HDX-MS

HDX - MS
  • 文章类型: Journal Article
    激活的FXII(FXIIa)是血浆接触系统的主要引发剂,并且可以激活促凝血和促炎途径。其活性在遗传性血管性水肿(HAE)的病理生理学中很重要。这里,我们描述了与garadacimab的Fab片段复合的FXIIa(βFXIIa)的β链的高分辨率低温电子显微镜(cryo-EM)结构。Garadacimab通过异常长的CDR-H3与βFXIIa结合,该CDR-H3以非规范方式插入S1口袋。这种结构机制可能是抑制HAE中活化的FXIIa蛋白水解活性的主要贡献者。GaradacimabFab-βFXIIa结构还揭示了garadacimab与活化的FXIIa的高亲和力结合的关键决定因素。用其他真正的FXIIa抑制剂进行结构分析,如苄脒和C1-INH,揭示了garadacimab抑制βFXIIa的惊人相似机制。总之,garadacimabFab-βFXIIa结构提供了对其作用机制的重要见解,并描述了主要和辅助互补位/表位。
    Activated FXII (FXIIa) is the principal initiator of the plasma contact system and can activate both procoagulant and proinflammatory pathways. Its activity is important in the pathophysiology of hereditary angioedema (HAE). Here, we describe a high-resolution cryoelectron microscopy (cryo-EM) structure of the beta-chain from FXIIa (βFXIIa) complexed with the Fab fragment of garadacimab. Garadacimab binds to βFXIIa through an unusually long CDR-H3 that inserts into the S1 pocket in a non-canonical way. This structural mechanism is likely the primary contributor to the inhibition of activated FXIIa proteolytic activity in HAE. Garadacimab Fab-βFXIIa structure also reveals critical determinants of high-affinity binding of garadacimab to activated FXIIa. Structural analysis with other bona fide FXIIa inhibitors, such as benzamidine and C1-INH, reveals a surprisingly similar mechanism of βFXIIa inhibition by garadacimab. In summary, the garadacimab Fab-βFXIIa structure provides crucial insights into its mechanism of action and delineates primary and auxiliary paratopes/epitopes.
    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
    Bak是一种成孔Bcl2蛋白,可诱导线粒体外膜细胞凋亡,可以通过Bak寡聚化进行或被抗凋亡Bcl2蛋白抑制,例如BclxL。BclxL在抑制Bak孔形成方面非常有效,但是这种首选互动的机制基础仍然是神秘的。这里,我们将Bakα1鉴定为BclxL的第二个结合位点,并显示其与BclxL的Bcl2同源性(BH)3结合槽特异性相互作用。BclxL和BKα1之间的亲和力比Bak-BH3弱,这表明BKα1在成孔轨迹的早期暴露,暂时捕获BclxL,随后过渡到近端BH3位点。其中与BclxL的初始瞬时相互作用被调节的Bak变体显示对BclxL抑制的显著改变的响应。这项工作有助于更好地理解Bcl2蛋白家族不同参与者之间的微调相互作用。
    Bak is a pore-forming Bcl2 protein that induces apoptosis at the outer mitochondrial membrane, which can either proceed via Bak oligomerization or be inhibited by anti-apoptotic Bcl2 proteins, such as BclxL. BclxL is very efficient in inhibiting Bak pore formation, but the mechanistic basis of this preferred interaction has remained enigmatic. Here, we identify Bakα1 as a second binding site for BclxL and show that it specifically interacts with the Bcl2-homology (BH)3 binding groove of BclxL. The affinity between BclxL and Bakα1 is weaker than with Bak-BH3, suggesting that Bakα1, being exposed early in the pore-forming trajectory, transiently captures BclxL, which subsequently transitions to the proximal BH3 site. Bak variants where the initial transient interaction with BclxL is modulated show a markedly altered response to BclxL inhibition. This work contributes to a better mechanistic understanding of the fine-tuned interactions between different players of the Bcl2 protein family.
    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
    内皮素受体B型(ETB)表现出与包括Gs在内的各种异源三聚体G蛋白亚型的混杂偶联,Gi/o,Gq/11和G12/13。最近的荧光和结构研究提出了有关这些G蛋白亚型的偶联效率和决定因素的问题。在这里,通过采用综合方法,结合氢/氘交换质谱和基于NanoLuc二进制技术的细胞系统,我们研究了Gs的构象变化,Gi,和由ETB激活触发的Gq。ETB与Gi和Gq耦合,但不与Gs耦合。我们强调了特定区域的关键作用,在ETB-Gi1或ETB-Gq偶联中包括Gα的C末端和ETB的胞内环2(ICL2)。虽然Gα的C末端对于ETB-Gi1和ETB-Gq偶联是必需的,ETBICL2影响Gq耦合但不影响Gi1耦合。我们的结果表明,ETB与Gs的耦合效率不同,Gi1和Gq,伴随着ETB诱导的激活后G蛋白的明显构象变化。
    The endothelin receptor type B (ETB) exhibits promiscuous coupling with various heterotrimeric G protein subtypes including Gs, Gi/o, Gq/11, and G12/13. Recent fluorescence and structural studies have raised questions regarding the coupling efficiencies and determinants of these G protein subtypes. Herein, by utilizing an integrative approach, combining hydrogen/deuterium exchange mass spectrometry and NanoLuc Binary Technology-based cellular systems, we investigated conformational changes of Gs, Gi, and Gq triggered by ETB activation. ETB coupled to Gi and Gq but not with Gs. We underscored the critical roles of specific regions, including the C terminus of Gα and intracellular loop 2 (ICL2) of ETB in ETB-Gi1 or ETB-Gq coupling. Although The C terminus of Gα is essential for ETB-Gi1 and ETB-Gq coupling, ETB ICL2 influences Gq-coupling but not Gi1-coupling. Our results suggest a differential coupling efficiency of ETB with Gs, Gi1, and Gq, accompanied by distinct conformational changes in G proteins upon ETB-induced activation.
    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
    非典型趋化因子受体3(ACKR3)属于G蛋白偶联受体家族,但不通过G蛋白发出信号。控制ACKR3活化的功能选择性和构象动力学的结构特性知之甚少。这里,我们结合了氢/氘交换质谱,定点诱变,和分子动力学模拟,以检查不同功效的ACKR3配体的结合模式和作用机理。我们的结果表明,ACKR3的激活或抑制受螺旋6,胞内环2和螺旋7的细胞内构象变化控制,而DRY基序在这两个过程中都受到保护。此外,我们确定了β-抑制蛋白1结合时ACKR3的结合位点和变构调节。总之,这项研究强调了小配体的结构-功能关系,β-抑制蛋白1的结合模式,激活动力学,以及ACKR3中的非典型动态特征可能导致其无法激活G蛋白。
    Atypical Chemokine Receptor 3 (ACKR3) belongs to the G protein-coupled receptor family but it does not signal through G proteins. The structural properties that govern the functional selectivity and the conformational dynamics of ACKR3 activation are poorly understood. Here, we combined hydrogen/deuterium exchange mass spectrometry, site-directed mutagenesis, and molecular dynamics simulations to examine the binding mode and mechanism of action of ACKR3 ligands of different efficacies. Our results show that activation or inhibition of ACKR3 is governed by intracellular conformational changes of helix 6, intracellular loop 2, and helix 7, while the DRY motif becomes protected during both processes. Moreover, we identified the binding sites and the allosteric modulation of ACKR3 upon β-arrestin 1 binding. In summary, this study highlights the structure-function relationship of small ligands, the binding mode of β-arrestin 1, the activation dynamics, and the atypical dynamic features in ACKR3 that may contribute to its inability to activate G proteins.
    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
    治疗性蛋白质的可逆自缔合(RSA)在高浓度制剂的开发中提出了主要挑战。尤其是那些打算皮下给药的。因此,了解自缔合机制对于设计和选择具有可接受的可开发性的候选人以进行临床试验至关重要。实验和计算机建模的结合提供了阐明自关联界面的强大工具。已经在不同的溶液条件下广泛研究了单克隆抗体的RSA,并且已经显示涉及抗原结合片段和可结晶片段两者的相互作用。新型模态,如双特异性抗体,抗原结合片段,单链可变片段,和双抗体构成了一类快速增长的基于抗体的治疗剂,与单克隆抗体相比具有独特的生理化学性质。在这项研究中,双抗体-白细胞介素22融合蛋白(FP-1)的RSA界面使用氢-氘交换耦合质谱(HDX-MS)结合计算机模拟研究.一起来看,结果表明,复杂的溶液行为是FP-1自缔合的基础,其界面可归因于双抗体可变轻链中的特定片段。这些发现还表明,HDX-MS与计算机建模的组合是指导新型生物治疗方式的设计和候选选择的强大工具。
    Reversible self-association (RSA) of therapeutic proteins presents major challenges in the development of high-concentration formulations, especially those intended for subcutaneous administration. Understanding self-association mechanisms is therefore critical to the design and selection of candidates with acceptable developability to advance to clinical trials. The combination of experiments and in silico modeling presents a powerful tool to elucidate the interface of self-association. RSA of monoclonal antibodies has been studied extensively under different solution conditions and have been shown to involve interactions for both the antigen-binding fragment and the crystallizable fragment. Novel modalities such as bispecific antibodies, antigen-binding fragments, single-chain-variable fragments, and diabodies constitute a fast-growing class of antibody-based therapeutics that have unique physiochemical properties compared to monoclonal antibodies. In this study, the RSA interface of a diabody-interleukin 22 fusion protein (FP-1) was studied using hydrogen-deuterium exchange coupled with mass spectrometry (HDX-MS) in combination with in silico modeling. Taken together, the results show that a complex solution behavior underlies the self-association of FP-1 and that the interface thereof can be attributed to a specific segment in the variable light chain of the diabody. These findings also demonstrate that the combination of HDX-MS with in silico modeling is a powerful tool to guide the design and candidate selection of novel biotherapeutic modalities.
    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
    抑制蛋白与磷酸化G蛋白偶联受体(GPCRs)相互作用,并调节GPCRs的同源脱敏和内化。抑制蛋白中的门环是稳定基础状态和与磷酸化受体相互作用的关键区域。我们使用β-抑制蛋白1和2型血管加压素受体(V2Rpp)的磷酸化C尾肽作为模型系统,研究了门环(K292,K294和H295)中特定残基的作用。我们测量了V2Rpp的结合亲和力并分析了β-抑制蛋白-1的构象动力学。我们的结果表明,K294在与V2Rpp的相互作用中起关键作用,而不影响V2Rpp结合状态的整体构象。残基K292和H295有助于极性核心在基础状态下的稳定性,并在V2Rpp结合状态下形成指环的特定构象。
    Arrestins interact with phosphorylated G protein-coupled receptors (GPCRs) and regulate the homologous desensitization and internalization of GPCRs. The gate loop in arrestins is a critical region for both stabilization of the basal state and interaction with phosphorylated receptors. We investigated the roles of specific residues in the gate loop (K292, K294, and H295) using β-arrestin-1 and phosphorylated C-tail peptide of vasopressin receptor type 2 (V2Rpp) as a model system. We measured the binding affinity of V2Rpp and analyzed conformational dynamics of β-arrestin-1. Our results suggest that K294 plays a critical role in the interaction with V2Rpp without influencing the overall conformation of the V2Rpp-bound state. The residues K292 and H295 contribute to the stability of the polar core in the basal state and form a specific conformation of the finger loop in the V2Rpp-bound state.
    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
    补体系统通过促进调理吞噬作用和细菌溶解而充当抵抗入侵病原体的第一道防线。补体的抗体依赖性激活通过经典途径发生,并且依赖于C1复合物的补体蛋白酶的启动活性,C1r和C1s。莱姆病的病原体,伯氏疏螺旋体,表达OspEF相关蛋白家族的两个旁系外表面脂蛋白,ElpB和ElpQ,作为经典途径激活的特异性抑制剂。我们先前已经证明ElpB和ElpQ以高亲和力直接结合C1r和C1s,并特异性抑制C1s对C2和C4的切割。为了进一步了解这些新型蛋白酶抑制剂的功能,我们使用ElpQ和全长激活的C1s作为Elp/蛋白酶相互作用的模型进行了一系列氢-氘交换质谱(HDX-MS)实验。未结合的ElpQ和ElpQ/C1s复合物之间的HDX-MS图谱的比较揭示了ElpQ上推定的C1s结合位点。HDX-MS引导,产生定点ElpQ突变体,并使用表面等离子体共振测试与C1r和C1s的直接结合。ElpQ的C端区域内的几个残基被鉴定为对蛋白酶结合重要,包括ElpQ-和ElpB-介导的补体抑制所需的单个保守酪氨酸残基。总的来说,我们的研究确定了Elp蛋白识别经典途径蛋白酶的关键分子决定因素。这项研究提高了我们对Elp蛋白所采用的独特补体抑制机制的理解,Elp蛋白是莱姆病螺旋体中复杂的补体逃避系统的一部分。
    The complement system serves as the first line of defense against invading pathogens by promoting opsonophagocytosis and bacteriolysis. Antibody-dependent activation of complement occurs through the classical pathway and relies on the activity of initiating complement proteases of the C1 complex, C1r and C1s. The causative agent of Lyme disease, Borrelia burgdorferi, expresses two paralogous outer surface lipoproteins of the OspEF-related protein family, ElpB and ElpQ, that act as specific inhibitors of classical pathway activation. We have previously shown that ElpB and ElpQ bind directly to C1r and C1s with high affinity and specifically inhibit C2 and C4 cleavage by C1s. To further understand how these novel protease inhibitors function, we carried out a series of hydrogen-deuterium exchange mass spectrometry (HDX-MS) experiments using ElpQ and full-length activated C1s as a model of Elp-protease interaction. Comparison of HDX-MS profiles between unbound ElpQ and the ElpQ/C1s complex revealed a putative C1s-binding site on ElpQ. HDX-MS-guided, site-directed ElpQ mutants were generated and tested for direct binding to C1r and C1s using surface plasmon resonance. Several residues within the C-terminal region of ElpQ were identified as important for protease binding, including a single conserved tyrosine residue that was required for ElpQ- and ElpB-mediated complement inhibition. Collectively, our study identifies key molecular determinants for classical pathway protease recognition by Elp proteins. This investigation improves our understanding of the unique complement inhibitory mechanism employed by Elp proteins which serve as part of a sophisticated complement evasion system present in Lyme disease spirochetes.
    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
    Brazzein(Brz)是一种由54个氨基酸组成的甜味蛋白质,被认为是潜在的糖替代品。目前获得Brazzein的方法很复杂,关于其热稳定性的信息有限。在这项研究中,我们成功地表达了重组Brazzein,达到15.2μg/mL的甜度阈值。随后,我们在80、90、95和100°C的温度下进行了2小时的热处理,以研究蛋白质的结构变化。此外,我们使用氢-氘交换耦合质谱(HDX-MS)来分析加热对蛋白质结构-甜度关系的影响。我们的结果表明,热灭活过程主要影响Brazzein的残基6-14和36-45,特别是关键残基Tyr8,Tyr11,Ser14,Glu36和Arg43,它们与甜味密切相关。这些发现对于提高Brazzein的热稳定性具有重要意义。
    Brazzein (Brz) is a sweet-tasting protein composed of 54 amino acids and is considered as a potential sugar substitute. The current methods for obtaining brazzein are complicated, and limited information is available regarding its thermal stability. In this study, we successfully expressed recombinant brazzein, achieving a sweetness threshold of 15.2 μg/mL. Subsequently, we conducted heat treatments at temperatures of 80, 90, 95, and 100 °C for a duration of 2 h to investigate the structural changes in the protein. Furthermore, we employed hydrogen-deuterium exchange coupled to mass spectrometry (HDX-MS) to analyze the effect of heating on the protein structure-sweetness relationships. Our results indicated that the thermal inactivation process primarily affects residues 6-14 and 36-45 of brazzein, especially key residues Tyr8, Tyr11, Ser14, Glu36, and Arg43, which are closely associated with its sweetness. These findings have significant implications for improving the thermal stability of brazzein.
    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
    传感器效应蛋白整合来自不同刺激的信息,并将其转化为细胞反应。一些感官领域,像红光反应性细菌色素,显示出调节各种效应器的显着模块化。一个效应子结构域是GGDEF二鸟苷酸环化酶,其催化细菌第二信使环-二聚体-鸟苷单磷酸的形成。虽然已经描述了不同植物色素的关键信号整合元素,对远距离信号处理和通信的广义理解,植物色素二鸟苷酸环化酶中大约100埃,不见了。在这里,我们表明动力学驱动的变构是在分子水平上理解信号整合的关键。我们产生了稳定在其远红色吸收Pfr状态的蛋白质变体,并通过使用氢-氘交换与质谱联用的构象动力学分析证明,单个氨基酸替换伴随着整个蛋白质中功能元件的动力学改变。我们表明构象动力学与这些变体的酶活性相关,还解释了非光致变色变体的活性增加。此外,我们证明了混合的Pfr/中间态二聚体的功能重要性,该二聚体使用了一种快速恢复的变体,该变体仍然能够通过红光实现酶刺激的野生型样倍数变化.这支持了植物色素中单一质子活化的功能作用,可能与许多植物色素系统观察到的非规范混合Pfr/中间状态光谱相关的属性。我们预计我们的结果将刺激对基于不同细菌色素的传感器效应物的动力学驱动的变构调节方向的研究。这最终将影响用于创建用于丰富光遗传学工具箱的新型传感器效应系统的设计策略。
    Sensor-effector proteins integrate information from different stimuli and transform this into cellular responses. Some sensory domains, like red-light responsive bacteriophytochromes, show remarkable modularity regulating a variety of effectors. One effector domain is the GGDEF diguanylate cyclase catalyzing the formation of the bacterial second messenger cyclic-dimeric-guanosine monophosphate. While critical signal integration elements have been described for different phytochromes, a generalized understanding of signal processing and communication over large distances, roughly 100 Å in phytochrome diguanylate cyclases, is missing. Here we show that dynamics-driven allostery is key to understanding signal integration on a molecular level. We generated protein variants stabilized in their far-red-absorbing Pfr state and demonstrated by analysis of conformational dynamics using hydrogen-deuterium exchange coupled to mass spectrometry that single amino acid replacements are accompanied by altered dynamics of functional elements throughout the protein. We show that the conformational dynamics correlate with the enzymatic activity of these variants, explaining also the increased activity of a non-photochromic variant. In addition, we demonstrate the functional importance of mixed Pfr/intermediate state dimers using a fast-reverting variant that still enables wild-type-like fold-changes of enzymatic stimulation by red light. This supports the functional role of single protomer activation in phytochromes, a property that might correlate with the non-canonical mixed Pfr/intermediate-state spectra observed for many phytochrome systems. We anticipate our results to stimulate research in the direction of dynamics-driven allosteric regulation of different bacteriophytochrome-based sensor-effectors. This will eventually impact design strategies for the creation of novel sensor-effector systems for enriching the optogenetic toolbox.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    人异柠檬酸脱氢酶1(IDH1)的突变通过用产生代谢产物的新形态活性代替其常规活性来驱动多种癌症中的肿瘤形成。对肿瘤驱动IDH1突变体之间的机制差异了解甚少。我们先前报道,R132Q突变体独特地保留了常规活性,同时催化了强大的代谢产物产生,允许在单个活性位点内比较这些反应机制的机会。这里,我们采用静态和动态结构方法,发现,与R132H相比,R132Q活性位点采用了为催化引发的构象,具有优化的底物结合和氢化物转移,以驱动R132H上改善的常规和新形态活性。这种活性位点重塑揭示了对选择性突变IDH1治疗性抑制剂的抗性的可能机制。这项工作增强了我们对基本IDH1机制的理解,同时精确定位了改善抑制剂选择性的区域。
    Mutations in human isocitrate dehydrogenase 1 (IDH1) drive tumor formation in a variety of cancers by replacing its conventional activity with a neomorphic activity that generates an oncometabolite. Little is understood of the mechanistic differences among tumor-driving IDH1 mutants. We previously reported that the R132Q mutant uniquely preserves conventional activity while catalyzing robust oncometabolite production, allowing an opportunity to compare these reaction mechanisms within a single active site. Here, we employed static and dynamic structural methods and found that, compared to R132H, the R132Q active site adopted a conformation primed for catalysis with optimized substrate binding and hydride transfer to drive improved conventional and neomorphic activity over R132H. This active site remodeling revealed a possible mechanism of resistance to selective mutant IDH1 therapeutic inhibitors. This work enhances our understanding of fundamental IDH1 mechanisms while pinpointing regions for improving inhibitor selectivity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号