G protein-coupled estrogen receptor

G 蛋白偶联雌激素受体
  • 文章类型: Journal Article
    全氟化合物(PFCs)属于全球环境污染物的重要类别。研究生物系统中PFC的毒理学效应在生命科学等各种学科中具有重要意义。环境科学,化学,和生态毒理学。在这项研究中,在模拟人体生理条件下(pH=7.4),采用多种光谱技术和计算模拟相结合的方法研究了全氟化合物(PFCs)对G蛋白偶联雌激素受体(GPER)的影响.此外,本研究主要从分子水平探讨PFCs与GPER的结合模式和毒理机制。所有三种全氟磺酸(PFSAs)都可以通过静态猝灭和非辐射能量转移诱导GPER荧光的猝灭。在不同温度下的稳态荧光计算显示表观结合常数约为106,证实了三种PFSA和GPER之间的强结合亲和力。分子对接研究表明,PFSA的结合位点位于GPER头区最大的疏水腔内,它们可以与空腔内的氨基酸残基进行氢键和疏水相互作用。傅里叶变换红外光谱,三维荧光,和分子动力学模拟共同表明,蛋白质在与小分子结合后变得更加稳定。疏水性整体增加,并观察到蛋白质二级结构的改变。本研究加深了对PFCs对内分泌系统影响的理解,帮助评估它们对人类健康的潜在影响。它为决策和环境管理提供了基础,同时也为开发新的污染监测方法和药物治疗提供了见解。
    Perfluorinated compounds (PFCs) belong to a significant category of global environmental pollutants. Investigating the toxicological effects of PFCs within biological systems is of critical significance in various disciplines such as life sciences, environmental science, chemistry, and ecotoxicology. In this study, under simulated human physiological conditions (pH = 7.4), a combination of multiple spectroscopic techniques and computational simulations was employed to investigate the impact of perfluorinated compounds (PFCs) on the G protein-coupled estrogen receptor (GPER). Additionally, the research focused on exploring the binding modes and toxicological mechanisms between PFCs and GPER at the molecular level. All three perfluorinated sulfonic acids (PFSAs) can induce quenching of GPER fluorescence through static quenching and non-radiative energy transfer. Steady-state fluorescence calculations at different temperatures revealed apparent binding constants in the order of 106, confirming a strong binding affinity between the three PFSAs and GPER. Molecular docking studies indicated that the binding sites of PFSAs are located within the largest hydrophobic cavity in the head region of GPER, where they can engage in hydrogen bonding and hydrophobic interactions with amino acid residues within the cavity. Fourier transform infrared spectroscopy, three-dimensional fluorescence, and molecular dynamics simulations collectively indicate that proteins become more stable upon binding with small molecules. There is an overall increase in hydrophobicity, and alterations in the secondary structure of the protein are observed. This study deepens the comprehension of the effects of PFCs on the endocrine system, aiding in evaluating their potential impact on human health. It provides a basis for policy-making and environmental management while also offering insights for developing new pollution monitoring methods and drug therapies.
    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
    雌激素是一组激素,与神经系统合作,影响所有性别的整体健康。它影响许多过程,包括发生在中枢神经系统的那些,影响学习和记忆,并在神经退行性疾病和精神障碍中发挥作用。激素的作用是由特异性受体介导的。经典雌激素受体的重要作用,ERα和ERβ,在各种疾病中已经知道很多年了,但是在确定了结构和位置不同的受体之后,G蛋白偶联雌激素受体(GPER),研究了其在人体生理和病理生理学中的作用。这篇评论汇编了与GPER相关的信息,强调其对稳态和疾病的影响,同时特别注意这种受体在神经生物学和生物行为过程中的功能和功能障碍。了解受体调节的可能性对治疗至关重要,受体的破坏会导致疾病或紊乱,不管正确的雌激素水平。我们得出的结论是,对GPER受体的研究有可能开发出调节雌激素并积极影响人类健康的疗法。
    Estrogen is a group of hormones that collaborate with the nervous system to impact the overall well-being of all genders. It influences many processes, including those occurring in the central nervous system, affecting learning and memory, and playing roles in neurodegenerative diseases and mental disorders. The hormone\'s action is mediated by specific receptors. Significant roles of classical estrogen receptors, ERα and ERβ, in various diseases were known since many years, but after identifying a structurally and locationally distinct receptor, the G protein-coupled estrogen receptor (GPER), its role in human physiology and pathophysiology was investigated. This review compiles GPER-related information, highlighting its impact on homeostasis and diseases, while putting special attention on functions and dysfunctions of this receptor in neurobiology and biobehavioral processes. Understanding the receptor modulation possibilities is essential for therapy, as disruptions in receptors can lead to diseases or disorders, irrespective of correct estrogen levels. We conclude that studies on the GPER receptor have the potential to develop therapies that regulate estrogen and positively impact human health.
    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
    三氯卡班(TCC)和三氯生(TCS)已在人体中普遍存在,并引起越来越多的关注。本研究旨在揭示TCC和TCS通过非基因组GPER介导的信号通路对三阴性乳腺癌的诱导风险。分子模拟表明,TCC在理论上表现出比TCS更高的GPER结合亲和力。钙动员测定显示TCC/TCS激活GPER信号传导途径,观察到的最低有效浓度(LOEC)为10nM/100nM。TCC和TCS也上调MMP-2/9,EGFR,MAPK3但通过GPER介导的信号通路下调MAPK8。增殖试验显示TCC/TCS诱导4T1乳腺癌细胞增殖,LOEC为100nM/1000nM。伤口愈合和transwell测定显示TCC/TCS以浓度依赖性方式促进4T1细胞迁移,LOEC为10nM。TCC对乳腺癌细胞增殖和迁移的影响强于TCS,且均受GPER调控。TCC/TCS诱导的迁移效应比增殖效应更显著。机制研究表明,TCC/TCS下调上皮标志物(E-cadherin)的表达,但上调间充质标志物(蜗牛和N-cadherin)的表达,被GPER抑制剂G15逆转。这些生物标志物结果表明,TCC/TCS诱导的4T1细胞迁移是GPER信号通路调控的一种经典的上皮间质转化机制。原位肿瘤模型证实,在人暴露水平为10mg/kg/d时,TCC通过GPER介导的信号通路促进乳腺癌原位肿瘤生长和远端组织转移。TCC诱导的乳腺癌组织转移比原位肿瘤生长更显著。总的来说,我们首次证明TCC/TCS可以激活GPER信号通路诱导乳腺癌进展.
    Triclocarban (TCC) and triclosan (TCS) have been detected ubiquitously in human body and evoked increasing concerns. This study aimed to reveal the induction risks of TCC and TCS on triple negative breast cancer through non-genomic GPER-mediated signaling pathways. Molecular simulation indicated that TCC exhibited higher GPER binding affinity than TCS theoretically. Calcium mobilization assay displayed that TCC/TCS activated GPER signaling pathway with the lowest observed effective concentrations (LOEC) of 10 nM/100 nM. TCC and TCS also upregulated MMP-2/9, EGFR, MAPK3 but downregulated MAPK8 via GPER-mediated signaling pathway. Proliferation assay showed that TCC/TCS induced 4 T1 breast cancer cells proliferation with the LOEC of 100 nM/1000 nM. Wound-healing and transwell assays showed that TCC/TCS promoted 4 T1 cells migration in a concentration-dependent manner with the LOEC of 10 nM. The effects of TCC on breast cancer cells proliferation and migration were stronger than TCS and both were regulated by GPER. TCC/TCS induced migratory effects were more significantly than proliferative effect. Mechanism study showed that TCC/TCS downregulated the expression of epithelial marker (E-cadherin) but upregulated mesenchymal markers (snail and N-cadherin), which was reversed by GPER inhibitor G15. These biomarkers results indicated that TCC/TCS-induced 4 T1 cells migration was a classic epithelial to mesenchymal transition mechanism regulated by GPER signaling pathway. Orthotopic tumor model verified that TCC promoted breast cancer in-situ tumor growth and distal tissue metastasis via GPER-mediated signaling pathway at human-exposure level of 10 mg/kg/d. TCC-induced tissue metastasis of breast cancer was more significantly than in-situ tumor growth. Overall, we demonstrated for the first time that TCC/TCS could activate the GPER signaling pathways to induce breast cancer progression.
    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
    雌激素在乳腺癌的发生和发展中起着至关重要的作用。雌激素受体(ER)α,ERβ,G蛋白偶联雌激素受体是乳腺癌中雌激素的主要受体。这些受体主要通过与雌激素结合而被激活。ER和膜生长因子受体之间的串扰产生了额外的途径,这些途径放大了其配体的作用并促进了肿瘤的生长。这种串扰可能会导致ERα阳性乳腺癌的内分泌治疗抵抗。此外,这可能解释了ERα-/HER2阳性乳腺癌对抗人表皮生长因子受体2(HER2)治疗的耐药和三阴性乳腺癌对化疗的耐药.因此,有必要了解ERs与生长因子受体之间的复杂串扰。在这次审查中,我们描述了乳腺癌中ER和膜生长因子受体之间的串扰.此外,本文综述了目前临床治疗的进展,并讨论了药物如何靶向串扰。最后,我们讨论了当前的挑战,并提出了关于通过药理抑制靶向串扰的潜在解决方案。总的来说,本综述提供了乳腺癌中ER和膜生长因子受体之间的串扰,以及对未来研究和临床治疗使用化疗方案改善患者生活质量的宝贵见解。
    Estrogens play a critical role in the initiation and progression of breast cancer. Estrogen receptor (ER)α, ERβ, and G protein-coupled estrogen receptor are the primary receptors for estrogen in breast cancer. These receptors are mainly activated by binding with estrogens. The crosstalk between ERs and membrane growth factor receptors creates additional pathways that amplify the effects of their ligands and promote tumor growth. This crosstalk may cause endocrine therapy resistance in ERα-positive breast cancer. Furthermore, this may explain the resistance to anti-human epidermal growth factor receptor-2 (HER2) treatment in ERα-/HER2-positive breast cancer and chemotherapy resistance in triple-negative breast cancer. Accordingly, it is necessary to understand the complex crosstalk between ERs and growth factor receptors. In this review, we delineate the crosstalk between ERs and membrane growth factor receptors in breast cancer. Moreover, this review highlights the current progress in clinical treatment and discusses how pharmaceuticals target the crosstalk. Lastly, we discuss the current challenges and propose potential solutions regarding the implications of targeting crosstalk via pharmacological inhibition. Overall, the present review provides a landscape of the crosstalk between ERs and membrane growth factor receptors in breast cancer, along with valuable insights for future studies and clinical treatments using a chemotherapy-sparing regimen to improve patient quality of life.
    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
    三阴性乳腺癌(TNBC)是一种生物学和临床异质性疾病。G蛋白偶联雌激素受体(GPER)在介导TNBC细胞中雌激素和雌激素样化合物的作用中起着至关重要的作用。与其他亚型相比,GPER在TNBC中有较高的表达。在雌激素受体α(ERα)阳性乳腺癌中,GPER机制已被彻底表征和分析。但不是在TNBC。我们以前的研究表明,GPERmRNA的高表达表明ERα阳性乳腺癌的预后较好;然而,其在TNBC中的作用不同。GPER是否可以作为TNBC的预测预后标志物或治疗靶标尚不清楚。在这次审查中,我们详细介绍了GPER的亚细胞定位,各种配体的不同作用,以及GPER与TNBC密切相关因素之间的相互作用。我们专注于TNBC特异性的内部分子机制,并深入探索了GPER在促进肿瘤发展中的作用。我们还讨论了GPER与特定细胞因子和趋化因子的相互作用,以及GPER与免疫逃避的关系。此外,我们在现有研究的背景下讨论了使用GPER作为治疗靶点的可行性.这篇全面的综述强调了GPER对TNBC的影响,为未来的研究提供了框架和方向。
    Triple-negative breast cancer (TNBC) is a biologically and clinically heterogeneous disease. The G protein-coupled estrogen receptor (GPER) plays a crucial role in mediating the effect of estrogen and estrogen-like compounds in TNBC cells. Compared with other subtypes, GPER has a higher expression in TNBC. The GPER mechanisms have been thoroughly characterized and analyzed in estrogen receptor α (ERα) positive breast cancer, but not in TNBC. Our previous work revealed that a higher expression of GPER mRNA indicates a better prognosis for ERα-positive breast cancer; however, its effects in TNBC differ. Whether GPER could serve as a predictive prognostic marker or therapeutic target for TNBC remains unclear. In this review, we provide a detailed introduction to the subcellular localization of GPER, the different effects of various ligands, and the interactions between GPER and closely associated factors in TNBC. We focused on the internal molecular mechanisms specific to TNBC and thoroughly explored the role of GPER in promoting tumor development. We also discussed the interaction of GPER with specific cytokines and chemokines, and the relationship between GPER and immune evasion. Additionally, we discussed the feasibility of using GPER as a therapeutic target in the context of existing studies. This comprehensive review highlights the effects of GPER on TNBC, providing a framework and directions for future research.
    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:越来越多的证据表明小胶质细胞介导的炎症和NLR家族含pyrin结构域3(NLRP3)炎性小体介导的焦亡在帕金森病(PD)的发病机制中的关键作用。宝火苷I,一种从淫羊藿中提取的天然类黄酮,已被证明具有抗炎作用,但其对PD的潜在神经保护作用和机制尚未被证明。
    方法:通过LPS诱导的BV2细胞或从宽型或G蛋白偶联雌激素受体(GPER)基因敲除小鼠中分离的原代小胶质细胞来评估保火苷I的抗炎作用。使用LPS诱导的GPER+/+或GPER-/-小鼠PD模型进一步探索与GPER介导的NLRP3炎性体抑制相关的潜在机制。通过westernblot分析检测保火苷I的神经保护作用。实时PCR,分子对接,小鼠行为测试,免疫荧光,和免疫组织化学。
    结果:保火苷I通过抑制NF-κB信号的激活和细胞凋亡水平的升高,可显着减轻LPS诱导的神经炎症,这可以通过细胞凋亡相关蛋白的表达下调来证明(NLRP3,ASC,小胶质细胞中的pro-Caspase-1,IL-1β)。保火苷I的胃内给药可防止LPS诱导的运动功能障碍和多巴胺能神经元的丢失,减少促炎细胞因子的表达,并抑制LPS诱导的PD小鼠模型中黑质纹状体途径中的小胶质细胞(Iba-1)和星形胶质细胞(GFAP)的激活。用GPER拮抗剂G15预处理小胶质细胞或小鼠GPER基因缺失可显著阻断保霍甙I对LPS诱导的神经炎症的抑制作用和NLRP3/ASC/Caspase-1通路的激活。分子对接进一步表明,宝火苷I可能以-10.4kcal/mol的结合能直接结合GPER。
    结论:保火苷I通过抑制NF-κB信号和NLRP3/ASC/Caspase-1通路的激活,对PD具有神经保护作用。其抗炎作用的分子靶标在PD小鼠模型中被证明是GPER。保火苷I可能是一种有价值的抗神经炎药和具有明确治疗PD靶标的药物。
    BACKGROUND: Accumulating evidence indicates the crucial role of microglia-mediated inflammation and the NLR family pyrin domain containing 3 (NLRP3) inflammasome-mediated pyroptosis in the pathogenesis of Parkinson\'s disease (PD). Baohuoside I, a natural flavonoid extracted from Herba Epimedii, has been shown to possess anti-inflammatory effects, but its potential neuroprotective effects and mechanism against PD have not been documented.
    METHODS: The anti-inflammatory effects of Baohuoside I were evaluated by LPS-induced BV2 cells or primary microglia isolated from wide type or G protein-coupled estrogen receptor (GPER) gene knockout mice. The underlying mechanism related to GPER-mediated NLRP3 inflammasome inhibition was further explored using LPS-induced GPER+/+ or GPER-/- mouse models of PD. The neuroprotective effects of Baohuoside I were detected through western blot analysis, real-time PCR, molecular docking, mouse behavioral tests, immunofluorescence, and immunohistochemistry.
    RESULTS: Baohuoside I significantly alleviated LPS-induced neuroinflammation by inhibiting the activation of NF-κB signal and the increase of pyroptosis levels as evidenced by the downregulated expression of pyroptosis-related proteins (NLRP3, ASC, pro-Caspase-1, IL-1β) in microglia cells. Intragastric administration of Baohuoside I protected against LPS-induced motor dysfunction and loss of dopaminergic neurons, reduced pro-inflammatory cytokines expressions, and inhibited microglial (Iba-1) and astrocyte (GFAP) activation in the nigrostriatal pathway in LPS-induced mouse model of PD. Pretreatment with GPER antagonist G15 in microglia cells or GPER gene deletion in mice significantly blocked the inhibitory effects of Baohuoside I on LPS-induced neuroinflammation and activation of the NLRP3/ASC/Caspase-1 pathway. Molecular docking further indicated that Baohuoside I might bind to GPER directly with a binding energy of -10.4 kcal/mol.
    CONCLUSIONS: Baohuoside I provides neuroprotective effects against PD by inhibiting the activation of the NF-κB signal and NLRP3/ASC/Caspase-1 pathway. The molecular target for its anti-inflammatory effects is proved to be GPER in the PD mouse model. Baohuoside I may be a valuable anti-neuroinflammatory agent and a drug with well-defined target for the treatment of PD.
    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
    文献中越来越多的证据表明,雄激素和雌激素之间的不平衡是与称为隐睾的常见犬生殖障碍相关的疾病。据推测,雌激素在调节睾丸细胞功能和生殖事件中的作用是由于两个核雌激素受体(ER)的广泛表达,睾丸中的ER-α和ER-β以及跨膜G蛋白偶联雌激素受体(GPER)。在这项研究中,免疫组织化学,Western印迹和qRT-PCR用于评估GPER在正常和隐睾犬睾丸-附睾复合物中的分布和表达。还评估了ER-α和ER-β以更好地表征所有三种受体的相对丰度。此外,在这些组织中,研究了两种通常与氧化应激相关的蛋白质SOD1和Nrf2的表达水平,以评估与ER的可能关系。我们的数据揭示了正常犬和隐睾犬之间GPER的分布和表达的变化。特别是,受隐睾影响的狗在检查的生殖道水平上显示GPER上调。还考虑所获得的SOD1和Nrf2表达式的调制结果,我们可以假设GPER参与了隐睾疾病。进一步的研究是,然而,必须表征GPER的作用及其特定的信号机制。
    There is growing evidence by the literature that the unbalance between androgens and estrogens is a relevant condition associated with a common canine reproductive disorder known as cryptorchidism. The role of estrogens in regulating testicular cell function and reproductive events is supposedly due to the wide expression of two nuclear estrogen receptors (ERs), ER-alpha and ER-beta and a trans-membrane G protein-coupled estrogen receptor (GPER) in the testis. In this study, immunohistochemistry, Western blotting and qRT-PCR were used to assess the distribution and expression of GPER in the testis-epididymal complex in the normal and cryptorchid dog. ER-alpha and ER-beta were also evaluated to better characterize the relative abundances of all three receptors. In addition, in these tissues, the expression level of two proteins as SOD1 and Nrf2 normally associated with oxidative stress was investigated to evaluate a possible relationship with ERs. Our data revealed changes in the distribution and expression of the GPER between the normal and cryptorchid dog. In particular, dogs affected by cryptorchidism showed an upregulation of GPER at level of the examined reproductive tract. Also considering the obtained result of a modulation of SOD1 and Nrf2 expression, we could hypothesize the involvement of GPER in the cryptorchid condition. Further studies are, however, necessary to characterize the role of GPER and its specific signaling mechanisms.
    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
    目的:G蛋白偶联雌激素受体(GPER)是位于质膜上的雌激素受体。我们之前报道了GPER特异性激动剂G-1的给药,在小鼠模型中抑制急性卵清蛋白(OVA)诱导的哮喘的发展。在这里,我们评估了GPER在慢性OVA哮喘小鼠模型中的作用.
    方法:对患有慢性OVA哮喘的BALB/c小鼠皮下给予G-1或生理盐水,并进行病理和免疫学评估。此外,使用流式细胞术测量脾脏中表达Foxp3的CD4阳性T细胞和肺中的ILC2。
    结果:我们观察到G-1治疗组的支气管肺泡灌洗液(BALF)中炎性细胞数量显着减少。在气道中,炎性细胞积累,Th2细胞因子(IL-4,IL-5,IL-13和eotaxin)和上皮细胞因子TSLP被抑制,而在BALF,抗炎细胞因子(IL-10和TGF-β)增加。此外,在脾单核细胞中,在G-1组中,表达Foxp3的CD4阳性T细胞增加,而用G-1治疗没有改变ILC2在肺中的百分比。
    结论:G-1给药抑制了慢性OVA哮喘小鼠的过敏性气道炎症。GPER可能是慢性过敏性哮喘的潜在治疗靶点。
    OBJECTIVE: G protein-coupled estrogen receptor (GPER) is an estrogen receptor located on the plasma membrane. We previously reported that the administration of G-1, a GPER-specific agonist, suppressed development of acute ovalbumin (OVA)-induced asthma in a mouse model. Herein, we evaluate the involvement of GPER in a mouse model of chronic OVA asthma.
    METHODS: G-1 or saline was administered subcutaneously to BALB/c mice with chronic OVA asthma, and pathological and immunological evaluation was performed. In addition, Foxp3-expressing CD4-positive T-cells in the spleen and ILC2 in the lungs were measured using flow cytometry.
    RESULTS: We observed a significant decrease in the number of inflammatory cells in the bronchoalveolar lavage fluid (BALF) in the G-1 treated group. In the airways, inflammatory cell accumulation, Th2 cytokines (IL-4, IL-5, IL-13, and eotaxin) and epithelial cytokine TSLP were suppressed, while in the BALF, anti-inflammatory cytokines (IL-10 and TGF-β) were increased. Furthermore, in splenic mononuclear cells, Foxp3-expressing CD4-positive T-cells were increased in the G-1 group, whereas treatment with G-1 did not change the percentage of ILC2 in the lungs.
    CONCLUSIONS: G-1 administration suppressed allergic airway inflammation in mice with chronic OVA asthma. GPER may be a potential therapeutic target for chronic allergic asthma.
    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: Randomized Controlled Trial, Veterinary
    本研究旨在探讨染料木素(GEN)对慢性热应激(HS)条件下肉鸡生产性能和代谢紊乱的预防作用及其机制。将总共120只3周龄的雄性罗斯肉鸡随机分为5组:保持在常温(每天21°C±1°C)的热中性区(TN)组,接受循环高温的HS组(每天8小时,每天32°C±1°C),和3组暴露于不同剂量的GEN(50、100或150mg/kg饮食)的HS。实验期持续3周。这里,HS导致生长性能参数下降和激素分泌紊乱(P<0.05),经100和150mg/kgGEN处理后,差异有统计学意义(P<0.05)。此外,150mg/kgGEN可以减轻HS引起的肝脏指数(P<0.01)和腹部脂肪率(P<0.05)的增加(P<0.05)。经100和150mg/kgGEN处理后,HS诱导的肝脏和血清脂质过度积累(P<0.01)得到改善(P<0.05)。此外,在100和150mg/kgGEN治疗后,HS诱导的肝脏脂解相关mRNA水平降低和脂质合成相关mRNA水平升高(P<0.01)有效减弱(P<0.05)。重要的是,HS刺激的肝线粒体能量功能障碍和过氧化物酶体增殖物激活受体-γ共激活因子1α(PGC-1α)的mRNA或蛋白质水平降低,150mg/kgGEN可改善肝脏中的核呼吸因子1和线粒体转录因子A(P<0.05)。此外,50至150mg/kgGEN治疗导致G蛋白偶联雌激素受体(GPR30)的mRNA或蛋白质水平显着增加,AMP激活蛋白激酶(AMPK)α1,磷酸化AMPKα,和磷酸化的乙酰辅酶A羧化酶α。总的来说,GEN缓解HS下的代谢紊乱和肝脏线粒体能量功能障碍,可能通过激活GPR30-AMPM-PGC-1α途径。这些数据为GEN作为添加剂减轻肉鸡HS提供了充分的基础。
    The objective of this study was to investigate the preventive effects and mechanisms of genistein (GEN) on production performance and metabolic disorders in broilers under chronic heat stress (HS). A total of 120 male 3-wk-old Ross broilers were randomly assigned to 5 groups: a thermoneutral zone (TN) group maintained at normal temperature (21°C ± 1°C daily), an HS group subjected to cyclic high temperature (32°C ± 1°C for 8 h daily), and 3 groups exposed to HS with varying doses of GEN (50, 100, or 150 mg/kg diet). The experimental period lasted for 3 wk. Here, HS led to a decline in growth performance parameters and hormone secretion disorders (P < 0.05), which were improved by 100 and 150 mg/kg GEN treatment (P < 0.05). Moreover, the HS-induced increases in the liver index (P < 0.01) and abdominal fat rate (P < 0.05) were attenuated by 150 mg/kg GEN (P < 0.05). The HS-induced excessive lipid accumulation in the liver and serum (P < 0.01) was ameliorated after 100 and 150 mg/kg GEN treatment (P < 0.05). Furthermore, the HS-induced decreases in lipolysis-related mRNA levels and increases in lipid synthesis-related mRNA levels in the liver (P < 0.01) were effectively blunted after 100 and 150 mg/kg GEN treatment (P < 0.05). Importantly, the HS-stimulated hepatic mitochondrial energetic dysfunction and decreases in the mRNA or protein levels of peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α), nuclear respiratory factor 1, and mitochondrial transcription factor A in the liver were ameliorated by 150 mg/kg GEN (P < 0.05). Moreover, 50 to 150 mg/kg GEN treatment resulted in a significant increase in the mRNA or protein levels of G protein-coupled estrogen receptor (GPR30), AMP-activated protein kinase (AMPK) α1, phosphorylated AMPKα, and phosphorylated acetyl-CoA carboxylase α. Collectively, GEN alleviated metabolic disorders and hepatic mitochondrial energetic dysfunction under HS, possibly through the activation of GPR30-AMPM-PGC-1α pathways. These data provide a sufficient basis for GEN as an additive to alleviate HS in broilers.
    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
    结直肠癌(CRC)是最高危的癌症之一;然而,有研究表明,CRC中的雌激素信号可能具有保护作用.因此,我们重点研究了CRC中雌激素受体中G蛋白偶联雌激素受体(GPER)的功能.在这项研究中,我们研究了白藜芦醇通过GPER在CRC(RKO和WiDr)细胞中的治疗效果,CRC细胞来源的异种移植模型,和类器官(30T和33T)。与低表达GPER的WiDr细胞相比,白藜芦醇显着抑制了高表达GPER的RKO细胞的细胞活力和增殖。在异种移植模型中,白藜芦醇还延迟肿瘤生长并显示高存活率,这取决于RKO衍生肿瘤中的GPER表达。此外,白藜芦醇显著抑制高GPER表达的类器官的活力。此外,白藜芦醇对CRC的抗癌作用表明白藜芦醇对GPER反应迅速,同时增加p-ERK和Bax的表达并裂解PARP蛋白。
    Colorectal cancer (CRC) is one of the most high-risk cancers; however, it has been suggested that estrogen signaling in CRC could have a protective effect. Therefore, we focused on the function of the G protein-coupled estrogen receptor (GPER) among the estrogen receptors in CRC. In this study, we investigated the therapeutic effect of resveratrol via GPER in CRC (RKO and WiDr) cells, CRC cell-derived xenograft models, and organoids (30T and 33T). Resveratrol significantly suppressed cell viability and proliferation in highly GPER-expressing RKO cells compared to that in low GPER-expressing WiDr cells. In xenograft models, resveratrol also delayed tumor growth and exhibited a high survival rate depending on GPER expression in RKO-derived tumors. Furthermore, resveratrol significantly inhibited the viability of organoids with high GPER expression. Additionally, the anticancer effect of resveratrol on CRC showed that resveratrol rapidly responded to GPER, while increasing the expression of p-ERK and Bax and cleaving PARP proteins.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号