G protein-coupled receptor

G 蛋白偶联受体
  • 文章类型: Journal Article
    血管性痴呆(VD),进行性血管性认知障碍,以存在脑灌注不足为特征,血脑屏障通透性增加,和白质病变。尽管目前的治疗策略主要集中在高血压等危险因素上,糖尿病,还有心脏病,目前缺乏有效的靶向治疗,VD的潜在机制尚不清楚.我们以前发现Apelin受体(APJ),它们是G蛋白偶联受体(GPCRs),可以同二聚化并产生与VD大鼠中APJ单体不同的信号。Apelin-13降低APJ同二聚体水平,导致海马齿状回区内源性神经干细胞增殖,表明它有神经保护作用.在这项研究中,我们建立了大鼠和细胞氧糖剥夺/复氧VD模型,以研究APJ同源二聚体对自噬的影响。我们发现APJ同二聚体通过Gαq和PI3K/Akt/mTOR通路抑制自噬来保护VD,体内和体外。这一发现为慢性脑缺血再灌注疾病提供了有希望的治疗靶标,并为开发靶向APJ同源二聚体的药物提供了实验基础。
    Vascular dementia (VD), a progressive vascular cognitive impairment, is characterised by the presence of cerebral hypoperfusion, increased blood-brain barrier permeability, and white matter lesions. Although current treatment strategies primarily focus on risk factors such as hypertension, diabetes, and heart disease, efficient and targeted therapies are lacking and the underlying mechanisms of VD remain unclear. We previously discovered that Apelin receptors (APJ), which are G protein-coupled receptors (GPCRs), can homodimerize and generate signals that are distinct from those of APJ monomers in VD rats. Apelin-13 reduces the level of APJ homodimers and leads to the proliferation of endogenous neural stem cells in the hippocampal dentate gyrus area, suggesting that it has a neuroprotective role. In this study, we established a rat and cellular oxygen-glucose deprivation/reoxygenation VD model to investigate the impact of APJ homodimerisation on autophagy. We found that APJ homodimers protect against VD by inhibiting autophagy through the Gαq and PI3K/Akt/mTOR pathways upon Gαi signalling, both in vivo and in vitro. This discovery provides a promising therapeutic target for chronic cerebral ischaemia-reperfusion diseases and an experimental foundation for the development of drugs that target APJ homodimers.
    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
    Morphinan拮抗剂,阻断阿片样物质对μ阿片样物质受体的作用,已经研究了它们的镇痛潜力。先前的研究已经表明,这些拮抗剂在突变型μ-阿片受体(MOR;S196A)存在的情况下引起镇痛作用较少的副作用。然而,在医学应用中引入突变受体代表了重大挑战。我们假设化学化合物与MOR的结合可能引起与S196A突变相当的作用。通过高通量筛选和构效关系研究,我们确定了一个调制器,4-(2-(4-氟苯基)-4-氧代噻唑烷-3-基)-3-甲基苯甲酸(BPRMU191),赋予小分子吗啡喃拮抗剂激动特性,诱导G蛋白依赖性MOR激活。BPRMU191和吗啡喃拮抗剂的共同应用导致MOR依赖性镇痛,副作用减少。包括胃肠功能紊乱,抗伤害耐受性,身体和心理上的依赖。结合BPRMU191和吗啡喃拮抗剂可以作为严重疼痛的潜在治疗策略,减少不良反应,并为研究G蛋白偶联受体调节提供途径。
    Morphinan antagonists, which block opioid effects at mu-opioid receptors, have been studied for their analgesic potential. Previous studies have suggested that these antagonists elicit analgesia with fewer adverse effects in the presence of the mutant mu-opioid receptor (MOR; S196A). However, introducing a mutant receptor for medical applications represents significant challenges. We hypothesize that binding a chemical compound to the MOR may elicit a comparable effect to the S196A mutation. Through high-throughput screening and structure-activity relationship studies, we identified a modulator, 4-(2-(4-fluorophenyl)-4-oxothiazolidin-3-yl)-3-methylbenzoic acid (BPRMU191), which confers agonistic properties to small-molecule morphinan antagonists, which induce G protein-dependent MOR activation. Co-application of BPRMU191 and morphinan antagonists resulted in MOR-dependent analgesia with diminished side effects, including gastrointestinal dysfunction, antinociceptive tolerance, and physical and psychological dependence. Combining BPRMU191 and morphinan antagonists could serve as a potential therapeutic strategy for severe pain with reduced adverse effects and provide an avenue for studying G protein-coupled receptor modulation.
    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
    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: Journal Article
    喂养行为,最基本的生理活动,由两组相对的因素控制,致食欲和厌食因素。sulfakinin家族,哺乳动物饱腹因子胆囊收缩素(CCK)的昆虫类似物,已被证明可以抑制各种昆虫的食物摄入。然而,sulfakinin调节摄食行为的机制仍然是一个生物学问题。本研究旨在阐明家蚕减食肽sulfakinin介导的信号通路。我们通过功能测定鉴定了Bombyxmori神经肽G蛋白偶联受体A9(BNGR-A9)作为sulfakinin的受体。用sulfakinin刺激引发了细胞内IP3,Ca2+的迅速增加,和ERK1/2磷酸化的显著增强,以对Gαq特异性抑制剂敏感的方式。用合成的磺胺酶治疗导致食物消耗和平均体重减少。此外,给蚕施用合成的磺胺基素显着升高血淋巴海藻糖水平,通过用BNGR-A9dsRNA预处理显著降低的效果。因此,我们的发现确立了sulfakinin/BNGR-A9信号通路作为家蚕摄食行为和血淋巴海藻糖稳态的关键调节因子,强调其在食物摄入的负控制和能量平衡的正调节中的作用。
    Feeding behavior, the most fundamental physiological activity, is controlled by two opposing groups of factors, orexigenic and anorexigenic factors. The sulfakinin family, an insect analogue of the mammalian satiety factor cholecystokinin (CCK), has been shown to suppress food intake in various insects. Nevertheless, the mechanisms through which sulfakinin regulates feeding behavior remain a biological question. This study aimed to elucidate the signaling pathway mediated by the anorexigenic peptide sulfakinin in Bombyx mori. We identified the Bombyx mori neuropeptide G protein-coupled receptor A9 (BNGR-A9) as the receptor for sulfakinin through functional assays. Stimulation with sulfakinin triggered a swift increase in intracellular IP3, Ca2+, and a notable enhancement of ERK1/2 phosphorylation, in a manner sensitive to a Gαq-specific inhibitor. Treatment with synthetic sulfakinin resulted in decreased food consumption and average body weight. Additionally, administering synthetic sulfakinin to silkworms significantly elevated hemolymph trehalose levels, an effect markedly reduced by pre-treatment with BNGR-A9 dsRNA. Consequently, our findings establish the sulfakinin/BNGR-A9 signaling pathway as a critical regulator of feeding behavior and hemolymph trehalose homeostasis in Bombyx mori, highlighting its roles in the negative control of food intake and the positive regulation of energy balance.
    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:卵巢癌(OV)是女性生殖系统常见的恶性肿瘤,5年生存率约为30%。无效的早期诊断和预后导致大多数患者的生存不良。G蛋白偶联受体(GPCRs,人类细胞表面受体的最大家族)与OV相关。我们旨在鉴定GPCR相关基因(GPCRRG)特征,并开发一种新的模型来预测OV预后。
    方法:我们从癌症基因组图谱(TCGA)和基因表达综合(GEO)数据库下载了数据。使用最小绝对收缩和选择算子(LASSO)Cox回归分析筛选预后GPCRRGs,并建立了预后模型。通过Kaplan-Meier(K-M)生存分析评估模型的预测能力。使用定量逆转录聚合酶链反应检查正常和OV细胞系中GPCRRG的水平。使用单样本基因集富集分析(ssGSEA)和CIBERSORT分析了高危和低危人群的免疫学特征。
    结果:根据风险评分,17个GPCRRGs与OV预后相关。CXCR4,GPR34,LGR6,LPAR3和RGS2在三个OV数据集中显着表达,并且能够进行准确的OV诊断。预后模型的K-M分析表明,它可以区分高危和低危患者,对应于较差和更好的预测,分别。GPCRRG表达与免疫浸润率相关。
    结论:我们的预后模型阐述了GPCRRGs在OV中的作用,为OV患者的预后和免疫反应预测提供了新的工具。
    BACKGROUND: Ovarian cancer (OV) is a common malignant tumor of the female reproductive system with a 5-year survival rate of ∼30 %. Inefficient early diagnosis and prognosis leads to poor survival in most patients. G protein-coupled receptors (GPCRs, the largest family of human cell surface receptors) are associated with OV. We aimed to identify GPCR-related gene (GPCRRG) signatures and develop a novel model to predict OV prognosis.
    METHODS: We downloaded data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Prognostic GPCRRGs were screened using least absolute shrinkage and selection operator (LASSO) Cox regression analysis, and a prognostic model was constructed. The predictive ability of the model was evaluated by Kaplan-Meier (K-M) survival analysis. The levels of GPCRRGs were examined in normal and OV cell lines using quantitative reverse-Etranscription polymerase chain reaction. The immunological characteristics of the high- and low-risk groups were analyzed using single-sample gene set enrichment analysis (ssGSEA) and CIBERSORT.
    RESULTS: Based on the risks scores, 17 GPCRRGs were associated with OV prognosis. CXCR4, GPR34, LGR6, LPAR3, and RGS2 were significantly expressed in three OV datasets and enabled accurate OV diagnosis. K-M analysis of the prognostic model showed that it could differentiate high- and low-risk patients, which correspond to poorer and better prognoses, respectively. GPCRRG expression was correlated with immune infiltration rates.
    CONCLUSIONS: Our prognostic model elaborates on the roles of GPCRRGs in OV and provides a new tool for prognosis and immune response prediction in patients with OV.
    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
    G蛋白偶联受体(GPCRs)是现有药物治疗多种人类疾病的主要靶标,并主导着药物市场。然而,超过50%的GPCRs仍然无法使用。追求突破,克服这种局面,在开发特异性靶向GPCRs的基于RNA的药物方面有重要的临床研究,但到目前为止还没有批准.RNA疗法代表了一种独特且有前途的方法,可以选择性地靶向以前无法药物的靶标。包括不可用的GPCR。然而,RNA治疗剂的开发在RNA稳定性和有效体内递送方面面临重大挑战。这篇综述概述了RNA疗法和各种类型的纳米颗粒RNA递送系统的进展。它还描述了GPCR靶向RNA药物在各种人类疾病中的潜在应用。
    G protein-coupled receptors (GPCRs) are the major targets of existing drugs for a plethora of human diseases and dominate the pharmaceutical market. However, over 50% of the GPCRs remain undruggable. To pursue a breakthrough and overcome this situation, there is significant clinical research for developing RNA-based drugs specifically targeting GPCRs, but none has been approved so far. RNA therapeutics represent a unique and promising approach to selectively targeting previously undruggable targets, including undruggable GPCRs. However, the development of RNA therapeutics faces significant challenges in areas of RNA stability and efficient in vivo delivery. This review presents an overview of the advances in RNA therapeutics and the diverse types of nanoparticle RNA delivery systems. It also describes the potential applications of GPCR-targeted RNA drugs for various human diseases.
    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)是细胞膜蛋白的一大家族,是药物发现的重要靶标。纳米抗体,也称为VHH(仅重链抗体的可变结构域,HcAbs)抗体,是源自骆驼科动物的小抗体片段,由于其优于常规抗体的优势,其作为靶向GPCRs的潜在治疗剂而获得了极大的关注。然而,在开发靶向GPCRs的纳米抗体方面存在挑战,其中表位的可及性是最重要的,因为细胞膜部分屏蔽了GPCR表面。我们开发了一种通用方案,用于使用GPCR过表达HEK293细胞的细胞膜提取物作为美洲驼/羊驼免疫抗原来制备靶向GPCR的纳米抗体。我们构建了免疫VHH文库,并通过噬菌体展示生物淘选鉴定了纳米抗体。单克隆纳米抗体在大肠杆菌中重组表达(E.大肠杆菌)并纯化以表征它们的结合效力。
    G protein-coupled receptors (GPCRs) are a large family of cell membrane proteins that are important targets for drug discovery. Nanobodies, also known as VHH (variable domains of heavy chain-only antibodies, HcAbs) antibodies, are small antibody fragments derived from camelids that have gained significant attention as potential therapeutics for targeting GPCRs due to their advantages over conventional antibodies. However, there are challenges in developing nanobodies targeting GPCRs, among which epitope accessibility is the most significant because the cell membrane partially shields the GPCR surface. We developed a universal protocol for making nanobodies targeting GPCRs using the cell membrane extract of GPCR-overexpressing HEK293 cells as the llama/alpaca immunization antigen. We constructed an immune VHH library and identified nanobodies by phage display bio-panning. The monoclonal nanobodies were recombinantly expressed in Escherichia coli (E. coli) and purified to characterize their binding potency.
    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),它们构成了人类最大的膜蛋白受体家族,是高度复杂的信号系统,具有复杂的结构和动态的构象和位置。在这些受体中,一个特定的子集被称为孤儿GPCRs(oGPCRs),由于它们在中枢和周围神经系统功能中的作用,在疼痛研究中引起了极大的兴趣。GPCR功能的多样性归因于多种因素,包括变构调节剂,信号偏差,低聚,本构信号,和分隔的信号。这篇综述主要集中在疼痛机制的oGPCR研究的最新进展。讨论特定oGPCR的作用,包括GPR34,GPR37,GPR65,GPR83,GPR84,GPR85,GPR132,GPR151,GPR160,GPR171,GPR177和GPR183。还简要描述了这些受体中与中枢神经系统疾病有关的孤儿受体。了解这些oGPCRs的功能不仅有助于更深入地了解疼痛机制,而且为发现疼痛治疗的新靶点提供参考。
    G protein-coupled receptors (GPCRs), which form the largest family of membrane protein receptors in humans, are highly complex signaling systems with intricate structures and dynamic conformations and locations. Among these receptors, a specific subset is referred to as orphan GPCRs (oGPCRs) and has garnered significant interest in pain research due to their role in both central and peripheral nervous system function. The diversity of GPCR functions is attributed to multiple factors, including allosteric modulators, signaling bias, oligomerization, constitutive signaling, and compartmentalized signaling. This review primarily focuses on the recent advances in oGPCR research on pain mechanisms, discussing the role of specific oGPCRs including GPR34, GPR37, GPR65, GPR83, GPR84, GPR85, GPR132, GPR151, GPR160, GPR171, GPR177, and GPR183. The orphan receptors among these receptors associated with central nervous system diseases are also briefly described. Understanding the functions of these oGPCRs can contribute not only to a deeper understanding of pain mechanisms but also offer a reference for discovering new targets for pain treatment.
    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
    几十年来,通过G蛋白偶联受体(GPCRs)的信号转导一直是细胞生物学的主要焦点。许多病症与利用Gi蛋白抑制腺苷酸环化酶(AC)以及调节其他效应物的GPCR相关。一些早期研究通过测量组成型活性Gαi突变体的推定区域的同源替换后的活性丧失,成功地定义了Gαi几个成员的AC相互作用域。然而,这些发现是否确实可以转化为受体激活的Gαi的背景尚未得到严格验证.为了解决这个问题,我们在Gαi1的GTP酶缺陷型Q204L(QL)和R178C(RC)突变体中引入了一系列已知和新的嵌合突变,然后对其抑制AC的能力进行了检查.令人惊讶的是,大多数嵌合体未能消除QL突变带来的组成活性,而一些能够消除RC突变体的抑制活性。在携带百日咳毒素(PTX)抗性C351I突变的相同嵌合构建体中类似地观察到受体介导的AC抑制。此外,带有RC的功能丧失嵌合体似乎被内源性RGS蛋白过度失活。分子对接揭示了AC与Gαi1的α3/β5环之间的潜在相互作用。随后的cAMP测定支持α3/β5环的协同作用,α4螺旋,和α4/β6环介导Gαi1-i3对AC的抑制。我们的结果揭示了组成型活性突变体和受体激活的Gαi1之间抑制AC的显着功能差异,并确定了以前未知的Gαi亚基的AC相互作用域。这些结果共同为细胞环境中AC抑制的机制提供了有价值的见解。
    Signal transduction through G protein-coupled receptors (GPCRs) has been a major focus in cell biology for decades. Numerous disorders are associated with GPCRs that utilize Gi proteins to inhibit adenylyl cyclase (AC) as well as regulate other effectors. Several early studies have successfully defined the AC-interacting domains of several members of Gαi by measuring the loss of activity upon homologous replacements of putative regions of constitutive active Gαi mutants. However, whether such findings can indeed be translated into the context of a receptor-activated Gαi have not been rigorously verified. To address this issue, an array of known and new chimeric mutations was introduced into GTPase-deficient Q204L (QL) and R178C (RC) mutants of Gαi1, followed by examinations on their ability to inhibit AC. Surprisingly, most chimeras failed to abolish the constitutive activity brought on by the QL mutation, while some were able to eliminate the inhibitory activity of RC mutants. Receptor-mediated inhibition of AC was similarly observed in the same chimeric constructs harbouring the pertussis toxin (PTX)-resistant C351I mutation. Moreover, RC-bearing loss-of-function chimeras appeared to be hyper-deactivated by endogenous RGS protein. Molecular docking revealed a potential interaction between AC and the α3/β5 loop of Gαi1. Subsequent cAMP assays support a cooperative action of the α3/β5 loop, the α4 helix, and the α4/β6 loop in mediating AC inhibition by Gαi1-i3. Our results unveiled a notable functional divergence between constitutively active mutants and receptor-activated Gαi1 to inhibit AC, and identified a previously unknown AC-interacting domain of Gαi subunits. These results collectively provide valuable insights on the mechanism of AC inhibition in the cellular environment.
    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
    在药物化学中使用特权支架是加速药物发现过程的有效途径,特别是在命中/领先优化阶段。2-苯基环丙基甲胺(PCPMA)是药物化学中不常用的化学支架,但是许多含有PCPMA的化合物对各种疾病都有治疗作用,特别是中枢神经系统(CNS)疾病,如抑郁症,精神分裂症,睡眠障碍,和帕金森病。PCPMA支架的骨架能够实现通过烷基接头与苯环连接的氨基的独特结构,使其成为设计生物活性化合物的有用模板,特别是用于CNS药物靶标,包括胺能GPCRs和转运蛋白。本文综述了含PCPMA的药物和药物样分子的药物化学研究,他们的行动机制,和生物活动。我们得出结论,PCPMA是CNS药物设计的独特且有用的特权支架。
    The use of privileged scaffolds in medicinal chemistry is an effective way to accelerate the drug discovery process, especially at the hit/lead optimization stage. 2-Phenylcyclopropylmethylamine (PCPMA) is a less commonly used chemical scaffold in medicinal chemistry, but many PCPMA-containing compounds exert therapeutic effects for various diseases, in particular central nervous system (CNS) diseases such as depression, schizophrenia, sleep disorder, and Parkinson\'s disease. The backbone of the PCPMA scaffold enables a unique structure of an amino group linked to a benzene ring through an alkyl linker, making it a useful template for the design of bioactive compounds especially for CNS drug targets including aminergic GPCRs and transporters. This review summarizes the medicinal chemistry studies of PCPMA-containing drugs and drug-like molecules, their mechanisms of action, and biological activities. We conclude that PCPMA is a unique and useful privileged scaffold for CNS drug design.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号