Estrogen Receptor alpha

雌激素受体 α
  • 文章类型: Journal Article
    UBE2M,一种NEDD8结合酶,在各种人类癌症中失调并促进肿瘤细胞增殖。然而,其在雌激素受体阳性(ER+)乳腺癌中的作用尚不清楚.我们发现UBE2M在ER+乳腺癌组织中的表达显著高于在ER阴性(ER-)乳腺癌组织中的表达。UBE2M的较高表达表明ER+乳腺癌患者的预后较差,但ER-乳腺癌患者的预后较差。感兴趣的,在UBE2M和ERα之间观察到正反馈回路。具体来说,ERα增强HIF-1α介导的UBE2M转录。反过来,UBE2M通过UBE2M-CUL3/4A-E6AP-ERα轴抑制其泛素化和降解来维持ERα表达。功能上,UBE2M的沉默通过诱导细胞周期停滞和凋亡来抑制乳腺癌细胞的生长,并在体外和体内提高其对氟维司群的敏感性。总之,我们的研究结果表明,UBE2M-ERα反馈回路驱动乳腺癌进展和氟维司群耐药,提示UBE2M作为ER+乳腺癌内分泌治疗的可行靶点。
    UBE2M, a NEDD8-conjugating enzyme, is dysregulated in various human cancers and promotes tumor cell proliferation. However, its role in estrogen receptor-positive (ER+) breast cancer remains unknown. We found that UBE2M expression was significantly higher in ER+ breast cancer tissues than in ER-negative (ER-) breast cancer tissues. Higher expression of UBE2M indicated a poorer prognosis in patients with ER+ breast cancer but not in those with ER- breast cancer. Of interest, a positive feedback loop was observed between UBE2M and ERα. Specifically, ERα enhanced the HIF-1α-mediated transcription of UBE2M. In turn, UBE2M maintained ERα expression by inhibiting its ubiquitination and degradation through UBE2M-CUL3/4A-E6AP-ERα axis. Functionally, silencing of UBE2M suppressed the growth of breast cancer cells by inducing cell cycle arrest and apoptosis and improved their sensitivity to fulvestrant both in vitro and in vivo. Altogether, our findings reveal that the UBE2M-ERα feedback loop drives breast cancer progression and fulvestrant resistance, suggesting UBE2M as a viable target for endocrine therapy of ER+ breast cancer.
    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
    确定控制癌细胞增殖和存活的主要表观遗传因素允许发现可利用的新分子靶标以克服对当前药理学方案的抗性。在乳腺癌(BC),对内分泌治疗(ET)的抵抗是由遗传和表观遗传事件引起的异常雌激素受体α(ERα)信号引起的。靶向ERα途径的关键上游组分提供了一种独立于任何其他下游事件干扰癌细胞中雌激素信号传导的方法。通过将全基因组“drop-out”筛选的计算分析与siRNA介导的基因敲除(kd)相结合,我们在腔样中发现了一组必需基因,ERα+BC,包括BRPF1,编码一种含溴结构域蛋白,属于一个表观遗传学读写器家族,作为染色质重塑体来控制基因转录。为了收集BRPF1在BC和ERα信号传导中的作用的机制见解,我们应用了染色质和转录组分析,基因消融和靶向药理抑制与细胞和功能测定相结合。结果表明,BRPF1与ERα结合到BC细胞染色质上,其阻断抑制细胞周期进程,通过调节染色质可及性减少细胞增殖并介导转录组变化。这种效应是由雌激素信号的广泛抑制引起的,由于ERα基因沉默,在抗雌激素(AE)敏感和耐药的BC细胞和临床前患者衍生模型(PDO)中。BRPF1与ERα的功能相互作用的表征揭示了雌激素反应性BC细胞存活的新调节剂,并表明该表观遗传因子是治疗这些肿瘤的潜在新靶标。
    Identifying master epigenetic factors controlling proliferation and survival of cancer cells allows to discover new molecular targets exploitable to overcome resistance to current pharmacological regimens. In breast cancer (BC), resistance to endocrine therapy (ET) arises from aberrant Estrogen Receptor alpha (ERα) signaling caused by genetic and epigenetic events still mainly unknown. Targeting key upstream components of the ERα pathway provides a way to interfere with estrogen signaling in cancer cells independently from any other downstream event. By combining computational analysis of genome-wide \'drop-out\' screenings with siRNA-mediated gene knock-down (kd), we identified a set of essential genes in luminal-like, ERα + BC that includes BRPF1, encoding a bromodomain-containing protein belonging to a family of epigenetic readers that act as chromatin remodelers to control gene transcription. To gather mechanistic insights into the role of BRPF1 in BC and ERα signaling, we applied chromatin and transcriptome profiling, gene ablation and targeted pharmacological inhibition coupled to cellular and functional assays. Results indicate that BRPF1 associates with ERα onto BC cell chromatin and its blockade inhibits cell cycle progression, reduces cell proliferation and mediates transcriptome changes through the modulation of chromatin accessibility. This effect is elicited by a widespread inhibition of estrogen signaling, consequent to ERα gene silencing, in antiestrogen (AE) -sensitive and -resistant BC cells and pre-clinical patient-derived models (PDOs). Characterization of the functional interplay of BRPF1 with ERα reveals a new regulator of estrogen-responsive BC cell survival and suggests that this epigenetic factor is a potential new target for treatment of these tumors.
    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
    性类固醇调节哺乳动物白色脂肪组织的分布。此外,WAT重塑需要脂肪细胞祖细胞。然而,调节脂肪细胞祖细胞的性别依赖性机制仍未确定。这里,我们发现Cxcr4以性二态方式作用,影响增殖细胞池,导致女性脂肪量受限。我们发现在Pparγ表达细胞中Cxcr4的缺失导致雌性,不是男性,脂肪营养不良,不能通过高脂肪饮食来恢复。此外,Cxcr4缺失与增殖的脂肪细胞祖细胞池的损失相关。Cxcr4损失伴随着与雌二醇超敏反应和脂肪生成停滞相关的脂肪来源的PPARγ标记细胞中雌激素受体α的上调。雌激素去除或抗雌激素的施用可恢复Cxcr4缺陷小鼠中WAT积累和脂肪来源细胞的动力学。这些发现暗示Cxcr4是女性脂肪生成变阻器,这可能会为针对女性肥胖的策略提供信息。
    Sex steroids modulate the distribution of mammalian white adipose tissues. Moreover, WAT remodeling requires adipocyte progenitor cells. Nevertheless, the sex-dependent mechanisms regulating adipocyte progenitors remain undetermined. Here, we uncover Cxcr4 acting in a sexually dimorphic manner to affect a pool of proliferating cells leading to restriction of female fat mass. We find that deletion of Cxcr4 in Pparγ-expressing cells results in female, not male, lipodystrophy, which cannot be restored by high-fat diet consumption. Additionally, Cxcr4 deletion is associated with a loss of a pool of proliferating adipocyte progenitors. Cxcr4 loss is accompanied by the upregulation of estrogen receptor alpha in adipose-derived PPARγ-labelled cells related to estradiol hypersensitivity and stalled adipogenesis. Estrogen removal or administration of antiestrogens restores WAT accumulation and dynamics of adipose-derived cells in Cxcr4-deficient mice. These findings implicate Cxcr4 as a female adipogenic rheostat, which may inform strategies to target female adiposity.
    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:雌激素受体在近70%的乳腺癌(ER阳性)中表达。雌激素受体α作为乳腺癌进展的重要因素在早期选择治疗方法中起着重要作用。因此,对非侵入性技术的关注激增,包括循环无细胞DNA(ccfDNA)或无细胞DNA(cfDNA),检测和跟踪ESR1基因型。因此,本研究旨在通过系统评价和综合荟萃分析,探讨无细胞DNA检测ESR1突变在乳腺癌患者中的诊断准确性.
    方法:PubMed,Embase,并在2022年4月6日之前搜索了WebofScience数据库。cfDNA对ESR1测量的诊断研究,这是用肿瘤组织活检证实的,已被纳入研究。敏感性,特异性,准确度,阳性预测值(PPV),负预测值(NPV),分析数据时考虑了正似然比(PLR)和负似然比(NLR).
    结果:在649篇论文中,13篇论文和15个队列,包括389名参与者,进入荟萃分析。综合荟萃分析表明其敏感性较高(75.52,95%CI60.19-90.85),特异性(88.20,95%CI80.99-95.40),血浆ESR1的高精度为88.96(95%CI83.23-94.69)。我们还发现中等PPV为56.94(95%CI41.70-72.18),但高NPV为88.53(95%CI82.61-94.44)。我们还发现NLR为0.443(95%CI0.09-0.79),PLR为1.60(95%CI1.20-1.99)。
    结论:本系统综述和综合荟萃分析显示,血浆cfDNA检测在检测乳腺癌患者的ESR1突变方面具有较高的敏感性和特异性。这表明该测试可能是有价值的诊断工具。它可以作为一种可靠的非侵入性技术来识别乳腺癌患者的ESR1突变。然而,需要更广泛的研究来证实其预后价值.
    BACKGROUND: Estrogen receptors express in nearly 70% of breast cancers (ER-positive). Estrogen receptor alpha plays a fundamental role as a significant factor in breast cancer progression for the early selection of therapeutic approaches. Accordingly, there has been a surge of attention to non-invasive techniques, including circulating Cell-free DNA (ccfDNA) or Cell-Free DNA (cfDNA), to detect and track ESR1 genotype. Therefore, this study aimed to examine the diagnosis accuracy of ESR1 mutation detection by cell-free DNA in breast cancer patientsthrough a systematic review and comprehensive meta-analysis.
    METHODS: PubMed, Embase, and Web of Science databases were searched up to 6 April 2022. Diagnostic studies on ESR1 measurement by cfDNA, which was confirmed using the tumour tissue biopsy, have been included in the study. The sensitivity, specificity, accuracy, positive predictive value (PPV), negative predictive value (NPV), positive likelihood ratio (PLR) and negative likelihood ratio (NLR) were considered to analyse the data.
    RESULTS: Out of 649 papers, 13 papers with 15 cohorts, including 389 participants, entered the meta-analyses. The comprehensive meta-analysis indicated a high sensitivity (75.52, 95% CI 60.19-90.85), specificity (88.20, 95% CI 80.99-95.40), and high accuracy of 88.96 (95% CI 83.23-94.69) for plasma ESR1. We also found a moderate PPV of 56.94 (95% CI 41.70-72.18) but a high NPV of 88.53 (95% CI 82.61-94.44). We also found an NLR of 0.443 (95% CI 0.09-0.79) and PLR of 1.60 (95% CI 1.20-1.99).
    CONCLUSIONS: This systematic review and comprehensive meta-analysis reveal that plasma cfDNA testing exhibits high sensitivity and specificity in detecting ESR1 mutations in breast cancer patients. This suggests that the test could be a valuable diagnostic tool. It may serve as a dependable and non-invasive technique for identifying ESR1 mutations in breast cancer patients. However, more extensive research is needed to confirm its prognostic value.
    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:雌激素受体阳性(ER+)乳腺癌约占病例的80%,他莫昔芬是选择新辅助化疗。然而,很大比例的患者出现化学耐药性,妥协的恢复。临床证据表明,高血浆水平的低密度脂蛋白(LDL)可以促进癌症进展。本研究分析了LDL对初级血浆活性他莫昔芬代谢产物抗性获得的影响,4-羟基他莫昔芬(4OH-Tam)和4-羟基-N-去甲基-他莫昔芬(endoxifen),乳腺癌ER+细胞(MCF-7)。
    方法:两种抗性细胞变体,MCF-7Var-H和MCF-7Var-I,通过一种新的策略产生,并评估了它们的表型特征。通过MTT测定进行表型评估,细胞术,免疫荧光显微镜,酶谱和蛋白质表达分析。
    结果:MCF-7Var-H,仅由他莫昔芬代谢物产生,显示激素受体的关键下调,增强的迁移能力,金属蛋白酶9胞外介质排泄,和与天然MCF-7相反的间质形态,表明向三阴性乳腺癌(TNBC)表型的转变。相比之下,MCF-7Var-I在高LDL培养基中产生,仅显示ER的轻微上调和其他不太明显的代谢适应。结果表明转录因子核因子红系2相关因子2(Nrf2)在变体之间观察到的表型差异中的潜在作用。
    结论:在他莫昔芬代谢产物化学抗性获取过程中,高或低浓度的LDL会导致与化学抗性相关的不同细胞机制。可能涉及与Nrf2活性相关的新型适应性细胞应答。
    OBJECTIVE: Estrogen receptor-positive (ER+) breast cancer represents about 80% of cases, tamoxifen is the election neoadjuvant chemotherapy. However, a large percentage of patients develop chemoresistance, compromising recovery. Clinical evidence suggests that high plasmatic levels of low-density lipoproteins (LDL) could promote cancer progression. The present study analyzed the effect of LDL on the primary plasmatic active Tamoxifen\'s metabolites resistance acquisition, 4-hydroxytamoxifen (4OH-Tam) and 4-hydroxy-N-desmethyl-tamoxifen (endoxifen), in breast cancer ERα + cells (MCF-7).
    METHODS: Two resistant cellular variants, MCF-7Var-H and MCF-7Var-I, were generated by a novel strategy and their phenotype features were evaluated. Phenotypic assessment was performed by MTT assays, cytometry, immunofluorescence microscopy, zymography and protein expression analysis.
    RESULTS: MCF-7Var-H, generated only with tamoxifen metabolites, showed a critical down-regulation in hormone receptors, augmented migration capacity, metalloprotease 9 extracellular medium excretion, and a mesenchymal morphology in contrast with native MCF-7, suggesting the transition towards Triple-negative breast cancer (TNBC) phenotype. In contrast, MCF-7Var-I which was generated in a high LDL media, showed only a slight upregulation in ER and other less noticeable metabolic adaptations. Results suggest a potential role of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) in phenotypic differences observed among variants.
    CONCLUSIONS: LDL high or low concentrations during Tamoxifen´s metabolites chemoresistance acquisition leads to different cellular mechanisms related to chemoresistance. A novel adaptative cellular response associated with Nrf2 activity could be implicated.
    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
    生物信号转导网络对于生命所有领域的信息处理和基因表达调控至关重要。众所周知,失调会导致各种各样的疾病,包括癌症。这里我介绍自洽的信号转导分析,它利用基因组规模的组学数据(特别是转录组学和/或蛋白质组学),以便以个性化的方式预测通过这些网络的信息流。我将该方法应用于乳腺癌患者内分泌治疗的研究,并表明抑制雌激素受体α的药物会引起广泛的抗肿瘤作用,并且它们最有临床影响力的是通过调节控制基因GREB1,HK1,AKT1,MAPK1,AKT2和NQO1的增殖信号。这种方法为研究人员提供了一个有价值的工具,可以理解失调是如何发生的,为什么会发生。以及对网络的扰动(如靶向治疗)如何影响网络本身,以及最终的患者结果。
    Biological signal transduction networks are central to information processing and regulation of gene expression across all domains of life. Dysregulation is known to cause a wide array of diseases, including cancers. Here I introduce self-consistent signal transduction analysis, which utilizes genome-scale -omics data (specifically transcriptomics and/or proteomics) in order to predict the flow of information through these networks in an individualized manner. I apply the method to the study of endocrine therapy in breast cancer patients, and show that drugs that inhibit estrogen receptor α elicit a wide array of antitumoral effects, and that their most clinically-impactful ones are through the modulation of proliferative signals that control the genes GREB1, HK1, AKT1, MAPK1, AKT2, and NQO1. This method offers researchers a valuable tool in understanding how and why dysregulation occurs, and how perturbations to the network (such as targeted therapies) effect the network itself, and ultimately patient outcomes.
    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
    癌症的发展和进展通常与基因失调有关。通常由转录因子(TF)序列或表达的变化引起。鉴定参与癌基因调控的关键TFs为潜在的新疗法提供了框架。本研究提出了一个大规模的癌基因TF-DNA相互作用网络,以及未来研究的广泛启动子克隆资源。高度连接的TFs与与癌症预后良好或不良相关的基因启动子结合,这表明,旨在改变这两个预后组之间基因表达平衡的策略可能本质上是复杂的。然而,我们确定了癌基因靶向治疗的潜力,一半的测试癌基因可能通过影响特定的激活剂或双功能TFs而被抑制。最后,我们研究了关键癌症相关TFESR1内在无序区域在DNA结合和转录活性中的作用,并发现这些区域在TF函数中可以有复杂的权衡。总之,我们的研究拓宽了我们对与癌基因调控有关的TFs的认识,并为未来的研究和治疗提供了宝贵的资源.
    Cancer development and progression are generally associated with gene dysregulation, often resulting from changes in the transcription factor (TF) sequence or expression. Identifying key TFs involved in cancer gene regulation provides a framework for potential new therapeutics. This study presents a large-scale cancer gene TF-DNA interaction network, as well as an extensive promoter clone resource for future studies. Highly connected TFs bind to promoters of genes associated with either good or poor cancer prognosis, suggesting that strategies aimed at shifting gene expression balance between these two prognostic groups may be inherently complex. However, we identified potential for oncogene-targeted therapeutics, with half of the tested oncogenes being potentially repressed by influencing specific activators or bifunctional TFs. Finally, we investigate the role of intrinsically disordered regions within the key cancer-related TF ESR1 in DNA binding and transcriptional activity, and found that these regions can have complex trade-offs in TF function. Altogether, our study broadens our knowledge of the TFs involved in cancer gene regulation and provides a valuable resource for future studies and therapeutics.
    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
    双重靶向染色质调节和DNA损伤修复信号为癌症治疗提供了有希望的途径。应用合理的药物设计,我们合成了一种有效的双靶向小分子,SP-1-303.这里,我们报道了SP-1-303作为I类同工型选择性组蛋白去乙酰化酶(HDAC)抑制剂和共济失调-毛细血管扩张突变蛋白(ATM)的激活剂.体外酶分析显示HDAC1和HDAC3的选择性抑制。细胞生长抑制研究表明,SP-1-303对MCF-7和T47D细胞的有效生长抑制浓度(EC50)为0.32至0.34μM,差异抑制雌激素受体阳性乳腺癌(ERBC)细胞的生长。与三阴性乳腺癌细胞的1.2-2.5μM相比,和约12μM的正常乳腺上皮细胞。Western分析显示,SP-1-303降低雌激素受体α(ER-α)表达并增加p53蛋白表达,同时诱导ATM及其底物的磷酸化,BRCA1和p53在ER+BC细胞中呈时间依赖性。药代动力学评价表明,在大鼠模型中静脉内给药后,曲线下面积(AUC)为5227.55ng/ml×h,消除半衰期为1.26h。总的来说,SP-1-303作为一种新型第二代I类(HDAC1和HDAC3)选择性HDAC抑制剂,和ATM激活器,能够调节ER表达,并抑制ER+BC细胞的生长。SP-1-303对I类HDAC和ATM的联合靶向为治疗ER+乳腺癌提供了有希望的治疗方法,并支持进一步的临床前评估。
    Dual-targeting chromatin regulation and DNA damage repair signaling presents a promising avenue for cancer therapy. Applying rational drug design, we synthesized a potent dual-targeting small molecule, SP-1-303. Here, we report SP-1-303 as a class I isoform selective histone deacetylase (HDAC) inhibitor and an activator of the ataxia-telangiectasia mutated protein (ATM). In vitro enzymatic assays demonstrated selective inhibition of HDAC1 and HDAC3. Cellular growth inhibition studies show that SP-1-303 differentially inhibits growth of estrogen receptor positive breast cancer (ER+ BC) cells with effective growth inhibition concentrations (EC50) for MCF-7 and T47D cells ranging from 0.32 to 0.34 μM, compared to 1.2-2.5 μM for triple negative breast cancer cells, and ~12 μM for normal breast epithelial cells. Western analysis reveals that SP-1-303 decreases estrogen receptor alpha (ER-α) expression and increases p53 protein expression, while inducing the phosphorylation of ATM and its substrates, BRCA1 and p53, in a time-dependent manner in ER+ BC cells. Pharmacokinetic evaluation demonstrates an area under the curve (AUC) of 5227.55 ng/ml × h with an elimination half-life of 1.26 h following intravenous administration in a rat model. Collectively, SP-1-303 emerges as a novel second generation class I (HDAC1 and HDAC3) selective HDAC inhibitor, and ATM activator, capable of modulating ER expression, and inhibiting growth of ER+ BC cells. Combined targeting of class I HDACs and ATM by SP-1-303 offers a promising therapeutic approach for treating ER+ breast cancers and supports further preclinical evaluation.
    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
    围绝经期对全球女性健康影响显著,尽管存在相关风险,但仍经常使用激素替代疗法(HRT)进行管理。这项研究探索了一种新颖的替代外泌体疗法,旨在刺激卵巢组织中雌激素的产生,因此提供了一种潜在的非激素治疗围绝经期症状。采用离体方法,用人脐带间充质干细胞来源的外泌体处理围绝经期女性卵巢皮质标本,并在特定条件下培养(专利号:PCT/US2022/073467).外泌体是在环磷酸鸟苷(cGMP)条件下产生的,确保高安全标准。使用酶联免疫吸附测定(ELISA)定量雌激素水平,通过定量聚合酶链反应(PCR)评估雌激素和卵泡刺激素(FSH)受体的基因表达变化。免疫组织化学(IHC)用于评估细胞增殖和凋亡标志物。结果表明,与对照组相比,治疗组织中的雌激素水平和雌激素受体α(Era)表达显着增加。此外,观察到凋亡标志物的减少和细胞增殖标志物的增加.这些发现表明,外泌体治疗可以有效增强围绝经期卵巢组织的雌激素产生并调节受体敏感性。这种方法可以作为HRT的更安全的替代品,与身体的自然调节机制保持一致,并可能为管理围绝经期症状提供更有效的治疗选择。
    Perimenopause significantly impacts women\'s health globally, often managed with hormone replacement therapy (HRT) despite the associated risks. This study explores a novel alternative exosome therapy, aimed at stimulating estrogen production in ovarian tissues, thus offering a potential non-hormonal treatment for perimenopausal symptoms. Employing ex vivo methodologies, ovarian cortex specimens from perimenopausal women were treated with exosomes derived from human umbilical cord mesenchymal stem cells and cultured under specific conditions (patent number: PCT/US2022/073467). The exosomes were produced under cyclic guanosine monophosphate (cGMP) conditions, ensuring high safety standards. Estrogen levels were quantified using enzyme-linked immunosorbent assay (ELISA), and gene expression changes in estrogen and follicle-stimulating hormone (FSH) receptors were assessed via quantitative polymerase chain reaction (PCR). Immunohistochemistry (IHC) was utilized to evaluate cellular proliferation and apoptotic markers. The results indicated a significant increase in estrogen levels and estrogen receptor-alpha (Erα) expression in treated tissues compared to controls. Additionally, a decrease in apoptotic markers and an increase in cellular proliferation markers were observed. These findings suggest that exosome therapy can effectively enhance estrogen production and modulate receptor sensitivity in perimenopausal ovarian tissues. This approach could serve as a safer alternative to HRT, aligning with the body\'s natural regulatory mechanisms and potentially offering a more effective treatment option for managing perimenopausal symptoms.
    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
    草甘膦,几种广谱除草剂的活性成分,在世界各地广泛使用,尽管已知许多不利影响。其中,它被认为是一种内分泌干扰物。这项工作旨在测试草甘膦对PNT1A人前列腺细胞的影响和潜在的内分泌干扰作用,永生化非肿瘤上皮细胞系,同时具有ERα和ERβ雌激素受体。结果表明,草甘膦诱导细胞毒性,线粒体功能障碍,并通过核易位快速激活ERα和ERβ。分子分析表明凋亡可能参与草甘膦诱导的细胞毒性。凋亡过程可归因于线粒体代谢的改变;因此,使用海马分析仪研究线粒体功能的主要参数。在草甘膦处理的细胞中观察到线粒体功能受损,随着ATP产量的减少,备用呼吸能力,和质子泄漏,随着线粒体偶联效率的提高。最后,免疫荧光分析的结果表明,草甘膦作为雌激素干扰物,决定了两种ER的核易位.核易位的发生与剂量无关,比特定的激素更快,并在整个治疗过程中坚持。总之,收集的结果表明,在非肿瘤前列腺细胞中,草甘膦可以引起细胞死亡,并充当异种雌激素,激活雌激素受体。激素功能的改变会对暴露动物的生殖健康产生负面影响,损害他们的生育能力。
    Glyphosate, the active ingredient of several broad-spectrum herbicides, is widely used throughout the world, although many adverse effects are known. Among these, it has been recognized as an endocrine disruptor. This work aimed to test the effects and potential endocrine disrupting action of glyphosate on PNT1A human prostate cells, an immortalized non-tumor epithelial cell line, possessing both ERα and ERβ estrogen receptors. The results showed that glyphosate induces cytotoxicity, mitochondrial dysfunction, and rapid activation of ERα and ERβ via nuclear translocation. Molecular analysis indicated a possible involvement of apoptosis in glyphosate-induced cytotoxicology. The apoptotic process could be attributed to alterations in mitochondrial metabolism; therefore, the main parameters of mitochondrial functionality were investigated using the Seahorse analyzer. Impaired mitochondrial function was observed in glyphosate-treated cells, with reductions in ATP production, spare respiratory capacity, and proton leakage, along with increased efficiency of mitochondrial coupling. Finally, the results of immunofluorescence analysis demonstrated that glyphosate acts as an estrogen disruptor determining the nuclear translocation of both ERs. Nuclear translocation occurred independent of dose, faster than the specific hormone, and persisted throughout treatment. In conclusion, the results collected show that in non-tumor prostate cells glyphosate can cause cell death and acts as a xenoestrogen, activating estrogen receptors. The consequent alteration of hormonal functions can have negative effects on the reproductive health of exposed animals, compromising their fertility.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号