glutathione peroxidase 4

谷胱甘肽过氧化物酶 4
  • 文章类型: Journal Article
    背景:铁凋亡是一种重要的细胞死亡类型,可导致心脏骤停(CA)和复苏后全身缺血再灌注引起的心肌功能障碍。萝卜硫素(SFN),被称为核因子E2相关因子2(Nrf2)的激活剂,已被证明能有效缓解局部心肌缺血再灌注损伤。本研究旨在探讨SFN是否可以通过抑制心肌细胞铁凋亡来改善复苏后心肌功能障碍及其潜在的调节机制。
    结果:建立了猪体内CA和复苏模型。构建缺氧/复氧(H/R)刺激的AC16心肌细胞作为体外模型,模拟CA和复苏过程。体外实验,SFN还原铁沉积相关的亚铁,脂质活性氧,和丙二醛,谷胱甘肽增加,并进一步促进AC16心肌细胞在H/R刺激后的细胞存活。机械上,用SFN激活Nrf2降低干扰素调节因子1(IRF1)的表达,然后降低其与谷胱甘肽过氧化物酶4(GPX4)启动子的结合,H/R刺激后AC16心肌细胞最终恢复后者的转录。体内实验,SFN逆转IRF1和GPX4的异常表达,抑制心脏铁性凋亡,改善猪CA和复苏后的心肌功能障碍。
    结论:SFN能有效改善CA和复苏后的心肌功能障碍,其机制可能与通过调节Nrf2/IRF1/GPX4通路抑制心肌细胞铁性凋亡有关。
    BACKGROUND: Ferroptosis is an important type of cell death contributing to myocardial dysfunction induced by whole body ischemia reperfusion following cardiac arrest (CA) and resuscitation. Sulforaphane (SFN), known as the activator of the nuclear factor E2-related factor 2 (Nrf2), has been proven to effectively alleviate regional myocardial ischemia reperfusion injury. The present study was designed to investigate whether SFN could improve post-resuscitation myocardial dysfunction by inhibiting cardiomyocytes ferroptosis and its potential regulatory mechanism.
    RESULTS: An in vivo pig model of CA and resuscitation was established. Hypoxia/reoxygenation (H/R)-stimulated AC16 cardiomyocytes was constructed as an in vitro model to simulate the process of CA and resuscitation. In vitro experiment, SFN reduced ferroptosis-related ferrous iron, lipid reactive oxygen species, and malondialdehyde, increased glutathione, and further promoted cell survival after H/R stimulation in AC16 cardiomyocytes. Mechanistically, the activation of Nrf2 with the SFN decreased interferon regulatory factor 1 (IRF1) expression, then reduced its binding to the promoter of glutathione peroxidase 4 (GPX4), and finally recovered the latter\'s transcription after H/R stimulation in AC16 cardiomyocytes. In vivo experiment, SFN reversed abnormal expression of IRF1 and GPX4, inhibited cardiac ferroptosis, and improved myocardial dysfunction after CA and resuscitation in pigs.
    CONCLUSIONS: SFN could effectively improve myocardial dysfunction after CA and resuscitation, in which the mechanism was potentially related to the inhibition of cardiomyocytes ferroptosis through the regulation of Nrf2/IRF1/GPX4 pathway.
    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
    新发现的雌激素受体,G蛋白偶联受体30(GPR30),在大脑中普遍存在,并已被证明可以提供显着的神经保护。最近的研究与铁中毒有关,一种新特征的程序性细胞死亡形式,与脑缺血再灌注损伤(CIRI)密切相关,强调它是一个主要的促成因素。因此,我们的研究旨在探索GPR30靶向在控制神经元铁性凋亡和减轻CIRI影响方面的潜力。结果表明,在小鼠模型中,GPR30激活不仅改善了神经系统预后并减少了梗死面积,而且减少了大脑中动脉闭塞(MCAO)后的铁积累和丙二醛形成。这种保护作用延伸到Nrf2和GPX4蛋白水平的增加。在使用GPR30特异性激动剂G1进行氧葡萄糖剥夺和再氧合(OGD/R)的PC12细胞中复制了类似的保护性结果。重要的是,用ML385抑制Nrf2削弱了GPR30激活的神经保护作用,表明GPR30主要通过上调Nrf2和GPX4来抑制神经元铁性凋亡来减轻CIRI。
    The newly identified estrogen receptor, G protein-coupled receptor 30 (GPR30), is prevalent in the brain and has been shown to provide significant neuroprotection. Recent studies have linked ferroptosis, a newly characterized form of programmed cell death, closely with cerebral ischemia-reperfusion injury (CIRI), highlighting it as a major contributing factor. Consequently, our research aimed to explore the potential of GPR30 targeting in controlling neuronal ferroptosis and lessening CIRI impacts. Results indicated that GPR30 activation not only improved neurological outcomes and decreased infarct size in a mouse model but also lessened iron accumulation and malondialdehyde formation post-middle cerebral artery occlusion (MCAO). This protective effect extended to increased levels of Nrf2 and GPX4 proteins. Similar protective results were replicated in PC12 cells subjected to Oxygen Glucose Deprivation and Reoxygenation (OGD/R) using the GPR30-specific agonist G1. Importantly, inhibition of Nrf2 with ML385 curtailed the neuroprotective effects of GPR30 activation, suggesting that GPR30 mitigates CIRI primarily through inhibition of neuronal ferroptosis via upregulation of Nrf2 and GPX4.
    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
    在骨科研究中,许多研究已经将维生素E用作保护性抗氧化剂或使用叔丁基过氧化氢诱导软骨细胞的氧化损伤。这些研究通常支持以下假设:关节病理学会导致氧化应激和脂质过氧化增加,脂质抗氧化剂可以预防这种情况,以改善细胞存活或功能和关节健康;然而,在临床试验中,补充脂质抗氧化剂对骨关节炎无效,动物数据不明确。此外,循环维生素E的增加与骨关节炎的发病率增加有关。台式和临床结果之间的这种脱节使我们假设氧化应激驱动的软骨细胞氧化还原功能范例不能捕获脂质抗氧化剂和促氧化剂对关节软骨细胞的代谢和生理作用。我们使用离体和体内软骨模型来研究脂质抗氧化剂对健康,小学,关节软骨细胞和应用免疫自旋捕获技术,以提供高水平的氧化应激独立于特定活性氧的广泛指标。关键发现表明脂质抗氧化剂是线粒体前,而脂质抗氧化剂降低了线粒体措施。在没有受伤的情况下,脂质抗氧化剂增加了自由基的形成;然而,在受伤的情况下,自由基形成减少。在无压力条件下,通过谷胱甘肽过氧化物酶4的过表达,软骨细胞线粒体与氧化还原调节之间的这种关系在体内得以再现。在18个月或以上的小鼠中,谷胱甘肽过氧化物酶4的过表达显着降低了线粒体过氧化物酶体增殖激活受体γ的存在,并破坏了线粒体与氧化还原环境之间的关系。这种复杂的相互作用表明,靶向关节软骨的策略可能受益于采用更细微的关节软骨细胞氧化还原代谢范例。
    In orthopedic research, many studies have applied vitamin E as a protective antioxidant or used tert-butyl hydroperoxide to induce oxidative injury to chondrocytes. These studies often support the hypothesis that joint pathology causes oxidative stress and increased lipid peroxidation that might be prevented with lipid antioxidants to improve cell survival or function and joint health; however, lipid antioxidant supplementation was ineffective against osteoarthritis in clinical trials and animal data have been equivocal. Moreover, increased circulating vitamin E is associated with increased rates of osteoarthritis. This disconnect between benchtop and clinical results led us to hypothesize that oxidative stress-driven paradigms of chondrocyte redox function do not capture the metabolic and physiologic effects of lipid antioxidants and prooxidants on articular chondrocytes. We used ex vivo and in vivo cartilage models to investigate the effect of lipid antioxidants on healthy, primary, articular chondrocytes and applied immuno-spin trapping techniques to provide a broad indicator of high levels of oxidative stress independent of specific reactive oxygen species. Key findings demonstrate lipid antioxidants were pro-mitochondrial while lipid prooxidants decreased mitochondrial measures. In the absence of injury, radical formation was increased by lipid antioxidants; however, in the presence of injury, radical formation was decreased. In unstressed conditions, this relationship between chondrocyte mitochondria and redox regulation was reproduced in vivo with overexpression of glutathione peroxidase 4. In mice aged 18 months or more, overexpression of glutathione peroxidase 4 significantly decreased the presence of pro-mitochondrial peroxisome proliferation activated receptor gamma and deranged the relationship between mitochondria and the redox environment. This complex interaction suggests strategies targeting articular cartilage may benefit from adopting more nuanced paradigms of articular chondrocyte redox metabolism.
    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
    砷,一种神经毒性的类金属,构成重大健康风险。然而,鞣花酸,以其抗氧化性能而闻名,已经显示出神经保护的潜力。本研究旨在探讨鞣花酸对砷诱导的神经元铁性凋亡和认知障碍的神经保护作用及其机制。使用砷暴露的Wistar大鼠模型和砷诱导的HT22细胞模型,我们评估了认知能力,测量血清和脑砷水平,并通过组织学分析和透射电子显微镜评估病理损伤。此外,我们使用GSH检查了氧化应激和铁离子水平,MDA,ROS和组织铁生化试剂盒,并使用westernblot和qRT-PCR分析铁凋亡相关标志物的表达。我们的结果显示,砷暴露会增加血清和大脑中的砷水平,导致海马病理性损伤和随后的学习和记忆能力下降。砷诱导的神经元铁凋亡是通过抑制xCT/GSH/GPX4/Nrf2信号轴和破坏铁代谢来介导的。值得注意的是,鞣花酸干预有效降低血清和脑砷水平,改善神经元损伤,和改善氧化应激,铁性凋亡,和认知障碍。这些有益作用与Nrf2/Keap1信号通路的激活有关。GPX4表达上调,和增强铁离子排泄。总之,鞣花酸通过减轻神经元的铁性凋亡和认知障碍,对砷诱导的神经毒性具有有希望的神经保护作用。
    Arsenic, a neurotoxic metalloid, poses significant health risks. However, ellagic acid, renowned for its antioxidant properties, has shown potential in neuroprotection. This study aimed to investigate the neuroprotective effects of ellagic acid against arsenic-induced neuronal ferroptosis and cognitive impairment and elucidate the underlying mechanisms. Using an arsenic-exposed Wistar rat model and an arsenic-induced HT22 cells model, we assessed cognitive ability, measured serum and brain arsenic levels, and evaluated pathological damage through histological analysis and transmission electron microscopy. Additionally, we examined oxidative stress and iron ion levels using GSH, MDA, ROS and tissue iron biochemical kits, and analyzed the expression of ferroptosis-related markers using western blot and qRT-PCR. Our results revealed that arsenic exposure increased both serum and brain arsenic levels, resulting in hippocampal pathological damage and subsequent decline in learning and memory abilities. Arsenic-induced neuronal ferroptosis was mediated by the inhibition of the xCT/GSH/GPX4/Nrf2 signaling axis and disruption of iron metabolism. Notably, ellagic acid intervention effectively reduced serum and brain arsenic levels, ameliorated neuronal damage, and improved oxidative stress, ferroptosis, and cognitive impairment. These beneficial effects were associated with the activation of the Nrf2/Keap1 signaling pathway, upregulation of GPX4 expression, and enhanced iron ion excretion. In conclusion, ellagic acid demonstrates promising neuroprotective effects against arsenic-induced neurotoxicity by mitigating neuronal ferroptosis and cognitive impairment.
    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
    乙型肝炎病毒相关的慢性急性肝衰竭(HBV-ACLF)是一种短期死亡率高的综合征,其预后在临床管理中至关重要。本研究旨在探讨谷胱甘肽过氧化物酶4(GPX4)在HBV-ACLF发生发展中的临床意义及其对90天死亡率的预后价值。
    通过RT-qPCR或ELISA检测289名参与者血清或外周血单个核细胞(PBMC)中GPX4、氧化应激相关分子和炎性细胞因子的表达水平,通过MethyLight测定PBMC中GPX4启动子的甲基化水平。
    HBV-ACLF患者PBMC和血清中GPX4的表达水平低于非HBV相关的慢加急性肝衰竭(非HBVACLF)患者,慢性乙型肝炎(CHB)和健康对照(HC)患者,而GPX4启动子的甲基化水平更高。在HBV-ACLF患者中,GPX4启动子的甲基化水平与氧化应激相关,炎症相关分子,和一些临床病理指标。GPX4启动子的甲基化水平被确定为HBV-ACLF患者90天死亡率的独立危险因素,并且在受试者工作特征曲线(AUROC)下产生了比终末期肝病模型更大的面积(MELD)评分预测90天死亡率。
    GPX4启动子甲基化水平具有作为HBV-ACLF患者90天死亡率预测指标的潜力。
    UNASSIGNED: Hepatitis B virus-associated acute-on-chronic liver failure (HBV-ACLF) is a syn-drome with a high short-term mortality rate, and its prognosis is critical in clinical management. This study aimed to investigate the clinical significance of glutathione peroxidase 4 (GPX4) in the occurrence and development of HBV-ACLF and its prognostic value for 90-day mortality.
    UNASSIGNED: The expression levels of GPX4, oxidative stress-related molecules and inflammatory cytokines in serum or peripheral blood mononuclear cells (PBMCs) of 289 participants were determined by RT-qPCR or ELISA, and the methylation level of GPX4 promoter in PBMCs was determined by MethyLight.
    UNASSIGNED: The expression levels of GPX4 in the PBMCs and serum of HBV-ACLF patients were lower than those in non-HBV-associated acute-on-chronic liver failure (non-HBV ACLF) patients, patients with chronic hepatitis B (CHB) and healthy control (HC) individuals, while the methylation level of the GPX4 promoter was greater. In HBV-ACLF patients, the methylation level of the GPX4 promoter is correlated with oxidative stress, inflammation-related molecules, and some clinicopathological indicators. The methylation level of the GPX4 promoter was identified as an independent risk factor for 90-day mortality in HBV-ACLF patients and yielded a larger area under the receiver operating characteristic curve (AUROC) than the model for end-stage liver disease (MELD) score in predicting 90-day mortality.
    UNASSIGNED: The GPX4 promoter methylation level has promising potential as a predictor of 90-day mortality in patients with HBV-ACLF.
    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:高尿酸(HUA)在心血管疾病的病理生理过程中发挥重要作用。研究表明,尿酸水平升高会对心血管健康产生不利影响。然而,高尿酸血症对心肌病的影响尚不确定.需要进一步的研究来阐明HUA与心肌病之间的关系。阐明其对心脏健康的潜在影响。
    结果:我们证明Uox-KO小鼠加速了心肌病的发展,导致心脏功能明显受损和心肌纤维化。同时,线粒体形态被破坏,脂质过氧化产物数量增加,抗氧化功能减弱。此外,我们评估了铁抑素-1(Fer-1)的作用,铁性凋亡抑制剂.Fer-1治疗可逆转HUA联合DOX治疗引起的心肌损伤。Benzbromarone,一种降尿酸的药物,通过DOX给药减轻Uox-KO小鼠的高尿酸血症,从而降低心肌纤维化和铁细胞凋亡。体外,我们观察到HUA联合DOX处理的心肌细胞活性显著降低,脂质活性氧(ROS)明显增加。之后,我们证明了HUA可以促进DOX的氧化应激,以线粒体ROS增加为特征,并下调谷胱甘肽过氧化物酶4(GPX4)的蛋白质水平。N-乙酰-L-半胱氨酸,抗氧化剂,抑制HUA通过增加GPX4表达促进DOX诱导的铁凋亡的过程。
    方法:我们证实HUA可加重心肌损伤。这对于治疗高尿酸血症患者的心脏损害具有临床意义。
    结论:我们的数据表明HUA促进心肌病。HUA通过增加氧化应激和下调GPX4促进DOX诱导的铁凋亡。
    Aims: High uric acid (HUA), as a pro-oxidant, plays a significant role in the pathophysiology of cardiovascular disease. Studies have indicated that elevated uric acid levels can adversely affect cardiovascular health. Nevertheless, the impact of hyperuricemia on cardiomyopathy remains uncertain. Further research is needed to elucidate the relationship between HUA and cardiomyopathy, shedding light on its potential implications for heart health. Results: We demonstrated that uricase knockout (Uox-KO) mice accelerated the development of cardiomyopathy, causing significantly impaired cardiac function and myocardial fibrosis. Meanwhile, the mitochondrial morphology was destroyed, the lipid peroxidation products increased in number and the antioxidant function was weakened. In addition, we evaluated the effects of ferrostatin-1 (Fer-1), the ferroptosis inhibitor. Myocardial damage can be reversed by the Fer-1 treatment caused by HUA combined with doxorubicin (DOX) treatment. Benzbromarone, a uric acid-lowering drug, decreases myocardial fibrosis, and ferroptosis by alleviating hyperuricemia in Uox-KO mice by DOX administration. In vitro, we observed that the activity of cardiomyocytes treated with HUA combined with DOX decreased significantly, and lipid reactive oxygen species (ROS) increased significantly. Afterward, we demonstrated that HUA can promote oxidative stress in DOX, characterized by increased mitochondrial ROS, and downregulate protein levels of glutathione peroxidase 4 (GPX4). N-acetyl-l-cysteine, an antioxidant, inhibits the process by which HUA promotes DOX-induced ferroptosis by increasing the GPX4 expression. Innovation: We verified that HUA can exacerbate myocardial damage. This has clinical implications for the treatment of cardiac damage in patients with hyperuricemia. Conclusions: Our data suggested that HUA promotes the cardiomyopathy. HUA promotes DOX-induced ferroptosis by increasing oxidative stress and downregulating GPX4. Antioxid. Redox Signal. 00, 00-00.
    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
    合成谷胱甘肽(GSH)需要半胱氨酸,辅酶A,其他含硫代谢物,大多数蛋白质。在大多数细胞中,半胱氨酸来自细胞外二硫键来源,包括胱氨酸,谷胱甘肽-二硫化物,和肽。硫氧还蛋白还原酶-1(TrxR1)-或谷胱甘肽-二硫化物还原酶(GSR)驱动的酶系统可以通过硫氧还蛋白促进胱氨酸还原,谷氧还蛋白,或其他硫氧还蛋白折叠蛋白。游离胱氨酸通过胱氨酸-谷氨酸反转运蛋白进入细胞,xCT,但系统性地,血浆谷胱甘肽-二硫化物可能作为胱氨酸来源占主导地位。Erastin,同时抑制xCT和电压依赖性阴离子通道,诱导铁细胞死亡,之所以如此命名,是因为这种类型的细胞死亡被铁螯合剂拮抗。许多癌细胞似乎倾向于铁性凋亡,这已被提议作为一个有针对性的癌症责任。铁凋亡与脂质过氧化和谷胱甘肽过氧化物酶4(GPX4)或铁凋亡抑制蛋白1(FSP1)的损失有关,它们都能防止脂质过氧化物的积累。已经提出,xCT抑制诱导的细胞半胱氨酸缺乏降低GSH水平,饥饿GPX4以减少功率并允许膜脂质过氧化物积累,从而导致铁中毒。然而,铁中毒的各个方面还没有完全理解,需要进一步审查,例如,既不破坏GSH合成,GSH的损失,也不破坏谷胱甘肽二硫化物还原酶(GSR),在动物模型中引发铁中毒。在这里,我们重新评估Erastin之间的关系,xCT,GPX4,细胞半胱氨酸和GSH,RSL3或ML162,和铁凋亡。我们的结论是,而Cys和铁蛋白都是癌症的潜在负担,他们彼此之间的关系仍然没有得到足够的理解。
    Cysteine is required for synthesis of glutathione (GSH), coenzyme A, other sulfur-containing metabolites, and most proteins. In most cells, cysteine comes from extracellular disulfide sources including cystine, glutathione-disulfide, and peptides. The thioredoxin reductase-1 (TrxR1)- or glutathione-disulfide reductase (GSR)-driven enzymatic systems can fuel cystine reduction via thioredoxins, glutaredoxins, or other thioredoxin-fold proteins. Free cystine enters cells thorough the cystine-glutamate antiporter, xCT, but systemically, plasma glutathione-disulfide might predominate as a cystine source. Erastin, inhibiting both xCT and voltage-dependent anion channels, induces ferroptotic cell death, so named because this type of cell death is antagonized by iron-chelators. Many cancer cells seem to be predisposed to ferroptosis, which has been proposed as a targetable cancer liability. Ferroptosis is associated with lipid peroxidation and loss of either glutathione peroxidase-4 (GPX4) or ferroptosis suppressor protein-1 (FSP1), which each prevent accumulation of lipid peroxides. It has been suggested that an xCT inhibition-induced cellular cysteine-deficiency lowers GSH levels, starving GPX4 for reducing power and allowing membrane lipid peroxides to accumulate, thereby causing ferroptosis. Aspects of ferroptosis are however not fully understood and need to be further scrutinized, for example that neither disruption of GSH synthesis, loss of GSH, nor disruption of glutathione disulfide reductase (GSR), triggers ferroptosis in animal models. Here we reevaluate the relationships between Erastin, xCT, GPX4, cellular cysteine and GSH, RSL3 or ML162, and ferroptosis. We conclude that, whereas both Cys and ferroptosis are potential liabilities in cancer, their relationship to each other remains insufficiently understood.
    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
    缺血再灌注损伤(IRI)可严重影响移植物的存活和预后,是肝移植过程中不可避免的事件。Ferroptosis是一种新的铁依赖性细胞死亡形式,其特征是铁积累和压倒性的脂质过氧化;它在形态上有所不同,基因,和其他众所周知的细胞死亡类型的生物化学(自噬,坏死,和凋亡)。越来越多的证据表明,铁死亡参与肝IRI的发病机制,靶向铁性凋亡可能是一种有前途的治疗方法。这里,我们回顾了铁死亡的途径和现象,探讨铁死亡与肝IRI的关系和意义,并讨论调节铁死亡减轻肝脏IRI的可能策略。
    Ischemia-reperfusion injury (IRI) can seriously affect graft survival and prognosis and is an unavoidable event during liver transplantation. Ferroptosis is a novel iron-dependent form of cell death characterized by iron accumulation and overwhelming lipid peroxidation; it differs morphologically, genetically, and biochemically from other well-known cell death types (autophagy, necrosis, and apoptosis). Accumulating evidence has shown that ferroptosis is involved in the pathogenesis of hepatic IRI, and targeting ferroptosis may be a promising therapeutic approach. Here, we review the pathways and phenomena involved in ferroptosis, explore the associations and implications of ferroptosis and hepatic IRI, and discuss possible strategies for modulating ferroptosis to alleviate the hepatic IRI.
    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: English Abstract
    目的:探讨钙连体苷E抑制肝癌细胞增殖和迁移的分子机制。
    方法:使用CCK-8测定和GPX4,SLC7A11,LC3,P62的表达以及Akt/mTOR的磷酸化检测了用钙杜洛苷E处理的HCC细胞系HepG2和Huh7的细胞活力变化。LY294002和雷帕霉素(自噬的抑制剂和激活剂,分别)使用EdU和Transwell测定法评估了钙杜洛苷E处理的HCC细胞的增殖和迁移。TCGA数据库用于探讨GPX4和SLC7A11在HCC和正常肝组织中的表达水平及其与患者生存结果的相关性。还使用RT-qPCR和Western印迹在HCC细胞和正常肝细胞中检测GPX4和SLC7A11表达。
    结果:卡伦杜洛苷E明显抑制肝癌细胞的活力。GPX4和SLC7A11在肝癌组织和细胞系中高表达,它们的表达水平与患者的生存率呈负相关。在HCC细胞系中,卡伦杜洛苷E显著抑制GPX4和SLC7A11蛋白的表达,激活Akt-mTOR通路,并增强了LC3Ⅱ的表达。雷帕霉素可显著增强钙连体苷E对GPX4和SLC7A11表达的抑制作用,而LY294002可减弱。抑制自噬通路明显减弱了钙连苷E对肝癌细胞增殖和迁移的抑制作用,而激活这一途径则产生相反的效果。
    结论:卡伦杜甙E通过自噬途径下调GPX4和SLC7A11的表达,从而抑制肝癌细胞的增殖和迁移。
    OBJECTIVE: To investigate the molecular mechanism through which calenduloside E inhibits hepatocellular carcinoma (HCC) cell proliferation and migration.
    METHODS: HCC cell lines HepG2 and Huh7 treated with calenduloside E were examined for changes in cell viability using CCK-8 assay and expressions of GPX4, SLC7A11, LC3, P62 and phosphorylation of Akt/mTOR using Western blotting. The effects LY294002 and Rapamycin (the inhibitor and activator of autophagy, respectively) on proliferation and migration of calenduloside E-treated HCC cells were evaluated using EdU and Transwell assays. The TCGA database was used to explore the expression levels of GPX4 and SLC7A11 in HCC and normal liver tissues and their correlation with the patients\'survival outcomes. GPX4 and SLC7A11 expressions were also detected in HCC cells and normal hepatocytes using RT-qPCR and Western blotting.
    RESULTS: Calenduloside E obviously inhibited the viability of HCC cells. GPX4 and SLC7A11 were highly expressed in HCC tissues and cell lines, and their expression levels were negatively correlated with the patients\'survival. In HCC cell lines, calenduloside E significantly inhibited the expressions of GPX4 and SLC7A11 proteins, activated the Akt-mTOR pathway, and enhanced the expression of LC3 Ⅱ. The inhibitory effect of calenduloside E on GPX4 and SLC7A11 expressions was significantly enhanced by rapamycin but attenuated by LY294002. Inhibiting the autophagy pathway obviously diminished the inhibitory effect of calenduloside E on proliferation and migration of HCC cells, while activating this pathway produced the opposite effect.
    CONCLUSIONS: Calenduside E inhibits the proliferation and migration of HCC cells by down-regulating GPX4 and SLC7A11 expression via the autophagy pathway.
    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
    三阴性乳腺癌(TNBC)的特点是预后严峻和挑战众多。我们研究的目的是研究胸苷酸合酶(TYMS)在TNBC中的作用及其对铁凋亡的影响。在数据库中分析了TYMS的表达,以及其预后相关性。通过免疫组织化学(IHC)鉴定TYMS阳性表达,而实时定量PCR(qRTPCR)用于测量各种细胞系中的TYMSmRNA水平。使用蛋白质印迹来评估蛋白质表达。细胞增殖,移动性,凋亡,和活性氧(ROS)水平使用CCK8,伤口划痕愈合试验,transwell分析,和流式细胞术,分别。此外,在BALB/c裸鼠中建立肿瘤异种移植模型用于进一步研究。测量肿瘤体积和重量,使用苏木精和曙红(H&E)染色进行组织病理学分析以评估肿瘤组织变化。IHC染色检测肿瘤组织中Ki67的表达。在TNBC中观察到TYMS的高表达,并发现与患者的不良预后有关。在各种细胞系中,TYMS在BT549细胞中表达最高。敲除TYMS导致细胞增殖和移动性的抑制,以及促进细胞凋亡。此外,敲除TYMS导致ROS和Fe2+水平的积累增加,随着ACLS4表达的上调和谷胱甘肽过氧化物酶4(GPX4)表达的下调。体内研究显示TYMS的敲低抑制肿瘤生长。此外,TYMS的敲低与mTOR的抑制有关,p-PI3K,和p-Akt表达式。我们的研究表明,TYMS的敲除通过铁凋亡抑制细胞增殖来抑制TNBC的进展。其潜在机制与PI3K/Akt通路有关。我们的研究为TYMS对TNBC的抑制作用提供了新的视野。
    Triple-negative breast cancer (TNBC) is characterized by a grim prognosis and numerous challenges. The objective of our study was to examine the role of thymidylate synthase (TYMS) in TNBC and its impact on ferroptosis. The expression of TYMS was analyzed in databases, along with its prognostic correlation. TYMS positive expression was identified through immunohistochemistry (IHC), while real-time quantitative PCR (qRTPCR) was employed to measure TYMS mRNA levels in various cell lines. Western blotting was utilized to assess protein expression. Cell proliferation, mobility, apoptosis, and reactive oxygen species (ROS) levels were evaluated using CCK8, wound scratch healing assay, transwell assay, and flow cytometry, respectively. Additionally, a tumor xenograft model was established in BALB/c nude mice for further investigation. Tumor volume and weight were measured, and histopathological analysis using hematoxylin and eosin (H&E) staining was conducted to assess tumor tissue changes. IHC staining was employed to detect the expression of Ki67 in tumor tissues. High expression of TYMS was observed in TNBC and was found to be correlated with poor prognosis in patients. Among various cell lines, TYMS expression was highest in BT549 cells. Knockdown of TYMS resulted in suppression of cell proliferation and mobility, as well as promotion of apoptosis. Furthermore, knockdown of TYMS led to increased accumulation of ROS and Fe2+ levels, along with upregulation of ACLS4 expression and downregulation of glutathione peroxidase 4 (GPX4) expression. In vivo studies showed that knockdown of TYMS inhibited tumor growth. Additionally, knockdown of TYMS was associated with inhibition of mTOR, p-PI3K, and p-Akt expression. Our research showed that the knockdown of TYMS suppressed the TNBC progression by inhibited cells proliferation via ferroptosis. Its underlying mechanism is related to the PI3K /Akt pathway. Our study provides a novel sight for the suppression effect of TYMS on TNBC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号