cryoelectron microscopy

冷冻电子显微镜
  • 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
    伏马菌素B1(FB1)靶向鞘脂生物合成,抑制神经酰胺合酶。在本期的结构中,Zhang等人1确定了与FB1及其酰化衍生物复合的酵母神经酰胺合酶的低温电子显微镜结构,酰基-FB1,揭示了FB1N-酰化的两步“乒乓”机制以及它如何抑制神经酰胺合酶。
    Fumonisin B1 (FB1) targets sphingolipid biosynthesis, inhibiting ceramide synthases. In this issue of Structure, Zhang et al.1 determined the cryoelectron microscopic structures of yeast ceramide synthase in complex with FB1 and its acylated derivative, acyl-FB1, revealing a two-step \"ping-pong\" mechanism for the N-acylation of FB1 and how it inhibits ceramide synthase.
    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
    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
    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
    病毒与细胞膜的融合是包膜病毒生命周期中的关键步骤。病毒融合蛋白促进了这一过程,其中许多是构象pH敏感的。pH变化如何引发这种融合的细节在很大程度上仍然难以捉摸。这项研究提出了原型III类融合蛋白的冷冻电子显微镜(cryo-EM)结构,GP64,在其融合前和早期中间状态下,揭示了伴随膜融合过程的结构中间体。这些结构确定了pH敏感开关的参与,包括H23、H245和H304,在感测触发膜融合的初始步骤的低pH时。该开关的pH传感作用通过细胞-细胞合胞体形成和双重染料标记的测定得到证实。研究结果表明,多个组氨酸残基之间的配位充当pH传感器和激活剂。多组氨酸开关在病毒融合中的参与适用于感染人的流感病毒和其他病毒的融合剂,这可能导致开发抗病毒疗法和疫苗的策略。
    The fusion of viruses with cellular membranes is a critical step in the life cycle of enveloped viruses. This process is facilitated by viral fusion proteins, many of which are conformationally pH-sensitive. The specifics of how changes in pH initiate this fusion have remained largely elusive. This study presents the cryo-electron microscopy (cryo-EM) structures of a prototype class III fusion protein, GP64, in its prefusion and early intermediate states, revealing the structural intermediates accompanying the membrane fusion process. The structures identify the involvement of a pH-sensitive switch, comprising H23, H245, and H304, in sensing the low pH that triggers the initial step of membrane fusion. The pH sensing role of this switch is corroborated by assays of cell-cell syncytium formation and dual dye-labeling. The findings demonstrate that coordination between multiple histidine residues acts as a pH sensor and activator. The involvement of a multi-histidine switch in viral fusion is applicable to fusogens of human-infecting thogotoviruses and other viruses, which could lead to strategies for developing anti-viral therapies and vaccines.
    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
    耐药结核病是一个全球性的公共卫生问题。对利福平的抗性,最有效的结核病治疗药物,是一个日益增长的主要问题。病原体,结核分枝杆菌(Mtb),具有一组ATP结合盒(ABC)转运蛋白,这些转运蛋白负责通过主动输出产生耐药性。这里,我们描述了将MtbRv1217c-1218c描述为ABC转运蛋白的研究,该转运蛋白可以介导分枝杆菌对利福平的耐药性,并确定了Rv1217c-1218c的低温电子显微镜结构。结构显示Rv1217c-1218c具有V型出口商折叠。如果没有ATP,Rv1217c-1218c通过来自每个跨膜结构域(TMD)的两个并列膜螺旋形成周质门,而核苷酸结合域(NBD)形成部分封闭的二聚体,该二聚体由四个盐桥保持在一起。腺苷酸-亚氨基二磷酸(AMPPNP)结合诱导结构变化,其中NBDs变得彼此进一步封闭,其下游转化为TMD的闭合构象。AMPPNP结合导致外部小叶腔的塌陷和周质门的打开,它被提议在底物出口中发挥作用。利福平结合结构显示疏水性和面向周质的腔参与利福平结合。在所有确定的结构中都观察到磷脂分子,并形成Rv1217c-1218c转运蛋白系统的组成部分。我们的结果为介导利福平耐药性的分枝杆菌ABC出口商提供了结构基础,这可能会导致对对抗利福平抵抗的不同见解。
    Drug-resistant Tuberculosis (TB) is a global public health problem. Resistance to rifampicin, the most effective drug for TB treatment, is a major growing concern. The etiological agent, Mycobacterium tuberculosis (Mtb), has a cluster of ATP-binding cassette (ABC) transporters which are responsible for drug resistance through active export. Here, we describe studies characterizing Mtb Rv1217c-1218c as an ABC transporter that can mediate mycobacterial resistance to rifampicin and have determined the cryo-electron microscopy structures of Rv1217c-1218c. The structures show Rv1217c-1218c has a type V exporter fold. In the absence of ATP, Rv1217c-1218c forms a periplasmic gate by two juxtaposed-membrane helices from each transmembrane domain (TMD), while the nucleotide-binding domains (NBDs) form a partially closed dimer which is held together by four salt-bridges. Adenylyl-imidodiphosphate (AMPPNP) binding induces a structural change where the NBDs become further closed to each other, which downstream translates to a closed conformation for the TMDs. AMPPNP binding results in the collapse of the outer leaflet cavity and the opening of the periplasmic gate, which was proposed to play a role in substrate export. The rifampicin-bound structure shows a hydrophobic and periplasm-facing cavity is involved in rifampicin binding. Phospholipid molecules are observed in all determined structures and form an integral part of the Rv1217c-1218c transporter system. Our results provide a structural basis for a mycobacterial ABC exporter that mediates rifampicin resistance, which can lead to different insights into combating rifampicin resistance.
    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
    WNT信号传导是发展和稳态的基础,但是卷曲受体(FZD)如何传播信号仍然是一个谜。这里,我们提出了FZD4的低温EM结构,该结构与Dishevelled2(DVL2)的DEP结构域接合,一个关键的WNT传感器。我们发现了一种独特的结合模式,其中DEP指环插入FZD4腔中以形成疏水界面。FZD4胞内环2(ICL2)还通过极性接触锚定复合物。诱变证实了结构观察。DEP接口在FZD中高度保守,指示FZD与DVD接合的通用机制。我们进一步揭示了DEP模拟G蛋白/β抑制蛋白/GRK以识别受体的活性构象,扩展当前的GPCR参与模式。最后,我们确定了一个独特的FZD4二聚化界面。我们的发现描述了控制FZD/DVL组装和WNT信号传播的分子决定因素,提供长期寻求的WNT信号转导基础答案。
    WNT signaling is fundamental in development and homeostasis, but how the Frizzled receptors (FZDs) propagate signaling remains enigmatic. Here, we present the cryo-EM structure of FZD4 engaged with the DEP domain of Dishevelled 2 (DVL2), a key WNT transducer. We uncover a distinct binding mode where the DEP finger-loop inserts into the FZD4 cavity to form a hydrophobic interface. FZD4 intracellular loop 2 (ICL2) additionally anchors the complex through polar contacts. Mutagenesis validates the structural observations. The DEP interface is highly conserved in FZDs, indicating a universal mechanism by which FZDs engage with DVLs. We further reveal that DEP mimics G-protein/β-arrestin/GRK to recognize an active conformation of receptor, expanding current GPCR engagement models. Finally, we identify a distinct FZD4 dimerization interface. Our findings delineate the molecular determinants governing FZD/DVL assembly and propagation of WNT signaling, providing long-sought answers underlying WNT signal transduction.
    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
    OSCA/TMEM63通道,它们具有类似于运输机的架构,是真正的机械敏感(MS)离子通道,可感知真核细胞中的高阈值机械力。这些转运蛋白样通道的激活机制尚未完全了解。在这里,我们报告了在去污剂环境中具有顺序的细胞外扩张孔的二聚体OSCA/TMEM63孔突变体OSCA1.1-F516A的冷冻EM结构。这些结构表明,细胞外孔的顺序扩张类似于花朵的绽放,并与每个单体亚基向二聚体界面的顺序收缩以及随后的二聚体界面脂质的挤出偶联。有趣的是,虽然OSCA1.1-F516A在天然脂质环境中保持不导电,它可以被具有降低的单通道电导的溶血磷脂酰胆碱(Lyso-PC)直接激活。在无溶血-PC和含有溶血-PC的脂质纳米盘中的OSCA1.1-F516A的结构分析表明,溶血-PC通过吸引M6b向上移动远离细胞内侧从而延伸细胞内孔而诱导细胞内孔扩张。进一步的功能研究表明,高阈值机械力完全激活MSOSCA/TMEM63二聚体通道还涉及细胞间和细胞外孔的开放。我们的结果提供了独特的转运蛋白样MSOSCA/TMEM63通道的基本激活范例,这可能适用于TMEM63/TMEM16/TMC超家族的功能分支。
    OSCA/TMEM63 channels, which have transporter-like architectures, are bona fide mechanosensitive (MS) ion channels that sense high-threshold mechanical forces in eukaryotic cells. The activation mechanism of these transporter-like channels is not fully understood. Here we report cryo-EM structures of a dimeric OSCA/TMEM63 pore mutant OSCA1.1-F516A with a sequentially extracellular dilated pore in a detergent environment. These structures suggest that the extracellular pore sequential dilation resembles a flower blooming and couples to a sequential contraction of each monomer subunit towards the dimer interface and subsequent extrusion of the dimer interface lipids. Interestingly, while OSCA1.1-F516A remains non-conducting in the native lipid environment, it can be directly activated by lyso-phosphatidylcholine (Lyso-PC) with reduced single-channel conductance. Structural analysis of OSCA1.1-F516A in lyso-PC-free and lyso-PC-containing lipid nanodiscs indicates that lyso-PC induces intracellular pore dilation by attracting the M6b to upward movement away from the intracellular side thus extending the intracellular pore. Further functional studies indicate that full activation of MS OSCA/TMEM63 dimeric channels by high-threshold mechanical force also involves the opening of both intercellular and extracellular pores. Our results provide the fundamental activation paradigm of the unique transporter-like MS OSCA/TMEM63 channels, which is likely applicable to functional branches of the TMEM63/TMEM16/TMC superfamilies.
    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
    黄热病病毒(YFV)感染可导致人类严重疾病,每年在非洲和美洲造成大量伤亡。分泌型NS1(sNS1)被认为是黄病毒感染的诊断标记,在黄病毒生命周期中起着至关重要的作用,但对YFVsNS1的组成和结构知之甚少。这里,我们提出了重组YFVsNS1存在于寡聚化的异质混合物中,主要以四聚体形式。冷冻EM结构表明,sNS1的YFV四聚体通过β-roll结构域和油腻指之间的疏水相互作用而堆叠。根据三维变异性分析,四聚体处于半稳定状态,可能包含动态变化的多种构象。我们相信,我们的研究为NS1的寡聚化提供了重要的见解,并将有助于开发基于NS1的诊断和治疗方法。
    Yellow fever virus (YFV) infections can cause severe diseases in humans, resulting in mass casualties in Africa and the Americas each year. Secretory NS1 (sNS1) is thought to be used as a diagnostic marker of flavivirus infections, playing an essential role in the flavivirus life cycle, but little is known about the composition and structure of YFV sNS1. Here, we present that the recombinant YFV sNS1 exists in a heterogeneous mixture of oligomerizations, predominantly in the tetrameric form. The cryoEM structures show that the YFV tetramer of sNS1 is stacked by the hydrophobic interaction between β-roll domains and greasy fingers. According to the 3D variability analysis, the tetramer is in a semi-stable state that may contain multiple conformations with dynamic changes. We believe that our study provides critical insights into the oligomerization of NS1 and will aid the development of NS1-based diagnoses and therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号