耐药性的理解,并为开发针对Cdr1的创新抗真菌药物以对抗唑类耐药性奠定了基础。
    In Candida albicans, Cdr1 pumps azole drugs out of the cells to reduce intracellular accumulation at detrimental concentrations, leading to azole-drug resistance. Milbemycin oxime, a veterinary anti-parasitic drug, strongly and specifically inhibits Cdr1. However, how Cdr1 recognizes and exports azole drugs, and how milbemycin oxime inhibits Cdr1 remain unclear. Here, we report three cryo-EM structures of Cdr1 in distinct states: the apo state (Cdr1Apo), fluconazole-bound state (Cdr1Flu), and milbemycin oxime-inhibited state (Cdr1Mil). Both the fluconazole substrate and the milbemycin oxime inhibitor are primarily recognized within the central cavity of Cdr1 through hydrophobic interactions. The fluconazole is suggested to be exported from the binding site into the environment through a lateral pathway driven by TM2, TM5, TM8 and TM11. Our findings uncover the inhibitory mechanism of milbemycin oxime, which inhibits Cdr1 through competition, hindering export, and obstructing substrate entry. These discoveries advance our understanding of Cdr1-mediated azole resistance in C. albicans and provide the foundation for the development of innovative antifungal drugs targeting Cdr1 to combat azole-drug resistance.
    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
    γ-氨基丁酸-A(GABAA)受体广泛分布于视网膜和脑中,是治疗视觉的潜在药物靶点,睡眠和认知障碍。内源性神经活性类固醇包括β-雌二醇和硫酸孕烯醇酮负调节ρ1GABAA受体的功能,但其抑制机制尚不清楚。通过将五种低温EM结构与电生理学和分子动力学模拟相结合,我们表征了β-雌二醇和硫酸孕烯醇酮在人ρ1GABAA受体上的结合位点和负调节机制。β-雌二醇结合在细胞外和跨膜结构域之间的界面的口袋中,显然特定于ρ亚族,并干扰将GABA结合与孔开放联系起来的变构构象转变。相比之下,硫酸孕烯醇酮在孔内结合以阻止离子渗透,优先考虑激活的结构。这些结果阐明了两种不同的神经活性类固醇抑制ρ1的对比机制,对亚型特异性门控和药理学设计有潜在影响。
    ρ-type γ-aminobutyric acid-A (GABAA) receptors are widely distributed in the retina and brain, and are potential drug targets for the treatment of visual, sleep and cognitive disorders. Endogenous neuroactive steroids including β-estradiol and pregnenolone sulfate negatively modulate the function of ρ1 GABAA receptors, but their inhibitory mechanisms are not clear. By combining five cryo-EM structures with electrophysiology and molecular dynamics simulations, we characterize binding sites and negative modulation mechanisms of β-estradiol and pregnenolone sulfate at the human ρ1 GABAA receptor. β-estradiol binds in a pocket at the interface between extracellular and transmembrane domains, apparently specific to the ρ subfamily, and disturbs allosteric conformational transitions linking GABA binding to pore opening. In contrast, pregnenolone sulfate binds inside the pore to block ion permeation, with a preference for activated structures. These results illuminate contrasting mechanisms of ρ1 inhibition by two different neuroactive steroids, with potential implications for subtype-specific gating and pharmacological design.
    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
    尽管胺能GPCRs是约25%的批准药物的靶标,由于其正构结合位点的高度序列保守性,开发亚型选择性药物是一个主要挑战。比特位配体共价连接正构和变构药效团,具有通过减少脱靶副作用来增强受体选择性和改善当前药物的潜力。然而,缺乏有关其绑定模式的结构信息阻碍了合理的设计。在这里,我们确定了与D3R选择性双位激动剂FOB02-04A结合的hD3R:GαOβγ复合物的低温-EM结构。结构,功能和计算分析提供了对其结合模式的见解,并指出了一个新的TM2-ECL1-TM1区域,这需要TM1的N端排序,作为胺能GPCRs亚型选择性的主要决定因素。该地区在药物开发方面未得到充分利用,扩展了胺能GPCRs中已建立的二级结合袋,并有可能用于设计新型和亚型选择性药物。
    Although aminergic GPCRs are the target for ~25% of approved drugs, developing subtype selective drugs is a major challenge due to the high sequence conservation at their orthosteric binding site. Bitopic ligands are covalently joined orthosteric and allosteric pharmacophores with the potential to boost receptor selectivity and improve current medications by reducing off-target side effects. However, the lack of structural information on their binding mode impedes rational design. Here we determine the cryo-EM structure of the hD3R:GαOβγ complex bound to the D3R selective bitopic agonist FOB02-04A. Structural, functional and computational analyses provide insights into its binding mode and point to a new TM2-ECL1-TM1 region, which requires the N-terminal ordering of TM1, as a major determinant of subtype selectivity in aminergic GPCRs. This region is underexploited in drug development, expands the established secondary binding pocket in aminergic GPCRs and could potentially be used to design novel and subtype selective drugs.
    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
    作为结核分枝杆菌(Mtb)中第一个确定的多药外排泵,EfpA是一种重要的蛋白质和有前途的药物靶标。然而,人们对EfpA的功能和抑制机制知之甚少。在这里,我们报告了EfpA在向外开放构象中的cryo-EM结构,与三种内源性脂质或抑制剂BRD-8000.3结合。EfpA内部的三种脂质从膜的内部小叶到外部小叶。BRD-8000.3占据内膜小叶水平的一个脂质位点,竞争性抑制脂质结合。EfpA在整体结构和脂质结合位点上类似于相关的溶血磷脂转运蛋白MFSD2A,并且可以充当脂质翻转酶。结合AlphaFold预测的EfpA结构,它是向内开放的,我们提出了一个完整的EfpA构象转变周期。一起,我们的研究结果为理解EfpA功能和开发EfpA靶向抗结核药物提供了结构和机制基础.
    As the first identified multidrug efflux pump in Mycobacterium tuberculosis (Mtb), EfpA is an essential protein and promising drug target. However, the functional and inhibitory mechanisms of EfpA are poorly understood. Here we report cryo-EM structures of EfpA in outward-open conformation, either bound to three endogenous lipids or the inhibitor BRD-8000.3. Three lipids inside EfpA span from the inner leaflet to the outer leaflet of the membrane. BRD-8000.3 occupies one lipid site at the level of inner membrane leaflet, competitively inhibiting lipid binding. EfpA resembles the related lysophospholipid transporter MFSD2A in both overall structure and lipid binding sites and may function as a lipid flippase. Combining AlphaFold-predicted EfpA structure, which is inward-open, we propose a complete conformational transition cycle for EfpA. Together, our results provide a structural and mechanistic foundation to comprehend EfpA function and develop EfpA-targeting anti-TB drugs.
    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
    低温电子显微镜(cryo-EM)样品制备过程中的空气-水界面(AWI)相互作用会导致明显的样品损失,阻碍结构生物学研究。线虫和缓步动物等生物体产生晚期胚胎发生丰富的(LEA)蛋白以承受干燥胁迫。这里我们展示了这些LEA蛋白,当在骤冷期间用作添加剂时,有效减轻AWI对脆性多亚基分子样品的损伤。所得的高分辨率低温EM图与使用现有的AWI损伤缓解方法获得的图相当或更好。低温电子层析成像显示粒子位于特定的界面,提示LEA蛋白在AWI处形成屏障。这种相互作用可以解释观察到的样品依赖性颗粒的优选取向。LEA蛋白提供了一个简单的,成本效益高,以及低温EM结构生物学家克服AWI相关样品损伤的适应性方法,可能振兴具有挑战性的项目和推进结构生物学领域。
    Air-water interface (AWI) interactions during cryo-electron microscopy (cryo-EM) sample preparation cause significant sample loss, hindering structural biology research. Organisms like nematodes and tardigrades produce Late Embryogenesis Abundant (LEA) proteins to withstand desiccation stress. Here we show that these LEA proteins, when used as additives during plunge freezing, effectively mitigate AWI damage to fragile multi-subunit molecular samples. The resulting high-resolution cryo-EM maps are comparable to or better than those obtained using existing AWI damage mitigation methods. Cryogenic electron tomography reveals that particles are localized at specific interfaces, suggesting LEA proteins form a barrier at the AWI. This interaction may explain the observed sample-dependent preferred orientation of particles. LEA proteins offer a simple, cost-effective, and adaptable approach for cryo-EM structural biologists to overcome AWI-related sample damage, potentially revitalizing challenging projects and advancing the field of structural biology.
    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
    SARS-CoV-2的Omicron亚变体,特别是BA.2.86和JN.1,已在多个国家迅速传播,在正在进行的COVID-19大流行中构成重大威胁。与BA.2的前身相比,Spike(S)蛋白上有34个额外的突变,BA.2.86及其进化后代的含义,JN.1在受体结合域(RBD)中有额外的L455S突变,是最重要的问题。在这项工作中,我们系统地检查了SARS-CoV-2Omicron亚变体的中和敏感性,并揭示了BA.2.86和JN.1的增强的抗体逃避。我们还确定了分别与宿主受体ACE2复合的BA.2.86和JN.1的三聚体S蛋白的冷冻EM结构。BA.2.86和JN.1的RBD内的突变诱导RBD和ACE2之间的相互作用网络的重塑。JN.1的L455S突变进一步诱导RBD-ACE2界面的显著转变,表明JN.1的结合亲和力明显低于BA.2.86。对具有核心中和表位的广泛中和抗体的分析揭示了OmicronBA.2.86亚变体进化的潜在抗体逃避机制。总的来说,我们构建了循环Omicron亚变体的病毒受体的进化景观。
    The Omicron subvariants of SARS-CoV-2, especially for BA.2.86 and JN.1, have rapidly spread across multiple countries, posing a significant threat in the ongoing COVID-19 pandemic. Distinguished by 34 additional mutations on the Spike (S) protein compared to its BA.2 predecessor, the implications of BA.2.86 and its evolved descendant, JN.1 with additional L455S mutation in receptor-binding domains (RBDs), are of paramount concern. In this work, we systematically examine the neutralization susceptibilities of SARS-CoV-2 Omicron subvariants and reveal the enhanced antibody evasion of BA.2.86 and JN.1. We also determine the cryo-EM structures of the trimeric S proteins from BA.2.86 and JN.1 in complex with the host receptor ACE2, respectively. The mutations within the RBDs of BA.2.86 and JN.1 induce a remodeling of the interaction network between the RBD and ACE2. The L455S mutation of JN.1 further induces a notable shift of the RBD-ACE2 interface, suggesting the notably reduced binding affinity of JN.1 than BA.2.86. An analysis of the broadly neutralizing antibodies possessing core neutralizing epitopes reveals the antibody evasion mechanism underlying the evolution of Omicron BA.2.86 subvariant. In general, we construct a landscape of evolution in virus-receptor of the circulating Omicron subvariants.
    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
    一旦新生链的N端在核糖体多肽隧道出口(PTE)处变得可接近,它们就会经历共翻译酶促加工。在真核生物中,甲硫氨酸氨基肽酶(MAP1和MAP2)的N端甲硫氨酸切除(NME),和N-乙酰转移酶A(NatA)的N-末端乙酰化(NTA),是对80S核糖体进行的后续修饰的最常见组合。如何在快速翻译核糖体的背景下协调这些酶促过程仍然难以捉摸。这里,我们报道了两种在空的人80S核糖体上组装的多酶复合物的低温EM结构,指示NME-NTA的两条路线。两个组件都在80S上形成,与新生的链基材无关。无论路线如何,NatA在核糖体上占据非侵入性“远端”结合位点,不干扰MAP1或MAP2结合,也不干扰大多数其他核糖体相关因子(RAF)。NatA可以参与协调,通过丰富的新生多肽相关复合物(NAC)的水泵样伴侣功能与MAP1动态组装。与MAP1相比,MAP2完全涵盖了PTE,因此与NAC和MAP1招募不相容。一起,我们的数据为蛋白质生物发生中NME和NTA的协调编排提供了结构框架.
    Nascent chains undergo co-translational enzymatic processing as soon as their N-terminus becomes accessible at the ribosomal polypeptide tunnel exit (PTE). In eukaryotes, N-terminal methionine excision (NME) by Methionine Aminopeptidases (MAP1 and MAP2), and N-terminal acetylation (NTA) by N-Acetyl-Transferase A (NatA), is the most common combination of subsequent modifications carried out on the 80S ribosome. How these enzymatic processes are coordinated in the context of a rapidly translating ribosome has remained elusive. Here, we report two cryo-EM structures of multi-enzyme complexes assembled on vacant human 80S ribosomes, indicating two routes for NME-NTA. Both assemblies form on the 80S independent of nascent chain substrates. Irrespective of the route, NatA occupies a non-intrusive \'distal\' binding site on the ribosome which does not interfere with MAP1 or MAP2 binding nor with most other ribosome-associated factors (RAFs). NatA can partake in a coordinated, dynamic assembly with MAP1 through the hydra-like chaperoning function of the abundant Nascent Polypeptide-Associated Complex (NAC). In contrast to MAP1, MAP2 completely covers the PTE and is thus incompatible with NAC and MAP1 recruitment. Together, our data provide the structural framework for the coordinated orchestration of NME and NTA in protein biogenesis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号