GPER1

GPER1
  • 文章类型: Journal Article
    雌激素被认为部分地通过调节皮层和海马中的突触传递来调节认知功能。给予17β-雌二醇(E2)可以通过激活G蛋白偶联的雌激素受体1(GPER1)迅速增强海马中的兴奋性突触传递,并促进大鼠外侧内嗅皮质中的兴奋性突触传递。为了评估GPER1激活促进突触传递的机制,我们评估了急性10nME2给药对II/III层内嗅神经元药理学分离的兴奋性和抑制性突触电流的影响.在出生后第63天(PD)和74天之间对雌性Long-Evans大鼠进行卵巢切除,并植入真皮下E2胶囊以维持E2的持续低水平。在卵巢切除术后7至20天之间获得电生理记录。应用E220分钟不会显着影响AMPA或NMDA受体介导的兴奋性突触电流。然而,GABA受体介导的抑制性突触电流(IPSC)被E2显着降低,并在20分钟的洗脱期恢复至基线水平。在GPER1受体拮抗剂G15存在下阻断GABA介导的IPSC的抑制。GPER1可以调节蛋白激酶A(PKA),但用细胞内KT5720阻断PKA并不能阻止E2诱导的IPSC减少。GPER1还可以刺激细胞外信号调节激酶(ERK),GABAA受体的负调节剂,用PD90859阻断ERK的激活阻止了E2诱导的IPSC的减少。因此,E2可以导致GABA介导的IPSC中GPER1和ERK信号传导介导的快速减少。这提供了一种新机制,通过该机制,E2可以快速调节内嗅层II/III神经元的突触兴奋性,并且还可能导致其他大脑区域的E2和ERK依赖性突触传递改变。
    Estrogens are believed to modulate cognitive functions in part through the modulation of synaptic transmission in the cortex and hippocampus. Administration of 17β-estradiol (E2) can rapidly enhance excitatory synaptic transmission in the hippocampus and facilitate excitatory synaptic transmission in rat lateral entorhinal cortex via activation of the G protein-coupled estrogen receptor-1 (GPER1). To assess the mechanisms through which GPER1 activation facilitates synaptic transmission, we assessed the effects of acute 10 nM E2 administration on pharmacologically isolated evoked excitatory and inhibitory synaptic currents in layer II/III entorhinal neurons. Female Long-Evans rats were ovariectomized between postnatal day (PD) 63 and 74 and implanted with a subdermal E2 capsule to maintain continuous low levels of E2. Electrophysiological recordings were obtained between 7 and 20 days after ovariectomy. Application of E2 for 20 min did not significantly affect AMPA or NMDA receptor-mediated excitatory synaptic currents. However, GABA receptor-mediated inhibitory synaptic currents (IPSCs) were markedly reduced by E2 and returned towards baseline levels during the 20-min washout period. The inhibition of GABA-mediated IPSCs was blocked in the presence of the GPER1 receptor antagonist G15. GPER1 can modulate protein kinase A (PKA), but blocking PKA with intracellular KT5720 did not prevent the E2-induced reduction in IPSCs. GPER1 can also stimulate extracellular signal-regulated kinase (ERK), a negative modulator of GABAA receptors, and blocking activation of ERK with PD90859 prevented the E2-induced reduction of IPSCs. E2 can therefore result in a rapid GPER1 and ERK signaling-mediated reduction in GABA-mediated IPSCs. This provides a novel mechanism through which E2 can rapidly modulate synaptic excitability in entorhinal layer II/III neurons and may also contribute to E2 and ERK-dependent alterations in synaptic transmission in other brain areas.
    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蛋白偶联雌激素受体1(GPER1)的激活在不同的动物模型中引起抗高血压作用。内皮素-1信号系统在血压调节中起着重要作用。缺乏功能性内皮素受体B受体(ETB)会引起高血压和盐敏感性。GPER1和ETB相互作用促进雌性大鼠尿钠排泄。我们假设GPER1的激活可以防止雌性大鼠中ETB拮抗作用引起的高血压和盐敏感性。雌性SpragueDawley大鼠植入放射性遥测术。然后,对动物进行卵巢切除术,同时植入微型泵以递送GPER1激动剂,G1或其相应的车辆(Veh)。手术后两周,我们开始用ETB拮抗剂治疗大鼠,A-192621。将动物维持在正常盐(NS)饮食下,然后用高盐(HS)饮食再攻击5天。平均动脉血压的评估显示Veh治疗的增加,但没有G1处理,大鼠对卵巢切除术的反应。A-192621在NS喂养的Veh和G1治疗的大鼠中增加血压。G1改善了在A-192621治疗的卵巢切除大鼠中被破坏的昼夜节律血压节律。因此,尽管全身性GPER1激活并不能保护卵巢切除大鼠免受ETB拮抗作用引起的高血压和盐敏感性的影响,它维持了昼夜节律的血压。功能性ETB是引起GPER1的抗高血压作用所必需的。需要进一步的研究来提高我们对G蛋白偶联受体在调节昼夜节律血压节律中的相互作用的理解。
    Activation of the G protein-coupled estrogen receptor 1 (GPER1) elicits antihypertensive actions in different animal models. The endothelin-1 signaling system plays a fundamental role in blood pressure regulation. Lack of functional endothelin receptor B receptors (ETB) evokes hypertension and salt sensitivity. GPER1 and ETB interact to promote urinary sodium excretion in female rats. We hypothesized that activation of GPER1 protects against hypertension and salt sensitivity induced by ETB antagonism in female rats. Female Sprague Dawley rats were implanted with radiotelemetry. Then, animals were subjected to ovariectomy and simultaneously implanted with minipumps to deliver either the GPER1 agonist, G1, or its corresponding vehicle (Veh). Two weeks post-surgery, we initiated treatment of rats with the ETB antagonist, A-192621. Animals were maintained on a normal salt (NS) diet then challenged with a high salt (HS) diet for an additional 5 days. Assessment of mean arterial blood pressure revealed an increase in Veh-treated, but not G1-treated, rats in response to ovariectomy. A-192621 increased blood pressure in NS-fed Veh and G1-treated rats. G1 improved the circadian blood pressure rhythms which were disrupted in A-192621-treated ovariectomized rats. Thus, although systemic GPER1 activation did not protect ovariectomized rats from hypertension and salt sensitivity induced by ETB antagonism, it maintained the circadian blood pressure rhythms. Functional ETB is required to elicit the antihypertensive actions of GPER1. Additional studies are needed to improve our understanding of the interaction between G protein-coupled receptors in regulating circadian blood pressure rhythm.
    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
    结直肠癌(CRC)患者的性别和疾病分期存在生存差异。然而,潜在的分子机制还没有很好的理解。在这里,我们表明天冬酰胺合成酶(ASNS)和G蛋白偶联雌激素受体1(GPER1)是营养消耗的关键传感器,并与女性CRC的较差结局有关。使用在有限营养供应下生长的等基因SW48KRAS野生型(WT)和G12A突变(MT)细胞的3D球体模型,我们发现谷氨酰胺消耗抑制了两种细胞系的细胞生长,而ASNS和GPER1表达在KRASMT和WT中上调。雌二醇降低了KRASWT的生长,但对MT细胞没有影响。选择性GPER1和ASNS抑制剂在谷氨酰胺耗竭条件下抑制细胞增殖,MT细胞的caspase-3活性增加,特别是在雌二醇存在下。在癌症基因组图谱的临床结肠癌队列中,仅在晚期肿瘤中,GPER1和ASNS高表达与女性总体生存率较差相关.这些结果表明,KRASMT细胞具有响应营养供应减少的机制,通常在晚期肿瘤中观察到,通过增加ASNS和GPER1的表达来驱动细胞生长。此外,KRASMT细胞在营养耗尽条件下对雌二醇的保护作用具有抗性。结果表明,GPER1和ASNS表达,随着营养供应和KRAS突变之间的相互作用,进一步阐明了CRC代谢和生长中性别差异的潜在机制,并对KRAS突变CRC的精确管理具有临床意义。
    Survival differences exist in colorectal cancer (CRC) patients by sex and disease stage. However, the potential molecular mechanism(s) are not well understood. Here we show that asparagine synthetase (ASNS) and G protein-coupled estrogen receptor-1 (GPER1) are critical sensors of nutrient depletion and linked to poorer outcomes for females with CRC. Using a 3D spheroid model of isogenic SW48 KRAS wild-type (WT) and G12A mutant (MT) cells grown under a restricted nutrient supply, we found that glutamine depletion inhibited cell growth in both cell lines, whereas ASNS and GPER1 expression were upregulated in KRAS MT versus WT. Estradiol decreased growth in KRAS WT but had no effect on MT cells. Selective GPER1 and ASNS inhibitors suppressed cell proliferation with increased caspase-3 activity of MT cells under glutamine depletion condition particularly in the presence of estradiol. In a clinical colon cancer cohort from The Cancer Genome Atlas, both high GPER1 and ASNS expression were associated with poorer overall survival for females only in advanced stage tumors. These results suggest KRAS MT cells have mechanisms in place that respond to decreased nutrient supply, typically observed in advanced tumors, by increasing the expression of ASNS and GPER1 to drive cell growth. Furthermore, KRAS MT cells are resistant to the protective effects of estradiol under nutrient deplete conditions. The findings indicate that GPER1 and ASNS expression, along with the interaction between nutrient supply and KRAS mutations shed additional light on the mechanisms underlying sex differences in metabolism and growth in CRC, and have clinical implications in the precision management of KRAS mutant CRC.
    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
    神经炎症是出血性中风后的关键致病事件。内质网(ER)应激诱导的细胞凋亡和核苷酸结合域,富含亮氨酸的重复,和含pyrin结构域的蛋白3(NLRP3)相关的焦亡可以促进神经炎症反应的升级,导致脑损伤增加。G蛋白偶联雌激素受体1(GPER1),作为最广泛特征的脑源性雌激素,据报道会触发神经保护作用。然而,GPER1激活的抗凋亡和抗发热作用及其潜在机制尚未完全阐明.我们通过血管内穿孔建立了实验性SAH模型。SAH后1小时静脉内施用GPER1选择性激动剂G1。对于机械勘探,单磷酸腺苷活化蛋白激酶(AMPK)的选择性抑制剂,dorsomorphin,在SAH诱导前30分钟通过侧脑室注射给药。SAH后评估包括SAH等级,短期和长期的神经系统结果,脑水肿,脑血流量,透射电子显微镜(TEM),蛋白质印迹(WB),ELISA,TUNEL染色,Fluoro-JadeC染色(FJC),和免疫荧光染色。观察到GPER1的表达在SAH后6小时升高,并在24小时达到峰值,主要与神经元共定位。G1治疗后显着改善SAH小鼠的短期和长期神经功能缺损,以及抑制神经元内质网应激相关凋亡蛋白的表达(即,CHOP,GRP78,caspase-12,cleavedcaspase-3,Bax,Bcl2)和焦亡相关蛋白(即,NLRP3,ASC,裂解的Caspase-1)。此外,我们的研究表明,用dorsomorphin抑制AMPK减弱了G1的神经保护作用。这伴随着与ER应激诱导的细胞凋亡和焦亡相关的分子途径的改变。本文的这些数据阐明了GPER1通过以AMPK依赖性方式减轻神经炎症而发挥神经保护作用。调节神经元内质网应激相关的凋亡和焦亡。通过激活GPER1增强抗凋亡和抗发热作用可能是SAH患者的有效治疗策略。
    Neuroinflammation is a critical pathogenic event following hemorrhagic stroke. Endoplasmic reticulum (ER) stress-induced apoptosis and nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 3(NLRP3)-associated pyroptosis can contribute to the escalation of neuroinflammatory responses, leading to increased brain damage. G protein-coupled estrogen receptor 1(GPER1), as the most extensively characterized brain-derived estrogen, was reported to trigger neuroprotective effects. However, the anti-apoptotic and anti-pyroptotic effect of GPER1 activation and the underlying mechanism has not been fully elucidated. We established the experimental SAH model by intravascular perforation. The GPER1 selective agonist G1 was intravenously administered 1 h following SAH. For mechanistic exploration, the selective inhibitor of adenosine monophosphate-activated protein kinase (AMPK), dorsomorphin, was administered via intracerebroventricular injection 30 min prior to SAH induction. Post-SAH assessments included SAH grade, the short-term and long-term neurological outcomes, brain edema, cerebral blood flow, transmission electron microscopy (TEM), western blot (WB), ELISA, TUNEL staining, Fluoro-Jade C staining (FJC), and immunofluorescence staining. The expression of GPER1 was observed to elevate at 6 h and peaked at 24 h subsequent to SAH, predominantly co-localized with neurons. Post-treatment with G1 markedly ameliorated both the short-term and long-term neurological deficits of SAH mouse, as well as inhibiting the expression of neuronal ER stress-associated apoptotic proteins (i.e., CHOP, GRP78, Caspase-12, Cleaved Caspase-3, Bax, Bcl2) and pyroptosis-associated proteins (i.e., NLRP3, ASC, Cleaved Caspase-1). Additionally, our research revealed that inhibition of AMPK with dorsomorphin attenuated the neuroprotective effects of G1. This was accompanied by modifications in the molecular pathways associated with ER stress-induced apoptosis and pyroptosis. These data herein elucidated that GPER1 exerted neuroprotective effects by mitigating neuroinflammation in an AMPK-dependent manner, which modulates neuronal ER stress-associated apoptosis and pyroptosis. Boosting the anti-apoptotic and anti-pyroptotic effect by activating GPER1 may be an efficient treatment strategy for SAH patients.
    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
    镉(Cd)是一种常见的环境污染物,硒(Se)可以改善重金属毒性。因此,本研究旨在探讨硒对镉致肝细胞损伤的保护作用及其机制。为了实现这一点,我们利用了东大沟-兴隆队列,BRL3A电池模型,和暴露于Cd和/或Se的大鼠模型。结果表明,硒可以抵抗环境镉暴露引起的肝功能损伤和GPER1水平的降低,各种方法证实,Se可以在体内和体外保护Cd诱导的肝毒性。机械上,Cd引起过度的线粒体自噬激活,LC3B的共定位证明,PINK1,Parkin,P62和TOMM20。mt-keima腺病毒转染BRL3A细胞表明Cd抑制自噬-溶酶体融合,从而阻碍了有丝分裂通量。重要的是,G1,GPER1的特异性激动剂,减轻Cd诱导的线粒体自噬过度激活和肝细胞毒性,而G15加剧了这些影响。值得注意的是,硒补充减弱Cd诱导的GPER1蛋白减少和过度的线粒体自噬激活,同时促进自噬-溶酶体融合,从而恢复有丝分裂通量。总之,这项研究提出了一种新的机制,其中硒减轻GPER1介导的线粒体自噬并促进自噬-溶酶体融合,从而恢复Cd诱导的有丝分裂通量损伤,防止肝细胞损伤。
    Cadmium (Cd) is a common environmental pollutant, while selenium (Se) can ameliorate heavy metal toxicity. Consequently, this study aimed to investigate the protective effects of Se against Cd-induced hepatocyte injury and its underlying mechanisms. To achieve this, we utilized the Dongdagou-Xinglong cohort, BRL3A cell models, and a rat model exposed to Cd and/or Se. The results showed that Se counteracted liver function injury and the decrease in GPER1 levels caused by environmental Cd exposure, and various methods confirmed that Se could protect against Cd-induced hepatotoxicity both in vivo and in vitro. Mechanistically, Cd caused excessive mitophagy activation, evidenced by the colocalization of LC3B, PINK1, Parkin, P62, and TOMM20. Transfection of BRL3A cells with mt-keima adenovirus indicated that Cd inhibited autophagosome-lysosome fusion, thereby impeding mitophagic flux. Importantly, G1, a specific agonist of GPER1, mitigated Cd-induced mitophagy overactivation and hepatocyte toxicity, whereas G15 exacerbates these effects. Notably, Se supplementation attenuated Cd-induced GPER1 protein reduction and excessive mitophagy activation while facilitating autophagosome-lysosome fusion, thereby restoring mitophagic flux. In conclusion, this study proposed a novel mechanism whereby Se alleviated GPER1-mediated mitophagy and promoted autophagosome-lysosome fusion, thus restoring Cd-induced mitophagic flux damage, and preventing hepatocyte injury.
    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
    头颈部鳞状细胞癌(HNSCC)是头颈部肿瘤(HNTs)最常见的组织学形式,起源于嘴唇和口腔的上皮,咽部,喉部,唾液腺,鼻腔,和鼻窦。主要风险因素包括消费各种形式的烟草和酒精,以及高危型人乳头瘤病毒或EB病毒感染。不管病因是什么,男性患不同类型HNTs的风险比女性高2至6倍以上。造成这种差异的原因可能在于生物和社会心理因素的结合。因此,假设暴露于女性性激素,主要是雌激素,为女性提供保护,防止HNTs的形成和转移。在这次审查中,我们合成了有关雌激素和雌激素受体(ERs)在HNTs发育和进展中的作用的现有知识,特别强调膜ER,这些研究要少得多。我们可以总结一下,除了流行病学研究明确指出雌激素对女性的保护作用之外,两个核ER的表达增加,ERα,和ERβ,和膜ER,ERα36、GPER1和NaV1.2存在于不同类型的HNSCC中,抗雌激素可作为一种有效的治疗方法。
    Head and neck squamous cell carcinoma (HNSCC) is the most common histological form of head and neck tumors (HNTs), which originate from the epithelium of the lips and oral cavity, pharynx, larynx, salivary glands, nasal cavity, and sinuses. The main risk factors include consumption of tobacco in all forms and alcohol, as well as infections with high-risk human papillomaviruses or the Epstein-Barr virus. Regardless of the etiological agent, the risk of developing different types of HNTs is from two to more than six times higher in males than in females. The reason for such disparities probably lies in a combination of both biological and psychosocial factors. Therefore, it is hypothesized that exposure to female sex hormones, primarily estrogen, provides women with protection against the formation and metastasis of HNTs. In this review, we synthesized available knowledge on the role of estrogen and estrogen receptors (ERs) in the development and progression of HNTs, with special emphasis on membrane ERs, which are much less studied. We can summarize that in addition to epidemiologic studies unequivocally pointing to the protective effect of estrogen in women, an increased expression of both nuclear ERs, ERα, and ERβ, and membrane ERs, ERα36, GPER1, and NaV1.2, was present in different types of HNSCC, for which anti-estrogens could be used as an effective therapeutic approach.
    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:肺癌是全球癌症相关死亡的主要原因。非小细胞肺癌(NSCLC)发病率和病死率的性别差异,以及它与雌激素依赖的关系,提示雌激素受体(ERs)有助于NSCLC的发生发展。然而,G蛋白偶联雌激素受体(GPER1)对NSCLC的影响尚待确定.铁性凋亡逃逸是近年来发现的肿瘤的标志之一。在这种情况下,本研究评估了GPER1是否通过预防铁性凋亡促进NSCLC进展,并探讨了GPER1防止铁凋亡的潜在机制。
    方法:使用CCK8测定和平板克隆评估GPER1对H2O2,铁凋亡诱导物RSL3和Erastin的细胞毒性的影响。基于MDA和BODIPY™581/591C11的水平测量脂质过氧化水平。进行GPER1过表达和敲低,并使用G1,检测SCD1和PI3K/AKT/mTOR信号因子的表达。对配对标本进行免疫荧光分析和免疫组织化学检测NSCLC组织中GPER1和SCD1表达的相关性。使用CCK8测定法在体外和使用异种移植肿瘤模型在体内测量GPER1对顺铂的细胞毒性的影响。
    结果:GPER1和G1减轻了H2O2的细胞毒性,降低了对RSL3的敏感性,并损害了NSCLC组织的脂质过氧化。此外,GPER1和G1促进SCD1蛋白和mRNA的表达和PI3K/AKT/mTOR信号的激活。GPER1和SCD1在NSCLC组织中表达升高并呈正相关。高GPER1表达预测预后不良。GPER1敲除增强了顺铂的体外和体内抗肿瘤活性。
    结论:GPER1通过促进PI3K/AKT/mTOR信号的激活来预防非小细胞肺癌的铁凋亡,从而诱导SCD1表达。因此,针对GPER1联合顺铂的治疗将表现出更好的抗肿瘤效果。
    BACKGROUND: Lung cancer is the leading cause of cancer-related death worldwide. The sex differences in the occurrence and fatality rates of non-small cell lung cancer (NSCLC), along with its association with estrogen dependence, suggest that estrogen receptors (ERs) contribute to the development of NSCLC. However, the influence of G protein-coupled estrogen receptor (GPER1) on NSCLC remains to be determined. Escape from ferroptosis is one of the hallmarks of tumor discovered in recent years. In this context, the present study evaluated whether GPER1 promotes NSCLC progression by preventing ferroptosis, and the underlying mechanism through which GPER1 protects against ferroptosis was also explored.
    METHODS: The effects of GPER1 on the cytotoxicity of H2O2, the ferroptosis inducer RSL3, and Erastin were assessed using the CCK8 assay and plate cloning. Lipid peroxidation levels were measured based on the levels of MDA and BODIPY™581/591C11. GPER1 overexpression and knockdown were performed and G1 was used, and the expression of SCD1 and PI3K/AKT/mTOR signaling factors was measured. Immunofluorescence analysis and immunohistochemistry were performed on paired specimens to measure the correlation between the expression of GPER1 and SCD1 in NSCLC tissues. The effect of GPER1 on the cytotoxicity of cisplatin was measured in vitro using the CCK8 assay and in vivo using xenograft tumor models.
    RESULTS: GPER1 and G1 alleviated the cytotoxicity of H2O2, reduced sensitivity to RSL3, and impaired lipid peroxidation in NSCLC tissues. In addition, GPER1 and G1 promoted the protein and mRNA expression of SCD1 and the activation of PI3K/AKT/mTOR signaling. GPER1 and SCD1 expression were elevated and positively correlated in NSCLC tissues, and high GPER1 expression predicted a poor prognosis. GPER1 knockdown enhanced the antitumor activity of cisplatin in vitro and in vivo.
    CONCLUSIONS: GPER1 prevents ferroptosis in NSCLC by promoting the activation of PI3K/AKT/mTOR signaling, thereby inducing SCD1 expression. Therefore, treatments targeting GPER1 combined with cisplatin would exhibit better antitumor effects.
    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
    脂肪组织是雌激素产生的第二重要部位,雄激素通过芳香化酶转化为雌激素。虽然胃癌患者经常发生富含脂肪细胞的腹膜转移,潜在机制尚不清楚.在这项研究中,我们确定G蛋白偶联雌激素受体(GPER1)是胃癌腹膜转移的启动子.功能性体外研究表明,β-雌二醇(E2)或GPER1激动剂G1抑制胃癌细胞中的失巢凋亡。此外,基因过度表达或敲除GPER1显著抑制或增强胃癌细胞的体外失巢凋亡和体内腹膜转移,分别。机械上,GPER1敲除破坏了NADPH池并增加了活性氧(ROS)的产生。相反,GPER1的过表达具有相反的作用。GPER1抑制烟酰胺腺嘌呤二核苷酸激酶1(NADK1)泛素化并促进其磷酸化,导致NADK1在蛋白质水平和活性上的表达升高,分别。此外,NADK1的遗传抑制破坏了NADPH和氧化还原稳态,导致高水平的ROS和显著的失巢,在基于细胞的异种移植模型中抑制肺和腹膜转移。总之,我们的研究提示,抑制GPER1介导的NADK1活性及其泛素化可能是胃癌腹膜转移的一种有前景的治疗策略.
    Adipose tissue is the second most important site of estrogen production, where androgens are converted into estrogen by aromatase. While gastric cancer patients often develop adipocyte-rich peritoneal metastasis, the underlying mechanism remains unclear. In this study, we identified the G-protein-coupled estrogen receptor (GPER1) as a promoter of gastric cancer peritoneal metastasis. Functional in vitro studies revealed that β-Estradiol (E2) or the GPER1 agonist G1 inhibited anoikis in gastric cancer cells. Additionally, genetic overexpression or knockout of GPER1 significantly inhibited or enhanced gastric cancer cell anoikis in vitro and peritoneal metastasis in vivo, respectively. Mechanically, GPER1 knockout disrupted the NADPH pool and increased reactive oxygen species (ROS) generation. Conversely, overexpression of GPER1 had the opposite effects. GPER1 suppressed nicotinamide adenine dinucleotide kinase 1(NADK1) ubiquitination and promoted its phosphorylation, which were responsible for the elevated expression of NADK1 at protein levels and activity, respectively. Moreover, genetic inhibition of NADK1 disrupted NADPH and redox homeostasis, leading to high levels of ROS and significant anoikis, which inhibited lung and peritoneal metastasis in cell-based xenograft models. In summary, our study suggests that inhibiting GPER1-mediated NADK1 activity and its ubiquitination may be a promising therapeutic strategy for peritoneal metastasis of gastric cancer.
    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
    大量研究表明,雌激素缺乏通过影响成骨信号通路抑制骨骼中前成骨细胞的增殖和分化,导致骨量减少和再生受损。探讨雌激素缺乏下维持骨再生的机制。我们随机选择1102例临床病例,其中18至75岁的女性患者在同济大学口腔医院接受了拔牙治疗,拔牙缺损的愈合效果差异不大,在某种程度上,下颌骨的再生对雌激素水平的降低不敏感。阐明雌激素缺乏下促进颌骨再生的机制,在接受卵巢切除术(OVX)或假手术的雌性大鼠的上颌骨中建立了拔牙缺损模型,并分离骨髓基质细胞(BMSCs)进行单细胞测序。进一步定量PCR,RNA干扰,茜素红染色,免疫组织化学和免疫印迹实验表明,在卵巢切除术的背景下,上颌骨缺损促进G蛋白偶联雌激素受体1(Gper1)的表达,刺激下游cAMP/PKA/pCREB信号,促进细胞增殖,从而为成骨提供了足够的祖细胞,并增强了颌骨的再生能力。相应地,Gper1基因的杂合缺失减弱了CREB的磷酸化,导致细胞增殖减少,并损害上颌骨缺损的修复。这项研究证明了Gper1在维持颌骨再生中的重要性,尤其是在雌激素缺乏的情况下。
    Numerous studies have demonstrated that estrogen deficiency inhibit the proliferation and differentiation of pre-osteoblasts in skeleton by affecting osteogenic signaling, lead to decreased bone mass and impaired regeneration. To explore the mechanisms maintaining bone regeneration under estrogen deficiency, we randomly selected 1102 clinical cases, in which female patients aged between 18 and 75 have underwent tooth extraction in Stomatological Hospital of Tongji University, there is little difference in the healing effect of extraction defects, suggesting that to some extent, the regeneration of jawbone is insensitive to the decreased estrogen level. To illuminate the mechanisms promoting jawbone regeneration under estrogen deficiency, a tooth extraction defect model was established in the maxilla of female rats who underwent ovariectomy (OVX) or sham surgery, and jawbone marrow stromal cells (BMSCs) were isolated for single-cell sequencing. Further quantitative PCR, RNA interference, alizarin red staining, immunohistochemistry and western blotting experiments demonstrated that in the context of ovariectomy, maxillary defects promoted G protein-coupled estrogen receptor 1 (Gper1) expression, stimulate downstream cAMP/PKA/pCREB signaling, and facilitate cell proliferation, and thus provided sufficient progenitors for osteogenesis and enhanced the regeneration capacity of the jawbone. Correspondingly, the heterozygous deletion of the Gper1 gene attenuated the phosphorylation of CREB, led to decreased cell proliferation, and impaired the restoration of maxillary defects. This study demonstrates the importance of Gper1 in maintaining jawbone regeneration, especially in the context of estrogen deficiency.
    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
    背景:G蛋白偶联雌激素受体(GPER)1介导非基因组雌激素相关的信号传导,并通过多个下游途径在调节细胞生长和程序性细胞死亡中起重要作用。尽管人们对GPER1在癌症发展中的作用越来越感兴趣,尚未对GPER1进行泛癌症分析.
    方法:在这项研究中,我们通过人蛋白图谱(HPA)对GPER1在泛癌症中的作用进行了全面分析,癌症基因组图谱(TCGA)加州大学,圣克鲁斯·塞纳(UCSCXENA),基因型-组织表达(GTEx),MethSurv,阿拉巴马大学伯明翰分校CANcer数据分析门户(UALCAN),cBioPortal,STRING和TISIB分离数据库,然后用R软件进行富集分析。
    结果:GPER1在组织和器官中广泛表达,并且在多种癌症中的表达与正常组织不同。在诊断评估中,在九种癌症类型中,曲线下面积(AUC)超过0.9。生存分析显示,GPER1与11种癌症的预后相关。此外,GPER1表达与多种癌症的免疫浸润有关。
    结论:总之,GPER1在各种恶性肿瘤中具有良好的诊断或预后价值。加上其与免疫成分的广泛相关性,上述结果表明,GPER1在肿瘤诊断和预后方面显示出希望,为精确和个性化的抗肿瘤策略提供新思路。
    The G protein-coupled oestrogen receptor (GPER) 1 mediates non-genomic oestrogen-related signalling and plays an important role in the regulation of cell growth and programmed cell death through multiple downstream pathways. Despite the increasing interest in the role of GPER1 in cancer development, no pan-cancer analysis has been available for GPER1.
    In this study we performed a comprehensive analysis of the role of GPER1 in pan-cancer via Human Protein Atlas (HPA), The Cancer Genome Atlas (TCGA), University of California, Santa Cruz Xena (UCSC XENA), Genotype-Tissue Expression (GTEx), MethSurv, The University of Alabama at Birmingham CANcer data analysis Portal (UALCAN), cBioPortal, STRING and TISIDB detabases, followed by enrichment analysis using R software.
    GPER1 was widely expressed in tissues and organs and differed in expression from normal tissue in a variety of cancers. In diagnostic assessment, it\'s Area Under the Curve (AUC) surpassed 0.9 in nine cancer types. Survival analysis showed that GPER1 was correlated with the prognosis of 11 cancer types. Moreover, GPER1 expression was associated with immune infiltration in multiple cancers.
    In summary, GPER1 has good diagnostic or prognostic value across various malignancies. Together with its extensive correlation with immune components, the aforementioned results suggests that GPER1 shows promise in tumour diagnosis and prognosis, providing new ideas for precise and personalised anti-tumour strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号