exosite

Exosite
  • 文章类型: Journal Article
    暂无摘要。
    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
    止血依赖于丝氨酸蛋白酶及其辅因子的反应网络以形成血凝块。凝血因子IXa(蛋白酶)在止血中起着至关重要的作用,这从与缺乏凝血因子IXa相关的出血疾病中可以明显看出。特异性靶向单个凝血因子的RNA适体具有作为抗凝剂和作为结构与功能之间关系的探针的潜力。这里,我们报道了人因子IXa的X射线结构,该结构没有以apo形式或与对因子IXa具有特异性的抑制性适体复合的形式与活性位点结合的配体。适体与催化结构域中的外位点结合并变构扭曲活性位点。我们的研究揭示了IXa态的构象集合,其中Trp215在活性位点附近的大移动驱动闭合(适体结合)之间的功能转变,潜伏(apo),和开放(底物结合)状态。与其他凝固蛋白酶相比,apo酶的潜伏状态可能会影响IXa独特的差催化活性。exosite,适体与之结合,与VIIIa和肝素的结合有关,两者都调节IXa功能。我们的发现揭示了exosite驱动的IXa功能变构调节的重要性以及重新平衡止血以获得治疗效果的新策略。
    Hemostasis relies on a reaction network of serine proteases and their cofactors to form a blood clot. Coagulation factor IXa (protease) plays an essential role in hemostasis as evident from the bleeding disease associated with its absence. RNA aptamers specifically targeting individual coagulation factors have potential as anticoagulants and as probes of the relationship between structure and function. Here, we report X-ray structures of human factor IXa without a ligand bound to the active site either in the apo-form or in complex with an inhibitory aptamer specific for factor IXa. The aptamer binds to an exosite in the catalytic domain and allosterically distorts the active site. Our studies reveal a conformational ensemble of IXa states, wherein large movements of Trp215 near the active site drive functional transitions between the closed (aptamer-bound), latent (apo), and open (substrate-bound) states. The latent state of the apo-enzyme may bear on the uniquely poor catalytic activity of IXa compared to other coagulation proteases. The exosite, to which the aptamer binds, has been implicated in binding VIIIa and heparin, both of which regulate IXa function. Our findings reveal the importance of exosite-driven allosteric modulation of IXa function and new strategies to rebalance hemostasis for therapeutic gain.
    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
    量子点(QD)的有利光学特性激发了它们在与成像和生物分析相关的各种应用中的用途。包括检测蛋白酶及其活性。最近的研究表明,量子点的表面化学能够调节蛋白酶的活性,但只是非特定的。这里,我们提出了一种策略,以选择性地加速特定靶蛋白酶的活性多达两个数量级。外位点结合“诱饵”肽来自跨越一系列生物学作用的蛋白质-底物,受体,和抑制剂-并用于增加QD-肽缀合物对凝血酶或因子Xa的亲和力,导致共结合底物的蛋白水解速率增加。与QD表面化学的影响不同,加速度对目标蛋白酶具有特异性,而其他蛋白酶的加速度可忽略不计。这种“诱饵和切割”传感方法的好处包括检测极限提高了一个数量级以上,在非目标蛋白水解的压倒性背景下重新启用目标蛋白酶的检测,和缓解抑制剂的作用。累积结果指向可推广的策略,加速的机制,设计诱饵肽和缀合物的考虑因素,并讨论了扩大这种方法范围的途径。总的来说,这项研究代表了在提高灵敏度和选择性的纳米颗粒酶传感器的合理设计方面迈出的重要一步.
    The advantageous optical properties of quantum dots (QDs) motivate their use in a wide variety of applications related to imaging and bioanalysis, including the detection of proteases and their activity. Recent studies have shown that surface chemistry on QDs is able to modulate protease activity, but only nonspecifically. Here, we present a strategy to selectively accelerate the activity of a particular target protease by as much as two orders of magnitude. Exosite-binding \"bait\" peptides were derived from proteins that span a range of biological roles─substrate, receptor, and inhibitor─and were used to increase the affinity of the QD-peptide conjugates for either thrombin or factor Xa, resulting in increased rates of proteolysis for coconjugated substrates. Unlike effects from QD surface chemistry, the acceleration was specific to the target protease with negligible acceleration of other proteases. Benefits of this \"bait and cleave\" sensing approach included detection limits that improved by more than an order of magnitude, reenabled detection of target protease against an overwhelming background of nontarget proteolysis, and mitigation of the action of inhibitors. The cumulative results point to a generalizable strategy, where the mechanism of acceleration, considerations for the design of bait peptides and conjugates, and routes to expanding the scope of this approach are discussed. Overall, this research represents a major step forward in the rational design of nanoparticle-based enzyme sensors that enhance sensitivity and selectivity.
    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
    SARS-CoV-2是造成最近全球大流行的病原体,这已经夺走了全球数十万的受害者。尽管在开发有效疫苗方面付出了巨大的努力,人们对新变种的实际保护提出了担忧。因此,研究人员渴望找到对抗这种病原体的替代策略。像其他机会主义实体一样,SARS-CoV-2生命周期中的关键步骤是细胞酶Furin在RARR685→基序处的包膜糖蛋白成熟。这种切割的抑制极大地影响病毒的繁殖,因此代表了一个理想的药物目标,以控制感染。重要的是,没有检测到弗林逃逸变种,这表明病原体不能以任何方式取代这种蛋白酶。这里,我们设计了一种新型的SARS-CoV-2衍生的荧光底物来筛选市售和定制的小分子文库,以鉴定新的Furin抑制剂。我们发现,与市售的Pyr-RTKR-AMC底物相比,模拟Omicron变体的包膜糖蛋白的切割位点的肽底物(QTQTKSHRRAR-AMC)是筛选Furin活性的优越工具。使用此设置,我们确定了能够在体外调节Furin活性并适合干扰SARS-CoV-2成熟的有前途的新型化合物。特别是,我们表明3-(5-((5-溴噻吩-2-基)亚甲基)-4-氧代-4,5二氢噻唑-2-基)(3-氯-4-甲基苯基)氨基)丙酸(P3,IC50=35μM)可能代表了一种有吸引力的化学支架,可通过一种作用机制来开发更有效的药物。总的来说,研究了SARS-CoV-2衍生肽作为弗林蛋白酶抑制剂体外高通量筛选(HTS)的新底物,并通过创新的化学支架将化合物P3鉴定为有希望的命中。鉴于Furin在感染中的关键作用以及缺乏任何食品和药物管理局(FDA)批准的Furin抑制剂,P3代表有趣的抗病毒候选物。
    SARS-CoV-2 is the pathogen responsible for the most recent global pandemic, which has claimed hundreds of thousands of victims worldwide. Despite remarkable efforts to develop an effective vaccine, concerns have been raised about the actual protection against novel variants. Thus, researchers are eager to identify alternative strategies to fight against this pathogen. Like other opportunistic entities, a key step in the SARS-CoV-2 lifecycle is the maturation of the envelope glycoprotein at the RARR685↓ motif by the cellular enzyme Furin. Inhibition of this cleavage greatly affects viral propagation, thus representing an ideal drug target to contain infection. Importantly, no Furin-escape variants have ever been detected, suggesting that the pathogen cannot replace this protease by any means. Here, we designed a novel fluorogenic SARS-CoV-2-derived substrate to screen commercially available and custom-made libraries of small molecules for the identification of new Furin inhibitors. We found that a peptide substrate mimicking the cleavage site of the envelope glycoprotein of the Omicron variant (QTQTKSHRRAR-AMC) is a superior tool for screening Furin activity when compared to the commercially available Pyr-RTKR-AMC substrate. Using this setting, we identified promising novel compounds able to modulate Furin activity in vitro and suitable for interfering with SARS-CoV-2 maturation. In particular, we showed that 3-((5-((5-bromothiophen-2-yl)methylene)-4-oxo-4,5 dihydrothiazol-2-yl)(3-chloro-4-methylphenyl)amino)propanoic acid (P3, IC50 = 35 μM) may represent an attractive chemical scaffold for the development of more effective antiviral drugs via a mechanism of action that possibly implies the targeting of Furin secondary sites (exosites) rather than its canonical catalytic pocket. Overall, a SARS-CoV-2-derived peptide was investigated as a new substrate for in vitro high-throughput screening (HTS) of Furin inhibitors and allowed the identification of compound P3 as a promising hit with an innovative chemical scaffold. Given the key role of Furin in infection and the lack of any Food and Drug Administration (FDA)-approved Furin inhibitor, P3 represents an interesting antiviral candidate.
    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
    Activation-by-inhibition is a biochemical paradox seldom observed in exosite enzymes, wherein active site-bound inhibitors unexpectedly lead to enzyme activation. This intriguing phenomenon occurs at low, undersaturating substrate concentrations, posing a significant challenge in drug discovery, especially when targeting enzymes such as protein kinases, proteases, and other posttranslational modification enzymes. These enzymes often rely on accessory recognition sites known as exosites, which contribute to complex substrate binding mechanisms and unique kinetic behaviors. This study aims to provide a theoretical kinetic explanation for nonallosteric mechanism-based activation-by-inhibition, shedding light on the complexities of inhibiting exosite enzymes solely through active site targeting. Notably, the dual activator-inhibitor behavior of active site-bound inhibitors manifests in a nonmonotonic biphasic dose-response, emphasizing the importance of understanding the role of the inhibitor concentration at low substrate levels. Our findings underscore the potential widespread occurrence of activation by inhibition, a phenomenon that may have been overlooked in the past, and thus advocate for novel strategies in drug design that consider the impact of exosites on enzyme behavior to effectively target exosite 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
    这项全面的审查涉及人类健康和疾病中肽酶活性的复杂和多方面的调节,提供全面的调查,远远超出了活动地点的边界。我们的审查侧重于多种机制,并强调了exosites的重要作用,变构位点,以及参与酶原激活的过程。这些机制在塑造肽酶功能的复杂世界中起着核心作用,并且是开发创新药物和治疗干预措施的有希望的潜在目标。该综述还简要讨论了糖胺聚糖和非抑制性结合蛋白对酶活性的影响。了解它们的作用可能是制定治疗策略的关键因素。通过阐明控制肽酶活性的调节机制的复杂网络,这篇综述加深了我们对这一领域的理解,并为影响和调节肽酶活性的各种策略提供了路线图。
    This comprehensive review addresses the intricate and multifaceted regulation of peptidase activity in human health and disease, providing a comprehensive investigation that extends well beyond the boundaries of the active site. Our review focuses on multiple mechanisms and highlights the important role of exosites, allosteric sites, and processes involved in zymogen activation. These mechanisms play a central role in shaping the complex world of peptidase function and are promising potential targets for the development of innovative drugs and therapeutic interventions. The review also briefly discusses the influence of glycosaminoglycans and non-inhibitory binding proteins on enzyme activities. Understanding their role may be a crucial factor in the development of therapeutic strategies. By elucidating the intricate web of regulatory mechanisms that control peptidase activity, this review deepens our understanding in this field and provides a roadmap for various strategies to influence and modulate peptidase activity.
    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
    通过O连接的N-乙酰葡糖胺(O-GlcNAc)修饰核质蛋白是细胞生理学的重要调节因子。O-GlcNAc仅通过一种酶就安装在一千多种蛋白质上,O-GlcNAc转移酶(OGT)。因此,如何对OGT进行监管是一个值得关注的话题。为了深入了解这些问题,我们使用OGT从长度为15个氨基酸的约109个肽的无偏倚文库中进行噬菌体展示选择。经过几轮选择和深度突变平移,我们确定了一个高保真肽共有序列,[Y/F]-x-P-x-Y-x-[I/M/F],驱动肽与OGT结合。含有该序列的肽以高纳摩尔至低微摩尔范围与OGT结合,并以低微摩尔效力以非竞争性方式抑制OGT。与含有该基序的肽复合的OGT的X射线结构分析令人惊讶地揭示了与OGT活性位点近侧的外位点结合。该结构定义了驱动肽结合的详细分子基础,并解释了对序列基序内特定残基的需求。对人类蛋白质组的分析揭示了该基序在52种核和细胞质蛋白中。总的来说,这些数据提示了OGT的调节模式,通过该模式,多肽可以结合到该外切位点以通过其活性位点的空间闭塞引起OGT的变构抑制。我们期望这些见解将推动对细胞内OGT调节的更好理解,并能够开发新的化学工具来对OGT活性进行精细控制。
    The modification of nucleocytoplasmic proteins by O-linked N-acetylglucosamine (O-GlcNAc) is an important regulator of cell physiology. O-GlcNAc is installed on over a thousand proteins by just one enzyme, O-GlcNAc transferase (OGT). How OGT is regulated is therefore a topic of interest. To gain insight into these questions, we used OGT to perform phage display selection from an unbiased library of ~109 peptides of 15 amino acids in length. Following rounds of selection and deep mutational panning, we identified a high-fidelity peptide consensus sequence, [Y/F]-x-P-x-Y-x-[I/M/F], that drives peptide binding to OGT. Peptides containing this sequence bind to OGT in the high nanomolar to low micromolar range and inhibit OGT in a noncompetitive manner with low micromolar potencies. X-ray structural analyses of OGT in complex with a peptide containing this motif surprisingly revealed binding to an exosite proximal to the active site of OGT. This structure defines the detailed molecular basis driving peptide binding and explains the need for specific residues within the sequence motif. Analysis of the human proteome revealed this motif within 52 nuclear and cytoplasmic proteins. Collectively, these data suggest a mode of regulation of OGT by which polypeptides can bind to this exosite to cause allosteric inhibition of OGT through steric occlusion of its active site. We expect that these insights will drive improved understanding of the regulation of OGT within cells and enable the development of new chemical tools to exert fine control over OGT activity.
    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
    植物豆科植物对于处理种子储存蛋白至关重要,并且是植物程序性细胞死亡的关键调节剂。尽管最近对豆类的研究有所增加,对他们的活动调节知之甚少。在我们的研究中,我们使用下拉实验来鉴定AtCYT6是拟南芥中豆科蛋白酶同工型β(AtLEGβ)的天然抑制剂。生化分析表明,AtCYT6通过两个独立的胱抑素结构域抑制AtLEGβ和木瓜蛋白酶样半胱氨酸蛋白酶。N端结构域抑制木瓜蛋白酶样蛋白酶,而C端结构域抑制AtLEGβ。此外,我们发现AtCYT6与legumain以类似底物的方式相互作用,由其反应中心环中保守的天冬酰胺残基促进。通过带电的外部位相互作用额外稳定了复合物的形成,有助于pH依赖性抑制。AtLEGβ对AtCYT6的加工提示了对植物生理学有影响的上下文特异性调节机制。发展,和程序性细胞死亡。这些发现增强了我们对AtLEGβ调节及其更广泛的生理意义的理解。
    Plant legumains are crucial for processing seed storage proteins and are critical regulators of plant programmed cell death. Although research on legumains boosted recently, little is known about their activity regulation. In our study, we used pull-down experiments to identify AtCYT6 as a natural inhibitor of legumain isoform β (AtLEGβ) in Arabidopsis thaliana. Biochemical analysis revealed that AtCYT6 inhibits both AtLEGβ and papain-like cysteine proteases through two separate cystatin domains. The N-terminal domain inhibits papain-like proteases, while the C-terminal domain inhibits AtLEGβ. Furthermore, we showed that AtCYT6 interacts with legumain in a substrate-like manner, facilitated by a conserved asparagine residue in its reactive center loop. Complex formation was additionally stabilized by charged exosite interactions, contributing to pH-dependent inhibition. Processing of AtCYT6 by AtLEGβ suggests a context-specific regulatory mechanism with implications for plant physiology, development, and programmed cell death. These findings enhance our understanding of AtLEGβ regulation and its broader physiological significance.
    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
    具有血小板反应蛋白1型基序(ADAMTS1)的整合素样和金属蛋白酶是参与受精的蛋白酶,癌症,心血管发育,和胸动脉瘤.蛋白聚糖如versican和聚集蛋白聚糖已被确定为ADAMTS1底物,Adamts1在小鼠中的消融通常会导致versican积累;然而,先前的定性研究表明,ADAMTS1蛋白聚糖酶活性弱于其他家族成员,如ADAMTS4和ADAMTS5。这里,我们研究了ADAMTS1蛋白聚糖酶活性的功能决定因素。我们发现ADAMTS1versicanase活性比ADAMTS5低约1000倍,比ADAMTS4低50倍,相对于全长versican的动力学常数(kcat/Km)为3.6x103M-1s-1。对结构域缺失变体的研究鉴定出间隔区和富含半胱氨酸的结构域是ADAMTS1杂色酶活性的主要决定因素。此外,我们证实这些C端结构域参与聚集蛋白聚糖和双糖蛋白酶解,一种富含亮氨酸的小蛋白多糖。谷氨酰胺扫描诱变间隔结构域环上暴露的带正电荷的残基,并用ADAMTS4进行环取代,鉴定了β3-β4中的底物结合残基(外位点)簇(R756Q/R759Q/R762Q),β9-β10(残基828-835),和β6-β7(K795Q)环。这项研究为理解ADAMTS1及其蛋白聚糖底物之间的相互作用提供了机理基础,并为开发ADAMTS1蛋白聚糖酶活性的选择性外位点调节剂铺平了道路。
    A disintegrin-like and metalloproteinase with thrombospondin type 1 motifs (ADAMTS1) is a protease involved in fertilization, cancer, cardiovascular development, and thoracic aneurysms. Proteoglycans such as versican and aggrecan have been identified as ADAMTS1 substrates, and Adamts1 ablation in mice typically results in versican accumulation; however, previous qualitative studies have suggested that ADAMTS1 proteoglycanase activity is weaker than that of other family members such as ADAMTS4 and ADAMTS5. Here, we investigated the functional determinants of ADAMTS1 proteoglycanase activity. We found that ADAMTS1 versicanase activity is approximately 1000-fold lower than ADAMTS5 and 50-fold lower than ADAMTS4 with a kinetic constant (kcat/Km) of 3.6 × 103 M-1 s-1 against full-length versican. Studies on domain-deletion variants identified the spacer and cysteine-rich domains as major determinants of ADAMTS1 versicanase activity. Additionally, we confirmed that these C-terminal domains are involved in the proteolysis of aggrecan as well as biglycan, a small leucine-rich proteoglycan. Glutamine scanning mutagenesis of exposed positively charged residues on the spacer domain loops and loop substitution with ADAMTS4 identified clusters of substrate-binding residues (exosites) in β3-β4 (R756Q/R759Q/R762Q), β9-β10 (residues 828-835), and β6-β7 (K795Q) loops. This study provides a mechanistic foundation for understanding the interactions between ADAMTS1 and its proteoglycan substrates and paves the way for development of selective exosite modulators of ADAMTS1 proteoglycanase activity.
    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
    链球菌C5a肽酶(ScpA)特异性灭活人补体因子hC5a,一种有效的过敏毒素最近被确定为治疗COVID-19感染的治疗靶标。设计ScpA以增强其作为治疗剂的潜力将需要详细检查其高选择性活性的基础。ScpA和相关枯草杆菌酶的新兴观点是,其底物的选择是动态的两步过程,涉及活性位点周围结构域和底物C-ter的灵活性。ScpA-hC5a相互作用的表面等离子体共振(SPR)分析表明,底物的高亲和力结合是由酶的Fn2结构域上的外位点与庞大的N-ter裂解产物(PN,C5a[1]的核心残基1-67)。在Fn2exosite中引入D783A突变,位于离催化丝氨酸大约50埃的位置,显示出显着降低酶的底物结合亲和力和kcat。对D783A突变体(ScpAD783A)进行了X射线晶体学研究,以更好地解释该突变对ScpA特异性和活性的影响。在这里,我们提供了ScpAD783A的1.9µAX射线衍射数据和结构的分子置换溶液。原始衍射图像和坐标均已在公共数据库中提供。Jain等人报道的ScpAD783A相关SPR和酶动力学分析的其他细节。[2].
    The Streptococcal C5a peptidase (ScpA) specifically inactivates the human complement factor hC5a, a potent anaphylatoxin recently identified as a therapeutic target for treatment of COVID-19 infections. Engineering of ScpA to enhance its potential as a therapeutic will require detailed examination of the basis for its highly selective activity. The emerging view of ScpA and related subtilases is that selection of their substrates is a dynamic two-step process involving flexibility in the domains around the active site and in the C-ter of the substrate. Surface plasmon resonance (SPR) analyses of the ScpA-hC5a interaction have shown that high affinity binding of the substrate is driven by electrostatic interactions between an exosite on the Fn2 domain of the enzyme and the bulky N-ter cleavage product (PN, \'core\' residues 1-67) of C5a [1]. Introduction of a D783A mutation in the Fn2 exosite, located approximately 50 Å from the catalytic serine, was shown to significantly reduce substrate binding affinity and k cat of the enzyme. X-ray crystallographic studies on the D783A mutant (ScpAD783A) were undertaken to better interpret the impact of this mutation on the specificity and activity of ScpA. Here we present the 1.9 Å X-ray diffraction data for ScpAD783A and the molecular replacement solution for the structure. Both raw diffraction images and coordinates have been made available on public databases. Additional details on the related SPR and enzyme kinetics analyses on ScpAD783A reported in Jain et al. [2].
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号