GPCRs

GPCRs
  • 文章类型: Journal Article
    胶质瘤,尤其是胶质母细胞瘤患者,呈现高度异质性和免疫抑制的微环境,导致他们对治疗和生存的反应不佳。靶向肿瘤微环境被认为是一种有希望的治疗策略。M2巨噬细胞在胶质瘤组织中高度浸润,甚至高达50%的整体组织细胞的总数。这里,我们通过WGCNA分析将GPR65鉴定为神经胶质瘤M2巨噬细胞相关模块的hub基因.GPR65的表达和预后分析提示,GPR65与胶质瘤的恶性程度和不良预后呈正相关。异质性分析发现GPR65在胶质瘤的血管增生区域高表达,与M2巨噬细胞的空间表达特征相匹配。通过单细胞数据分析和免疫荧光,我们进一步验证了GPR65在神经胶质瘤微环境中的巨噬细胞而不是肿瘤细胞中高表达。最重要的是,我们发现抑制GPR65足以降低巨噬细胞对神经胶质瘤细胞的极化反应,并打破与神经胶质瘤细胞的恶性合作。我们的研究报道了GPR65在胶质瘤微环境中的表达特征和恶性行为。为胶质瘤微环境的治疗提供了新的替代靶点。
    Glioma, especially glioblastoma patients, present highly heterogeneous and immunosuppressive microenvironment, leading to their poor response to treatment and survival. Targeting the tumor microenvironment is considered a promising therapeutic strategy. M2 macrophages are highly infiltrated in glioma tissue, even up to 50% of the total number of bulk tissue cells. Here, we identified GPR65 as the hub gene of the M2 macrophage-related module in glioma through WGCNA analysis. The expression and prognosis analysis suggested that GPR65 was positively correlated with the malignancy and poor prognosis of glioma, and the heterogeneity analysis found that GPR65 was highly expressed in the vascular proliferation area of glioma, which matched the spatial expression characteristics of M2 macrophages. We further verified that GPR65 was highly expressed in macrophages but not tumor cells in the glioma microenvironment by single-cell data analysis and immunofluorescence. Most importantly, we found that inhibition of GPR65 was sufficient to reduce macrophages\' polarization response to glioma cell and break the malignant cooperation with glioma cells. Our study reports the expression characteristics and malignant behavior of GPR65 in the glioma microenvironment, which provides a new alternative target of treatment to glioma 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
    褪黑激素是一种具有多种生物学作用的多功能分子,作为一种促进健康的生物活性分子,在任何食品中都具有巨大的价值,酵母生产它的能力在过去十年中得到了广泛的证明。然而,它的定量提出了昂贵的分析挑战,由于通常的低浓度发现的结果,酵母代谢。本研究通过优化基于G蛋白偶联受体(GPCR)的酵母生物传感器来进行褪黑激素检测和定量,从而解决了这些分析挑战。战略遗传修饰被用来显著提高其灵敏度和荧光信号输出,使其适用于检测酵母产生的褪黑激素。优化的生物传感器显示出显着提高的灵敏度和荧光,能够筛选101个酵母菌株和检测各种葡萄酒样品中的褪黑激素。这种生物传感器在酵母生长培养基中定量褪黑激素的功效强调了其在探索褪黑激素生产动态和在功能性食品开发中的潜在应用中的实用性。这项研究提供了一种新的分析方法,可以进行快速且具有成本效益的褪黑激素分析,以更深入地了解发酵产品中褪黑激素的生物活性及其对人类健康的影响。这些发现凸显了生物传感器技术在简化发酵科学分析过程中的更广泛潜力。
    Melatonin is a multifunctional molecule with diverse biological roles that holds great value as a health-promoting bioactive molecule in any food product and yeast\'s ability to produce it has been extensively demonstrated in the last decade. However, its quantification presents costly analytical challenges due to the usual low concentrations found as the result of yeast metabolism. This study addresses these analytical challenges by optimizing a yeast biosensor based on G protein-coupled receptors (GPCR) for melatonin detection and quantitation. Strategic genetic modifications were employed to significantly enhance its sensitivity and fluorescent signal output, making it suitable for detection of yeast-produced melatonin. The optimized biosensor demonstrated significantly improved sensitivity and fluorescence, enabling the screening of 101 yeast strains and the detection of melatonin in various wine samples. This biosensor\'s efficacy in quantifying melatonin in yeast growth media underscores its utility in exploring melatonin production dynamics and potential applications in functional food development. This study provides a new analytical approach that allows a rapid and cost-effective melatonin analysis to reach deeper insights into the bioactivity of melatonin in fermented products and its implications for human health. These findings highlight the broader potential of biosensor technology in streamlining analytical processes in fermentation science.
    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
    蛋白质-蛋白质界面中的合作相互作用证明了相互依赖或链接的网络样行为及其对蛋白质偶联的影响。协同相互作用也可能在蛋白质-蛋白质界面的远处引起波纹或变构效应。尽管它们在蛋白质-蛋白质界面中至关重要,确定哪些氨基酸对相互作用是合作的是具有挑战性的。在这项工作中,我们使用了贝叶斯网络建模,一种可解释的机器学习方法,结合分子动力学轨迹,以鉴定在G蛋白偶联受体(GPCR)复合物与Gα亚基的界面中显示高协同性及其变构效应的残基对。我们的结果揭示了六个GPCR:Gα接触,这些接触对于不同的Gα亚型是共同的,并且在界面形成中显示出强的协同性。C端helix5和G蛋白的核心都是相互依赖的实体,在GPCR偶联中起重要作用。我们表明,混杂的GPCR偶联到不同的Gα亚型,使所有特定于每个Gα亚型的GPCR:Gα接触(Gαs,Gαi和Gαq)。这项工作强调了数据驱动的贝叶斯网络建模在阐明GPCR:G蛋白复合物中复杂的依赖性和选择性决定因素方面的潜力。为这些基本细胞信号传导成分的动态性质提供有价值的见解。
    Cooperative interactions in protein-protein interfaces demonstrate the interdependency or the linked network-like behavior and their effect on the coupling of proteins. Cooperative interactions also could cause ripple or allosteric effects at a distance in protein-protein interfaces. Although they are critically important in protein-protein interfaces, it is challenging to determine which amino acid pair interactions are cooperative. In this work, we have used Bayesian network modeling, an interpretable machine learning method, combined with molecular dynamics trajectories to identify the residue pairs that show high cooperativity and their allosteric effect in the interface of G protein-coupled receptor (GPCR) complexes with Gα subunits. Our results reveal six GPCR:Gα contacts that are common to the different Gα subtypes and show strong cooperativity in the formation of interface. Both the C terminus helix5 and the core of the G protein are codependent entities and play an important role in GPCR coupling. We show that a promiscuous GPCR coupling to different Gα subtypes, makes all the GPCR:Gα contacts that are specific to each Gα subtype (Gαs, Gαi, and Gαq). This work underscores the potential of data-driven Bayesian network modeling in elucidating the intricate dependencies and selectivity determinants in GPCR:G protein complexes, offering valuable insights into the dynamic nature of these essential cellular signaling components.
    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
    NanoLuc介导的生物发光共振能量转移(NanoBRET)因其能够均匀地测量配体与G蛋白偶联受体(GPCRs)的结合而受到欢迎。包括趋化因子受体亚家族.这些受体,如ACKR3,CXCR4,CXCR3,在免疫系统的调节中起着至关重要的作用,与炎症性疾病和癌症有关,并被视为有希望的药物靶标。这项研究的目的是使用不同的荧光标记的趋化因子CXCL12类似物优化基于NanoBRET的配体与NLuc-ACKR3和NLuc-CXCR4的结合,以及它们在同时两种趋化因子受体的多重NanoBRET结合测定中的用途。四种荧光CXCL12类似物(CXCL12-AZD488、-AZD546、-AZD594、-AZD647)显示与NLuc-ACKR3和NLuc-CXCR4的高亲和力可饱和结合,具有相对低水平的非特异性结合。此外,所有AZDye标记的CXCL12s与Nluc受体的结合被药理学相关的未标记趋化因子和小分子抑制。还成功建立了CXCL10-AZD488与Nluc-CXCR3结合的NanoBRET结合测定,并成功用于同时测量未标记的小分子与NLuc-CXCR3和NLuc-CXCR4的结合。总之,多路复用基于NanoBRET的竞争结合测定是用于同时针对多个GPCRs测试未标记(小)分子的有前途的工具。
    NanoLuc-mediated bioluminescence resonance energy transfer (NanoBRET) has gained popularity for its ability to homogenously measure ligand binding to G protein-coupled receptors (GPCRs), including the subfamily of chemokine receptors. These receptors, such as ACKR3, CXCR4, CXCR3, play a crucial role in the regulation of the immune system, are associated with inflammatory diseases and cancer, and are seen as promising drug targets. The aim of this study was to optimize NanoBRET-based ligand binding to NLuc-ACKR3 and NLuc-CXCR4 using different fluorescently labeled chemokine CXCL12 analogs and their use in a multiplex NanoBRET binding assay of two chemokine receptors at the same time. The four fluorescent CXCL12 analogs (CXCL12-AZD488, -AZD546, -AZD594, -AZD647) showed high-affinity saturable binding to both NLuc-ACKR3 and NLuc-CXCR4, with relatively low levels of non-specific binding. Additionally, the binding of all AZDye-labeled CXCL12s to Nluc receptors was inhibited by pharmacologically relevant unlabeled chemokines and small molecules. The NanoBRET binding assay for CXCL10-AZD488 binding to Nluc-CXCR3 was also successfully established and successfully employed for the simultaneous measurement of the binding of unlabeled small molecules to NLuc-CXCR3 and NLuc-CXCR4. In conclusion, multiplexing the NanoBRET-based competition binding assay is a promising tool for testing unlabeled (small) molecules against multiple GPCRs simultaneously.
    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
    磷酸肌醇3-激酶(PI3K)β(PI3Kβ)在功能上具有独特的整合来自受体酪氨酸激酶(RTK)的信号的能力,G蛋白偶联受体,和Rho家族GTPases。PI3Kβ优先考虑与各种膜束缚信号输入的相互作用的机制,然而,尚不清楚。先前的实验没有确定与膜束缚蛋白的相互作用是否主要控制PI3Kβ定位而不是直接调节脂质激酶活性。为了解决我们知识的差距,我们建立了一种检测方法,可以直接观察三种不同的蛋白质相互作用如何调节PI3Kβ,当PI3Kβ以生物学相关的构型在支持的脂质双层上呈现给激酶时.使用单分子全内反射荧光(TIRF)显微镜,我们确定了控制PI3Kβ膜定位的机制,信令输入的优先级排序,和脂质激酶激活。我们发现,在参与GβGγ或Rac1(GTP)之前,自抑制的PI3Kβ优先考虑与RTK衍生的酪氨酸磷酸化(pY)肽的相互作用。尽管pY肽强烈地将PI3Kβ定位于膜上,脂质激酶活性的刺激是适度的。在存在pY/GβGγ或pY/Rac1(GTP)的情况下,PI3Kβ活性显著增强,超出了简单增加膜定位所能解释的范围。相反,PI3Kβ通过与变构调节一致的机制被pY/GβGγ和pY/Rac1(GTP)协同激活。
    Phosphoinositide 3-kinase (PI3K) beta (PI3Kβ) is functionally unique in the ability to integrate signals derived from receptor tyrosine kinases (RTKs), G-protein coupled receptors, and Rho-family GTPases. The mechanism by which PI3Kβ prioritizes interactions with various membrane-tethered signaling inputs, however, remains unclear. Previous experiments did not determine whether interactions with membrane-tethered proteins primarily control PI3Kβ localization versus directly modulate lipid kinase activity. To address this gap in our knowledge, we established an assay to directly visualize how three distinct protein interactions regulate PI3Kβ when presented to the kinase in a biologically relevant configuration on supported lipid bilayers. Using single molecule Total Internal Reflection Fluorescence (TIRF) Microscopy, we determined the mechanism controlling PI3Kβ membrane localization, prioritization of signaling inputs, and lipid kinase activation. We find that auto-inhibited PI3Kβ prioritizes interactions with RTK-derived tyrosine phosphorylated (pY) peptides before engaging either GβGγ or Rac1(GTP). Although pY peptides strongly localize PI3Kβ to membranes, stimulation of lipid kinase activity is modest. In the presence of either pY/GβGγ or pY/Rac1(GTP), PI3Kβ activity is dramatically enhanced beyond what can be explained by simply increasing membrane localization. Instead, PI3Kβ is synergistically activated by pY/GβGγ and pY/Rac1 (GTP) through a mechanism consistent with allosteric regulation.
    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:G蛋白偶联受体(GPCRs)的脱敏是指通过长时间或间歇性暴露于激动剂来减弱受体反应性。β-抑制蛋白与磷酸化受体的细胞质腔的结合,与G蛋白竞争,已被广泛接受为解释GPCRs脱敏的广泛模型。然而,对各种GPCR的研究,包括多巴胺D2样受体(D2R,D3R,D4R),提出了其他脱敏机制的存在。本研究采用了具有不同脱敏特性的D2R/D3R变体,并利用功能丧失方法来揭示GPCRs同源脱敏的潜在机制。关注调节AKT泛素化的信号级联。
    结果:AKT通过TRAF6进行K8/14泛素化,这发生在细胞核中并促进其膜募集,受体脱敏条件下的磷酸化和激活。TRAF6的核进入依赖于输入蛋白复合物的存在。Src通过介导TRAF6与输入蛋白β1之间的相互作用来调节TRAF6的核进入。泛素化的AKT易位到质膜,在那里它与Mdm2结合以在S166和S186残基处磷酸化它。此后,磷酸化的Mdm2被募集到细胞核,导致β-Arr2的去泛素化。然后去泛素化的β-Arr2与Gβγ形成复合物,作为GPCRs脱敏的生物标志物。像在D3R,AKT的泛素化也参与β2肾上腺素受体的脱敏。
    结论:我们的研究表明,导致TRAF6从细胞质到细胞核的亚细胞定位变化以介导AKT泛素化的受体的特性可以启动GPCRs的脱敏。
    BACKGROUND: Desensitization of G protein-coupled receptors (GPCRs) refers to the attenuation of receptor responsiveness by prolonged or intermittent exposure to agonists. The binding of β-arrestin to the cytoplasmic cavity of the phosphorylated receptor, which competes with the G protein, has been widely accepted as an extensive model for explaining GPCRs desensitization. However, studies on various GPCRs, including dopamine D2-like receptors (D2R, D3R, D4R), have suggested the existence of other desensitization mechanisms. The present study employed D2R/D3R variants with different desensitization properties and utilized loss-of-function approaches to uncover the mechanisms underlying GPCRs homologous desensitization, focusing on the signaling cascade that regulates the ubiquitination of AKT.
    RESULTS: AKT undergoes K8/14 ubiquitination by TRAF6, which occurs in the nucleus and promotes its membrane recruitment, phosphorylation and activation under receptor desensitization conditions. The nuclear entry of TRAF6 relies on the presence of the importin complex. Src regulates the nuclear entry of TRAF6 by mediating the interaction between TRAF6 and importin β1. Ubiquitinated AKT translocates to the plasma membrane where it associates with Mdm2 to phosphorylate it at the S166 and S186 residues. Thereafter, phosphorylated Mdm2 is recruited to the nucleus, resulting in the deubiquitination of β-Arr2. The deubiquitinated β-Arr2 then forms a complex with Gβγ, which serves as a biomarker for GPCRs desensitization. Like in D3R, ubiquitination of AKT is also involved in the desensitization of β2 adrenoceptors.
    CONCLUSIONS: Our study proposed that the property of a receptor that causes a change in the subcellular localization of TRAF6 from the cytoplasm to the nucleus to mediate AKT ubiquitination could initiate the desensitization of GPCRs.
    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
    G蛋白偶联kisspeptin受体(GPR54或KISS1R)是生殖中的重要介质,代谢和癌症生物学;然而,有有限的荧光探针或抗体直接成像的这些受体在细胞和完整的组织,这可以帮助询问他们的多种生物学角色。在这里,我们描述了一种新的耐酸的基于BODIPY的氨基酸(Trp-BODIPYPLUS)的合理设计和表征,及其用于固相合成荧光生物活性肽的实现。Trp-BODIPYPLUS保留了短线性和环状肽的结合能力,并在靶标结合时显示出明显的开启荧光发射,用于无洗涤成像。最后,我们使用Trp-BODIPYPLUS制备了一些首批基于kisspeptin的荧光探针,并通过荧光成像观察了GPR54受体在人细胞和整个小鼠胰岛中的表达和定位.
    我们将Trp-BODIPYPLUS描述为一种新的耐酸结构单元,用于直接固相合成荧光肽。我们证明了Trp-BODIPYPLUS与第一个荧光探针的实用性,可直接可视化和定量分析人细胞和完整小鼠胰岛中G蛋白偶联受体54(GPR54或kisspeptin)的表达和定位。
    The G protein-coupled kisspeptin receptor (GPR54 or KISS1R) is an important mediator in reproduction, metabolism and cancer biology; however, there are limited fluorescent probes or antibodies for direct imaging of these receptors in cells and intact tissues, which can help to interrogate their multiple biological roles. Herein, we describe the rational design and characterization of a new acid-resistant BODIPY-based amino acid (Trp-BODIPY PLUS), and its implementation for solid-phase synthesis of fluorescent bioactive peptides. Trp-BODIPY PLUS retains the binding capabilities of both short linear and cyclic peptides and displays notable turn-on fluorescence emission upon target binding for wash-free imaging. Finally, we employed Trp-BODIPY PLUS to prepare some of the first fluorogenic kisspeptin-based probes and visualized the expression and localization of GPR54 receptors in human cells and in whole mouse pancreatic islets by fluorescence imaging.
    We describe Trp‐BODIPY PLUS as a new acid‐resistant building block for straightforward solid‐phase synthesis of fluorogenic peptides. We demonstrate the utility of Trp‐BODIPY PLUS with the first fluorogenic probe for direct visualization and quantitative analysis of the expression and localization of the G protein‐coupled receptors 54 (GPR54 or kisspeptin) in human cells and intact mouse pancreatic islets.
    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
    G蛋白偶联受体(GPCRs)构成了最大的受体超家族,占蛋白质编码基因的4%。尽管这种跨膜受体普遍存在,仍有相当数量的孤儿,缺乏确定的内源性配体。自从他们的概念,反向药理学方法已用于表征此类受体。然而,GPCR信号的多面性和细致入微的性质对其药理学阐明提出了巨大挑战。考虑到它们的治疗相关性,继续寻找天然孤儿GPCR配体。尽管在3D结晶结构方面的结构输入有限,随着机器学习方法的进步,在准确预测配体方面取得了很大进展。尽管这种方法证明是有价值的,因为配体稀缺是孤儿GPCR去甲的最大障碍,剩余的孤儿GPCR的未来配对可能不一定采取一刀切的方法,但应更全面地考虑许多细微差别的可能性,以涵盖GPCR信号的全部范围.
    G protein-coupled receptors (GPCRs) make up the largest receptor superfamily, accounting for 4% of protein-coding genes. Despite the prevalence of such transmembrane receptors, a significant number remain orphans, lacking identified endogenous ligands. Since their conception, the reverse pharmacology approach has been used to characterize such receptors. However, the multifaceted and nuanced nature of GPCR signaling poses a great challenge to their pharmacological elucidation. Considering their therapeutic relevance, the search for native orphan GPCR ligands continues. Despite limited structural input in terms of 3D crystallized structures, with advances in machine-learning approaches, there has been great progress with respect to accurate ligand prediction. Though such an approach proves valuable given that ligand scarcity is the greatest hurdle to orphan GPCR deorphanization, the future pairings of the remaining orphan GPCRs may not necessarily take a one-size-fits-all approach but should be more comprehensive in accounting for numerous nuanced possibilities to cover the full spectrum of GPCR signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    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
    G蛋白偶联受体(GPCRs)是细胞信号传导和生理功能的主要介质。尽管它们具有生物学意义,GPCR在毒理学领域尚未广泛研究。在这里,我们使用126例非嗅觉GPCRs的高通量药物筛选试验研究了这些受体作为塑料化学物质的新靶标。在第一遍屏幕中,我们测试了磷酸三酚的活性,双酚A,和邻苯二甲酸二乙酯,以及从塑料食品包装中提取的三种实际混合物,涵盖所有主要聚合物类型。我们发现了11个GPCR-化学相互作用,其中化学混合物对腺苷受体1(ADORA1)和褪黑激素受体1(MTNR1A)表现出最强大的活性。我们进一步证实聚氯乙烯和聚氨酯产品含有ADORA1或MTNRA1激动剂,使用证实的二次筛选和药理学敲除实验。最后,对相关基因本体论术语的分析表明,ADORA1和MTNR1A的激活可能与生理和代谢过程的下游效应有关.这项工作强调了由塑料化学物质引起的信号中断比以前认为的更广泛,并证明了非基因组途径的相关性。其中有,到目前为止,仍未探索。
    G protein-coupled receptors (GPCRs) are central mediators of cell signaling and physiological function. Despite their biological significance, GPCRs have not been widely studied in the field of toxicology. Herein, we investigated these receptors as novel targets of plastic chemicals using a high-throughput drug screening assay with 126 human non-olfactory GPCRs. In a first-pass screen, we tested the activity of triphenol phosphate, bisphenol A, and diethyl phthalate, as well as three real-world mixtures of chemicals extracted from plastic food packaging covering all major polymer types. We found 11 GPCR-chemical interactions, of which the chemical mixtures exhibited the most robust activity at adenosine receptor 1 (ADORA1) and melatonin receptor 1 (MTNR1A). We further confirm that polyvinyl chloride and polyurethane products contain ADORA1 or MTNRA1 agonists using a confirmatory secondary screen and pharmacological knockdown experiments. Finally, an analysis of the associated gene ontology terms suggests that ADORA1 and MTNR1A activation may be linked to downstream effects on circadian and metabolic processes. This work highlights that signaling disruption caused by plastic chemicals is broader than that previously believed and demonstrates the relevance of nongenomic pathways, which have, thus far, remained unexplored.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号