Protein allostery

  • 文章类型: Journal Article
    低分子量肝素和合成模拟物如磺达肝素显示不同的结合动力学,蛋白酶特异性,和临床效果。已经提出了变构和模板介导的桥接机制的组合来解释速率加速和特异性的差异。使用异质肝素物种的困难使模拟物和天然肝素之间抗凝血酶激活差异的晶体学解释变得难以接近。在这项研究中,我们检查了结合磺达肝素引起的抗凝血酶的变构变化,使用毫秒氢氘交换质谱(TRESI-HDXMS)的依诺肝素和解聚的天然肝素,并使用碰撞诱导的解折叠(CIU)将这些构象变化与气相中的复合物稳定性联系起来。这一探索表明,除了磺达肝素引起的动态变化外,长链肝素会降低Arg393附近的结构灵活性,Arg393是蛋白质反应中心环中的可切割残基。蛋白质动力学的这些局部变化与随着肝素链长度而增加的总体复合物稳定性的增加有关。最终,这些结果揭示了肝素模拟物和天然肝素之间活性和特异性差异的分子机制。
    Low molecular weight heparin and synthetic mimetics such as fondaparinux show different binding kinetics, protease specificity, and clinical effects. A combination of allosteric and template-mediated bridging mechanisms have been proposed to explain the differences in rate acceleration and specificity. The difficulty in working with heterogeneous heparin species has rendered a crystallographic interpretation of the differences in antithrombin activation between mimetics and natural heparin inaccessible. In this study, we examine the allosteric changes in antithrombin caused by binding fondaparinux, enoxaparin and depolymerized natural heparins using millisecond hydrogen deuterium exchange mass spectrometry (TRESI-HDX MS) and relate these conformational changes to complex stability in the gas phase using collision induced unfolding (CIU). This exploration reveals that in addition to the dynamic changes caused by fondaparinux, long chain heparins reduce structural flexibility proximal to Arg393, the cleavable residue in the reactive centre loop of the protein. These local changes in protein dynamics are associated with an increase in overall complex stability that increases with heparin chain length. Ultimately, these results shed light on the molecular mechanisms underlying differences in activity and specificity between heparin mimetics and natural heparins.
    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
    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
    背景:肌萎缩侧索硬化症(ALS)是一种严重的神经退行性疾病,影响大脑和脊髓中的神经细胞,由超氧化物歧化酶1(SOD1)酶的突变引起。ALS相关突变会导致错误折叠,二聚化不稳定性,并增加了聚集体的形成。潜在的变构机制,然而,就其基本原子结构的细节而言,仍然晦涩难懂。因此,这种知识上的差距限制了新型SOD1抑制剂的开发以及对远端位点疾病相关突变如何影响酶活性的理解.
    方法:我们将基于微秒尺度的无偏分子动力学(MD)模拟与网络分析相结合,以阐明SOD1Apo(未金属化形式)中的局部和全局构象变化和变构通讯,霍洛,Apo_CallA(突变体和未金属化形式),和Holo_CallA(突变形式)系统。为了鉴定参与SOD1信号传导和变构通讯的热点残基,我们表现出网络中心性,社区网络,和路径分析。
    结果:结构分析表明,未金属化的SOD1系统和半胱氨酸突变在催化位点显示出很大的结构变化,影响结构稳定性。间和内H键分析确定了几个重要的残基对于保持界面稳定性至关重要,结构稳定性,和酶催化。动态运动分析表明,在Holo系统中,原子位移更加平衡,运动高度相关。使用距离和二面体概率分布分析阐明了在Apo和Holo系统中二硫键形成和R143构型中观察到的结构差异的基本原理。
    结论:我们的研究强调了将广泛的MD模拟与网络分析相结合以揭示蛋白质变形性特征的效率。
    Amyotrophic lateral sclerosis (ALS) is a serious neurodegenerative disorder affecting nerve cells in the brain and spinal cord that is caused by mutations in the superoxide dismutase 1 (SOD1) enzyme. ALS-related mutations cause misfolding, dimerisation instability, and increased formation of aggregates. The underlying allosteric mechanisms, however, remain obscure as far as details of their fundamental atomistic structure are concerned. Hence, this gap in knowledge limits the development of novel SOD1 inhibitors and the understanding of how disease-associated mutations in distal sites affect enzyme activity.
    We combined microsecond-scale based unbiased molecular dynamics (MD) simulation with network analysis to elucidate the local and global conformational changes and allosteric communications in SOD1 Apo (unmetallated form), Holo, Apo_CallA (mutant and unmetallated form), and Holo_CallA (mutant form) systems. To identify hotspot residues involved in SOD1 signalling and allosteric communications, we performed network centrality, community network, and path analyses.
    Structural analyses showed that unmetallated SOD1 systems and cysteine mutations displayed large structural variations in the catalytic sites, affecting structural stability. Inter- and intra H-bond analyses identified several important residues crucial for maintaining interfacial stability, structural stability, and enzyme catalysis. Dynamic motion analysis demonstrated more balanced atomic displacement and highly correlated motions in the Holo system. The rationale for structural disparity observed in the disulfide bond formation and R143 configuration in Apo and Holo systems were elucidated using distance and dihedral probability distribution analyses.
    Our study highlights the efficiency of combining extensive MD simulations with network analyses to unravel the features of protein allostery.
    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
    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
    溶液中蛋白质构象异构体的直接结构和动态表征是非常需要的,但目前是不切实际的。在这里,我们开发了一个单分子金等离子体纳米孔系统,用于观察蛋白质变构,使我们能够通过离子电流检测和SERS光谱测量来监测蛋白质的易位动力学和构象转换,分别。钙调蛋白(CaM)的变构转变被纳米孔系统精心探测。使用离子电流阻断信号和SERS光谱在单分子水平上很好地分辨了CaM的两个构象异构体。收集的SERS光谱提供了结构证据,以证实CaM和金等离子体纳米孔之间的相互作用,这是两个构象异构体不同的易位行为的原因。SERS光谱揭示了参与钙结合时CaM构象变化的氨基酸残基。结果表明,出色的光谱表征为单分子纳米孔技术提供了先进的直接结构分析能力。
    Direct structural and dynamic characterization of protein conformers in solution is highly desirable but currently impractical. Herein, we developed a single molecule gold plasmonic nanopore system for observation of protein allostery, enabling us to monitor translocation dynamics and conformation transition of proteins by ion current detection and SERS spectrum measurement, respectively. Allosteric transition of calmodulin (CaM) was elaborately probed by the nanopore system. Two conformers of CaM were well-resolved at a single-molecule level using both the ion current blockage signal and the SERS spectra. The collected SERS spectra provided structural evidence to confirm the interaction between CaM and the gold plasmonic nanopore, which was responsible for the different translocation behaviors of the two conformers. SERS spectra revealed the amino acid residues involved in the conformational change of CaM upon calcium binding. The results demonstrated that the excellent spectral characterization furnishes a single-molecule nanopore technique with an advanced capability of direct structure analysis.
    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
    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)

公众号