Receptors, G-Protein-Coupled

受体,G - 蛋白偶联
  • 文章类型: Journal Article
    卡路里限制增加寿命。在热量限制的组织特异性保护作用中,对胃肠道的影响尚不清楚.我们报告了嗜铬粒蛋白A阳性(+)的数量增加,包括促食性生长素释放肽+细胞,在热量限制小鼠的胃中。这种作用伴随着Notch靶标Hes1和Notch配体Jag1的增加,并通过用DAPT阻断Notch而逆转,γ-分泌酶抑制剂.原代培养和遗传修饰的报告小鼠表明,内分泌细胞丰度的增加是由于Lgr5干细胞和Neurog3内分泌祖细胞增殖的改变。与肠道不同,热量限制减少胃Lgr5+干细胞,同时以Notch依赖性方式增加内分泌祖细胞的FOXO1/Neurog3亚群。Further,FOXO1的激活足以促进独立于Notch的内分泌细胞分化。Notch抑制剂PF-03084014或生长素释放肽受体拮抗剂GHRP-6逆转了小鼠热量限制的表型效应。Tirzepatide还在小鼠中扩增生长素释放肽+细胞。总之,卡路里限制促进Notch依赖,FOXO1调节胃内分泌细胞分化。
    Calorie restriction increases lifespan. Among the tissue-specific protective effects of calorie restriction, the impact on the gastrointestinal tract remains unclear. We report increased numbers of chromogranin A-positive (+), including orexigenic ghrelin+ cells, in the stomach of calorie-restricted mice. This effect was accompanied by increased Notch target Hes1 and Notch ligand Jag1 and was reversed by blocking Notch with DAPT, a gamma-secretase inhibitor. Primary cultures and genetically modified reporter mice show that increased endocrine cell abundance is due to altered Lgr5+ stem and Neurog3+ endocrine progenitor cell proliferation. Different from the intestine, calorie restriction decreased gastric Lgr5+ stem cells, while increasing a FOXO1/Neurog3+ subpopulation of endocrine progenitors in a Notch-dependent manner. Further, activation of FOXO1 was sufficient to promote endocrine cell differentiation independent of Notch. The Notch inhibitor PF-03084014 or ghrelin receptor antagonist GHRP-6 reversed the phenotypic effects of calorie restriction in mice. Tirzepatide additionally expanded ghrelin+ cells in mice. In summary, calorie restriction promotes Notch-dependent, FOXO1-regulated gastric endocrine cell differentiation.
    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)主要受GPCR激酶(GRK)磷酸化和随后的β-抑制蛋白募集调节。广泛表达的GRKs分为胞浆GRK2/3和膜束缚的GRK5/6亚家族。GRK2/3与活化的G蛋白βγ亚基相互作用以转运至膜。然而,这种需要并不是偏见的一个因素,影响β-抑制素偏向激动剂创建的有效性。使用多种方法,如GRK2/3突变体不能与Gβγ相互作用,GRK2/3/5/6敲除细胞中的膜束缚GRKs和G蛋白抑制剂,我们表明,G蛋白激活将先于GRK2/3介导的β-arrestin2募集到激活的受体。这与游离Gβγ的来源无关,并且对于G-,Gi和Gq耦合的GPCR。因此,GRK2/3调节受体的β-抑制蛋白相互作用与G蛋白激活密不可分。我们概述了GRK对游离Gβγ的依赖性如何确定GPCR的潜在偏向性激动的理论框架。由于GRK2/3募集和受体磷酸化的这种固有细胞机制,我们预计β-抑制蛋白偏向的配体的产生对于仅由GRK2/3调节的GPCRs亚组具有机械挑战性,但对于GRK5/6调节的受体可实现,不需要释放的Gβγ。因此,任何GPCR的GRK特异性是开发抑制蛋白偏向的配体的基础。
    G protein-coupled receptors (GPCRs) are mainly regulated by GPCR kinase (GRK) phosphorylation and subsequent β-arrestin recruitment. The ubiquitously expressed GRKs are classified into cytosolic GRK2/3 and membrane-tethered GRK5/6 subfamilies. GRK2/3 interact with activated G protein βγ-subunits to translocate to the membrane. Yet, this need was not linked as a factor for bias, influencing the effectiveness of β-arrestin-biased agonist creation. Using multiple approaches such as GRK2/3 mutants unable to interact with Gβγ, membrane-tethered GRKs and G protein inhibitors in GRK2/3/5/6 knockout cells, we show that G protein activation will precede GRK2/3-mediated β-arrestin2 recruitment to activated receptors. This was independent of the source of free Gβγ and observable for Gs-, Gi- and Gq-coupled GPCRs. Thus, β-arrestin interaction for GRK2/3-regulated receptors is inseparably connected with G protein activation. We outline a theoretical framework of how GRK dependence on free Gβγ can determine a GPCR\'s potential for biased agonism. Due to this inherent cellular mechanism for GRK2/3 recruitment and receptor phosphorylation, we anticipate generation of β-arrestin-biased ligands to be mechanistically challenging for the subgroup of GPCRs exclusively regulated by GRK2/3, but achievable for GRK5/6-regulated receptors, that do not demand liberated Gβγ. Accordingly, GRK specificity of any GPCR is foundational for developing arrestin-biased ligands.
    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
    血吸虫病(SM)是由曼氏血吸虫引起的寄生虫病。SM引起寄生虫卵引起的慢性炎症,胶原/纤维化沉积在肝脏肉芽肿过程中,脾,脾中枢神经系统,肾脏,还有肺.肺动脉高压(PAH)是以肺循环高压和右心室超负荷为特征的临床表现。这项研究调查了在存在肝和PAH形式的人SM的情况下,针对G蛋白偶联受体(GPCR)的第二个环的功能性自身抗体(fAAB)的产生。
    呈现急性和慢性表现的未感染和感染个体(例如,肝肠,肝脾无PAH,并对SM的PAH)进行临床评估,并收集其血液以鉴定能够识别内皮素1,血管紧张素II的fAAB/GPCRs,和a-1肾上腺素能受体。在受体拮抗剂乌拉地尔存在下培养的大鼠心肌细胞中分析了人血清,氯沙坦,BQ123
    来自慢性肝和PAHSM个体的fAAB/GPCRs,但不是来自急性SM个体,识别三个受体。在拮抗剂的存在下,培养的心肌细胞的搏动率变化减少。此外,鉴定了fAAB的胞外域功能上的结合位点,发现IgG1和/或IgG3抗体与fAAB相关。
    我们的数据表明,抗GPCR的fAAB在慢性SM(肝和PAH)的血管活动中起重要作用,并且可能参与SM的高血压形式的发展。
    UNASSIGNED: Schistosomiasis (SM) is a parasitic disease caused by Schistosoma mansoni. SM causes chronic inflammation induced by parasitic eggs, with collagen/fibrosis deposition in the granuloma process in the liver, spleen, central nervous system, kidneys, and lungs. Pulmonary arterial hypertension (PAH) is a clinical manifestation characterized by high pressure in the pulmonary circulation and right ventricular overload. This study investigated the production of functional autoantibodies (fAABs) against the second loop of the G-protein-coupled receptor (GPCR) in the presence of hepatic and PAH forms of human SM.
    UNASSIGNED: Uninfected and infected individuals presenting acute and chronic manifestations (e.g., hepatointestinal, hepato-splenic without PAH, and hepato-splenic with PAH) of SM were clinically evaluated and their blood was collected to identify fAABs/GPCRs capable of recognizing endothelin 1, angiotensin II, and a-1 adrenergic receptor. Human serum was analyzed in rat cardiomyocytes cultured in the presence of the receptor antagonists urapidil, losartan, and BQ123.
    UNASSIGNED: The fAABs/GPCRs from chronic hepatic and PAH SM individuals, but not from acute SM individuals, recognized the three receptors. In the presence of the antagonists, there was a reduction in beating rate changes in cultured cardiomyocytes. In addition, binding sites on the extracellular domain functionality of fAABs were identified, and IgG1 and/or IgG3 antibodies were found to be related to fAABs.
    UNASSIGNED: Our data suggest that fAABs against GPCR play an essential role in vascular activity in chronic SM (hepatic and PAH) and might be involved in the development of hypertensive forms of SM.
    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
    胆汁酸(BAs)代谢对阿尔茨海默病(AD)的发病机制有重要影响。我们发现脱氧胆酸(DCA)在早期阶段在AD小鼠的大脑中增加。DCA的产生增强诱导胆汁酸受体武田G蛋白偶联受体(TGR5)的上调,在早期AD小鼠大脑的神经元中也特别增加。外源性DCA的积累损害野生型小鼠的认知功能,但不是在TGR5敲除小鼠中。这表明TGR5是介导DCA的这些作用的主要受体。此外,兴奋性神经元特异性敲除TGR5可改善AD小鼠的Aβ病理和认知障碍。连接TGR5和AD病理的潜在机制依赖于TGR5的下游效应子和APP的产生,简明扼要地总结为“p-STAT3-APH1-γ-分泌酶”信号通路。我们的研究确定了TGR5在AD病理发展中的关键作用。
    Bile acids (BAs) metabolism has a significant impact on the pathogenesis of Alzheimer\'s disease (AD). We found that deoxycholic acid (DCA) increased in brains of AD mice at an early stage. The enhanced production of DCA induces the up-regulation of the bile acid receptor Takeda G protein-coupled receptor (TGR5), which is also specifically increased in neurons of AD mouse brains at an early stage. The accumulation of exogenous DCA impairs cognitive function in wild-type mice, but not in TGR5 knockout mice. This suggests that TGR5 is the primary receptor mediating these effects of DCA. Furthermore, excitatory neuron-specific knockout of TGR5 ameliorates Aβ pathology and cognition impairments in AD mice. The underlying mechanism linking TGR5 and AD pathology relies on the downstream effectors of TGR5 and the APP production, which is succinctly concluded as a \"p-STAT3-APH1-γ-secretase\" signaling pathway. Our studies identified the critical role of TGR5 in the pathological development of AD.
    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
    ADGRF5(GPR116)已被确定为乳腺癌细胞迁移和转移的促进因子,然而,潜在的机制在很大程度上仍然难以捉摸。我们目前的研究表明,乳腺癌细胞中缺乏ADGRF5会损害细胞外基质(ECM)相关的细胞运动并阻碍体内肿瘤生长。这与基质金属蛋白酶8(MMP8)的表达增加有关,一种特征明确的抗肿瘤性MMP,以及肿瘤相关中性粒细胞(TAN)的极化向肿瘤微环境(TME)中的抗肿瘤N1表型转移。机械上,ADGRF5通过增强RhoA激活抑制ERK1/2活性,导致Thr235的C/EBPβ磷酸化降低,阻碍其核易位和随后的激活。至关重要的是,在其启动子区域内鉴定了MMP8转录必需的两个C/EBPβ结合基序。因此,ADGRF5沉默促进MMP8表达和CXCL8分泌,吸引增加的TAN渗透;同时,MMP8在核心蛋白分裂中起作用,这导致TME中TGF-β的捕获失活,从而将TAN极化为抗肿瘤N1中性粒细胞表型并减轻TGF-β增强的乳腺癌细胞运动。我们的发现揭示了ADGRF5之间的一种新的连接,一种粘附G蛋白偶联受体,和TME的编排,这决定了恶性肿瘤的进展。总的来说,数据强调ADGRF5是乳腺癌干预的有希望的治疗靶点.
    ADGRF5 (GPR116) has been identified as a facilitator of breast cancer cell migration and metastasis, yet the underlying mechanisms remain largely elusive. Our current study reveals that the absence of ADGRF5 in breast cancer cells impairs extracellular matrix (ECM)-associated cell motility and impedes in vivo tumor growth. This correlates with heightened expression of matrix metalloproteinase 8 (MMP8), a well-characterized antitumorigenic MMP, and a shift in the polarization of tumor-associated neutrophils (TANs) towards the antitumor N1 phenotype in the tumor microenvironment (TME). Mechanistically, ADGRF5 inhibits ERK1/2 activity by enhancing RhoA activation, leading to decreased phosphorylation of C/EBPβ at Thr235, hindering its nuclear translocation and subsequent activation. Crucially, two C/EBPβ binding motifs essential for MMP8 transcription are identified within its promoter region. Consequently, ADGRF5 silencing fosters MMP8 expression and CXCL8 secretion, attracting increased infiltration of TANs; simultaneously, MMP8 plays a role in decorin cleavage, which leads to trapped-inactivation of TGF-β in the TME, thereby polarizing TANs towards the antitumor N1 neutrophil phenotype and mitigating TGF-β-enhanced cell motility in breast cancer. Our findings reveal a novel connection between ADGRF5, an adhesion G protein-coupled receptor, and the orchestration of the TME, which dictates malignancy progression. Overall, the data underscore ADGRF5 as a promising therapeutic target for breast cancer intervention.
    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
    琥珀酸盐,传统上被视为只是三羧酸(TCA)循环的中间体,已经成为炎症的关键介质。TCA循环内的中断导致琥珀酸在线粒体基质中的积累。这种过量的琥珀酸盐随后扩散到细胞质中并释放到细胞外空间中。胞质琥珀酸水平升高通过抑制脯氨酸羟化酶稳定缺氧诱导因子-1α,增强炎症反应。值得注意的是,琥珀酸还通过将琥珀酸受体1接合在免疫细胞上而在细胞外充当信号分子,从而调节它们的促炎或抗炎活性。琥珀酸水平的改变与各种炎症性疾病有关,包括类风湿性关节炎,炎症性肠病,肥胖,和动脉粥样硬化。这些关联主要是由于过度的免疫细胞应答。鉴于其在炎症中的核心作用,针对琥珀酸途径为这些疾病提供了有希望的治疗途径。本文提供了琥珀酸参与炎症过程的广泛综述,并强调了未来研究和治疗可能性开发的潜在目标。
    Succinate, traditionally viewed as a mere intermediate of the tricarboxylic acid (TCA) cycle, has emerged as a critical mediator in inflammation. Disruptions within the TCA cycle lead to an accumulation of succinate in the mitochondrial matrix. This excess succinate subsequently diffuses into the cytosol and is released into the extracellular space. Elevated cytosolic succinate levels stabilize hypoxia-inducible factor-1α by inhibiting prolyl hydroxylases, which enhances inflammatory responses. Notably, succinate also acts extracellularly as a signaling molecule by engaging succinate receptor 1 on immune cells, thus modulating their pro-inflammatory or anti-inflammatory activities. Alterations in succinate levels have been associated with various inflammatory disorders, including rheumatoid arthritis, inflammatory bowel disease, obesity, and atherosclerosis. These associations are primarily due to exaggerated immune cell responses. Given its central role in inflammation, targeting succinate pathways offers promising therapeutic avenues for these diseases. This paper provides an extensive review of succinate\'s involvement in inflammatory processes and highlights potential targets for future research and therapeutic possibilities development.
    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
    代谢相关脂肪性肝病(MAFLD)是严重COVID-19的危险因素。这项研究探讨了肠道激素受体和免疫反应基因表达对MAFLD患者COVID-19预后的潜在影响。
    方法:我们研究了AHR的基因表达水平,FFAR2FXR,与对照组相比,MAFLD和COVID-19患者的TGR5。我们检查了基因表达与临床结果之间的关联。
    结果:COVID-19患者显示AHR表达改变,可能影响免疫反应和恢复。MAFLD患者的AHR下调与凝血参数增加相关。MAFLD患者FFAR2表达升高与特异性免疫细胞群和住院时间相关。在MAFLD和重度COVID-19中均观察到FXR表达显着降低。
    结论:我们的研究结果表明AHR的潜在调节作用,COVID-19和MAFLD中的FFAR2和FXR。
    Metabolic-associated fatty liver disease (MAFLD) is a risk factor for severe COVID-19. This study explores the potential influence of gut hormone receptor and immune response gene expression on COVID-19 outcomes in MAFLD patients.
    METHODS: We investigated gene expression levels of AHR, FFAR2, FXR, and TGR5 in patients with MAFLD and COVID-19 compared to controls. We examined associations between gene expression and clinical outcomes.
    RESULTS: COVID-19 patients displayed altered AHR expression, potentially impacting immune response and recovery. Downregulated AHR in patients with MAFLD correlated with increased coagulation parameters. Elevated FFAR2 expression in patients with MAFLD was linked to specific immune cell populations and hospital stay duration. A significantly lower FXR expression was observed in both MAFLD and severe COVID-19.
    CONCLUSIONS: Our findings suggest potential modulatory roles for AHR, FFAR2, and FXR in COVID-19 and MAFLD.
    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
    了解有偏倚的味道T1R2/T1R3G蛋白偶联受体(GPCR)激动剂对糖基化受体信号传导的作用可能会提供对人工和天然甜味剂介导的相反作用的见解。特别是在癌症和转移中。甜味剂味GPCRs可以通过几种活性状态激活,这些状态包括偏向激动作用,功能选择性,或配体导向的信号。然而,越来越多的甜味剂配体阵列具有不同程度的变构偏向调制,可以在结合和信号特异性方式上发生巨大变化。这里,新出现的证据表明味觉GPCRs参与了有偏倚的GPCR信号串扰,涉及基质金属蛋白酶-9(MMP-9)和神经氨酸酶-1(Neu-1)通过修饰唾液酸激活糖基化受体.研究结果表明,大多数天然和人造甜味剂在RAW-Blue和PANC-1细胞中以剂量依赖性方式显着激活Neu-1唾液酸酶。为了确认这种有偏差的GPCR信号串扰,BIM-23127(neuromedinB受体抑制剂,MMP-9i(特异性MMP-9抑制剂),和磷酸奥司他韦(特异性Neu-1抑制剂)显著阻断甜味剂激动剂诱导的Neu-1唾液酸酶活性。为了评估人工和天然甜味剂对胰腺癌进展关键生存途径的影响,我们分析了上皮间质标志物的表达,CD24,ADLH-1,E-cadherin,和N-cadherin在PANC-1细胞中,并在划痕伤口闭合试验中评估细胞迁移侵袭性,和隧道纳米管(TNTs)在转移细胞间通讯中的作用。人工和天然甜味剂诱导PANC-1胰腺癌细胞转移表型,促进细胞间迁移通讯和侵袭。使用分泌性碱性磷酸酶(SEAP)测定,甜味剂还诱导下游NFκB活化。这些发现阐明了一种新的味道T1R2/T1R3GPCR功能选择性的信号平台,其中甜味剂激活下游信号,通过提出的NFκB诱导的表观遗传重编程模型促进肿瘤发生和转移。
    Understanding the role of biased taste T1R2/T1R3 G protein-coupled receptors (GPCR) agonists on glycosylated receptor signaling may provide insights into the opposing effects mediated by artificial and natural sweeteners, particularly in cancer and metastasis. Sweetener-taste GPCRs can be activated by several active states involving either biased agonism, functional selectivity, or ligand-directed signaling. However, there are increasing arrays of sweetener ligands with different degrees of allosteric biased modulation that can vary dramatically in binding- and signaling-specific manners. Here, emerging evidence proposes the involvement of taste GPCRs in a biased GPCR signaling crosstalk involving matrix metalloproteinase-9 (MMP-9) and neuraminidase-1 (Neu-1) activating glycosylated receptors by modifying sialic acids. The findings revealed that most natural and artificial sweeteners significantly activate Neu-1 sialidase in a dose-dependent fashion in RAW-Blue and PANC-1 cells. To confirm this biased GPCR signaling crosstalk, BIM-23127 (neuromedin B receptor inhibitor, MMP-9i (specific MMP-9 inhibitor), and oseltamivir phosphate (specific Neu-1 inhibitor) significantly block sweetener agonist-induced Neu-1 sialidase activity. To assess the effect of artificial and natural sweeteners on the key survival pathways critical for pancreatic cancer progression, we analyzed the expression of epithelial-mesenchymal markers, CD24, ADLH-1, E-cadherin, and N-cadherin in PANC-1 cells, and assess the cellular migration invasiveness in a scratch wound closure assay, and the tunneling nanotubes (TNTs) in staging the migratory intercellular communication. The artificial and natural sweeteners induced metastatic phenotype of PANC-1 pancreatic cancer cells to promote migratory intercellular communication and invasion. The sweeteners also induced the downstream NFκB activation using the secretory alkaline phosphatase (SEAP) assay. These findings elucidate a novel taste T1R2/T1R3 GPCR functional selectivity of a signaling platform in which sweeteners activate downstream signaling, contributing to tumorigenesis and metastasis via a proposed NFκB-induced epigenetic reprogramming modeling.
    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)是将信号从细胞外环境传递到细胞内部的跨膜蛋白。它们采用各种构象状态的能力,影响它们的功能,使它们在药物蛋白质组学研究中至关重要。虽然许多药物靶向特异性GPCR状态以发挥其作用,从而调节蛋白质的活性,但由于在整个过程中发生了大量的中间转化,因此解开激活途径仍然具有挑战性。并内在地影响受体的动力学。在这种情况下,计算建模,特别是分子动力学(MD)模拟,可以为GPCR转化的动力学和能量学提供有价值的见解,特别是与机器学习(ML)方法和技术相结合,以实现知识生成的模型可解释性。当前的研究建立在先前的工作的基础上,在先前的工作中,采用了层相关传播(LRP)技术来解释有关MD模拟中β2-肾上腺素能(β2AR)受体构象状态的多类分类问题中的预测。这里,我们解决了类不平衡带来的挑战,并通过评估不同不平衡缓解技术下基于深度学习(DL)的预测的鲁棒性和稳定性来扩展以前的分析。通过仔细评估可解释性和不平衡策略,我们的目标是产生可靠和强大的见解。
    G-protein coupled receptors (GPCRs) are transmembrane proteins that transmit signals from the extracellular environment to the inside of the cells. Their ability to adopt various conformational states, which influence their function, makes them crucial in pharmacoproteomic studies. While many drugs target specific GPCR states to exert their effects-thereby regulating the protein\'s activity-unraveling the activation pathway remains challenging due to the multitude of intermediate transformations occurring throughout this process, and intrinsically influencing the dynamics of the receptors. In this context, computational modeling, particularly molecular dynamics (MD) simulations, may offer valuable insights into the dynamics and energetics of GPCR transformations, especially when combined with machine learning (ML) methods and techniques for achieving model interpretability for knowledge generation. The current study builds upon previous work in which the layer relevance propagation (LRP) technique was employed to interpret the predictions in a multi-class classification problem concerning the conformational states of the β2-adrenergic (β2AR) receptor from MD simulations. Here, we address the challenges posed by class imbalance and extend previous analyses by evaluating the robustness and stability of deep learning (DL)-based predictions under different imbalance mitigation techniques. By meticulously evaluating explainability and imbalance strategies, we aim to produce reliable and robust insights.
    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蛋白偶联雌激素受体(GPER;G蛋白偶联雌激素受体30,也称为GPR30)是一种新型雌激素受体,已成为卵巢癌的有希望的靶标。GPER,七跨膜受体,抑制研究的卵巢癌细胞的细胞活力和迁移。然而,它对输卵管的影响,这是高级浆液性(HGSC)卵巢癌的潜在起源,尚未解决。这项研究是为了评估GPER的关系,卵巢癌亚型,即,高级浆液细胞系(OV90和OVCAR420),以及HGSC卵巢癌潜在起源的细胞类型(即,输卵管细胞系FT190)。此处评估的选择性配体是激动剂G-1,其用于体外研究以表征其对细胞活力和迁移的影响。因此,这项研究解决了特定GPER激动剂对细胞活力的影响,提供了更好地了解这种化合物对我们不同的研究细胞系的影响。引人注目的是,在G-1存在下观察到减弱的细胞增殖和迁移行为。因此,我们的体外研究揭示了HGSC卵巢癌起源的影响,并强调了GPER激动剂G-1作为卵巢癌的潜在疗法.
    The G-protein-coupled estrogen receptor (GPER; G-protein-coupled estrogen receptor 30, also known as GPR30) is a novel estrogen receptor and has emerged as a promising target for ovarian cancer. GPER, a seven-transmembrane receptor, suppresses cellular viability and migration in studied ovarian cancer cells. However, its impact on the fallopian tube, which is the potential origin of high-grade serous (HGSC) ovarian cancer, has not been addressed. This study was conducted to evaluate the relationship of GPER, ovarian cancer subtypes, i.e., high-grade serous cell lines (OV90 and OVCAR420), as well as the cell type that is the potential origin of HGSC ovarian cancer (i.e., the fallopian tube cell line FT190). The selective ligand assessed here is the agonist G-1, which was utilized in an in vitro study to characterize its effects on cellular viability and migration. As a result, this study has addressed the effect of a specific GPER agonist on cell viability, providing a better understanding of the effects of this compound on our diverse group of studied cell lines. Strikingly, attenuated cell proliferation and migration behaviors were observed in the presence of G-1. Thus, our in vitro study reveals the impact of the origin of HGSC ovarian cancers and highlights the GPER agonist G-1 as a potential therapy for ovarian cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号