Thioredoxin reductase 1

硫氧还蛋白还原酶 1
  • 文章类型: Journal Article
    卵巢KGN颗粒样肿瘤细胞系通常用作人类颗粒细胞的模型,尤其是它能产生类固醇激素.为了进一步探索这一点,我们使用三个公共RNA序列数据集鉴定了KGN细胞与原代人颗粒细胞相比差异表达的基因.意义重大,我们发现抗氧化剂基因TXNRD1(硫氧还蛋白还原酶1)在KGN细胞中的表达非常高。这是不祥的,因为细胞色素P450酶在类固醇激素的生物合成过程中泄漏电子并产生活性氧。基因本体论(GO)分析确定类固醇生物合成和胆固醇代谢过程在原代颗粒细胞中更活跃,而在KGN单元格中,DNA处理,染色体分离和动粒途径更为突出。细胞色素P450胆固醇侧链裂解(CYP11A1)和细胞色素P450芳香化酶(CYP19A1)的表达,这对类固醇激素孕激素和雌激素的生物合成很重要,加上他们的电子传输链成员(FDXR,在培养的KGN细胞中测量FDX1,POR)。KGN细胞用1mM二丁酰基cAMP(dbcAMP)或10μM毛喉素处理,有或没有TXNRD1的siRNA敲低。我们还检查了抗氧化基因的表达,通过AmplexRed测定产生H2O2,并通过γH2Ax染色对DNA进行损伤。通过dbcAMP或毛喉素处理观察到CYP11A1和CYP19A1的显着增加。然而,没有发现H2O2水平或DNA损伤的显着变化。通过siRNA抑制TXNRD1的表达阻断了dbcAMP对CYP11A1和CYP19A1表达的刺激。因此,TXNRD1在KGN细胞的类固醇生成中起着如此关键的作用,并且它是如此的高表达,我们得出的结论是,KGN细胞可能不是研究卵巢类固醇生成之间相互作用的最适合的原代颗粒细胞模型,活性氧和抗氧化剂。
    The ovarian KGN granulosa-like tumour cell line is commonly used as a model for human granulosa cells, especially since it produces steroid hormones. To explore this further, we identified genes that were differentially expressed by KGN cells compared to primary human granulosa cells using three public RNA sequence datasets. Of significance, we identified that the expression of the antioxidant gene TXNRD1 (thioredoxin reductase 1) was extremely high in KGN cells. This is ominous since cytochrome P450 enzymes leak electrons and produce reactive oxygen species during the biosynthesis of steroid hormones. Gene Ontology (GO) analysis identified steroid biosynthetic and cholesterol metabolic processes were more active in primary granulosa cells, whilst in KGN cells, DNA processing, chromosome segregation and kinetochore pathways were more prominent. Expression of cytochrome P450 cholesterol side-chain cleavage (CYP11A1) and cytochrome P450 aromatase (CYP19A1), which are important for the biosynthesis of the steroid hormones progesterone and oestrogen, plus their electron transport chain members (FDXR, FDX1, POR) were measured in cultured KGN cells. KGN cells were treated with 1 mM dibutyryl cAMP (dbcAMP) or 10 μM forskolin, with or without siRNA knockdown of TXNRD1. We also examined expression of antioxidant genes, H2O2 production by Amplex Red assay and DNA damage by γH2Ax staining. Significant increases in CYP11A1 and CYP19A1 were observed by either dbcAMP or forskolin treatments. However, no significant changes in H2O2 levels or DNA damage were found. Knockdown of expression of TXNRD1 by siRNA blocked the stimulation of expression of CYP11A1 and CYP19A1 by dbcAMP. Thus, with TXNRD1 playing such a pivotal role in steroidogenesis in the KGN cells and it being so highly overexpressed, we conclude that KGN cells might not be the most appropriate model of primary granulosa cells for studying the interplay between ovarian steroidogenesis, reactive oxygen species and antioxidants.
    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
    硫氧还蛋白还原酶经常在各种实体瘤中过表达,作为对抗氧化应激增强的保护机制。该系统的抑制剂,比如Auranofin,能有效根除癌细胞.然而,硫氧还蛋白还原酶1(TrxR1)在肺癌中的临床意义,以及其拮抗剂作为治疗选择的潜力,需要进一步的实验验证。在这项研究中,我们观察到TrxR1在非小细胞肺癌(NSCLC)中的显著上调,而不是小细胞肺癌。此外,TrxR1表达与生存率相关,肿瘤体积,和组织学分类。我们开发了一种名为LW-216的新型TrxR1抑制剂,并评估了其在NSCLC中的抗肿瘤功效。我们的结果表明,LW-216在R371和G442位点与细胞内TrxR1有效结合,促进TrxR1泛素化并抑制TrxR1表达,而不影响TrxR2表达。通过产生活性氧(ROS)处理LW-216诱导的NSCLC细胞DNA损伤和细胞凋亡。重要的是,补充N-乙酰半胱氨酸(NAC)或异位TrxR1表达可逆转LW-216诱导的细胞凋亡。此外,LW-216在植入NSCLC细胞的小鼠中显示出有效的肿瘤生长抑制作用,减少异种移植物中的TrxR1表达。值得注意的是,与Auranofin相比,LW-216在体内表现出优异的抗肿瘤活性。总的来说,我们的研究提供了令人信服的证据,支持LW-216靶向TrxR1作为NSCLC有前景的治疗策略的潜力.
    Thioredoxin reductases are frequently overexpressed in various solid tumors as a protective mechanism against heightened oxidative stress. Inhibitors of this system, such as Auranofin, are effective in eradicating cancer cells. However, the clinical significance of thioredoxin reductase 1 (TrxR1) in lung cancer, as well as the potential for its antagonist as a treatment option, necessitated further experimental validation. In this study, we observed significant upregulation of TrxR1 specifically in non-small cell lung cancer (NSCLC), rather than small cell lung cancer. Moreover, TrxR1 expression exhibited associations with survival rate, tumor volume, and histological classification. We developed a novel TrxR1 inhibitor named LW-216 and assessed its antitumor efficacy in NSCLC. Our results revealed that LW-216 is effectively bound with intracellular TrxR1 at sites R371 and G442, facilitating TrxR1 ubiquitination and suppressing TrxR1 expression, while not affecting TrxR2 expression. Treatment of LW-216-induced DNA damage and cell apoptosis in NSCLC cells through the generation of reactive oxygen species (ROS). Importantly, supplementation with N-acetylcysteine (NAC) or ectopic TrxR1 expression reversed LW-216-induced apoptosis. Furthermore, LW-216 displayed potent tumor growth inhibition in NSCLC cell-implanted mice, reducing TrxR1 expression in xenografts. Remarkably, LW-216 exhibited superior antitumor activity compared to Auranofin in vivo. Collectively, our research provides compelling evidence supporting the potential of targeting TrxR1 by LW-216 as a promising therapeutic strategy for NSCLC.
    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
    癌症是一种致命的疾病,全世界成千上万的人死亡。尽管癌症治疗研究产生了信息,在癌症治疗中的应用是有限的。因此,科学家们继续努力开发更有效的治疗方法。在研究中,我们旨在确定氨基噻唑化合物对人胶质母细胞瘤(U-87MG)和人皮肤成纤维细胞(HDFa)细胞的抗癌潜力,以及它们对引起癌细胞多药耐药的酶的抑制作用.进行了3-(4,5-二甲基噻唑-2-基)-2,5-二苯基-2H-四唑溴化物细胞活力测试,以了解噻唑衍生物的细胞毒性。使用流式细胞术分析研究了噻唑应用背后的细胞死亡机制。根据细胞活力分析,在细胞毒性方面,噻唑衍生物对U-87MG的影响大于HDFa细胞系。流式细胞术显示,U-87MG细胞的凋亡细胞死亡高于HDFa细胞系。可以得出结论,噻唑化合物对U-87MG和HDFa具有抗癌作用,并具有凋亡特性。它们对硫氧还蛋白还原酶1(TrxR1)的影响,谷胱甘肽S-转移酶(GST),和谷胱甘肽还原酶(GR)活性,这对化疗方法的发展很重要,也被检查过。从获得的结果来看,确定2-氨基-4-(对甲苯基)噻唑(T7)化合物显着抑制TrxR1和GST活性,2-氨基-6-甲基苯并噻唑(T8)化合物可显着抑制TrxR1和GST活性。化合物T7被确定为TrxR1和GST靶向的选择性抑制剂,和化合物T8被确定为靶向TrxR1和GR的成胶质细胞瘤治疗的选择性抑制剂。
    Cancer is a fatal disease that kills thousands of people worldwide. Despite the information produced by research on cancer treatment, applications in cancer treatment are limited. Therefore, scientists\' efforts to develop more effective treatment approaches continue. In the study, we aimed to determine the anticancer potential of amino thiazole compounds on human glioblastoma (U-87 MG) and human dermal fibroblast (HDFa) cells and their inhibition effects on enzymes that cause multidrug resistance in cancer cells. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide cell viability test was performed to understand the cytotoxic properties of thiazole derivatives. The cellular death mechanisms behind thiazole application were investigated using flow cytometry analysis. According to cell viability analysis, thiazole derivatives exhibited a greater effect on U-87 MG than the HDFa cell line in terms of cytotoxicity. Flow cytometry showed higher apoptotic cell death in U-87 MG cells than in the HDFa cell line. It can be concluded that thiazole compounds exert anticancer effects on U-87 MG and HDFa as well as show apoptotic properties. Their effects on thioredoxin reductase 1 (TrxR1), glutathione S-transferase (GST), and glutathione reductase (GR) activities, which are important in the development of chemotherapeutic methods, were also examined. From the results obtained, it was determined that the 2-amino-4-(p-tolyl)thiazole (T7) compound significantly suppressed both TrxR1 and GST activities, and the 2-amino-6-methylbenzothiazole (T8) compound significantly suppressed both TrxR1 and GST activities. Compound T7 was determined to be a selective inhibitor for TrxR1 and GST targeting, and compound T8 was determined to be a selective inhibitor for TrxR1 and GR targeting glioblastoma treatment.
    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)面临抗PD-1免疫疗法的低反应率,强调需要加强治疗策略。Auranofin,通过其金基成分抑制硫氧还蛋白还原酶(TrxR),在癌症治疗中显示出潜力。它的目标是TrxR系统,对于保护细胞免受氧化应激至关重要。TrxR在癌细胞中的过量产生支持它们的增殖。然而,auranofin对该系统的干扰会破坏细胞的氧化还原平衡,提高活性氧的水平,并引发癌细胞死亡。这项研究首次强调TXNRD1是导致HNSCC抗PD-1治疗耐药的关键因素。在这项研究中,我们确定了HNSCC中免疫疗法诱导的铁细胞凋亡耐药的靶向调节因子.我们观察到HNSCC中硫氧还蛋白还原酶1(TXNRD1)与肿瘤PD-L1表达和铁凋亡抑制的联系。此外,具有异常TXNRD1表达的HNSCC肿瘤表现出缺乏PD-1反应,NRF2过表达,和PD-L1上调。TXNRD1抑制促进具有NRF2激活的HNSCC细胞和缺乏PD-1反应的患者来源的类器官肿瘤的铁凋亡。机械上,TXNRD1通过与核糖核苷酸还原酶调节亚基M2(RRM2)结合来调节PD-L1转录并维持氧化还原平衡。TXNRD1表达破坏使HNSCC细胞对抗PD-1介导的JurkatT细胞活化敏感,通过铁性凋亡促进肿瘤杀伤。此外,通过auranofin共治疗抑制TXNRD1与抗PD-1治疗协同作用,通过介导CD8+T细胞浸润和下调PD-L1表达来增强免疫治疗介导的铁细胞凋亡。我们的发现表明,靶向TXNRD1是改善HNSCC患者免疫治疗结果的有希望的治疗策略。
    Head and neck squamous cell carcinoma (HNSCC) faces low response rates to anti-PD-1 immunotherapies, highlighting the need for enhanced treatment strategies. Auranofin, which inhibits thioredoxin reductase (TrxR) through its gold-based composition, has shown potential in cancer treatment. It targets the TrxR system, essential for safeguarding cells from oxidative stress. The overproduction of TrxR in cancerous cells supports their proliferation. However, auranofin\'s interference with this system can upset the cellular redox equilibrium, boost levels of reactive oxygen species, and trigger the death of cancer cells. This study is the first to highlight TXNRD1 as a crucial factor contributing to resistance to anti-PD-1 treatment in HNSCC. In this study, we identified targetable regulators of resistance to immunotherapy-induced ferroptosis in HNSCC. We observed a link of thioredoxin reductase 1 (TXNRD1) with tumoral PD-L1 expression and ferroptosis suppression in HNSCC. Moreover, HNSCC tumors with aberrant TXNRD1 expression exhibited a lack of PD-1 response, NRF2 overexpression, and PD-L1 upregulation. TXNRD1 inhibition promoted ferroptosis in HNSCC cells with NRF2 activation and in organoid tumors derived from patients lacking a PD-1 response. Mechanistically, TXNRD1 regulated PD-L1 transcription and maintained the redox balance by binding to ribonucleotide reductase regulatory subunit M2 (RRM2). TXNRD1 expression disruption sensitized HNSCC cells to anti-PD-1-mediated Jurkat T-cell activation, promoting tumor killing through ferroptosis. Moreover, TXNRD1 inhibition through auranofin cotreatment synergized with anti-PD-1 therapy to potentiate immunotherapy-mediated ferroptosis by mediating CD8+ T-cell infiltration and downregulating PD-L1 expression. Our findings indicate that targeting TXNRD1 is a promising therapeutic strategy for improving immunotherapy outcomes in patients with HNSCC.
    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
    谷胱甘肽过氧化物酶4(GPX4)是一个很有前途的抗癌治疗靶点;然而,GPX4抑制剂(GPX4i)的应用由于固有或获得性耐药性而受到限制。因此,了解潜在的耐药机制和发现可以克服耐药的分子是至关重要的。在这里,我们证明GPX4i通过诱导脂质活性氧介导的铁凋亡和凋亡来杀死膀胱癌细胞,顺铂耐药的膀胱癌细胞也对GPX4i耐药,代表比亲本膀胱癌细胞更高的半最大抑制浓度值。此外,硫氧还蛋白还原酶1(TrxR1)过表达是导致顺铂耐药膀胱癌细胞中GPX4i耐药的原因,和抑制TrxR1恢复了这些细胞对GPX4i的敏感性。体外和体内研究表明,JolkinolideB(JB),一种天然的二萜,以前被鉴定为TrxR1抑制剂,增强了GPX4i(RSL3和ML162)对顺铂耐药的膀胱癌细胞的抗增殖功效。此外,GPX4的敲除和抑制可以增加JB诱导的凋亡和凋亡。我们的结果表明,抑制TrxR1可以有效改善基于GPX4抑制的抗癌治疗。JB和GPX4i的组合,它具有良好的耐受性,并具有多种抗癌机制,可能是治疗膀胱癌的一种有希望的疗法。
    Glutathione peroxidase 4 (GPX4) is a promising anticancer therapeutic target; however, the application of GPX4 inhibitors (GPX4i) is limited owing to intrinsic or acquired drug resistance. Hence, understanding the mechanisms underlying drug resistance and discovering molecules that can overcome drug resistance are crucial. Herein, we demonstrated that GPX4i killed bladder cancer cells by inducing lipid reactive oxygen species-mediated ferroptosis and apoptosis, and cisplatin-resistant bladder cancer cells were also resistant to GPX4i, representing a higher half-maximal inhibitory concentration value than that of parent bladder cancer cells. In addition, thioredoxin reductase 1 (TrxR1) overexpression was responsible for GPX4i resistance in cisplatin-resistant bladder cancer cells, and inhibiting TrxR1 restored the sensitivity of these cells to GPX4i. In vitro and in vivo studies revealed that Jolkinolide B (JB), a natural diterpenoid and previously identified as a TrxR1 inhibitor, potentiated the antiproliferative efficacy of GPX4i (RSL3 and ML162) against cisplatin-resistant bladder cancer cells. Furthermore, GPX4 knockdown and inhibition could augment JB-induced paraptosis and apoptosis. Our results suggest that inhibiting TrxR1 can effectively improve GPX4 inhibition-based anticancer therapy. A combination of JB and GPX4i, which is well-tolerated and has several anticancer mechanisms, may serve as a promising therapy for treating bladder 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
    硫氧还蛋白还原酶1(TrxR1)已成为癌症治疗的有希望的靶标。在我们之前的研究中,我们发现了几种新的TrxR1抑制剂,并发现它们都具有优异的抗肿瘤活性。同时,我们发现这些TrxR1抑制剂都导致癌细胞中AKT磷酸化的增加,但AKT磷酸化在TrxR1抑制剂介导的细胞死亡中的具体作用尚不清楚.在这项研究中,我们确定AKT和TrxR1抑制剂的组合在结肠癌细胞中显示出很强的协同作用.此外,我们证明了金诺芬(TrxR1抑制剂)和MK-2206(AKT抑制剂)的协同作用是由ROS积累引起的。重要的是,我们发现ATM抑制剂KU-55933可以阻断auranofin引起的AKT磷酸化增加,并与金诺芬表现出协同作用。一起来看,我们的研究表明,ATM/AKT通路的激活是应对TrxR1抑制剂诱导的ROS积累的代偿机制,TrxR1和ATM/AKT通路的协同靶向是治疗结肠癌的有前景的策略。
    Thioredoxin reductase 1 (TrxR1) has emerged as a promising target for cancer therapy. In our previous research, we discovered several new TrxR1 inhibitors and found that they all have excellent anti-tumor activity. At the same time, we found these TrxR1 inhibitors all lead to an increase in AKT phosphorylation in cancer cells, but the detailed role of AKT phosphorylation in TrxR1 inhibitor-mediated cell death remains unclear. In this study, we identified the combination of AKT and TrxR1 inhibitor displayed a strong synergistic effect in colon cancer cells. Furthermore, we demonstrated that the synergistic effect of auranofin (TrxR1 inhibitor) and MK-2206 (AKT inhibitor) was caused by ROS accumulation. Importantly, we found that ATM inhibitor KU-55933 can block the increase of AKT phosphorylation caused by auranofin, and exhibited a synergistic effect with auranofin. Taken together, our study demonstrated that the activation of ATM/AKT pathway is a compensatory mechanism to cope with ROS accumulation induced by TrxR1 inhibitor, and synergistic targeting of TrxR1 and ATM/AKT pathway is a promising strategy for treating colon 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
    背景:索拉非尼(Sora),多靶点酪氨酸激酶抑制剂,被广泛认为是晚期肝细胞癌(HCC)的标准化学疗法。然而,耐药机制阻碍了其抗癌功效。来自Withaniasomnifera,WithaferinA(WA)作为天然生物活性化合物具有显着的抗肿瘤特性。这项研究旨在研究Sora和WA共同治疗对HCC影响的机制。
    方法:通过集落形成和MTT测定评价细胞增殖。流式细胞术用于确定细胞凋亡和活性氧(ROS)水平。细胞凋亡相关蛋白水平的评价,DNA损伤,利用IHC染色和蛋白质印迹法进行内质网应激。此外,caspase抑制剂Z-VAD-FMK,ATF4siRNA,ROS清除剂N-乙酰半胱氨酸(NAC),和TrxR1shRNA用于阐明潜在的信号通路。为了验证Sora/WA联合治疗的抗肿瘤作用,最终使用Huh7异种移植物进行体内实验。
    结果:Sora/WA共治疗在体内和体外均显示出显著的协同抗肿瘤作用。机械上,通过抑制TrxR1活性来增强WA对Sora的抗肿瘤作用,导致ROS积累。此外,ROS产生诱导DNA损伤和内质网(ER)应激途径的激活,最终引发细胞凋亡。抗氧化剂NAC预处理显著抑制ROS的生成,ER压力,DNA损伤,和Sora/WA共同处理诱导的细胞凋亡。此外,小干扰RNA(siRNA)对ATF4的抑制减弱了Sora/WA共同治疗诱导的细胞凋亡。在体内,Sora/WA共同治疗可显着抑制HCC异种移植模型中的肿瘤生长,并降低肿瘤组织中的TrxR1活性。
    结论:我们的研究表明,WA协同增强了Sora的抗肿瘤作用,为不断发展的HCC治疗方法提供了有希望的意义。
    BACKGROUND: Sorafenib (Sora), a multi-target tyrosine kinase inhibitor, is widely recognized as a standard chemotherapy treatment for advanced hepatocellular carcinoma (HCC). However, drug resistance mechanisms hinder its anticancer efficacy. Derived from Withania somnifera, Withaferin A (WA) exhibits remarkable anti-tumor properties as a natural bioactive compound. This study aimed to examine the mechanisms that underlie the impacts of Sora and WA co-treatment on HCC.
    METHODS: Cell proliferation was evaluated through colony formation and MTT assays. Flow cytometry was employed to determine cellular apoptosis and reactive oxygen species (ROS) levels. The evaluation of apoptosis-related protein levels, DNA damage, and endoplasmic reticulum stress was conducte utilizing IHC staining and western blotting. Moreover, the caspase inhibitor Z-VAD-FMK, ATF4 siRNA, ROS scavenger N-acetyl cysteine (NAC), and TrxR1 shRNA were used to elucidate the underlying signaling pathways. To validate the antitumor effects of Sora/WA co-treatment, in vivo experiments were ultimately executed using Huh7 xenografts.
    RESULTS: Sora/WA co-treatment demonstrated significant synergistic antitumor impacts both in vivo and in vitro. Mechanistically, the enhanced antitumor impact of Sora by WA was achieved through the inhibition of TrxR1 activity, resulting in ROS accumulation. Moreover, ROS generation induced the activation of DNA damage and endoplasmic reticulum (ER) stress pathways, eventually triggering cellular apoptosis. Pre-treatment with the antioxidant NAC significantly inhibited ROS generation, ER stress, DNA damage, and apoptosis induced by Sora/WA co-treatment. Additionally, the inhibition of ATF4 by small interfering RNA (siRNA) attenuated Sora/WA co-treatment-induced apoptosis. In vivo, Sora/WA co-treatment significantly suppressed tumor growth in HCC xenograft models and decreased TrxR1 activity in tumor tissues.
    CONCLUSIONS: Our study suggests that WA synergistically enhances the antitumor effect of Sora, offering promising implications for evolving treatment approaches for HCC.
    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:氧化应激通过诱导骨细胞死亡在许多骨骼疾病的发病机制中起着至关重要的作用。转录因子核因子红系2相关因子2(Nrf2)是通过抗氧化反应元件(ARE)抵抗细胞氧化应激的各种抗氧化基因表达的主要调节因子,可被各种兴奋剂诱导,包括植物化学物质甲基粘菌素(MET)和L-萝卜硫烷(SFN)。本研究旨在建立骨细胞体外模型,探讨MET和SFN对Nrf2/ARE通路的药理作用。
    方法:使用MLO-Y4鼠骨细胞和稳定转导的MLO-Y4-SIN-lenti-ARE报告基因细胞系。MET和SFN用作Nrf2诱导物。MET的细胞毒性,SFN,和过氧化氢(H2O2)使用CytoTox-Glo™测定进行评估。通过单亮氨酸酶测定检查时间和剂量依赖性的ARE诱导。Nrf2靶标记物的mRNA和蛋白表达,如血红素加氧酶1(Ho-1),NADPH醌脱氢酶1(Nqo1),和硫氧还蛋白还原酶1(Txnrd1),通过RT-qPCR检测,西方印迹,免疫荧光染色,分别。成骨标志物,骨桥蛋白,通过免疫荧光染色比较了是否治疗和未治疗的骨钙蛋白。
    结果:实验数据显示,与媒介物处理的对照相比,MET和SFN以时间和剂量依赖性方式诱导ARE活性,并增加了抗氧化剂标记的mRNA和蛋白质表达。用SFN处理的样本中骨桥蛋白和骨钙蛋白的表达明显高于未处理的样本。在H2O2诱导的应激条件下,用SFN处理的细胞死亡数显着低于未处理的细胞死亡数。
    结论:Nrf2诱导物MET和SFN通过Nrf2/ARE途径增加骨细胞中抗氧化基因的mRNA表达。值得注意的是,在H2O2诱导的应激条件下,SFN增加骨细胞相关成骨标志物的蛋白表达并抑制细胞死亡。因此,Nrf2刺激剂可以对骨细胞发挥缓解应力和成骨作用。
    BACKGROUND: Oxidative stress plays a crucial role in the pathogenesis of many skeletal diseases by inducing osteocyte death. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator of various antioxidant gene expressions through antioxidant response element (ARE) against cellular oxidative stress and can be induced by various stimulants, including the phytochemicals methysticin (MET) and L-sulforaphane (SFN). This study aimed to establish an osteocyte in vitro model to investigate the pharmacological effects of MET and SFN on the Nrf2/ARE pathway.
    METHODS: MLO-Y4 murine osteocytes and the stably transduced MLO-Y4-SIN-lenti-ARE reporter gene cell line were used. MET and SFN were used as Nrf2 inducers. The cytotoxicity of MET, SFN, and hydrogen peroxide (H2O2) was evaluated using the CytoTox-Glo™ Assay. Time- and dose-dependent ARE induction was examined by Monoluciferase Assay. The mRNA and protein expressions of Nrf2 target markers, such as heme-oxygenase 1 (Ho-1), NADPH quinone dehydrogenase 1 (Nqo1), and thioredoxin reductase 1 (Txnrd1), were detected by RT-qPCR, Western Blot, and immunofluorescence staining, respectively. Osteogenesis markers, osteopontin, and osteocalcin were compared with and without treatment by immunofluorescence staining.
    RESULTS: The experimental data showed that MET and SFN induced ARE activity in a time- and dose-dependent manner and increased the mRNA and protein expression of antioxidant markers compared to vehicle-treated controls. The protein expression of osteopontin and osteocalcin in the samples treated with SFN were significantly higher than without treatment, and the number of cell death treated with SFN was significantly lower than without treatment under H2O2-induced stress conditions.
    CONCLUSIONS: Nrf2 inducers MET and SFN increased the mRNA expression of antioxidant genes through the Nrf2/ARE pathway in osteocytes. Notably, SFN increased the protein expression of osteocyte-associated osteogenic markers and suppressed cell death under H2O2-induced stress condition. Thus, Nrf2 stimulators can exert stress-relieving and osteogenic effects on osteocytes.
    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
    肝细胞癌(HCC)是全球最常见的恶性肿瘤之一。由于现有药物数量有限及其副作用,肝癌治疗新的化疗策略的开发变得越来越重要。这项研究的目的是调查是否衍射酸(DA),地衣的次生代谢产物之一,对HepG2细胞具有潜在的抗癌作用,以及其抗癌作用是否通过抑制硫氧还蛋白还原酶1(TRXR1)介导,由于在包括HCC在内的肿瘤细胞中过度表达,这是化疗策略的目标。XTT分析结果表明,DA对HepG2细胞表现出强烈的细胞毒性,在48h时IC50值为78.07µg/mL。流式细胞仪分析结果表明,DA对HepG2细胞表现出晚期凋亡和坏死作用。与这些发现一致,实时荧光定量PCR结果显示,DA并未改变HepG2细胞中BAX/BCL2的比例,但上调了P53基因。此外,伤口愈合试验结果表明,DA在HepG2细胞中具有很强的抗迁移作用。实时PCR和Westernblot分析表明,DA增加了TRXR1基因和蛋白质的表达水平,而酶活性研究表明,DA抑制TRXR1。这些发现表明DA通过靶向TRXR1的酶抑制对HepG2细胞具有抗癌作用。总之,作为TRXR1抑制剂的DA可以被认为是有效的化学治疗剂,其可以是用于治疗HCC的有用的先导化合物。
    Hepatocellular carcinoma (HCC) represents one of the most common malignant tumors worldwide. Due to the limited number of available drugs and their side effects, the development of new chemotherapeutic strategies for HCC treatment has become increasingly important. This study is aimed at investigating whether diffractaic acid (DA), one of the secondary metabolites of lichen, exhibits a potential anticancer effect on HepG2 cells and whether its anticancer effect is mediated by inhibition of thioredoxin reductase 1 (TRXR1), which is a target of chemotherapeutic strategies due to overexpression in tumor cells including HCC. XTT assay results showed that DA exhibited strong cytotoxicity on HepG2 cells with an IC50 value of 78.07 µg/mL at 48 h. Flow cytometric analysis results revealed that DA displayed late apoptotic and necrotic effects on HepG2 cells. Consistent with these findings, real-time PCR results showed that DA did not alter the BAX/BCL2 ratio in HepG2 cells but upregulated the P53 gene. Moreover, the wound healing assay results revealed a strong anti-migratory effect of DA in HepG2 cells. Real-time PCR and Western blot analyses demonstrated that DA increased TRXR1 gene and protein expression levels, whereas enzyme activity studies disclosed that DA inhibited TRXR1. These findings suggest that DA has an anticancer effect on HepG2 cells by targeting the enzymatic inhibition of TRXR1. In conclusion, DA as a TRXR1 inhibitor can be considered an effective chemotherapeutic agent which may be a useful lead compound for the treatment of HCC.
    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
    硫氧还蛋白还原酶(TXNRD)是一种硒蛋白,在细胞抗氧化防御中起着至关重要的作用。以前,在TXNRD1中发现了一个独特的导杆基序,它影响电子的转移。在这项研究中,利用单氨基酸取代和激发发射矩阵(EEM)荧光光谱分析,我们发现导杆与FAD连通并调节酶的电子流。差示扫描荧光分析法(DSF)分析表明,导杆中的芳香族氨基酸是TXNRD1的稳定剂。动力学分析显示,导向棒对于二硫化物还原酶活性至关重要,但阻碍了TXNRD1的硒代半胱氨酸依赖性还原活性。同时,引导杆保护TXNRD1的硒代半胱氨酸残基免受亲电试剂的攻击。我们还发现,caveolin-1支架结构域(CSD)肽和化合物LCS3对TXNRD1的抑制作用不与引导条基序结合。总之,所获得的结果突出了指导条的新方面,这些方面限制了C端氧化还原基序的灵活性,并控制了从抗氧化剂到促氧化剂的转变。
    Thioredoxin reductase (TXNRD) is a selenoprotein that plays a crucial role in cellular antioxidant defense. Previously, a distinctive guiding bar motif was identified in TXNRD1, which influences the transfer of electrons. In this study, utilizing single amino acid substitution and Excitation-Emission Matrix (EEM) fluorescence spectrum analysis, we discovered that the guiding bar communicates with the FAD and modulates the electron flow of the enzyme. Differential Scanning Fluorimetry (DSF) analysis demonstrated that the aromatic amino acid in guiding bar is a stabilizer for TXNRD1. Kinetic analysis revealed that the guiding bar is vital for the disulfide reductase activity but hinders the selenocysteine-independent reduction activity of TXNRD1. Meanwhile, the guiding bar shields the selenocysteine residue of TXNRD1 from the attack of electrophilic reagents. We also found that the inhibition of TXNRD1 by caveolin-1 scaffolding domain (CSD) peptides and compound LCS3 did not bind to the guiding bar motif. In summary, the obtained results highlight new aspects of the guiding bar that restrict the flexibility of the C-terminal redox motif and govern the transition from antioxidant to pro-oxidant.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号