G protein-coupled receptor kinase

G 蛋白偶联受体激酶
  • 文章类型: Journal Article
    G蛋白偶联受体(GPCRs)介导细胞对无数激素和神经递质的反应,这些激素和神经递质在调节血压等生理过程中起着至关重要的作用。在动脉和肾脏等器官中,激素或神经递质,例如血管紧张素II(AngII),多巴胺,肾上腺素,去甲肾上腺素通过它们的受体发挥它们的功能,最终的效果是保持正常的血管反应性,正常的身体钠,和正常的血压。GPCR激酶(GRKs)发挥其生物学功能,通过介导激动剂占据的GPCRs的调节,非GPCRs,或非受体底物。特别是,越来越多的研究表明,心血管系统和肾脏中GRKs的异常表达和活性抑制或刺激GPCRs(例如,多巴胺受体,AngII受体,和α-和β-肾上腺素能受体),导致高血压。目前的研究集中在选择性GRK抑制剂在心血管疾病中的作用。包括高血压.此外,基因研究表明GRK基因变异与原发性高血压,降压药物对血压的反应,和抗高血压治疗的不良心血管结局。在这次审查中,我们对GRK介导的血压调节进行了全面的概述,GRKs在高血压发病机制中的作用,并强调治疗高血压的潜在策略。GPCR脱敏过程的示意图。GPCR的激活始于激动剂与其相应受体的结合。然后G蛋白激活由各种信号通路介导的下游效应子。GPCR信号通过GRK介导的受体磷酸化停止,通过β抑制蛋白引起受体内化。
    G protein-coupled receptors (GPCRs) mediate cellular responses to a myriad of hormones and neurotransmitters that play vital roles in the regulation of physiological processes such as blood pressure. In organs such as the artery and kidney, hormones or neurotransmitters, such as angiotensin II (Ang II), dopamine, epinephrine, and norepinephrine exert their functions via their receptors, with the ultimate effect of keeping normal vascular reactivity, normal body sodium, and normal blood pressure. GPCR kinases (GRKs) exert their biological functions, by mediating the regulation of agonist-occupied GPCRs, non-GPCRs, or non-receptor substrates. In particular, increasing number of studies show that aberrant expression and activity of GRKs in the cardiovascular system and kidney inhibit or stimulate GPCRs (e.g., dopamine receptors, Ang II receptors, and α- and β-adrenergic receptors), resulting in hypertension. Current studies focus on the effect of selective GRK inhibitors in cardiovascular diseases, including hypertension. Moreover, genetic studies show that GRK gene variants are associated with essential hypertension, blood pressure response to antihypertensive medicines, and adverse cardiovascular outcomes of antihypertensive treatment. In this review, we present a comprehensive overview of GRK-mediated regulation of blood pressure, role of GRKs in the pathogenesis of hypertension, and highlight potential strategies for the treatment of hypertension. Schematic representation of GPCR desensitization process. Activation of GPCRs begins with the binding of an agonist to its corresponding receptor. Then G proteins activate downstream effectors that are mediated by various signaling pathways. GPCR signaling is halted by GRK-mediated receptor phosphorylation, which causes receptor internalization through β-arrestin.
    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: Preprint
    GPCR信号传导的规范范例将G蛋白和β-抑制素识别为促进GPCR信号传导的两个主要换能器。最近的证据表明,非典型趋化因子受体3(ACKR3)不与G蛋白偶联,和β-抑制素对于其某些功能是可有可无的。这里,我们使用邻近标记来鉴定缺乏β-抑制蛋白的细胞中与ACKR3相互作用的蛋白质。我们鉴定了参与胞吞机制的蛋白质,并评估了几个基于GPCR的邻近标记实验中保守的蛋白质子集。我们发现骨形态发生蛋白2诱导激酶(BMP2K)与许多不同的GPCRs相互作用,对β-抑制蛋白的依赖性不同。一起,我们的工作强调了可以独立于G蛋白和β-抑制素调节GPCR信号传导的调节剂的存在,并为可能调节GPCR信号传导的其他靶标提供了重要证据.
    The canonical paradigm of GPCR signaling recognizes G proteins and β-arrestins as the two primary transducers that promote GPCR signaling. Recent evidence suggests the atypical chemokine receptor 3 (ACKR3) does not couple to G proteins, and β-arrestins are dispensable for some of its functions. Here, we employed proximity labeling to identify proteins that interact with ACKR3 in cells devoid of β-arrestin. We identified proteins involved in the endocytic machinery and evaluated a subset of proteins conserved across several GPCR-based proximity labeling experiments. We discovered that the bone morphogenic protein 2-inducible kinase (BMP2K) interacts with many different GPCRs with varying dependency on β-arrestin. Together, our work highlights the existence of modulators that can act independently of G proteins and β-arrestins to regulate GPCR signaling and provides important evidence for other targets that may regulate 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: Journal Article
    25-羟基胆固醇(25HC)是一种具有生物活性的氧固醇,在巨噬细胞和树突状细胞的炎症过程中,其产量大大增加。炎症反应经常伴随着心脏调节的变化,例如心脏β-肾上腺素能受体(AR)信号的钝化。这里,在小鼠心房中研究了25HC依赖性调节β-AR激活反应的机制。25HC在亚微摩尔水平上降低了β-AR介导的正性肌力作用和Ca2瞬时振幅的增强,不改变生产。25HC显着减弱了对β1-AR(而不是β2-AR)激活的正性肌力反应。选择性β3-AR拮抗剂以及Gi蛋白抑制剂可阻止25HC对β1-AR介导的反应的抑制作用,Gβγ,G蛋白偶联受体激酶2/3或β抑制蛋白。同时,蛋白激酶D和C的阻断剂以及磷酸二酯酶抑制剂并未排除25HC对β-AR激活的正性肌力反应的负作用。因此,25HC可以通过参与β3-AR来抑制β1-AR依赖性效应,Gi蛋白,Gβγ,G蛋白偶联受体激酶,和β-抑制蛋白。这种25HC依赖性机制可以促进心房β-肾上腺素能信号的炎症相关改变。
    25-Hydroxycholesterol (25HC) is a biologically active oxysterol, whose production greatly increases during inflammation by macrophages and dendritic cells. The inflammatory reactions are frequently accompanied by changes in heart regulation, such as blunting of the cardiac β-adrenergic receptor (AR) signaling. Here, the mechanism of 25HC-dependent modulation of responses to β-AR activation was studied in the atria of mice. 25HC at the submicromolar levels decreased the β-AR-mediated positive inotropic effect and enhancement of the Ca2+ transient amplitude, without changing NO production. Positive inotropic responses to β1-AR (but not β2-AR) activation were markedly attenuated by 25HC. The depressant action of 25HC on the β1-AR-mediated responses was prevented by selective β3-AR antagonists as well as inhibitors of Gi protein, Gβγ, G protein-coupled receptor kinase 2/3, or β-arrestin. Simultaneously, blockers of protein kinase D and C as well as a phosphodiesterase inhibitor did not preclude the negative action of 25HC on the inotropic response to β-AR activation. Thus, 25HC can suppress the β1-AR-dependent effects via engaging β3-AR, Gi protein, Gβγ, G protein-coupled receptor kinase, and β-arrestin. This 25HC-dependent mechanism can contribute to the inflammatory-related alterations in the atrial β-adrenergic signaling.
    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
    骨关节炎(OA)是一种慢性骨骼退行性疾病,是老年人群残疾的主要原因。含有锌指和BTB结构域的16(ZBTB16)是先前已揭示在人OA组织中受损的转录因子。本研究旨在阐述ZBTB16对OA的潜在影响,并可能评估任何潜在的调节机制。使用基因表达系列(GSE)数据库(https://www.ncbi.nlm.nih.gov/geo/query/acc。cgi?acc=GSE169077),而软骨细胞中的ZBTB16表达使用逆转录定量PCR(RT-qPCR)和蛋白质印迹法进行检查。使用细胞计数试剂盒-8测定检查细胞活力。TUNEL分析和蛋白质印迹用于评估细胞凋亡和凋亡相关标志物。包括Bcl-2,Bax和裂解的caspase-3。炎症因子的水平和表达,包括TNF-α,通过ELISA和蛋白质印迹法测定IL-1β和IL-6。RT-qPCR和蛋白质印迹法也用于分析细胞外基质(ECM)降解酶的表达水平,包括MMP-13,具有血小板反应蛋白1型基序5,聚集蛋白聚糖和II型胶原蛋白α1的解整合素样和金属蛋白酶。在使用CistromeDB数据库预测ZBTB16与G蛋白偶联受体激酶2型(GRK2)启动子的潜在结合后,通过RT-qPCR和蛋白质印迹确认GRK2表达。然后使用染色质免疫沉淀和荧光素酶报告基因测定来确定ZBTB16和GRK2启动子之间的潜在相互作用。通过共转染GRK2和ZBTB16过表达质粒,在ZBTB16过表达的软骨细胞中GRK2过表达后,再次进行上述功能实验。与正常软骨组织和脂多糖(LPS)刺激的软骨细胞相比,人OA组织中ZBTB16的表达降低。ZBTB16过表达增加细胞活力,同时减少细胞凋亡,LPS处理的软骨细胞的炎症和ECM降解。此外,发现在LPS刺激的软骨细胞中GRK2表达增加。ZBTB16成功结合GRK2启动子,负调节GRK2表达。GRK2上调逆转了ZBTB16过表达对细胞活力的影响,凋亡,LPS攻击的软骨细胞的炎症和ECM降解。总之,这些数据表明ZBTB16可能通过GRK2的转录失活抑制OA的发展。
    Osteoarthritis (OA) is a chronic degenerative disease of the bone that is a major contributor of disability in the elderly population. Zinc finger and BTB domain-containing 16 (ZBTB16) is a transcription factor that has been previously revealed to be impaired in human OA tissues. The present study was designed to elaborate the potential impact of ZBTB16 on OA and to possibly assess any latent regulatory mechanism. ZBTB16 expression in human OA tissues was examined using the Gene Expression Series (GSE) database (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE169077) whereas ZBTB16 expression in chondrocytes was examined using reverse transcription-quantitative PCR (RT-qPCR) and western blotting. Cell viability was examined using a Cell Counting Kit-8 assay. A TUNEL assay and western blotting were used to assess cell apoptosis and apoptosis-related markers, including Bcl-2, Bax and cleaved caspase-3. The levels and expression of inflammatory factors, including TNF-α, IL-1β and IL-6, were determined by ELISA and western blotting. RT-qPCR and western blotting were also used to analyze the expression levels of extracellular matrix (ECM)-degrading enzymes, including MMP-13, a disintegrin-like and metalloproteinase with thrombospondin type-1 motifs-5, aggrecan and collagen type II α1. After the potential binding of ZBTB16 with the G protein coupled receptor kinase type 2 (GRK2) promoter was predicted using the Cistrome DB database, GRK2 expression was confirmed by RT-qPCR and western blotting. Chromatin immunoprecipitation and luciferase reporter assays were then used to determine the potential interaction between ZBTB16 and the GRK2 promoter. Following GRK2 overexpression in ZBTB16-overexpressing chondrocytes by co-transfection of GRK2 and ZBTB16 overexpression plasmids, the aforementioned functional experiments were performed again. ZBTB16 expression was found to be reduced in human OA tissues compared with in normal cartilage tissues and lipopolysaccharide (LPS)-stimulated chondrocytes. ZBTB16 overexpression increased cell viability whilst decreasing apoptosis, inflammation and ECM degradation by LPS-treated chondrocytes. In addition, GRK2 expression was found to be increased in LPS-stimulated chondrocytes. ZBTB16 successfully bound to the GRK2 promoter, which negatively modulated GRK2 expression. GRK2 upregulation reversed the effects of ZBTB16 overexpression on the viability, apoptosis, inflammation and ECM degradation by LPS-challenged chondrocytes. In conclusion, these data suggest that ZBTB16 may inhibit the development of OA through the transcriptional inactivation of GRK2.
    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
    Gq蛋白偶联组胺H1受体在变态反应和炎症反应中发挥关键作用,其中细胞外信号调节激酶(ERK)的磷酸化似乎介导炎性细胞因子的产生。ERK磷酸化受G蛋白和抑制蛋白介导的信号转导途径调节。这里,我们的目的是探讨H1受体介导的ERK磷酸化过程如何被Gq蛋白和抑制素差异调节.为此,我们评估了H1受体介导的ERK磷酸化的调节机制在中国仓鼠卵巢细胞表达Gq蛋白和抑制蛋白的人H1受体,S487TR和S487A,其中C末端的Ser487残基被截短并突变为丙氨酸,分别。免疫印迹分析表明,组胺诱导的ERK磷酸化在表达Gq蛋白偏向S487TR的细胞中是迅速和短暂的,而在表达抑制蛋白偏向S487A的细胞中缓慢且持续。Gq蛋白(YM-254890)和蛋白激酶C(PKC)(GF109203X)的抑制剂,和细胞内Ca2+螯合剂(BAPTA-AM)抑制组胺诱导的ERK磷酸化细胞表达S487TR,但不是那些表达S487A的人。相反,G蛋白偶联受体激酶抑制剂(GRK2/3)(cmpd101),β-arrestin2(β-arrestin2siRNA),网格蛋白(高渗蔗糖),Raf(LY3009120),和MEK(U0126)在表达S487A的细胞中抑制组胺诱导的ERK磷酸化,但不是那些表达S487TR的人。这些结果表明,H1受体介导的ERK磷酸化可能受到Gq蛋白/Ca2/PKC和GRK/抑制蛋白/网格蛋白/Raf/MEK途径的差异调节,可能决定组胺诱导的变态反应和炎症反应的早期和晚期。分别。
    Gq protein-coupled histamine H1 receptors play crucial roles in allergic and inflammatory reactions, in which the phosphorylation of extracellular signal-regulated kinase (ERK) appears to mediate the production of inflammatory cytokines. ERK phosphorylation is regulated by G protein- and arrestin-mediated signal transduction pathways. Here, we aimed to explore how H1 receptor-mediated processes of ERK phosphorylation might be differentially regulated by Gq proteins and arrestins. For this purpose, we evaluated the regulatory mechanism(s) of H1 receptor-mediated ERK phosphorylation in Chinese hamster ovary cells expressing Gq protein- and arrestin-biased mutants of human H1 receptors, S487TR and S487A, in which the Ser487 residue in the C-terminal was truncated and mutated to alanine, respectively. Immunoblotting analysis indicated that histamine-induced ERK phosphorylation was prompt and transient in cells expressing Gq protein-biased S487TR, whereas it was slow and sustained in cells expressing arrestin-biased S487A. Inhibitors of Gq proteins (YM-254890) and protein kinase C (PKC) (GF109203X), and an intracellular Ca2+ chelator (BAPTA-AM) suppressed histamine-induced ERK phosphorylation in cells expressing S487TR, but not those expressing S487A. Conversely, inhibitors of G protein-coupled receptor kinases (GRK2/3) (cmpd101), β-arrestin2 (β-arrestin2 siRNA), clathrin (hypertonic sucrose), Raf (LY3009120), and MEK (U0126) suppressed histamine-induced ERK phosphorylation in cells expressing S487A, but not those expressing S487TR. These results suggest that H1 receptor-mediated ERK phosphorylation might be differentially regulated by the Gq protein/Ca2+/PKC and GRK/arrestin/clathrin/Raf/MEK pathways to potentially determine the early and late phases of histamine-induced allergic and inflammatory responses, respectively.
    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
    Paget骨病(PDB)是一种代谢性骨病,其特征是破骨细胞功能失调,导致骨重建的局灶性异常。它会导致疼痛,骨折,骨畸形.G蛋白偶联受体激酶3(GRK3)是G蛋白偶联受体(GPCR)信号的重要负调节因子。已知GRK3在成骨细胞和前成骨细胞中调节GPCR功能,但其在破骨细胞中的调节功能尚不明确。这里,我们报道了在人和小鼠原代细胞和已建立的细胞系中破骨细胞分化过程中,Grk3表达增加。我们还表明,缺乏Grk3的老年小鼠会出现与人类PDB和其他Paget疾病小鼠模型相似的骨损伤。我们表明,Grk3表达的缺乏增强了体外破骨细胞的生成和体内造血破骨细胞前体的增殖,但不影响破骨细胞介导的骨吸收功能或细胞衰老途径。值得注意的是,我们还观察到,与年龄和性别匹配的健康对照组相比,PDB患者外周血单个核细胞中Grk3的表达降低.我们的数据表明,GRK3与破骨细胞分化的调节有关,并且可能与PDB和其他与破骨细胞活化相关的代谢性骨疾病的发病机理有关。
    Paget\'s Disease of Bone (PDB) is a metabolic bone disease that is characterized by dysregulated osteoclast function leading to focal abnormalities of bone remodeling. It can lead to pain, fracture, and bone deformity. G protein-coupled receptor kinase 3 (GRK3) is an important negative regulator of G protein-coupled receptor (GPCR) signaling. GRK3 is known to regulate GPCR function in osteoblasts and preosteoblasts, but its regulatory function in osteoclasts is not well defined. Here, we report that Grk3 expression increases during osteoclast differentiation in both human and mouse primary cells and established cell lines. We also show that aged mice deficient in Grk3 develop bone lesions similar to those seen in human PDB and other Paget\'s Disease mouse models. We show that a deficiency in Grk3 expression enhances osteoclastogenesis in vitro and proliferation of hematopoietic osteoclast precursors in vivo but does not affect the osteoclast-mediated bone resorption function or cellular senescence pathway. Notably, we also observe decreased Grk3 expression in peripheral blood mononuclear cells of patients with PDB compared with age- and gender-matched healthy controls. Our data suggest that GRK3 has relevance to the regulation of osteoclast differentiation and that it may have relevance to the pathogenesis of PDB and other metabolic bone diseases associated with osteoclast activation.
    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
    大麻素受体1(CB1R),G蛋白偶联受体,在突触可塑性中起着至关重要的作用。CB1R信号传导的异常活性和失调导致广谱的病理状况。CB1R信号传导受到受体脱敏的调节,包括G蛋白偶联受体激酶(GRK)对细胞内C末端残基的磷酸化,这可能导致内吞作用。此外,CB1R信号传导受阻碍受体内化的蛋白Src同源性3结构域生长因子受体结合2样(SGIP1)调节,同时增强CB1R与β抑制蛋白的关联。据推测,丝氨酸/苏氨酸残基的两个簇的磷酸化,425SMGDS429和460TMSVSTDTS468,在CB1R中,C-tail控制受体与其参与脱敏的相互作用伙伴之间的关联动力学.这些事件的几个分子决定因素仍然没有得到很好的理解。我们假设这些相互作用的动力学受到SGIP1的调节。使用一组在上述丝氨酸和苏氨酸残基中突变的CB1R,以及一系列基于生物发光能量转移(BRET)的传感器,我们发现GRK3与G蛋白的Gβγ亚基形成复合物,这在很大程度上不依赖于GRK3与CB1R的相互作用。此外,CB1R仅与激活的GRK3相互作用。有趣的是,CB1R上两个特定残基的磷酸化引发GRK3与脱敏受体的解离。SGIP1增加GRK3与G蛋白的Gβγ亚基的关联,和CB1R。总之,我们的数据表明,CB1R信号体复合物由受体C尾的顺序磷酸化动态控制,并且还被SGIP1修饰.
    Cannabinoid receptor 1 (CB1R), a G protein-coupled receptor, plays a fundamental role in synaptic plasticity. Abnormal activity and deregulation of CB1R signaling result in a broad spectrum of pathological conditions. CB1R signaling is regulated by receptor desensitization including phosphorylation of residues within the intracellular C terminus by G protein-coupled receptor kinases (GRKs) that may lead to endocytosis. Furthermore, CB1R signaling is regulated by the protein Src homology 3-domain growth factor receptor-bound 2-like (SGIP1) that hinders receptor internalization, while enhancing CB1R association with β-arrestin. It has been postulated that phosphorylation of two clusters of serine/threonine residues, 425 SMGDS429 and 460 TMSVSTDTS468 , within the CB1R C-tail controls dynamics of the association between receptor and its interaction partners involved in desensitization. Several molecular determinants of these events are still not well understood. We hypothesized that the dynamics of these interactions are modulated by SGIP1. Using a panel of CB1Rs mutated in the aforementioned serine and threonine residues, together with an array of Bioluminescence energy transfer-based (BRET) sensors, we discovered that GRK3 forms complexes with Gβγ subunits of G proteins that largely independent of GRK3\'s interaction with CB1R. Furthermore, CB1R interacts only with activated GRK3. Interestingly, phosphorylation of two specific residues on CB1R triggers GRK3 dissociation from the desensitized receptor. SGIP1 increases the association of GRK3 with Gβγ subunits of G proteins, and with CB1R. Altogether, our data suggest that the CB1R signalosome complex is dynamically controlled by sequential phosphorylation of the receptor C-tail and is also modified by SGIP1.
    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
    目的:大麻素CB2受体(CB2)是调节炎症的一个有希望的治疗靶点。然而,围绕CB2脱敏和调节的机制知之甚少,特别是G蛋白偶联受体激酶(GRKs)的作用。这里,我们评估了6种GRK亚型在β抑制蛋白募集到CB2中的作用.还进行了几个远端C末端天冬氨酸残基的诱变,以试图描绘参与CB2调节的其他结构元件。
    方法:在表达CB2的HEK293细胞中,使用实时BRET测定测量β-抑制蛋白易位。进行G蛋白解离BRET测定以评估在β-抑制蛋白2存在下CB2的活化和脱敏。
    结果:GRK同工型1-6的过表达未能显着改善β-抑制素1或β-抑制素2向CB2的易位。与此一致,内源性GRK2/3的抑制并未显著减少β-抑制蛋白2易位.C端天冬氨酸残基的诱变导致β-抑制蛋白2易位的减弱,这转化为G蛋白激活的脱敏减少。
    结论:我们的研究结果表明,CB2不符合经典的GPCR调控范式,需要GRK-和β-抑制蛋白介导的脱敏。相反,C-末端天冬氨酸残基可作为磷酸模拟物诱导β-抑制蛋白活化。这项研究为CB2的调控机制提供了新的见解,这可能有助于我们对药物耐受性和依赖性的理解。
    OBJECTIVE: The cannabinoid CB2 receptor (CB2 ) is a promising therapeutic target for modulating inflammation. However, little is known surrounding the mechanisms underpinning CB2 desensitisation and regulation, particularly the role of G protein-coupled receptor kinases (GRKs). Here, we evaluated the role of six GRK isoforms in β-arrestin recruitment to CB2 . Mutagenesis of several distal C-terminal aspartic acid residues was also performed in an attempt to delineate additional structural elements involved in the regulation of CB2 .
    METHODS: In CB2 -expressing HEK 293 cells, β-arrestin translocation was measured using real-time BRET assays. G protein dissociation BRET assays were performed to assess the activation and desensitisation of CB2 in the presence of β-arrestin 2.
    RESULTS: Overexpression of GRK isoforms 1-6 failed to considerably improve translocation of either β-arrestin 1 or β-arrestin 2 to CB2 . Consistent with this, inhibition of endogenous GRK2/3 did not substantially reduce β-arrestin 2 translocation. Mutagenesis of C-terminal aspartic acid residues resulted in attenuation of β-arrestin 2 translocation, which translated to a reduction in desensitisation of G protein activation.
    CONCLUSIONS: Our findings suggest that CB2 does not adhere to the classical GPCR regulatory paradigm, entailing GRK- and β-arrestin-mediated desensitisation. Instead, C-terminal aspartic acid residues may act as phospho-mimics to induce β-arrestin activation. This study provides novel insights into the regulatory mechanisms of CB2 , which may aid in our understanding of drug tolerance and dependence.
    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
    The β-adrenergic receptors (βARs) are members of G protein-coupled receptor (GPCR) family and have been one of the most important GPCRs for studying receptor endocytosis and signaling pathway. Agonist binding of βARs leads to an activation of G proteins and their canonical effectors. In a parallel way, βAR stimulation triggers the termination of its signals by receptor desensitization. This termination process is initiated by G protein-coupled receptor kinase (GRK)-induced βAR phosphorylation that promotes the recruitment of β-arrestins to phosphorylated βAR. The uncoupled βARs which formed a complex with GRK and β-arrestin subsequently internalize into the cytosol. In addition, GRKs and β-arrestins also act as scaffolding proteins and signal transducers in their own functions to modulate various downstream effectors. Upon translocation to the βAR, β-arrestin is believed to undergo an important conformational change in the structure that is necessary for its signal transduction. The bioluminescence resonance energy transfer (BRET) technique involves the fusion of donor (luciferase) and acceptor (fluorescent) molecules to the interested proteins. Co-expression of these fusion proteins enables direct detection of their interactions in living cells. Here we describe the use of our established BRET technique to track the interaction of βAR with both GRK and β-arrestin. The assay described here allows the measurement of the BRET signal for detecting the interaction of β2AR with GRK2 and the conformational change of β-arrestin2 following βAR stimulation.
    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
    阿尔茨海默病(AD)的特征是淀粉样蛋白斑块中淀粉样蛋白β(Aβ)的细胞外沉积以及神经原纤维缠结(NFT)中过度磷酸化tau的细胞内聚集和积累。尽管已经确定了几种激酶有助于tau的病理磷酸化,针对AD的激酶靶向治疗在临床试验中尚未成功.严重的,负责许多确定的tau磷酸化位点的激酶仍然未知。G蛋白偶联受体(GPCR)激酶(GRKs)最近与非GPCR底物的磷酸化有关。例如,微管蛋白和α-突触核蛋白,在神经系统疾病中,包括精神分裂症和帕金森病。因此,我们研究了GRKs在AD病理生理学中的参与。
    我们对广泛表达的GRKs进行了全面的免疫组织化学和生化分析,即,对照受试者和AD患者的死后人脑组织中的GRK2、3、5和6。
    GRKs在神经元中显示出独特的细胞类型特异性表达模式,星形胶质细胞和小胶质细胞。在AD海马的CA1区域中,GRKs2、5和6的水平特异性降低。生化证据表明,GRKs与总量有区别联系,AD脑中的可溶性和不溶性tau库。补充免疫组织化学研究表明,GRKs与总tau差异共定位,磷酸化tau和NFT。值得注意的是,GRKs3和5也与淀粉样蛋白斑共同定位。
    这些研究建立了GRK与AD大脑中tau和淀粉样蛋白病理学的病理磷酸化和积累之间的联系,并提示了这些激酶在AD病理标志调节中的新作用。
    Alzheimer\'s disease (AD) is characterised by extracellular deposition of amyloid-β (Aβ) in amyloid plaques and intracellular aggregation and accumulation of hyperphosphorylated tau in neurofibrillary tangles (NFTs). Although several kinases have been identified to contribute to the pathological phosphorylation of tau, kinase-targeted therapies for AD have not been successful in clinical trials. Critically, the kinases responsible for numerous identified tau phosphorylation sites remain unknown. G protein-coupled receptor (GPCR) kinases (GRKs) have recently been implicated in phosphorylation of non-GPCR substrates, for example, tubulin and α-synuclein, and in neurological disorders, including schizophrenia and Parkinson\'s disease. Accordingly, we investigated the involvement of GRKs in the pathophysiology of AD.
    We performed a comprehensive immunohistochemical and biochemical analysis of the ubiquitously expressed GRKs, namely, GRK2, 3, 5 and 6, in postmortem human brain tissue of control subjects and AD patients.
    GRKs display unique cell-type-specific expression patterns in neurons, astrocytes and microglia. Levels of GRKs 2, 5 and 6 are specifically decreased in the CA1 region of the AD hippocampus. Biochemical evidence indicates that the GRKs differentially associate with total, soluble and insoluble pools of tau in the AD brain. Complementary immunohistochemical studies indicate that the GRKs differentially colocalise with total tau, phosphorylated tau and NFTs. Notably, GRKs 3 and 5 also colocalise with amyloid plaques.
    These studies establish a link between GRKs and the pathological phosphorylation and accumulation of tau and amyloid pathology in AD brains and suggest a novel role for these kinases in regulation of the pathological hallmarks of AD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号