Binding, Competitive

绑定,Competitive
  • 文章类型: Journal Article
    细胞中的微管组织依赖于靶向机制。细胞质连接蛋白(CLIP)和CLIP相关蛋白(CLASPs)是微管组织的关键调节因子。然而,潜在的机制仍然难以捉摸。这里,我们揭示了CLASP2的C端结构域与几种CLASP结合蛋白中发现的共同基序相互作用。这种相互作用驱动CLASP2动态定位到不同的细胞区室,CLASP2积累在细胞皮质或微管加端的蛋白质凝聚物中。这些缩合物通过CLASP2介导的竞争性结合彼此物理接触,确定皮质微管靶向。CLASP2的磷酸化调节冷凝物-冷凝物相互作用的动力学,并时空导航微管生长。此外,我们确定了以CLASP2依赖性方式参与缩合物接触的其他CLASP相互作用蛋白,揭示了控制微管靶向的一般机制。我们的发现不仅揭示了调节微管组织的可调多相系统,而且还提供了在中尺度水平上复杂的蛋白质-蛋白质相互作用的一般机制见解。
    Microtubule organization in cells relies on targeting mechanisms. Cytoplasmic linker proteins (CLIPs) and CLIP-associated proteins (CLASPs) are key regulators of microtubule organization, yet the underlying mechanisms remain elusive. Here, we reveal that the C-terminal domain of CLASP2 interacts with a common motif found in several CLASP-binding proteins. This interaction drives the dynamic localization of CLASP2 to distinct cellular compartments, where CLASP2 accumulates in protein condensates at the cell cortex or the microtubule plus end. These condensates physically contact each other via CLASP2-mediated competitive binding, determining cortical microtubule targeting. The phosphorylation of CLASP2 modulates the dynamics of the condensate-condensate interaction and spatiotemporally navigates microtubule growth. Moreover, we identify additional CLASP-interacting proteins that are involved in condensate contacts in a CLASP2-dependent manner, uncovering a general mechanism governing microtubule targeting. Our findings not only unveil a tunable multiphase system regulating microtubule organization, but also offer general mechanistic insights into intricate protein-protein interactions at the mesoscale level.
    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
    荧光配体已被证明是研究活细胞中G蛋白偶联受体的有力工具。在这里,我们已经表征了一种新的荧光配体PSB603-BY630,它对人腺苷A2B受体(A2BR)具有高选择性。A2BR似乎在调节肿瘤微环境中的免疫应答中起重要作用。在这里,我们使用PSB603-BY630来监测源自CD14+人单核细胞的M1-和M2-样巨噬细胞中与A2BR的特异性结合。PSB603-BY630以高亲和力(18.3nM)结合在HEK293G细胞中稳定表达的纳米荧光素酶标记的A2BR。配体对A2BR表现出非常高的选择性,在NLuc-A2AR检测到的特异性结合可忽略不计,NLuc-A1R,或NLuc-A3R受体,浓度高达500nM。竞争结合研究显示了A2BR与A2BR选择性配体PSB603和MRS-1706的预期药理学,证明了对50nMPSB603-BY630与A2BR的特异性结合的有效抑制。使用Glosensor监测Gs偶联的环状AMP反应对HEK293G细胞进行的功能研究表明,PSB603-BY630对BAY60-6583的激动剂反应具有负变构规律。此外,流式细胞术分析证实PSB603-BY630可用于选择性标记在人巨噬细胞上表达的内源性A2BRs。该配体应该是对不同腺苷受体亚型具有选择性的荧光配体库的重要补充。并将能够研究A2BRs在肿瘤微环境中对免疫细胞的作用。
    Fluorescent ligands have proved to be powerful tools in the study of G protein-coupled receptors in living cells. Here we have characterized a new fluorescent ligand PSB603-BY630 that has high selectivity for the human adenosine A2B receptor (A2BR). The A2BR appears to play an important role in regulating immune responses in the tumor microenvironment. Here we have used PSB603-BY630 to monitor specific binding to A2BRs in M1- and M2-like macrophages derived from CD14+ human monocytes. PSB603-BY630 bound with high affinity (18.3 nM) to nanoluciferase-tagged A2BRs stably expressed in HEK293G cells. The ligand exhibited very high selectivity for the A2BR with negligible specific-binding detected at NLuc-A2AR, NLuc-A1R, or NLuc-A3R receptors at concentrations up to 500 nM. Competition binding studies showed the expected pharmacology at A2BR with the A2BR-selective ligands PSB603 and MRS-1706 demonstrating potent inhibition of the specific binding of 50 nM PSB603-BY630 to A2BR. Functional studies in HEK293G cells using Glosensor to monitor Gs-coupled cyclic AMP responses indicated that PSB603-BY630 acted as a negative allosteric regular of the agonist responses to BAY 60-6583. Furthermore, flow cytometry analysis confirmed that PSB603-BY630 could be used to selectively label endogenous A2BRs expressed on human macrophages. This ligand should be an important addition to the library of fluorescent ligands which are selective for the different adenosine receptor subtypes, and will enable study of the role of A2BRs on immune cells in the tumor microenvironment.
    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
    我们得出具有完全和部分竞争性抑制的Michaelis-Menten酶动力学方案的速度-底物-抑制剂空间的稳态前和稳态后状态的近似表达式。我们对当前可用的标准准稳态近似(sQSSA)的改进似乎在宽范围的酶与底物以及酶与抑制剂的浓度比上有效。Further,我们表明,在某些条件下,酶-抑制剂-底物系统可以相对于酶-底物和酶-抑制剂复合物表现出暂时分离的两种不同稳态。我们定义比率fS=vmax/(KMSe0)和fI=umax/(KMIe0)作为底物和抑制剂催化转化为各自产物的加速因子。这里KMS和KMI是分别与底物和抑制剂与酶的结合相关的Michaelis-Menten参数,vmax和umax是各自的最大反应速度,e0、s0和i0是总酶,底物和抑制剂水平。当(fS/fI)<1时,则酶-底物复合物将显示多个稳态,并且仅在酶-抑制剂复合物耗尽之后才达到完全稳态。当(fS/fI)>1时,则酶-抑制剂复合物将显示多个稳态,并且仅在酶-底物复合物耗尽之后才达到完全稳态。这种多稳态行为,尤其是当(fS/fI)辛1时,是使用sQSSA估计完全和部分竞争抑制方案的各种动力学参数时出现大量误差的根本原因。值得注意的是,我们表明,与目前可用的sQSSA表达式相比,我们对酶-底物-抑制剂空间上的反应速度的改进表达式可以更明显地控制此误差。
    We derive approximate expressions for pre- and post-steady state regimes of the velocity-substrate-inhibitor spaces of the Michaelis-Menten enzyme kinetic scheme with fully and partial competitive inhibition. Our refinement over the currently available standard quasi steady state approximation (sQSSA) seems to be valid over wide range of enzyme to substrate and enzyme to inhibitor concentration ratios. Further, we show that the enzyme-inhibitor-substrate system can exhibit temporally well-separated two different steady states with respect to both enzyme-substrate and enzyme-inhibitor complexes under certain conditions. We define the ratios fS = vmax/(KMS + e0) and fI = umax/(KMI + e0) as the acceleration factors with respect to the catalytic conversion of substrate and inhibitor into their respective products. Here KMS and KMI are the Michaelis-Menten parameters associated respectively with the binding of substrate and inhibitor with the enzyme, vmax and umax are the respective maximum reaction velocities and e0, s0, and i0 are total enzyme, substrate and inhibitor levels. When (fS/fI) < 1, then enzyme-substrate complex will show multiple steady states and it reaches the full-fledged steady state only after the depletion of enzyme-inhibitor complex. When (fS/fI) > 1, then the enzyme-inhibitor complex will show multiple steady states and it reaches the full-fledged steady state only after the depletion of enzyme-substrate complex. This multi steady-state behavior especially when (fS/fI) ≠ 1 is the root cause of large amount of error in the estimation of various kinetic parameters of fully and partial competitive inhibition schemes using sQSSA. Remarkably, we show that our refined expressions for the reaction velocities over enzyme-substrate-inhibitor space can control this error more significantly than the currently available sQSSA expressions.
    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
    委内瑞拉马脑炎病毒(VEEV)是一种高毒力病原体,其衣壳蛋白的核定位信号(NLS)序列与宿主importin-α转运蛋白结合并阻断核输入。我们研究了两个小配体的分子机制,称为I1和I2,干扰VEEV的NLS肽与importin-α蛋白的结合。为此,我们进行了全原子复制交换分子动力学模拟,探索了VEEV核心NLS肽和I1或I2配体与importin-α主要NLS结合位点的竞争性结合。作为参考,我们用之前的模拟,检查了coreNLS肽或抑制剂与importin-α的非竞争性结合。我们发现这两种抑制剂都完全消除了核心NLS肽的天然结合,迫使其在importin-α主要NLS结合位点内采用多种非天然松散结合的姿势。两种抑制剂主要通过掩蔽其氨基酸而不是与其竞争结合importin-α来使天然coreNLS结合不稳定。因为I2与I1相反,结合位点外定位在主要NLS结合位点的边缘,与I1相比,它抑制较少的coreNLS天然结合相互作用。结构分析得到了在有或没有来自抑制剂的竞争的情况下与importin-α结合的核NLS肽的自由能的计算的支持。具体来说,这两种抑制剂都降低了核心NLS结合的自由能增益,I1造成的损失明显大于I2。为了测试我们的模拟,我们进行了AlphaScreen实验,测量两种抑制剂的IC50值.与计算机模拟结果一致,发现I1的IC50值低于I2的IC50值。我们假设I1和I2配体的抑制作用可能对VEEV衣壳蛋白的NLS具有特异性。
    Venezuelan equine encephalitis virus (VEEV) is a highly virulent pathogen whose nuclear localization signal (NLS) sequence from capsid protein binds to the host importin-α transport protein and blocks nuclear import. We studied the molecular mechanisms by which two small ligands, termed I1 and I2, interfere with the binding of VEEV\'s NLS peptide to importin-α protein. To this end, we performed all-atom replica exchange molecular dynamics simulations probing the competitive binding of the VEEV coreNLS peptide and I1 or I2 ligand to the importin-α major NLS binding site. As a reference, we used our previous simulations, which examined noncompetitive binding of the coreNLS peptide or the inhibitors to importin-α. We found that both inhibitors completely abrogate the native binding of the coreNLS peptide, forcing it to adopt a manifold of nonnative loosely bound poses within the importin-α major NLS binding site. Both inhibitors primarily destabilize the native coreNLS binding by masking its amino acids rather than competing with it for binding to importin-α. Because I2, in contrast to I1, binds off-site localizing on the edge of the major NLS binding site, it inhibits fewer coreNLS native binding interactions than I1. Structural analysis is supported by computations of the free energies of the coreNLS peptide binding to importin-α with or without competition from the inhibitors. Specifically, both inhibitors reduce the free energy gain from coreNLS binding, with I1 causing significantly larger loss than I2. To test our simulations, we performed AlphaScreen experiments measuring IC50 values for both inhibitors. Consistent with in silico results, the IC50 value for I1 was found to be lower than that for I2. We hypothesize that the inhibitory action of I1 and I2 ligands might be specific to the NLS from VEEV\'s capsid protein.
    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
    蓖麻毒素是已知的毒性最强的物质之一,是一种B型生物试剂。由大肠杆菌(STEC)和痢疾志贺氏菌产生的志贺毒素(Stxs)是食源性病原体。没有针对蓖麻毒素或STEC的有效疗法,并且迫切需要抑制剂。蓖麻毒素A亚基(RTA)和Stx2a的A1亚基(Stx2A1)与核糖体P-茎蛋白的C末端结构域(CTD)结合,以对sarcin/蓖麻毒素环进行纯化。尚未探索调节毒素-核糖体相互作用作为抑制策略。因此,检测毒素-核糖体相互作用抑制剂的检测方法的开发仍然是一个关键的需求。在这里,我们描述了一种基于荧光各向异性(FA)的竞争性结合测定,使用衍生自P茎CTD的BODIPY-TMR标记的11聚体肽(P11)来测量3-11个氨基酸的肽对RTA和Stx2A1的P茎袋的结合亲和力。与表面等离子体共振(SPR)测定的亲和力比较表明,尽管两种方法的等级顺序相同,FA测定可以更好地区分通过SPR显示非特异性相互作用的肽。FA测定仅检测与标记的P11竞争的相互作用,并且可以验证抑制剂特异性和作用机制。
    Ricin is one of the most toxic substances known and a type B biothreat agent. Shiga toxins (Stxs) produced by E. coli (STEC) and Shigella dysenteriae are foodborne pathogens. There is no effective therapy against ricin or STEC and there is an urgent need for inhibitors. Ricin toxin A subunit (RTA) and A1 subunit of Stx2a (Stx2A1) bind to the C-terminal domain (CTD) of the ribosomal P-stalk proteins to depurinate the sarcin/ricin loop. Modulation of toxin-ribosome interactions has not been explored as a strategy for inhibition. Therefore, development of assays that detect inhibitors targeting toxin-ribosome interactions remains a critical need. Here we describe a fluorescence anisotropy (FA)-based competitive binding assay using a BODIPY-TMR labeled 11-mer peptide (P11) derived from the P-stalk CTD to measure the binding affinity of peptides ranging from 3 to 11 amino acids for the P-stalk pocket of RTA and Stx2A1. Comparison of the affinity with the surface plasmon resonance (SPR) assay indicated that although the rank order was the same by both methods, the FA assay could differentiate better between peptides that show nonspecific interactions by SPR. The FA assay detects only interactions that compete with the labeled P11 and can validate inhibitor specificity and mechanism of action.
    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
    人血清白蛋白(HSA)是循环系统中最丰富的血浆蛋白。它是一个多域,多功能蛋白质,结合了不同的亲和力和广泛的特异性,绑定,商店,运输各种生物化合物,pharmacores,和脂肪酸。HSA由于其携带官能化配体和前药的能力而在药物递送中发现越来越多的用途。所有这些都提出了在多个配体的情况下结合位点占用的竞争问题,这反过来又会影响蛋白质的结构/动态/功能关系,并对生物医学应用产生影响。在这项工作中,棕榈酸(PA)的相互作用结合的影响,用DSC研究了华法林(War)和布洛芬(Ibu)对HSA的热稳定性,ATR-FTIR,和EPR。PA是一种高亲和力的生理配体,虽然这两种药物因其抗凝(War)和抗炎(Ibu)的功效而被广泛使用,并且是外源化合物,其适应于分别与一些脂肪酸结合位点重叠的被拒绝的药物位点DS1和DS2。结果表明,当用PA完全饱和时,HSA获得最高的热稳定性。该生理配体的结合不妨碍War或Ibu与蛋白质的天然状态的结合。此外,三个配体同时结合,表明由于变构效应的协同作用。二元和多种配体结合后增加的热稳定性缓和了蛋白质聚集倾向并限制了蛋白质动力学。生物物理学的发现提供了有关蛋白质稳定性的有趣特征,聚合,以及与多种配体相互作用的动力学,并且与药物递送有关。
    Human serum albumin (HSA) is the most abundant plasma protein of the circulatory system. It is a multidomain, multifunctional protein that, combining diverse affinities and wide specificity, binds, stores, and transports a variety of biological compounds, pharmacores, and fatty acids. HSA is finding increasing uses in drug-delivery due to its ability to carry functionalized ligands and prodrugs. All this raises the question of competition for binding sites occupancy in case of multiple ligands, which in turn influences the protein structure/dynamic/function relationship and also has an impact on the biomedical applications. In this work, the effects of interactive binding of palmitic acid (PA), warfarin (War) and ibuprofen (Ibu) on the thermal stability of HSA were studied using DSC, ATR-FTIR, and EPR. PA is a high-affinity physiological ligand, while the two drugs are widely used for their anticoagulant (War) and anti-inflammatory (Ibu) efficacy, and are exogenous compounds that accommodate in the deputed drug site DS1 and DS2, respectively overlapping with some of the fatty acid binding sites. The results indicate that HSA acquires the highest thermal stability when it is fully saturated with PA. The binding of this physiological ligand does not hamper the binding of War or Ibu to the native state of the protein. In addition, the three ligands bind simultaneously, suggesting a synergic cooperative influence due to allosteric effects. The increased thermal stability subsequent to binary and multiple ligands binding moderates protein aggregation propensity and restricts protein dynamics. The biophysics findings provide interesting features about protein stability, aggregation, and dynamics in interaction with multiple ligands and are relevant in drug-delivery.
    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
    ASCT2(丙氨酸丝氨酸半胱氨酸转运蛋白2),SLC1(溶质载体1)家族的成员,介导跨细胞膜的小中性氨基酸的Na依赖性交换。ASCT2在肿瘤细胞中高表达,使其成为抗癌治疗的有希望的目标。在这项研究中,我们使用电生理学和快速动力学方法,探索了高亲和力竞争性抑制剂Lc-BPE与ASCT2的结合机制.我们的研究表明,Lc-BPE结合需要一个或两个最初以高亲和力与apo转运蛋白结合的Na离子,Na1位点占用对抑制剂结合更为关键。与氨基酸底物结合形式相反,最后,第三个Na+离子不能结合,由于其结合位点(Na2)的扭曲,从而防止易位能力复合体的形成。基于快速动力学分析,Lc-BPE的应用产生了向外瞬态电流,表明,尽管其净中性性质,ASCT2中Lc-BPE的结合是弱电的,很可能是由于抑制剂的氨基酸部分内的不对称电荷分布。与Lc-BPE的预孵育还导致底物交换的转换率降低和底物诱导的阴离子电流的活化延迟,表明Lc-BPE解离动力学相对较慢。总的来说,我们的结果为原型竞争性抑制剂与ASCT转运蛋白的结合机制提供了新的见解.
    ASCT2 (alanine serine cysteine transporter 2), a member of the solute carrier 1 family, mediates Na+-dependent exchange of small neutral amino acids across cell membranes. ASCT2 was shown to be highly expressed in tumor cells, making it a promising target for anticancer therapies. In this study, we explored the binding mechanism of the high-affinity competitive inhibitor L-cis hydroxyproline biphenyl ester (Lc-BPE) with ASCT2, using electrophysiological and rapid kinetic methods. Our investigations reveal that Lc-BPE binding requires one or two Na+ ions initially bound to the apo-transporter with high affinity, with Na1 site occupancy being more critical for inhibitor binding. In contrast to the amino acid substrate bound form, the final, third Na+ ion cannot bind, due to distortion of its binding site (Na2), thus preventing the formation of a translocation-competent complex. Based on the rapid kinetic analysis, the application of Lc-BPE generated outward transient currents, indicating that despite its net neutral nature, the binding of Lc-BPE in ASCT2 is weakly electrogenic, most likely because of asymmetric charge distribution within the amino acid moiety of the inhibitor. The preincubation with Lc-BPE also led to a decrease of the turnover rate of substrate exchange and a delay in the activation of substrate-induced anion current, indicating relatively slow Lc-BPE dissociation kinetics. Overall, our results provide new insight into the mechanism of binding of a prototypical competitive inhibitor to the ASCT transporters.
    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:疫苗诱导的免疫性血栓性血小板减少症(VITT)是基于腺病毒的针对SARS-COV-2的疫苗的并发症,这是由于针对血小板因子4(PF4)的血栓前IgG抗体,在接受肝素治疗的患者中,可能难以与肝素诱导的血小板减少症(HIT)区分。
    目的:在这种情况下,我们评估了竞争性抗PF4酶免疫测定(EIA)的有用性。
    方法:1E12,1C12和2E1的F(ab\')2片段,3单克隆抗PF4抗体的能力,使用EIA评估抑制人VITT或HIT抗体与PF4的结合。进行丙氨酸扫描诱变以确定参与单克隆抗体和PF4之间相互作用的氨基酸(AA)。
    结果:用1E12测量VITTIgG与PF4结合的强抑制作用(中位抑制作用93%,n=8),而它对HIT抗体的结合没有影响(中位数:6%,n=8)。相比之下,1C12和2E1抑制VITT(中位数:74%和76%,分别)和HIT抗体(中位数:68%和53%,分别)与PF4结合。当用1E12对19个额外的VITT样品进行竞争性抗PF4EIA时,它强烈抑制IgG与PF4的结合,除了一个病人,根据临床病史,谁实际上发展了HIT。表位图谱显示,1E12与PF4上的5个关键AA相互作用,其中4个也是人类VITT抗体结合所必需的,从而解释了竞争性抑制。
    结论:使用1E12的简单竞争性抗PF4EIA可以帮助确认VITT诊断,并在两种诊断都可能的情况下将其与患者的HIT区分开。
    BACKGROUND: Vaccine-induced immune thrombotic thrombocytopenia (VITT) is a complication of adenoviral-based vaccine against SARS-CoV-2 due to prothrombotic immunoglobulin (Ig) G antibodies to platelet factor 4 (PF4) and may be difficult to distinguish from heparin-induced thrombocytopenia (HIT) in patients treated with heparin.
    OBJECTIVE: We assessed the usefulness of competitive anti-PF4 enzyme immunoassays (EIAs) in this context.
    METHODS: The ability of F(ab\')2 fragments of 1E12, 1C12, and 2E1, 3 monoclonal anti-PF4 antibodies, to inhibit the binding of human VITT or HIT antibodies to PF4 was evaluated using EIAs. Alanine-scanning mutagenesis was performed to define the amino acids involved in the interactions between the monoclonal antibodies and PF4.
    RESULTS: A strong inhibition of VITT IgG binding to PF4 was measured with 1E12 (median inhibition, 93%; n = 8), whereas it had no effect on the binding of HIT antibodies (median, 6%; n = 8). In contrast, 1C12 and 2E1 inhibited VITT (median, 74% and 76%, respectively) and HIT antibodies (median, 68% and 53%, respectively) binding to PF4. When a competitive anti-PF4 EIA was performed with 1E12 for 19 additional VITT samples, it strongly inhibited IgG binding to PF4, except for 1 patient, who had actually developed HIT according to the clinical history. Epitope mapping showed that 1E12 interacts with 5 key amino acids on PF4, of which 4 are also required for the binding of human VITT antibodies, thus explaining the competitive inhibition.
    CONCLUSIONS: A simple competitive anti-PF4 EIA with 1E12 could help confirm VITT diagnosis and distinguish it from HIT in patients when both diagnoses are possible.
    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
    了解蛋白质-配体相互作用的热力学和动力学对于生物学家和药理学家至关重要。为了可视化具有1:1化学计量且没有协同性的结合反应的平衡和动力学,我们获得了二阶结合过程中蛋白质-配体复合物浓度与时间的精确关系,并对竞争结合过程进行了数值模拟。首先,测量蛋白质-配体相互作用的两个常见问题集中在如何避免滴定方案和如何建立适当的孵育时间。然后,我们举例说明了常用的实验条件[L]0[P]0和[I]0[P]0如何影响动力学和热力学性质的估计。计算理论抑制曲线,并在预定条件下相应地估计表观IC50和IC50。使用估计的表观IC50,我们比较了通过使用Cheng-Prusoff方程计算的表观Ki和Ki,林-里格斯方程,和王群方程。我们还应用我们的工具来模拟高通量筛选,并比较实际实验的结果。用于模拟饱和度实验的可视化工具,结合和竞争结合的动力学实验,和抑制曲线,\"绑定曲线查看器,“可在www上获得。eplatton.net/binding-curve-viewer。
    Understanding the thermodynamics and kinetics of the protein-ligand interaction is essential for biologists and pharmacologists. To visualize the equilibrium and kinetics of the binding reaction with 1:1 stoichiometry and no cooperativity, we obtained the exact relationship of the concentration of the protein-ligand complex and the time in the second-order binding process and numerically simulated the process of competitive binding. First, two common concerns in measuring protein-ligand interactions were focused on how to avoid the titration regime and how to establish the appropriate incubation time. Then, we gave examples of how the commonly used experimental conditions of [L]0 ≫ [P]0 and [I]0 ≫ [P]0 affected the estimation of the kinetic and thermodynamic properties. Theoretical inhibition curves were calculated, and the apparent IC50 and IC50 were estimated accordingly under predefined conditions. Using the estimated apparent IC50, we compared the apparent Ki and Ki calculated by using the Cheng-Prusoff equation, Lin-Riggs equation, and Wang\'s group equation. We also applied our tools to simulate high-throughput screening and compare the results of real experiments. The visualization tool for simulating the saturation experiment, kinetic experiments of binding and competitive binding, and inhibition curve, \"Binding Curve Viewer,\" is available at www.eplatton.net/binding-curve-viewer.
    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
    现在已知超过3000种蛋白质与糖胺聚糖(GAG)结合。然而,GAG-蛋白质系统在识别的选择性方面知之甚少,分子作用机制,和翻译承诺。高通量筛选(HTS)技术对于研究GAG生物学和开发基于GAG的疗法是至关重要的。微阵列,在过去的二十年里发展起来的,现在已经改进到成为生物分子HTS中首选工具的程度。GAG微阵列,其中GAG序列固定在载玻片上,虽然类似于其他微阵列,有自己的挑战和考虑。GAG微阵列正迅速成为研究GAG-蛋白质系统的首选。这里,我们回顾了迄今为止GAG微阵列的不同模式和应用。我们讨论了这种技术的优点和缺点,解释使用不同化学反应基团的共价和非共价固定策略,并提出了定性和定量解释的各种分析格式,包括选择性筛选,结合亲和力研究,竞争性绑定研究等。我们还强调了在实施这项技术方面的最新进展,数据编目,并投射其未来的承诺。总的来说,GAG微阵列技术显示出巨大的潜力,它不仅仅是研究GAG-蛋白质系统的筛选工具。
    More than 3000 proteins are now known to bind to glycosaminoglycans (GAGs). Yet, GAG-protein systems are rather poorly understood in terms of selectivity of recognition, molecular mechanism of action, and translational promise. High-throughput screening (HTS) technologies are critically needed for studying GAG biology and developing GAG-based therapeutics. Microarrays, developed within the past two decades, have now improved to the point of being the preferred tool in the HTS of biomolecules. GAG microarrays, in which GAG sequences are immobilized on slides, while similar to other microarrays, have their own sets of challenges and considerations. GAG microarrays are rapidly becoming the first choice in studying GAG-protein systems. Here, we review different modalities and applications of GAG microarrays presented to date. We discuss advantages and disadvantages of this technology, explain covalent and non-covalent immobilization strategies using different chemically reactive groups, and present various assay formats for qualitative and quantitative interpretations, including selectivity screening, binding affinity studies, competitive binding studies etc. We also highlight recent advances in implementing this technology, cataloging of data, and project its future promise. Overall, the technology of GAG microarray exhibits enormous potential of evolving into more than a mere screening tool for studying GAG - protein systems.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号