Protein allostery

  • 文章类型: Journal Article
    在这项研究中,我们利用蛋白质残基网络(PRN),使用局部空间模式(LSP)对齐构建,探讨代谢物激活蛋白(CAP)与cAMP序贯结合的动力学行为。我们采用这些PRN的程度中心性来研究亚纳秒级时间尺度上的蛋白质动力学,假设它将反映与热运动相关的CAP熵的变化。我们表明,第一个cAMP的结合导致环核苷酸结合域A(CNBD-A)的稳定性增加和CNBD-B的不稳定,与以前的报告一致,这些报告解释了cAMP结合在熵驱动变形法方面的负协同性。基于LSP的PRN还允许研究中间性中心性,PRN的另一个图论特征,提供对CAP内全球残留物连通性的见解。使用这种方法,我们能够正确鉴定在介导CAP变构相互作用中起关键作用的氨基酸.我们的研究和以前的实验报告之间的协议验证了我们的方法,特别是关于度中心性作为与蛋白质热动力学相关的熵的代理的可靠性。因为基于LSP的PRN可以很容易地扩展到包括有机小分子的动力学,多核苷酸,或其他变构蛋白,这里提出的方法标志着该领域的重大进步,将它们定位为快速的重要工具,成本效益高,熵驱动变构的准确分析和变构热点的识别。
    In this study, we utilize Protein Residue Networks (PRNs), constructed using Local Spatial Pattern (LSP) alignment, to explore the dynamic behavior of Catabolite Activator Protein (CAP) upon the sequential binding of cAMP. We employed the Degree Centrality of these PRNs to investigate protein dynamics on a sub-nanosecond time scale, hypothesizing that it would reflect changes in CAP\'s entropy related to its thermal motions. We show that the binding of the first cAMP led to an increase in stability in the Cyclic-Nucleotide Binding Domain A (CNBD-A) and destabilization in CNBD-B, agreeing with previous reports explaining the negative cooperativity of cAMP binding in terms of an entropy-driven allostery. LSP-based PRNs also allow for the study of Betweenness Centrality, another graph-theoretical characteristic of PRNs, providing insights into global residue connectivity within CAP. Using this approach, we were able to correctly identify amino acids that were shown to be critical in mediating allosteric interactions in CAP. The agreement between our studies and previous experimental reports validates our method, particularly with respect to the reliability of Degree Centrality as a proxy for entropy related to protein thermal dynamics. Because LSP-based PRNs can be easily extended to include dynamics of small organic molecules, polynucleotides, or other allosteric proteins, the methods presented here mark a significant advancement in the field, positioning them as vital tools for a fast, cost-effective, and accurate analysis of entropy-driven allostery and identification of allosteric hotspots.
    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
    变构在调节蛋白质活性中起着至关重要的作用,使其成为药物开发中备受追捧的目标。变构药物研究的主要挑战之一是变构位点的鉴定。近年来,许多计算模型已被开发用于准确的变构位点预测。这些模型中的大多数专注于设计可应用于来自不同家族的蛋白质口袋的一般规则。在这项研究中,我们提出了一种使用学习排名(LTR)概念的新方法。LTR模型根据口袋与变构位点的相关性对口袋进行排名,也就是说,口袋符合已知变构位点特征的程度。在对两个数据集进行训练和验证之后,变构数据库(ASD)和CASBench,LTR模型能够在83.6%和80.5%的测试蛋白质中排名前三个位置的变构口袋,分别。该模型优于其他常见的机器学习模型,具有更高的F1分数(ASD中的0.662和CASBench中的0.608)和马修斯相关系数(ASD中的0.645和CASBench中的0.589)。经训练的模型可在PASSer平台(https://passer.smu.edu)以协助药物发现研究。
    Allostery plays a crucial role in regulating protein activity, making it a highly sought-after target in drug development. One of the major challenges in allosteric drug research is the identification of allosteric sites. In recent years, many computational models have been developed for accurate allosteric site prediction. Most of these models focus on designing a general rule that can be applied to pockets of proteins from various families. In this study, we present a new approach using the concept of Learning to Rank (LTR). The LTR model ranks pockets based on their relevance to allosteric sites, that is, how well a pocket meets the characteristics of known allosteric sites. After the training and validation on two datasets, the Allosteric Database (ASD) and CASBench, the LTR model was able to rank an allosteric pocket in the top three positions for 83.6% and 80.5% of test proteins, respectively. The model outperforms other common machine learning models with higher F1 scores (0.662 in ASD and 0.608 in CASBench) and Matthews correlation coefficients (0.645 in ASD and 0.589 in CASBench). The trained model is available on the PASSer platform (https://passer.smu.edu) to aid in drug discovery research.
    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
    中性突变漂移是生物多样性的重要来源,在蛋白质生物物理学的基础研究中仍未得到充分利用。本研究使用合成转录电路研究蛋白酪氨酸磷酸酶1B(PTP1B)的中性漂移,构象变化是限速的哺乳动物信号酶。纯化突变体的动力学分析表明催化活性,而不是热力学稳定性,引导在中性漂移下富集,中性或轻度激活突变可以减轻有害突变的影响。总的来说,突变体表现出中等的活性-稳定性权衡,这表明PTP1B活性的微小改善不需要伴随其稳定性的损失。大型突变池的多重测序表明,在生物选择下清除了变构影响位点的替换,富集位于活性位点之外的突变。研究结果表明,漂移种群中中性突变的位置依赖性可以揭示变构网络,并说明了使用合成转录系统探索调节酶中这些突变的方法。本文受版权保护。保留所有权利。
    Neutral mutational drift is an important source of biological diversity that remains underexploited in fundamental studies of protein biophysics. This study uses a synthetic transcriptional circuit to study neutral drift in protein tyrosine phosphatase 1B (PTP1B), a mammalian signaling enzyme for which conformational changes are rate limiting. Kinetic assays of purified mutants indicate that catalytic activity, rather than thermodynamic stability, guides enrichment under neutral drift, where neutral or mildly activating mutations can mitigate the effects of deleterious ones. In general, mutants show a moderate activity-stability tradeoff, an indication that minor improvements in the activity of PTP1B do not require concomitant losses in its stability. Multiplexed sequencing of large mutant pools suggests that substitutions at allosterically influential sites are purged under biological selection, which enriches for mutations located outside of the active site. Findings indicate that the positional dependence of neutral mutations within drifting populations can reveal the presence of allosteric networks and illustrate an approach for using synthetic transcriptional systems to explore these mutations in regulatory enzymes.
    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
    变构机制是蛋白质常用的调节工具,用于协调复杂的生化过程并控制细胞中的通讯。变构分子事件的定量理解和表征是现代生物学中的主要挑战之一,需要集成创新的计算实验方法来获得原子级的变构状态知识。互动,和动态构象景观。新兴的人工智能(AI)技术赋予了越来越多的计算和实验研究,为从第一原理探索和学习蛋白质变构宇宙开辟了新的范式。在这篇综述中,我们分析了变构蛋白功能的高通量深度突变扫描的最新进展;Alpha-fold结构预测方法在蛋白质动力学和变性反应研究中的应用和最新适应;集成机器学习和增强采样技术以表征变性反应的新领域;以及变性反应系统研究的结构生物学方法的最新进展。我们还重点介绍了SARS-CoV-2尖峰(S)蛋白的最新计算和实验研究,揭示了驱动功能构象变化的变构调节的重要且通常隐藏的作用。与宿主受体的结合相互作用,以及对病毒感染至关重要的S蛋白的突变逃逸机制。最后,我们对未来方向进行了总结和展望,表明AI增强的生物物理和计算机模拟方法开始将蛋白质变构的研究转变为变构景观的系统表征,隐藏的变构状态,以及可能带来分子生物学和药物发现新革命的机制。
    Allosteric mechanisms are commonly employed regulatory tools used by proteins to orchestrate complex biochemical processes and control communications in cells. The quantitative understanding and characterization of allosteric molecular events are among major challenges in modern biology and require integration of innovative computational experimental approaches to obtain atomistic-level knowledge of the allosteric states, interactions, and dynamic conformational landscapes. The growing body of computational and experimental studies empowered by emerging artificial intelligence (AI) technologies has opened up new paradigms for exploring and learning the universe of protein allostery from first principles. In this review we analyze recent developments in high-throughput deep mutational scanning of allosteric protein functions; applications and latest adaptations of Alpha-fold structural prediction methods for studies of protein dynamics and allostery; new frontiers in integrating machine learning and enhanced sampling techniques for characterization of allostery; and recent advances in structural biology approaches for studies of allosteric systems. We also highlight recent computational and experimental studies of the SARS-CoV-2 spike (S) proteins revealing an important and often hidden role of allosteric regulation driving functional conformational changes, binding interactions with the host receptor, and mutational escape mechanisms of S proteins which are critical for viral infection. We conclude with a summary and outlook of future directions suggesting that AI-augmented biophysical and computer simulation approaches are beginning to transform studies of protein allostery toward systematic characterization of allosteric landscapes, hidden allosteric states, and mechanisms which may bring about a new revolution in molecular biology and drug discovery.
    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
    变构调节1型菌毛粘附素FimH与甘露糖基化受体的结合,以增强尿路感染(UTI)期间尿路致病性大肠杆菌(UPEC)的适应性。位于甘露糖结合口袋外的两个FimH结构域(菌毛蛋白和凝集素)的突变已显示影响甘露糖结合亲和力。然而变构机制的细节还没有完全阐明。在这里,我们使用分子动力学(MD)模拟技术表征了天然FimH变体的不同FimH构象状态(称为低亲和力时态和高亲和力松弛构象),并报告了它们之间的关键结构动力学差异。来自大流行多药抗性大肠杆菌ST131谱系的临床显性FimH30变体含有R166H突变,其削弱FimH域间相互作用并允许与预先存在的高亲和力松弛构象增强的甘露糖相互作用。当在等基因ST131菌株背景中表达时,FimH30介导的高细胞粘附和侵袭,与其他变体相比,生物膜的形成增强。总的来说,我们的计算和实验结果支持FimH蛋白变构模型,该模型是由FimH预先存在的构象平衡的变化介导的,除了在蛋白质内从一个位点传递到另一个位点的结构扰动的顺序逐步过程。重要的是,这是首次揭示临床显性FimH变体中的自然突变如何影响蛋白质的构象格局,从而优化其在肠道和肠外生态位的ST131适应性功能的研究。
    The binding of the type 1 fimbrial adhesin FimH to mannosylated receptors is allosterically regulated to enhance the fitness of uropathogenic Escherichia coli (UPEC) during urinary tract infection (UTI). Mutations in the two FimH domains (pilin and lectin) located outside the mannose binding pocket have been shown to influence mannose binding affinity, yet the details of the allostery mechanism are not fully elucidated. Here we characterised different FimH conformational states (termed low-affinity tense and high-affinity relaxed conformations) of natural FimH variants using molecular dynamics (MD) simulation techniques and report key structural dynamics differences between them. The clinically dominant FimH30 variant from the pandemic multidrug resistant E. coli ST131 lineage contains an R166H mutation that weakens FimH interdomain interactions and allows enhanced mannose interactions with pre-existing high-affinity relaxed conformations. When expressed in an isogenic ST131 strain background, FimH30 mediated high human cell adhesion and invasion, and enhanced biofilm formation over other variants. Collectively, our computational and experimental findings support a model of FimH protein allostery that is mediated by shifts in the pre-existing conformational equilibrium of FimH, additional to the sequential step-wise process of structural perturbations transmitted from one site to another within the protein. Importantly, it is the first study to shed light into how natural mutations in a clinically dominant FimH variant influence the protein\'s conformational landscape optimising its function for ST131 fitness at intestinal and extraintestinal niches.
    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
    The human high-temperature requirement A2 (HtrA2) mitochondrial protease is critical for cellular proteostasis, with mutations in this enzyme closely associated with the onset of neurodegenerative disorders. HtrA2 forms a homotrimeric structure, with each subunit composed of protease and PDZ (PSD-95, DLG, ZO-1) domains. Although we had previously shown that successive ligand binding occurs with increasing affinity, and it has been suggested that allostery plays a role in regulating catalysis, the molecular details of how this occurs have not been established. Here, we use cysteine-based chemistry to generate subunits in different conformational states along with a protomer mixing strategy, biochemical assays, and methyl-transverse relaxation optimized spectroscopy-based NMR studies to understand the role of interprotomer allostery in regulating HtrA2 function. We show that substrate binding to a PDZ domain of one protomer increases millisecond-to-microsecond timescale dynamics in neighboring subunits that prime them for binding substrate molecules. Only when all three PDZ-binding sites are substrate bound can the enzyme transition into an active conformation that involves significant structural rearrangements of the protease domains. Our results thus explain why when one (or more) of the protomers is fixed in a ligand-binding-incompetent conformation or contains the inactivating S276C mutation that is causative for a neurodegenerative phenotype in mouse models of Parkinson\'s disease, transition to an active state cannot be formed. In this manner, wild-type HtrA2 is only active when substrate concentrations are high and therefore toxic and unregulated proteolysis of nonsubstrate proteins can be suppressed.
    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
    Studies of protein allostery increasingly reveal an involvement of the back and forth order-disorder transitions in this mechanism of protein activity regulation. Here, we investigate the allosteric mechanisms mediated by structural disorder using the structure-based statistical mechanical model of allostery (SBSMMA) that we have previously developed. We show that SBSMMA accounts for the energetics and causality of allosteric communication underlying dimerization of the BirA biotin repressor, activation of the sortase A enzyme, and inhibition of the Rac1 GTPase. Using the SBSMMA, we also show that introducing structural order or disorder in various regions of esterases can originate tunable allosteric modulation of the catalytic triad. On the basis of obtained results, we propose that operating with the order-disorder continuum allows one to establish an allosteric control scale for achieving desired modulation of the protein activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    G proteins play a central role in signal transduction and pharmacology. Signaling is initiated by cell-surface receptors, which promote guanosine triphosphate (GTP) binding and dissociation of Gα from the Gβγ subunits. Structural studies have revealed the molecular basis of subunit association with receptors, RGS proteins, and downstream effectors. In contrast, the mechanism of subunit dissociation is poorly understood. We use cell signaling assays, molecular dynamics (MD) simulations, and biochemistry and structural analyses to identify a conserved network of amino acids that dictates subunit release. In the presence of the terminal phosphate of GTP, a glycine forms a polar network with an arginine and glutamate, putting torsional strain on the subunit binding interface. This \"G-R-E motif\" secures GTP and, through an allosteric link, discharges the Gβγ dimer. Replacement of network residues prevents subunit dissociation regardless of agonist or GTP binding. These findings reveal the molecular basis of the final committed step of G protein activation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    脂氧合酶(LOX)催化作为细胞信号传导和炎症的重要介质的脂肪酸的(全)氧化。这些反应由在植物和动物酶中被变构调节的C-H活化步骤引发。来自高等真核生物的LOX配备了N末端PLAT(Polycystin-1,脂氧合酶,与小分子变构效应子结合的α-毒素)结构域,这反过来又调节底物特异性和催化的限速步骤。在这里,总结了描述脂肪酸及其衍生物对植物和动物脂肪加氧酶化学的变构调节的动力学和结构证据。
    Lipoxygenases (LOXs) catalyze the (per) oxidation of fatty acids that serve as important mediators for cell signaling and inflammation. These reactions are initiated by a C-H activation step that is allosterically regulated in plant and animal enzymes. LOXs from higher eukaryotes are equipped with an N-terminal PLAT (Polycystin-1, Lipoxygenase, Alpha-Toxin) domain that has been implicated to bind to small molecule allosteric effectors, which in turn modulate substrate specificity and the rate-limiting steps of catalysis. Herein, the kinetic and structural evidence that describes the allosteric regulation of plant and animal lipoxygenase chemistry by fatty acids and their derivatives are summarized.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    旋转细菌鞭毛马达因其在旋转感觉切换过程中的高度同步操作和放大而在生物化学方面非常出色。马达是鞭毛基体的一部分,复杂的多蛋白质组装。感官和能量转导取决于六种蛋白质的核心,这些蛋白质在不同物种中适应以调节扭矩并产生不同的开关。对趋化和环境刺激的运动反应由核心与小信号蛋白的相互作用驱动。最初的蛋白质相互作用通过多亚基细胞质环传播以切换扭矩。扭矩反转触发鞭毛丝中的结构转变以改变运动行为。核心组件的细微变化会反转或阻止开关操作。已经通过多种方法研究了鞭毛开关的力学,从蛋白质动力学到单分子和细胞生物物理学。建筑,在电子冷冻显微镜的最新进展的推动下,可用于多种物种。计算方法与遗传和生化数据库具有相关结构。开关超灵敏度及其对电机转矩的依赖基础的设计原理仍然难以捉摸,但是诱人的线索已经出现了。这篇综述旨在将最新的知识整合到一个统一的平台中,以激发新的研究策略。
    The rotary bacterial flagellar motor is remarkable in biochemistry for its highly synchronized operation and amplification during switching of rotation sense. The motor is part of the flagellar basal body, a complex multi-protein assembly. Sensory and energy transduction depends on a core of six proteins that are adapted in different species to adjust torque and produce diverse switches. Motor response to chemotactic and environmental stimuli is driven by interactions of the core with small signal proteins. The initial protein interactions are propagated across a multi-subunit cytoplasmic ring to switch torque. Torque reversal triggers structural transitions in the flagellar filament to change motile behavior. Subtle variations in the core components invert or block switch operation. The mechanics of the flagellar switch have been studied with multiple approaches, from protein dynamics to single molecule and cell biophysics. The architecture, driven by recent advances in electron cryo-microscopy, is available for several species. Computational methods have correlated structure with genetic and biochemical databases. The design principles underlying the basis of switch ultra-sensitivity and its dependence on motor torque remain elusive, but tantalizing clues have emerged. This review aims to consolidate recent knowledge into a unified platform that can inspire new research strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号