G protein-coupled receptor

G 蛋白偶联受体
  • 文章类型: Journal Article
    松弛素-2是一种肽类激素,在人类心血管和生殖生物学中具有重要作用。它激活细胞反应如血管舒张的能力,血管生成,抗炎和抗纤维化作用引起了人们对使用松弛素-2作为心力衰竭和几种纤维化病症的治疗剂的极大兴趣。然而,重组松弛素-2具有非常短的血清半衰期,限制其临床应用。这里,我们提出了针对松弛素-2激素的蛋白质工程努力,以增加其血清半衰期,同时保持其激活G蛋白偶联受体RXFP1的能力。为了实现这一点,我们优化了松弛素-2和抗体Fc片段之间的融合,在小鼠体内产生一种循环半衰期约为3至5天的激素,同时在体内和体外均保留对RXFP1受体的有效激动剂活性。
    Relaxin-2 is a peptide hormone with important roles in human cardiovascular and reproductive biology. Its ability to activate cellular responses such as vasodilation, angiogenesis, and anti-inflammatory and antifibrotic effects has led to significant interest in using relaxin-2 as a therapeutic for heart failure and several fibrotic conditions. However, recombinant relaxin-2 has a very short serum half-life, limiting its clinical applications. Here, we present protein engineering efforts targeting the relaxin-2 hormone in order to increase its serum half-life while maintaining its ability to activate the G protein-coupled receptor RXFP1. To achieve this, we optimized a fusion between relaxin-2 and an antibody Fc fragment, generating a version of the hormone with a circulating half-life of around 3 to 5 days in mice while retaining potent agonist activity at the RXFP1 receptor both in vitro and in vivo.
    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
    促胰液素受体激动剂对心血管疾病有潜在的应用,胃肠,和代谢系统,然而,尚未开发出具有临床活性的该受体的非肽基激动剂。在目前的工作中,我们已经确定了一种新的小分子先导化合物具有这种药理学特征。我们已经围绕该噻二唑支架制备并表征了系统的结构-活性系列,以更好地了解其活性的分子决定因素。我们能够增强体外活性并保持母体化合物的特异性。我们发现最活跃的候选物在血浆中相当稳定,尽管它被肝微粒体代谢。这种化学探针应该可用于体外研究,需要测试体内药理活性。这可能是开发一流的促胰液素受体口服活性激动剂的重要线索,这可能对多种疾病状态有用。
    Agonists of the secretin receptor have potential applications for diseases of the cardiovascular, gastrointestinal, and metabolic systems, yet no clinically-active non-peptidyl agonists of this receptor have yet been developed. In the current work, we have identified a new small molecule lead compound with this pharmacological profile. We have prepared and characterized a systematic structure-activity series around this thiadiazole scaffold to better understand the molecular determinants of its activity. We were able to enhance the in vitro activity and to maintain the specificity of the parent compound. We found the most active candidate to be quite stable in plasma, although it was metabolized by hepatic microsomes. This chemical probe should be useful for in vitro studies and needs to be tested for in vivo pharmacological activity. This could be an important lead toward the development of a first-in-class orally active agonist of the secretin receptor, which could be useful for multiple disease states.
    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
    药物遗传学领域,研究一种或多种序列变异对药物反应表型的影响,是药物基因组学的一个特例,采用全基因组方法的学科。大规模平行,下一代测序(NGS),允许药物基因组学将药物遗传学纳入与应答者和非应答者相关的变体的鉴定,最佳药物反应,以及药物不良反应。在来自整个基因组的信号的背景下,必须考虑大量罕见和常见的天然存在的GPCR变体。针对G蛋白偶联受体(GPCR)基因建立了许多药物遗传学基础,因为它们是大量治疗药物的主要靶标。功能研究,证明可能致病性和致病性GPCR变异,已成为建立用于计算机分析的模型不可或缺的一部分。GPCR基因的变体包括编码和非编码单核苷酸变体以及影响细胞表面表达的插入或缺失(indel)(运输,二聚化,和脱敏/下调),配体结合和G蛋白偶联,和导致可变剪接编码同种型/可变表达的变体。随着GPCR基因组数据广度的增加,我们可能会预期增加药物标签的使用,这些标签指出对GPCR靶向药物的临床使用有显著影响的变体.我们讨论了GPCR药物基因组数据的含义,这些数据来自对受体结构和功能以及受体-配体相互作用进行了良好表型鉴定的个体的基因组。以及优化药物选择对患者的潜在益处。讨论的例子包括SARS-CoV-2(COVID-19)感染中的肾素-血管紧张素系统,趋化因子受体在细胞因子风暴中的可能作用,和潜在的蛋白酶激活受体(PAR)干预。专用于GPCRs的资源,包括公开可用的计算工具,也讨论了。
    The field of pharmacogenetics, the investigation of the influence of one or more sequence variants on drug response phenotypes, is a special case of pharmacogenomics, a discipline that takes a genome-wide approach. Massively parallel, next generation sequencing (NGS), has allowed pharmacogenetics to be subsumed by pharmacogenomics with respect to the identification of variants associated with responders and non-responders, optimal drug response, and adverse drug reactions. A plethora of rare and common naturally-occurring GPCR variants must be considered in the context of signals from across the genome. Many fundamentals of pharmacogenetics were established for G protein-coupled receptor (GPCR) genes because they are primary targets for a large number of therapeutic drugs. Functional studies, demonstrating likely-pathogenic and pathogenic GPCR variants, have been integral to establishing models used for in silico analysis. Variants in GPCR genes include both coding and non-coding single nucleotide variants and insertion or deletions (indels) that affect cell surface expression (trafficking, dimerization, and desensitization/downregulation), ligand binding and G protein coupling, and variants that result in alternate splicing encoding isoforms/variable expression. As the breadth of data on the GPCR genome increases, we may expect an increase in the use of drug labels that note variants that significantly impact the clinical use of GPCR-targeting agents. We discuss the implications of GPCR pharmacogenomic data derived from the genomes available from individuals who have been well-phenotyped for receptor structure and function and receptor-ligand interactions, and the potential benefits to patients of optimized drug selection. Examples discussed include the renin-angiotensin system in SARS-CoV-2 (COVID-19) infection, the probable role of chemokine receptors in the cytokine storm, and potential protease activating receptor (PAR) interventions. Resources dedicated to GPCRs, including publicly available computational tools, are also discussed.
    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
    初级纤毛是从大多数哺乳动物细胞的表面突出的薄薄的毛发样细胞器。它们充当专门的细胞天线,可以响应特定的刺激而变化很大。然而,纤毛长度的变化对细胞信号和行为的影响尚不清楚.因此,我们旨在表征由不同化学试剂诱导的细长初级纤毛,氯化锂(LiCl),氯化钴(CoCl2),和鱼藤酮,使用表达睫状G蛋白偶联受体(GPCR)的人视网膜色素上皮1(hRPE1)细胞,黑色素浓缩激素(MCH)受体1(MCHR1)。MCH主要通过MCHR1介导的Akt磷酸化诱导纤毛缩短。因此,我们验证了MCH-MCHR1轴在细长纤毛中的正常功能。尽管MCH缩短了LiCl和鱼藤酮延长的纤毛,它没有缩短CoCl2诱导的细长纤毛,表现出较少的Akt磷酸化。此外,发现血清重新添加会延迟CoCl2诱导的细长纤毛的纤毛缩短。相比之下,鱼藤酮诱导的细长纤毛通过纤毛尖端的切碎机制迅速缩短。最后,我们发现,在纤毛延长的细胞中,每种化学物质对纤毛GPCR信号传导和血清介导的纤毛结构动力学产生不同的影响.这些结果为理解纤毛长度变化的功能后果提供了基础。
    Primary cilia are thin hair-like organelles that protrude from the surface of most mammalian cells. They act as specialized cell antennas that can vary widely in response to specific stimuli. However, the effect of changes in cilia length on cellular signaling and behavior remains unclear. Therefore, we aimed to characterize the elongated primary cilia induced by different chemical agents, lithium chloride (LiCl), cobalt chloride (CoCl2), and rotenone, using human retinal pigmented epithelial 1 (hRPE1) cells expressing ciliary G protein-coupled receptor (GPCR), melanin-concentrating hormone (MCH) receptor 1 (MCHR1). MCH induces cilia shortening mainly via MCHR1-mediated Akt phosphorylation. Therefore, we verified the proper functioning of the MCH-MCHR1 axis in elongated cilia. Although MCH shortened cilia that were elongated by LiCl and rotenone, it did not shorten CoCl2-induced elongated cilia, which exhibited lesser Akt phosphorylation. Furthermore, serum readdition was found to delay cilia shortening in CoCl2-induced elongated cilia. In contrast, rotenone-induced elongated cilia rapidly shortened via a chopping mechanism at the tip of the cilia. Conclusively, we found that each chemical exerted different effects on ciliary GPCR signaling and serum-mediated ciliary structure dynamics in cells with elongated cilia. These results provide a basis for understanding the functional consequences of changes in ciliary length.
    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
    1型大麻素受体(CB1R)介导中枢神经体系神经递质释放和突触可塑性。内源性,植物衍生的,合成大麻素与CB1R结合,启动抑制性G蛋白(Gi)和β-抑制蛋白信号通路。在Gi信号通路中,CB1R激活G蛋白门控,向内整流钾(GIRK)通道。β-抑制蛋白途径通过受体内化减少细胞表面的CB1R表达。由于它们与镇痛和药物耐受性有关,GIRK通道和受体内化对药物的开发具有重要意义。这项研究使用了具有pH敏感性的永生化小鼠垂体细胞,荧光标记的人CB1R(AtT20-SEPCB1)以测量GIRK通道活性和CB1R内化。通过使用荧光膜电位敏感染料测量大麻素诱导的GIRK通道活性。我们开发了一种动力学成像测定法,可可视化和测量CB1R内化。所有大麻素刺激GIRK通道反应的排序效能为WIN55,212-2>(±)CP55,940>Δ9-THC>AEA。功效相对于(±)CP55,940表示,其排序功效为(±)CP55,940>WIN55,212-2>AEA>Δ9-THC。所有大麻素均以(±)CP55,940>WIN55,212-2>AEA>Δ9-THC的等级顺序刺激CB1R内化。内化功效归一化为(±)CP55,940,排序功效为WIN55,212-2>AEA>(±)CP55,940>Δ9-THC。(±)CP55,940在刺激GIRK通道反应方面比AEA和Δ9-THC显着更有效和有效;CB1R内化在效力和功效之间没有观察到显着差异。比较大麻素的GIRK通道和CB1R内化反应时,没有发现显着差异。总之,AtT20-SEPCB1细胞可用于评估大麻素诱导的CB1R内化。虽然大麻素显示差异Gi信号时,彼此比较,这并没有扩展到CB1R内化.
    The type 1 cannabinoid receptor (CB1R) mediates neurotransmitter release and synaptic plasticity in the central nervous system. Endogenous, plant-derived, synthetic cannabinoids bind to CB1R, initiating the inhibitory G-protein (Gi) and the β-arrestin signaling pathways. Within the Gi signaling pathway, CB1R activates G protein-gated, inwardly-rectifying potassium (GIRK) channels. The β-arrestin pathway reduces CB1R expression on the cell surface through receptor internalization. Because of their association with analgesia and drug tolerance, GIRK channels and receptor internalization are of interest to the development of pharmaceuticals. This research used immortalized mouse pituitary gland cells transduced with a pH-sensitive, fluorescently-tagged human CB1R (AtT20-SEPCB1) to measure GIRK channel activity and CB1R internalization. Cannabinoid-induced GIRK channel activity is measured by using a fluorescent membrane-potential sensitive dye. We developed a kinetic imaging assay that visualizes and measures CB1R internalization. All cannabinoids stimulated a GIRK channel response with a rank order potency of WIN55,212-2 > (±)CP55,940 > Δ9-THC > AEA. Efficacy was expressed relative to (±)CP55,940 with a rank order efficacy of (±)CP55,940 > WIN55, 212-2 > AEA > Δ9-THC. All cannabinoids stimulated CB1R internalization with a rank order potency of (±)CP55,940 > WIN55, 212-2 > AEA > Δ9-THC. Internalization efficacy was normalized to (±)CP55,940 with a rank order efficacy of WIN55,212-2 > AEA > (±)CP55,940 > Δ9-THC. (±)CP55,940 was significantly more potent and efficacious than AEA and Δ9-THC at stimulating a GIRK channel response; no significant differences between potency and efficacy were observed with CB1R internalization. No significant differences were found when comparing a cannabinoid\'s GIRK channel and CB1R internalization response. In conclusion, AtT20-SEPCB1 cells can be used to assess cannabinoid-induced CB1R internalization. While cannabinoids display differential Gi signaling when compared to each other, this did not extend to CB1R internalization.
    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
    CC趋化因子受体2和CCL2高度参与癌症的生长和转移,免疫逃逸。实体瘤中钠离子浓度的升高也与转移和免疫调节相关。钠离子可以通过钠离子结合位点调节A类G蛋白偶联受体,其特征是高度保守的天冬氨酸残基(D2.50),也存在于CCR2中。因此,我们通过放射性配体结合研究和诱变进一步探索了CCR2中的这个结合位点。研究了钠离子和阿米洛利衍生物对三种明显结合的放射性配体的调节。观察到钠离子是拮抗剂结合的相对弱的调节剂,但显著增加了125I-CCL2与CCR2的解离。6-取代的六亚甲基阿米洛利(HMA)调节所有测试的放射性配体。HMA在CCR2的假定钠离子结合位点中的诱导拟合对接证实了其结合位点。最后,对钠离子结合位点(癌症相关)突变的研究显示,与野生型相比,表达显着降低。只有两个变种人,G123A3.35和G127K3.39能够被[3H]INCB3344和[3H]CCR2-RA-[R]结合。因此,诱变表明,钠离子结合位点残基,与其他A类GPCRs不同,与趋化因子受体进化有关,对受体完整性至关重要。此外,测试的突变似乎对HMA观察到的调节没有影响,或者对测试的放射性配体的氯化钠调节有较小的影响。总而言之,这些结果邀请进一步探索(癌症)生物学中的CCR2钠离子结合位点,并可能作为第三个药物结合位点。
    CC chemokine receptor 2 and CCL2 are highly involved in cancer growth and metastasis, and immune escape. Raised sodium ion concentrations in solid tumours have also been correlated to metastasis and immune modulation. Sodium ions can modulate class A G protein-coupled receptors through the sodium ion binding site characterized by a highly conserved aspartic acid residue (D2.50), also present in CCR2. Hence, we further explored this binding site in CCR2 by radioligand binding studies and mutagenesis. Modulation of three distinctly binding radioligands by sodium ions and amiloride derivates was investigated. Sodium ions were observed to be relatively weak modulators of antagonist binding, but substantially increased 125I-CCL2 dissociation from CCR2. 6-Substituted Hexamethylene Amiloride (HMA) modulated all tested radioligands. Induced-fit docking of HMA in the presumed sodium ion binding site of CCR2 confirmed its binding site. Finally, investigation of (cancer-associated) mutations in the sodium ion binding site showed a markedly decreased expression compared to wild type. Only two mutants, G123A3.35 and G127K3.39, were able to be bound by [3H]INCB3344 and [3H]CCR2-RA-[R]. Thus, mutagenesis showed that the sodium ion binding site residues, which are distinct from other class A GPCRs and related to chemokine receptor evolution, are crucial for receptor integrity. Moreover, the tested mutations appeared to have no effect on modulation observed by HMA or a minor effect on sodium chloride modulation on the tested radioligands. All in all, these results invite further exploration of the CCR2 sodium ion binding site in (cancer) biology, and potentially as a third druggable binding site.
    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
    虽然阿片类镇痛药在治疗疼痛中是不可或缺的,这些药物伴随着危及生命的副作用。虽然临床相关阿片类药物靶向µ阿片受体(MOR),MOR和δ阿片受体(DOR)之间的异二聚体已成为开发更安全镇痛药的另一个目标。尽管到目前为止已经报道了一些异二聚体优选的激动剂,在MOR或DOR单体/同二聚体的存在下选择性地激活MOR/DOR异二聚体仍然是困难的。为了获得开发MOR/DOR选择性激动剂的见解,在本文中,我们制备了CYM51010的类似物,CYM51010是已报道的异二聚体优选激动剂之一,并收集结构-活动关系信息。我们发现乙氧羰基是异二聚体活性所必需的,虽然这个基团可以用相似大小的官能团取代,例如乙氧基羰基。至于乙酰氨基苯基,不是一种取代基,相反,位于特定位置(对位)的取代基对活性至关重要。改变乙酰氨基苯基和哌啶部分之间的接头长度也对活性具有有害影响。另一方面,用三氟乙酰氨基取代乙酰氨基和用苄基取代苯乙基降低了单体/同二聚体的活性,同时保持了MOR/DOR的活性,这增强了选择性。我们在本文中的发现将在开发MOR/DOR的选择性激动剂以及阐明该异二聚体在镇痛过程中和副作用的建立中的生理作用中起重要作用。
    Although opioid analgesics are indispensable in treating pain, these drugs are accompanied by life-threatening side effects. While clinically relevant opioid drugs target the µ opioid receptor (MOR), a heterodimer between the MOR and the δ opioid receptor (DOR) has emerged as another target to develop safer analgesics. Although some heterodimer-preferring agonists have been reported so far, it is still difficult to activate the MOR/DOR heterodimer selectively in the presence of MOR or DOR monomers/homodimers. To gain insights to develop selective agonists for MOR/DOR, herein we prepared analogs of CYM51010, one of the reported heterodimer-preferring agonists, and collected structure-activity relationship information. We found that the ethoxycarbonyl group was needed for the activity for the heterodimer, although this group could be substituted with functional groups with similar sizes, such as an ethoxycarbonyl group. As for the acetylaminophenyl group, not a type of substituent, but rather a substituent located at a specific position (para-position) was essential for the activity. Changing the linker length between the acetylaminophenyl group and the piperidine moiety also had deleterious effects on the activity. On the other hand, the substitution of the acetylamino group with a trifluoroacetylamino group and the substitution of the phenethyl group with a benzyl group diminished the activities for the monomers/homodimers while keeping the activity for MOR/DOR, which enhanced the selectivity. Our findings herein will play an important role in developing selective agonists for MOR/DOR and for elucidating the physiological roles of this heterodimer in analgesic processes and in the establishment of side effects.
    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
    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
    当前免疫学中的一个关键问题是先天免疫系统如何产生高水平的特异性。我们先前在秀丽隐杆线虫中的研究表明,NMUR-1是一种神经元G蛋白偶联受体,与神经肽神经蛋白U(NMU)的哺乳动物受体同源,调节对不同细菌病原体的不同先天免疫反应。这里,通过使用定量蛋白质组学和功能测定,我们发现NMUR-1调节F1FOATP合酶和ATP的产生,以响应病原体感染,并且这种调节有助于NMUR-1介导的先天免疫特异性。我们进一步证明,ATP的生物合成及其对防御的贡献受NMUR-1配体CAPA-1及其表达神经元ASG的神经控制。这些发现表明,NMUR-1神经信号通过控制能量稳态作为防御病原体的一部分来调节先天免疫的特异性。我们的研究提供了对NMU信号在整个动物门免疫中的新兴作用的机制见解。
    A key question in current immunology is how the innate immune system generates high levels of specificity. Our previous study in Caenorhabditis elegans revealed that NMUR-1, a neuronal G protein-coupled receptor homologous to mammalian receptors for the neuropeptide neuromedin U (NMU), regulates distinct innate immune responses to different bacterial pathogens. Here, by using quantitative proteomics and functional assays, we discovered that NMUR-1 regulates F1FO ATP synthase and ATP production in response to pathogen infection, and that such regulation contributes to NMUR-1-mediated specificity of innate immunity. We further demonstrated that ATP biosynthesis and its contribution to defense is neurally controlled by the NMUR-1 ligand CAPA-1 and its expressing neurons ASG. These findings indicate that NMUR-1 neural signaling regulates the specificity of innate immunity by controlling energy homeostasis as part of defense against pathogens. Our study provides mechanistic insights into the emerging roles of NMU signaling in immunity across animal phyla.
    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
    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

公众号