membrane estrogen receptors

膜雌激素受体
  • 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
    雌激素受体(ER)在脑功能中起着多种作用,并与各种脑疾病有关。使用正电子发射断层扫描(PET)示踪剂可视化ERs的复杂景观已在肿瘤学中显示出希望,但在脑部疾病的背景下仍然受到限制。尽管最近在鉴定和开发针对各种ER亚型的更具选择性的配体方面取得了进展,进一步优化是必要的,以使这些受体的可靠和有效的成像。从这个角度来看,我们简要探讨了脑内雌激素信号传导的重要性,并提出了与PET示踪剂开发相关的挫折,用于识别脑内特定ERs亚型.然后,我们提出了开发有效的PET示踪剂的途径,以便非侵入性地研究大脑中ER的动力学,以及与纵向故障相关的神经精神疾病。这种观点提出了几个潜在的候选人,并强调了未满足的需求和需要进一步研究的领域,以释放PET示踪剂在ER成像中的全部潜力。最终有助于加深我们对ER的理解,并为潜在的治疗策略开辟新的途径。
    Estrogen receptors (ERs) play a multitude of roles in brain function and are implicated in various brain disorders. The use of positron emission tomography (PET) tracers for the visualization of ERs\' intricate landscape has shown promise in oncology but remains limited in the context of brain disorders. Despite recent progress in the identification and development of more selective ligands for various ERs subtypes, further optimization is necessary to enable the reliable and efficient imaging of these receptors. In this perspective, we briefly touch upon the significance of estrogen signaling in the brain and raise the setbacks associated with the development of PET tracers for identification of specific ERs subtypes in the brain. We then propose avenues for developing efficient PET tracers to non-invasively study the dynamics of ERs in the brain, as well as neuropsychiatric diseases associated with their malfunction in a longitudinal manner. This perspective puts several potential candidates on the table and highlights the unmet needs and areas requiring further research to unlock the full potential of PET tracers for ERs imaging, ultimately aiding in deepening our understanding of ERs and forging new avenues for potential therapeutic strategies.
    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
    核和膜启动的雌激素信号协同协调雌激素的多效性作用。经典的雌激素受体(ER)在转录中起作用,并控制着绝大多数的激素作用,而膜ER(mERs)能够急性调节雌激素信号,并且最近被证明具有强大的神经保护能力,而没有与核ER活性相关的负面副作用。近年来,GPER1是最广泛表征的mER。尽管触发了神经保护作用,认知改善,和血管保护作用和维持代谢稳态,GPER1已经成为争议的话题,特别是由于它参与了肿瘤发生。这就是为什么兴趣最近转向非GPER依赖的MER,即,mERα和mERβ。根据现有数据,非GPER依赖性mERs引起对脑损伤的保护作用,突触可塑性损伤,记忆和认知功能障碍,代谢失衡,血管功能不全.我们推测,这些特性是设计可用于治疗中风和神经退行性疾病的新疗法的新兴平台。由于mER具有干扰非编码RNA并通过影响组蛋白来调节脑组织的翻译状态的能力,非GPER依赖性mERs似乎是神经系统疾病现代药物治疗的有吸引力的靶点.
    Nuclear- and membrane-initiated estrogen signaling cooperate to orchestrate the pleiotropic effects of estrogens. Classical estrogen receptors (ERs) act transcriptionally and govern the vast majority of hormonal effects, whereas membrane ERs (mERs) enable acute modulation of estrogenic signaling and have recently been shown to exert strong neuroprotective capacity without the negative side effects associated with nuclear ER activity. In recent years, GPER1 was the most extensively characterized mER. Despite triggering neuroprotective effects, cognitive improvements, and vascular protective effects and maintaining metabolic homeostasis, GPER1 has become the subject of controversy, particularly due to its participation in tumorigenesis. This is why interest has recently turned toward non-GPER-dependent mERs, namely, mERα and mERβ. According to available data, non-GPER-dependent mERs elicit protective effects against brain damage, synaptic plasticity impairment, memory and cognitive dysfunctions, metabolic imbalance, and vascular insufficiency. We postulate that these properties are emerging platforms for designing new therapeutics that may be used in the treatment of stroke and neurodegenerative diseases. Since mERs have the ability to interfere with noncoding RNAs and to regulate the translational status of brain tissue by affecting histones, non-GPER-dependent mERs appear to be attractive targets for modern pharmacotherapy for nervous system diseases.
    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
    雌激素受体最初是在子宫中发现的,后来整个大脑和身体作为细胞内,影响配体结合时基因组变化的配体调节转录因子。然而,在细胞核外启动的快速雌激素受体信号也是通过尚不清楚的机制发生的。最近的研究表明,这些传统的受体,雌激素受体-α和雌激素受体-β,也可以贩运到表面膜上。来自这些膜结合雌激素受体(mERs)的信号级联不仅快速影响细胞兴奋性,但最终会影响基因表达,通过CREB的磷酸化可以看出。神经元mER作用的主要机制是通过代谢型谷氨酸受体(mGluRs)的谷氨酸非依赖性反式激活,这引发了多种信号结果。mERs与mGluRs的相互作用已被证明在女性的许多不同功能中很重要,包括,但不限于,繁殖和动机。在这里,我们回顾了女性的膜启动雌激素受体信号,重点关注这些mERs和mGluRs之间的相互作用。
    Estrogen receptors were initially identified in the uterus, and later throughout the brain and body as intracellular, ligand-regulated transcription factors that affect genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor-α and estrogen receptor-β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) not only rapidly effect cellular excitability, but can and do ultimately affect gene expression, as seen through the phosphorylation of CREB. A principal mechanism of neuronal mER action is through glutamate-independent transactivation of metabotropic glutamate receptors (mGluRs), which elicits multiple signaling outcomes. The interaction of mERs with mGluRs has been shown to be important in many diverse functions in females, including, but not limited to, reproduction and motivation. Here we review membrane-initiated estrogen receptor signaling in females, with a focus on the interactions between these mERs and mGluRs.
    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
    新合成途径优先雌激素-1(PaPE-1)选择性激活膜雌激素受体(mERs),即,mERα和mERβ,并已被证明能唤起神经保护作用;然而,其在保护脑组织免受缺氧和缺血方面的有效性尚未在后处理范例中得到证实。这是第一个研究表明,6小时延迟后处理与PaPE-1抑制缺氧/缺血诱导的神经元死亡,如体外小鼠原代细胞培养物中的中性红摄取所示。该效应伴随着损伤细胞的LDH释放和Fluoro-JadeC染色方面的神经毒性和神经变性的显著降低,分别。PaPE-1的神经保护作用机制还涉及通过线粒体膜电位和凋亡相关基因和蛋白质如Fas的表达水平的正常化证明的凋亡抑制。Fasl,Bcl2,FAS,FASL,BCL2,BAX,和GSK3β。此外,PaPE-1诱发的神经保护是通过减少ROS形成和恢复由于缺氧和缺血而变得失调的细胞代谢活性来介导的。这些数据提供了证据,用PaPE-1靶向膜非GPER雌激素受体是保护脑神经元免受缺氧/缺血性损伤的有效疗法,即使从损伤开始延迟6小时。
    Newly synthesized Pathway Preferential Estrogen-1 (PaPE-1) selectively activates membrane estrogen receptors (mERs), namely, mERα and mERβ, and has been shown to evoke neuroprotection; however, its effectiveness in protecting brain tissue against hypoxia and ischemia has not been verified in a posttreatment paradigm. This is the first study showing that a 6-h delayed posttreatment with PaPE-1 inhibited hypoxia/ischemia-induced neuronal death, as indicated by neutral red uptake in mouse primary cell cultures in vitro. The effect was accompanied by substantial decreases in neurotoxicity and neurodegeneration in terms of LDH release and Fluoro-Jade C staining of damaged cells, respectively. The mechanisms of the neuroprotective action of PaPE-1 also involved apoptosis inhibition demonstrated by normalization of both mitochondrial membrane potential and expression levels of apoptosis-related genes and proteins such as Fas, Fasl, Bcl2, FAS, FASL, BCL2, BAX, and GSK3β. Furthermore, PaPE-1-evoked neuroprotection was mediated through a reduction in ROS formation and restoration of cellular metabolic activity that had become dysregulated due to hypoxia and ischemia. These data provide evidence that targeting membrane non-GPER estrogen receptors with PaPE-1 is an effective therapy that protects brain neurons from hypoxic/ischemic damage, even when applied with a 6-h delay from injury onset.
    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
    雌激素发挥迅速,通过它们对质膜雌激素受体(mERs)的作用产生核外作用。与细胞膜相关的Gα蛋白参与雌激素调节的许多重要过程。然而,对Gα在mER介导的信号传导中的作用和所涉及的信号传导途径知之甚少。本文旨在概述Gα在mER介导的信号传导中的作用。免疫印迹,免疫荧光,免疫共沉淀,使用血管内皮细胞(EC)和人乳腺癌细胞系作为实验模型进行RNA干扰。使用豚鼠作为动物模型进行电生理学和免疫细胞化学。最近的进展表明,mERα通过Gα的信号传导是血管EC迁移或内皮H2S释放所必需的,而Gα13参与雌激素诱导的乳腺癌细胞侵袭。此外,Gαq偶联的PLC-PKC-PKA通路对于能量稳态的神经调节至关重要。本文综述了Gα对mER介导的信号传导的贡献。包括心血管保护,乳腺癌转移,稳态功能的神经调节,和成骨。
    Estrogens exert rapid, extranuclear effects by their action on the plasma membrane estrogen receptors (mERs). Gα protein associated with the cell membrane is involved in many important processes regulated by estrogens. However, the Gα\'s role in the mER-mediated signaling and the signaling pathways involved are poorly understood. This review aims to outline the Gα\'s role in the mER-mediated signaling. Immunoblotting, immunofluorescence, co-immunoprecipitation, and RNA interference were carried out using vascular endothelial cells (ECs) and human breast carcinoma cell lines as experimental models. Electrophysiology and immunocytochemistry were carried out using guinea pigs as animal models. Recent advances suggest that the signaling of mERα through Gα is required for vascular EC migration or endothelial H2S release, while Gα13 is involved in estrogen-induced breast cancer cell invasion. Besides, the Gαq-coupled PLC-PKC-PKA pathway is critical for the neural regulation of energy homeostasis. This review summarizes the contributions of Gα to mER-mediated signaling, including cardiovascular protection, breast cancer metastasis, neural regulation of homeostatic functions, and osteogenesis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    This review provides information on the structure of estrogen receptors (ERs), their localization and functions in mammalian cells. Additionally, the structure of proteasomes and mechanisms of protein ubiquitination and cleavage are described. According to the modern concept, the ubiquitin proteasome system (UPS) is involved in the regulation of the activity of ERs in several ways. First, UPS performs the ubiquitination of ERs with a change in their functional activity. Second, UPS degrades ERs and their transcriptional regulators. Third, UPS affects the expression of ER genes. In addition, the opportunity of the regulation of proteasome functioning by ERs-in particular, the expression of immune proteasomes-is discussed. Understanding the complex mechanisms underlying the regulation of ERs and proteasomes has great prospects for the development of new therapeutic agents that can make a significant contribution to the treatment of diseases associated with the impaired function of these biomolecules.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    永生化小鼠促性腺激素细胞系αT3-1和LβT2细胞已成为原代促性腺激素的替代模型。这些细胞系提供了同质的细胞群,与分离的前垂体相比,其中含有可能对雌二醇17β(E2)有反应的异质细胞群。假定E2的非经典作用通过质膜雌激素受体1(ESR1,也称为ERalpha)发生。这些作用包括抑制促性腺激素释放激素(GnRH)诱导的细胞内钙浓度增加以及在绵羊垂体(包括初级促性腺激素)中p44/42促分裂原活化蛋白激酶(ERK-1/2)的磷酸化。本实验的目的是确定alphaT3-1和LbetaT2是否是具有限制的细胞模型,无法检查促性腺激素中E2的非经典作用。进行实验以使用生物素化细胞和分离表面蛋白并用荧光标记的E2缀合物染色来确定细胞是否在质膜处具有ESRl。αT3-1细胞含有与质膜的脂筏相关但不富集的ESRl,并且在结合E2时不易位到脂筏。相比之下,LbetaT2细胞缺乏与质膜相关的ESR1。对于两种细胞类型,用E2预处理均不会抑制GnRH刺激的细胞内钙浓度增加。在任一细胞类型中,E2均未刺激ERK-1/2的磷酸化。尽管这些细胞系已被广泛用于研究GnRH信号,E2的非经典作用的体外或体内作用不能在任一细胞系中复制。
    The immortalized mouse gonadotrope cell lines alphaT3-1 and LbetaT2 cells have been a substitute model for primary gonadotropes. These cell lines have provided a homogeneous cell population, as compared to the dissociated anterior pituitaries, which contain a heterogeneous population of cells potentially responsive to estradiol-17beta (E2). Nonclassical actions of E2 assumed to occur through the plasma membrane estrogen receptor 1 (ESR1, also known as ERalpha). These actions have included inhibition of gonadotropin-releasing hormone (GnRH)-induced increases in intracellular calcium concentrations and phosphorylation of p44/42 mitogen-activated protein kinase (ERK-1/2) in ovine pituitaries including primary gonadotropes in vitro. The objective of the present experiment was to determine if alphaT3-1 and LbetaT2 are cell models with limitations to examine the nonclassical actions of E2 occurring in gonadotropes. Experiments were conducted to determine if the cells have ESR1 at the plasma membrane using biotinylation cell and isolation of surface protein and staining with a fluorescently labeled E2 conjugate. The alphaT3-1 cells contain ESR1 associated with but not enriched within lipid rafts of the plasma membrane and do not translocate to lipid rafts upon binding of E2. In contrast, LbetaT2 cells lack ESR1 associated with the plasma membrane. Pretreatment with E2 did not cause inhibition of GnRH-stimulated increases in intracellular concentrations of calcium for either cell type. Phosphorylation of ERK-1/2 was not stimulated by E2 in either cell type. Although these cells lines have been used extensively to study GnRH signaling, in vitro or in vivo effects of nonclassical actions of E2 cannot be replicated in either cell line.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    膜相关雌激素受体(mER)的激活会降低雌性大鼠的食物和水摄入量。其他研究表明,这些影响是介导的,至少在某种程度上,通过膜相关雌激素受体α(ERα)。然而,ERα摄取作用所需的关键作用位点和细胞内信号仍不清楚.给予内侧视前区(mPOA)的雌二醇可减少摄食行为,和膜相关的ERα已被证明通过与代谢型谷氨酸受体(mGluR)亚型的相互作用影响细胞内信号,但是这个信号通路的参与,在MPOA中,在摄食行为方面仍未测试。为了解决这些悬而未决的问题,我们首先表明,mPOA中mER的激活降低了过夜食物和水的摄入量,并且在与基因组作用机制一致的时间过程中这样做。接下来,我们测试了mPOA中mGluR1a信号传导对雌二醇的厌食和抗二异基因作用的需求。不出所料,mPOA中的雌二醇减少了食物摄入量,但仅在没有mGluR1a拮抗剂的情况下。雌二醇对水摄入量的影响并非如此,其不受mGluR1a拮抗剂的影响。这些结果表明,雌激素需要mGluR激活,至少它们对摄取行为的一些影响,并表明mPOA是一个关键的行动点。结果还揭示了雌激素对摄取行为的控制存在有趣的分歧,mPOA中的mGluR信号在控制食物摄入中起作用,但不是水的摄入量。
    Activation of membrane-associated estrogen receptors (mER) decreases food and water intake in female rats. Additional studies suggest these effects are mediated, at least in part, by membrane-associated estrogen receptor alpha (ERα). Nevertheless, the critical site of action and the intracellular signaling required for the ingestive effects of ERα remain unclear. Estradiol given to the medial preoptic area (mPOA) decreases ingestive behaviors, and membrane-associated ERα has been shown to affect intracellular signaling through interactions with metabotropic glutamate receptor (mGluR) subtypes, but an involvement of this signaling pathway, in the mPOA, in ingestive behavior remains untested. To address these open questions, we first showed that activation of mER in the mPOA decreased both overnight food and water intake, and did so in a time course consistent with a genomic mechanism of action. Next, we tested the requirement of mGluR1a signaling in the mPOA for the anorexigenic and anti-dipsogenic effects of estradiol. As expected, estradiol in the mPOA decreased food intake, but only in the absence of an mGluR1a antagonist. The same was not true for estradiol effects on water intake, which were unaffected by an mGluR1a antagonist. These results suggest that estrogens require mGluR activation for at least some of their effects on ingestive behaviors, and indicate that the mPOA is a critical site of action. The results also reveal an interesting divergence in the estrogenic control of ingestive behavior by which mGluR signaling in the mPOA plays a role in the control of food intake, but not water intake.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Rapid nongenomic signaling by estrogens (Es), initiated near the cell membrane, provides new explanations for the potent actions of environmental chemicals that imperfectly mimic physiological Es. These pathways can affect tumor growth, stabilization, or shrinkage via a number of signaling streams such as activation/inactivation of mitogen-activated protein kinases and caspases, generation of second messengers, and phospho-triggering of cyclin instability. Though prostate cancers are better known for their responsiveness to androgen deprivation, ∼17% of late stage tumors regress in response to high dose natural or pharmaceutical Es; however, the mechanisms at the cellular level are not understood. More accurate recent measurements show that estradiol (E2) levels decline in aging men, leading to the hypothesis that maintaining young male levels of E2 may prevent the growth of prostate cancers. Major contributions to reducing prostate cancer cell numbers included low E2 concentrations producing sustained ERK phospho-activation correlated with generation of reactive oxygen species causing cancer cell death, and phospho-activation of cyclin D1 triggering its rapid degradation by interrupting cell cycle progression. These therapeutic actions were stronger in early stage tumor cells (with higher membrane estrogen receptor levels), and E2 was far more effective compared to diethylstilbestrol (the most frequently prescribed E treatment). Xenoestrogens (XEs) exacerbated the growth of prostate cancer cells, and as we know from previous studies in pituitary cancer cells, can interfere with the nongenomic signaling actions of endogenous Es. Therefore, nongenomic actions of physiological levels of E2 may be important deterrents to the growth of prostate cancers, which could be undermined by the actions of XEs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号