auranofin

Auranofin
  • 文章类型: Journal Article
    犬乳腺肿瘤(CMT)是完整雌性犬中最常见的自发性肿瘤,通常会发生转移。Auranofin(AF)是一种用于治疗风湿病的金复合物。在各种类型的人和犬肿瘤中已经证明了AF的优异抗肿瘤能力。在这项研究中,五种CMT细胞系(CIPp,CMT-7364,CHMp,CIPm和CTBp)和三个CMT原代细胞(G7894,L1883和L6783)用于探索AF对CMT的抗肿瘤作用。使用两种CMT细胞系(CIPp和CMT-7364)来搜索AF对CMT的影响的潜在机制。结果表明,AF抑制了生长,迁移,入侵,和CMT细胞的集落形成能力。此外,在3D细胞培养模型中CMT的生长被AF有效抑制。此外,AF经由过程PI3K/AKT通路引诱CMT细胞凋亡。总之,AF通过调节PI3K/AKT通路有效诱导CMT凋亡,这表明房颤应在未来的研究中作为一种潜在的CMT治疗方法加以探讨。
    Canine mammary gland tumour (CMT) is the most common spontaneous tumour in intact female dogs and often exhibits metastases. Auranofin (AF) is a gold complex used for treating rheumatism. The excellent anti-tumour ability of AF has been demonstrated in various types of human and canine tumours. In this study, five CMT cell lines (CIPp, CMT-7364, CHMp, CIPm and CTBp) and three CMT primary cells (G7894, L1883 and L6783) were used to explore the anti-tumour effect of AF on CMT. Two CMT cell lines (CIPp and CMT-7364) were used to search the underlying mechanism of the effect of AF on CMT. The results showed that AF inhibited the growth, migration, invasion, and colony formation abilities of CMT cells. Additionally, the growth of CMT in a 3D cell culture model was effectively suppressed by AF. Furthermore, AF induced cell apoptosis of CMT cells via the PI3K/AKT pathway. In conclusion, AF effectively induces CMT apoptosis by regulating the PI3K/AKT pathway, indicating that AF should be explored as a potential CMT treatment in future studies.
    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
    对蛋白质组的多方面询问加深了对生物系统的全系统理解;然而,迄今为止,蛋白质组的氧化还原变化比表达和溶解度/稳定性分析更具挑战性.这里,设计了第一个与表达分析(REX)整合的高通量氧化还原蛋白质组学方法,并将其与蛋白质组积分溶解度改变(PISA)测定相结合。具有多达四个生物学重复的整个PISA-REX实验可以多路复用成单个串联质量标签TMTpro组。为了对这个紧凑的工具进行基准测试,分析了用金诺芬处理的HCT116细胞,与以前的研究相比有很大的改进。然后将PISA-REX用于研究干扰素α(IFN-α)刺激人单核细胞后的蛋白质组重塑。应用该工具研究从用干扰素α处理的野生型与Ncf1突变小鼠中分离的浆细胞样树突状细胞(pDC)的蛋白质组变化,显示NCF1缺乏增强STAT1途径并调节表达,溶解度,和干扰素诱导蛋白的氧化还原状态。提供关于蛋白质组的全面多方面的信息,紧凑型PISA-REX有可能成为蛋白质组学的行业标准,并为健康和疾病生物学打开新的窗口。
    Multifaceted interrogation of the proteome deepens the system-wide understanding of biological systems; however, mapping the redox changes in the proteome has so far been significantly more challenging than expression and solubility/stability analyses. Here, the first high-throughput redox proteomics approach integrated with expression analysis (REX) is devised and combined with the Proteome Integral Solubility Alteration (PISA) assay. The whole PISA-REX experiment with up to four biological replicates can be multiplexed into a single tandem mass tag TMTpro set. For benchmarking this compact tool, HCT116 cells treated with auranofin are analyzed, showing great improvement compared with previous studies. PISA-REX is then applied to study proteome remodeling upon stimulation of human monocytes by interferon α (IFN-α). Applying this tool to study the proteome changes in plasmacytoid dendritic cells (pDCs) isolated from wild-type versus Ncf1-mutant mice treated with interferon α, shows that NCF1 deficiency enhances the STAT1 pathway and modulates the expression, solubility, and redox state of interferon-induced proteins. Providing comprehensive multifaceted information on the proteome, the compact PISA-REX has the potential to become an industry standard in proteomics and to open new windows into the biology of health and disease.
    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
    急性髓性白血病(AML)是一种异质性疾病,其特征是复杂的分子和细胞遗传学异常。促氧化剂细胞氧化还原状态是AML细胞的常见标志,为基于氧化还原的抗癌策略提供了理论基础。我们之前发现了金诺芬(AUF),最初用于治疗类风湿性关节炎,并重新定位其抗癌活性,可以与药理学浓度的维生素C(VC)协同对抗乳腺癌细胞系模型。在这项研究中,我们观察到,这种药物组合协同有效地杀死了由不同骨髓亚型建立的白血病细胞系的细胞。除了诱导活性氧和ATP消耗的升高,4E-BP1和p70S6K的快速去磷酸化,对AUF/VC治疗的早期事件以及对蛋白质合成的强烈抑制,表明它们在AUF/VC诱导的细胞毒性中的意义。重要的是,一项对来自不同AML亚型的22个原发性AML样本的研究表明,在药理学上可达到的浓度下,AUF/VC组合可有效根除大多数样本中的原发性白血病CD34+细胞,同时对正常脐带血CD34+细胞毒性较小。我们的发现表明,针对AML的氧化还原脆弱性与AUF/VC组合可以提出一个潜在的抗AML治疗方法。
    Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by complex molecular and cytogenetic abnormalities. Pro-oxidant cellular redox status is a common hallmark of AML cells, providing a rationale for redox-based anticancer strategy. We previously discovered that auranofin (AUF), initially used for the treatment of rheumatoid arthritis and repositioned for its anticancer activity, can synergize with a pharmacological concentration of vitamin C (VC) against breast cancer cell line models. In this study, we observed that this drug combination synergistically and efficiently killed cells of leukaemic cell lines established from different myeloid subtypes. In addition to an induced elevation of reactive oxygen species and ATP depletion, a rapid dephosphorylation of 4E-BP1 and p70S6K, together with a strong inhibition of protein synthesis were early events in response to AUF/VC treatment, suggesting their implication in AUF/VC-induced cytotoxicity. Importantly, a study on 22 primary AML specimens from various AML subtypes showed that AUF/VC combinations at pharmacologically achievable concentrations were effective to eradicate primary leukaemic CD34+ cells from the majority of these samples, while being less toxic to normal cord blood CD34+ cells. Our findings indicate that targeting the redox vulnerability of AML with AUF/VC combinations could present a potential anti-AML therapeutic approach.
    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
    Auranofin(AF)是一种金基化合物,具有众所周知的药理和毒理学特征,目前用于治疗一些严重的类风湿性关节炎。在过去的二十年里,AF也被重新用于抗病毒,抗肿瘤,和抗菌药物。在这篇综述中,我们专注于AF的抗菌性能,专门研究迄今为止文献报道的单胚层和双胚层细菌中AF的最小抑制浓度(MIC)。AF被证明对单胚层细菌非常有效,虽然双皮肤不太容易受到影响,可能是由于外膜屏障。我们还报道了当前有关AF抗菌性能的机理假设,尽管目前还没有关于其抗菌作用方式的确切描述。即使其作用机制尚未完全阐明,需要进一步研究以优化其交付策略,AF因其独特的作用方式和对多种病原体的高效,值得进一步研究。这可能导致在对抗抗菌素耐药性和改善传染病治疗结果方面的潜在应用。
    Auranofin (AF) is a gold-based compound with a well-known pharmacological and toxicological profile, currently used in the treatment of some severe forms of rheumatoid arthritis. Over the last twenty years, AF has also been repurposed as antiviral, antitumor, and antibacterial drug. In this review we focused on the antibacterial properties of AF, specifically researching the minimal inhibitory concentrations (MIC) of AF in both mono- and diderm bacteria reported so far in literature. AF proves to be highly effective against monoderm bacteria, while diderm are far less susceptible, probably due to the outer membrane barrier. We also reported the current mechanistic hypotheses concerning the antimicrobial properties of AF, although a conclusive description of its antibacterial mode of action is not yet available. Even if its mechanism of action has not been fully elucidated yet and further studies are required to optimize its delivery strategy, AF deserves additional investigation because of its unique mode of action and high efficacy against a wide range of pathogens, which could lead to potential applications in fighting antimicrobial resistance and improving therapeutic outcomes in infectious diseases.
    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
    脓毒症是严重影响危重患者生存的常见感染并发症。目前,缺乏有效的药物治疗策略。Auranofin,被称为硫氧还蛋白还原酶(TrxR)的抑制剂,表现出抗炎活性,但其在脓毒症中的作用尚不清楚。这里,在盲肠结扎穿孔(CLP)小鼠模型中,我们证明了Auranofin对脓毒症的显著抑制作用.体外,Auranofin抑制由Caspase-11激活触发的焦亡。进一步的研究表明,抑制TrxR1可以抑制大肠杆菌诱导的巨噬细胞凋亡,而TrxR2不表现出这种效果。TrxR1,作为还原酶,调节硫氧还蛋白-1(Trx-1)的氧化还原状态。机械上,TrxR1对Trx-1还原活性的调节可能与Caspase-11活化诱导的细胞凋亡有关。此外,抑制TrxR1维持Trx-1在其氧化状态。Trx-1的氧化形式与Caveolin-1(CAV1)相互作用,调节外膜囊泡(OMV)内化。总之,我们的研究表明,抑制TrxR1通过维持Trx-1的氧化形式抑制OMV内化,从而限制Caspase-11的激活并减轻脓毒症.
    Sepsis is a common complication of infections that significantly impacts the survival of critically patients. Currently, effective pharmacological treatment strategies are lacking. Auranofin, known as an inhibitor of Thioredoxin reductase (TrxR), exhibits anti-inflammatory activity, but its role in sepsis is not well understood. Here, we demonstrate the significant inhibitory effect of Auranofin on sepsis in a cecal ligation and puncture (CLP) mouse model. In vitro, Auranofin inhibits pyroptosis triggered by Caspase-11 activation. Further investigations reveal that inhibiting TrxR1 suppresses macrophage pyroptosis induced by E. coli, while TrxR2 does not exhibit this effect. TrxR1, functioning as a reductase, regulates the oxidative-reductive status of Thioredoxin-1 (Trx-1). Mechanistically, the modulation of Trx-1\'s reductive activity by TrxR1 may be involved in Caspase-11 activation-induced pyroptosis. Additionally, inhibiting TrxR1 maintains Trx-1 in its oxidized state. The oxidized form of Trx-1 interacts with Caveolin-1 (CAV1), regulating outer membrane vesicle (OMV) internalization. In summary, our study suggests that inhibiting TrxR1 suppresses OMV internalization by maintaining the oxidized form of Trx-1, thereby restricting Caspase-11 activation and alleviating sepsis.
    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(TrxR)在过氧化物酶介导的氢过氧化物代谢过程中具有回收硫氧还蛋白(Trx)的功能,目前正在使用FDA批准的抗风湿药物作为目标。极诺芬(AF),选择性地使癌细胞对治疗敏感。AF治疗降低了小细胞肺癌(SCLC)细胞系(DMS273和DMS53)以及H727非典型肺类癌细胞系中的TrxR活性和克隆性存活率。AF处置还显著致敏DMS273和H727细胞系的体外索拉非尼,一种FDA批准的多激酶抑制剂,可耗尽细胞内谷胱甘肽(GSH)。药代动力学,药效学,在DMS273异种移植物的裸鼠中,以2mg/kg或4mg/kg(IP)腹膜内给药AF一次(QD)或每天两次(BID),持续1-5天,检查了AF的安全性。AF的血浆水平为10-20μM(通过金的质谱测定),并且在每天一次4mg/kg时获得最佳的TrxR活性抑制,对谷胱甘肽过氧化物酶1活性无影响。这种房颤治疗延长了14天,抑制TrxR(>75%),并导致中位总生存期从19天显著延长至23天(p=0.04,N=30个对照,28AF).在这个实验中,没有观察到动物体重的变化,全血细胞计数(CBC),骨髓毒性,血尿素氮,或肌酐。这些结果支持以下假设:AF在SCLC中在体外和体内均有效抑制TrxR,使NETs和SCLC对索拉非尼敏感,并且可以重新用作诱导硫醇代谢破坏的靶向药物的辅助治疗。
    Thioredoxin Reductase (TrxR) functions to recycle thioredoxin (Trx) during hydroperoxide metabolism mediated by peroxiredoxins and is currently being targeted using the FDA-approved anti-rheumatic drug, auranofin (AF), to selectively sensitize cancer cells to therapy. AF treatment decreased TrxR activity and clonogenic survival in small cell lung cancer (SCLC) cell lines (DMS273 and DMS53) as well as the H727 atypical lung carcinoid cell line. AF treatment also significantly sensitized DMS273 and H727 cell lines in vitro to sorafenib, an FDA-approved multi-kinase inhibitor that depleted intracellular glutathione (GSH). The pharmacokinetic, pharmacodynamic, and safety profile of AF was examined in nude mice with DMS273 xenografts administered AF intraperitoneally at 2 mg/kg or 4 mg/kg (IP) once (QD) or twice daily (BID) for 1-5 d. Plasma levels of AF were 10-20 μM (determined by mass spectrometry of gold), and the optimal inhibition of TrxR activity was obtained at 4 mg/kg once daily, with no effect on glutathione peroxidase 1 activity. This AF treatment extended for 14 d, inhibited TrxR (>75%), and resulted in a significant prolongation of median overall survival from 19 to 23 d (p = .04, N = 30 controls, 28 AF). In this experiment, there were no observed changes in animal bodyweight, complete blood counts (CBCs), bone marrow toxicity, blood urea nitrogen, or creatinine. These results support the hypothesis that AF effectively inhibits TrxR both in vitro and in vivo in SCLC, sensitizes NETs and SCLC to sorafenib, and could be repurposed as an adjuvant therapy with targeted agents that induce disruptions in thiol 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
    胶质母细胞瘤(GBM)是成人中最常见和晚期的恶性原发性脑肿瘤。GBM经常携带表皮生长因子受体(EGFR)野生型(EGFRwt)基因扩增和/或EGFRvIII激活突变。EGFR驱动的GBM依赖于硫氧还蛋白(Trx)和/或谷胱甘肽(GSH)抗氧化剂系统来抵抗活性氧(ROS)的过量产生。EGFRwt或EGFRvIII过表达对对Trx/GSH共靶向策略的反应的影响是未知的。在这项研究中,我们在GBM中EGFR过度表达的背景下研究了Trx/GSH共靶向。Auranofin是一种硫氧还蛋白还原酶(TrxR)抑制剂,FDA批准用于类风湿性关节炎。L-丁硫氨酸-磺基肟(L-BSO)通过靶向谷氨酸-半胱氨酸连接酶催化(GCLC)酶亚基来抑制GSH合成。我们分析了金诺芬的细胞毒性机制以及金诺芬与U87MG中L-BSO的相互作用,U87/EGFRwt,和U87/EGFRvIIIGBM等基因GBM细胞系。使用抗氧化剂N-乙酰半胱氨酸评估ROS依赖性作用。我们表明,金诺芬降低了TrxR1活性,增加了ROS。Auranofin通过ROS依赖性机制降低细胞活力和集落形成并增加蛋白质聚泛素化,提示ROS在金诺芬诱导的三种细胞系中的细胞毒性中的作用。ROS依赖性PARP-1裂解与U87/EGFRvIII细胞中EGFRvIII下调相关。值得注意的是,金诺芬和L-BSO联合诱导了细胞内GSH的显着消耗和协同细胞毒性,而与EGFR的过度表达无关。然而,与细胞毒性相关的分子机制在三种细胞系中受到不同程度的调节。U87/EGFRvIII表现出最显著的ROS增加,P-AKT(Ser-473),AKT随着EGFRvIII的大幅下调而下降。与U87MG细胞相比,U87/EGFRwt和U87/EGFRvIII显示较低的基础细胞内GSH水平和协同的ROS依赖性DNA损伤。我们的研究提供了体外GBM中金诺芬和L-BSO组合的ROS依赖性协同细胞毒性的证据。揭示EGFR过表达细胞对单独的金诺芬的敏感性,和协同金诺芬和L-BSO组合,支持在GBM中重新应用这种有前途的促氧化剂治疗策略的理由。
    Glioblastoma (GBM) is the most prevalent and advanced malignant primary brain tumor in adults. GBM frequently harbors epidermal growth factor receptor (EGFR) wild-type (EGFRwt) gene amplification and/or EGFRvIII activating mutation. EGFR-driven GBM relies on the thioredoxin (Trx) and/or glutathione (GSH) antioxidant systems to withstand the excessive production of reactive oxygen species (ROS). The impact of EGFRwt or EGFRvIII overexpression on the response to a Trx/GSH co-targeting strategy is unknown. In this study, we investigated Trx/GSH co-targeting in the context of EGFR overexpression in GBM. Auranofin is a thioredoxin reductase (TrxR) inhibitor, FDA-approved for rheumatoid arthritis. L-buthionine-sulfoximine (L-BSO) inhibits GSH synthesis by targeting the glutamate-cysteine ligase catalytic (GCLC) enzyme subunit. We analyzed the mechanisms of cytotoxicity of auranofin and the interaction between auranofin and L-BSO in U87MG, U87/EGFRwt, and U87/EGFRvIII GBM isogenic GBM cell lines. ROS-dependent effects were assessed using the antioxidant N-acetylsteine. We show that auranofin decreased TrxR1 activity and increased ROS. Auranofin decreased cell vitality and colony formation and increased protein polyubiquitination through ROS-dependent mechanisms, suggesting the role of ROS in auranofin-induced cytotoxicity in the three cell lines. ROS-dependent PARP-1 cleavage was associated with EGFRvIII downregulation in U87/EGFRvIII cells. Remarkably, the auranofin and L-BSO combination induced the significant depletion of intracellular GSH and synergistic cytotoxicity regardless of EGFR overexpression. Nevertheless, molecular mechanisms associated with cytotoxicity were modulated to a different extent among the three cell lines. U87/EGFRvIII exhibited the most prominent ROS increase, P-AKT(Ser-473), and AKT decrease along with drastic EGFRvIII downregulation. U87/EGFRwt and U87/EGFRvIII displayed lower basal intracellular GSH levels and synergistic ROS-dependent DNA damage compared to U87MG cells. Our study provides evidence for ROS-dependent synergistic cytotoxicity of auranofin and L-BSO combination in GBM in vitro. Unraveling the sensitivity of EGFR-overexpressing cells to auranofin alone, and synergistic auranofin and L-BSO combination, supports the rationale to repurpose this promising pro-oxidant treatment strategy in GBM.
    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
    基于质谱的组学技术越来越多地用于扰动研究,以通过识别重要的分子事件将药物作用映射到生物途径。显著性受每个分子参数的倍数变化和变化的影响,而且还通过多次测试校正。虽然折叠变化很大程度上是由生物系统决定的,变化是由实验工作流程决定的。这里,结果表明,使用两种结肠癌细胞系SW480和HCT116,先前传代培养的记忆效应可以影响扰动谱的变化。这些记忆效应很大程度上是由扰动实验中持续存在的生长状态差异驱动的。在SW480单元格中,记忆效应与中度治疗效应相结合,放大了多个组学水平的变异,包括eicosadomics,蛋白质组学,和磷酸蛋白质组学。具有更强的治疗效果,记忆效应不太明显,如在HCT116细胞中所证明的。通过实时监测细胞生长来控制传代培养的同质性。基于组合的蛋白质组和磷酸蛋白质组数据,受控的均质继代培养产生了321个因果猜想的扰动网络,与仅58个因果猜想相比,而没有控制SW480细胞中的继代同质性。确定了一些细胞反应和调节事件,这些反应和调节事件仅在考虑这些记忆效应时扩展了三氧化二砷(ATO)的作用方式。先前受控的继代培养导致发现ATO与硫氧还蛋白还原酶1抑制剂金诺芬的协同组合治疗,这可能证明对NRF2介导的抗性机制的管理有用。
    Mass spectrometry-based omics technologies are increasingly used in perturbation studies to map drug effects to biological pathways by identifying significant molecular events. Significance is influenced by fold change and variation of each molecular parameter, but also by multiple testing corrections. While the fold change is largely determined by the biological system, the variation is determined by experimental workflows. Here, it is shown that memory effects of prior subculture can influence the variation of perturbation profiles using the two colon carcinoma cell lines SW480 and HCT116. These memory effects are largely driven by differences in growth states that persist into the perturbation experiment. In SW480 cells, memory effects combined with moderate treatment effects amplify the variation in multiple omics levels, including eicosadomics, proteomics, and phosphoproteomics. With stronger treatment effects, the memory effect was less pronounced, as demonstrated in HCT116 cells. Subculture homogeneity was controlled by real-time monitoring of cell growth. Controlled homogeneous subculture resulted in a perturbation network of 321 causal conjectures based on combined proteomic and phosphoproteomic data, compared to only 58 causal conjectures without controlling subculture homogeneity in SW480 cells. Some cellular responses and regulatory events were identified that extend the mode of action of arsenic trioxide (ATO) only when accounting for these memory effects. Controlled prior subculture led to the finding of a synergistic combination treatment of ATO with the thioredoxin reductase 1 inhibitor auranofin, which may prove useful in the management of NRF2-mediated resistance 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
    阿尔茨海默病仍然是一个尚未解决的神经难题,无法治愈。目前的疗法只能缓解症状,通过血脑屏障的有限摄取阻碍。Auranofin,FDA批准的化合物,具有针对脑部疾病的有效抗氧化和抗炎特性。然而,其口服生物利用度的挑战促使探索基于纳米制剂的解决方案,提高血脑屏障的穿透性。本研究旨在探讨金诺芬纳米颗粒对链脲佐菌素诱导的AD大鼠的神经保护作用。通过多重乳液溶剂蒸发法配制了含金诺芬的聚乳酸-共-乙醇酸纳米颗粒。通过确定包封效率来进行表征,粒度分布,表面电荷,和形态学。通过施用链脲佐菌素(3mg/kg/i.c.v.,第1天和第3天),金诺芬(5和10毫克/千克),金诺芬纳米颗粒(2.5和5毫克/千克),和多奈哌齐(2mg/kg)口服14天。行为缺陷使用野外试验进行评估,莫里斯水迷宫,客观识别测试,氧化应激水平的变化,和大脑中的AD标记。老鼠被斩首后,切除大脑以分离海马体。随后的分析包括生化和神经炎症标志物的定量,以及神经递质水平的评估。金诺芬纳米粒子的表征显示了98%的包封率,平均粒径为101.5±10.3nm,表面电荷为27.5±5.10mV,多分散指数为0.438±0.12。在体内,服用金诺芬和金诺芬纳米颗粒可显着逆转链脲佐菌素诱导的认知缺陷,生化改变,神经炎症标记物,和神经递质水平。目前的发现表明,金诺芬纳米颗粒比单独的金诺芬具有更显著的神经保护潜力。治疗功效可能归因于其抗氧化和抗炎特性,以及其积极的神经调节作用。因此,我们的研究结果表明,它可能是阿尔茨海默病治疗的一个有希望的候选人。
    Alzheimer\'s disease remains an unsolved neurological puzzle with no cure. Current therapies offer only symptomatic relief, hindered by limited uptake through the blood-brain barrier. Auranofin, an FDA-approved compound, exhibits potent antioxidative and anti-inflammatory properties targeting brain disorders. Yet, its oral bioavailability challenge prompts the exploration of nanoformulation-based solutions enhancing blood-brain barrier penetrability. The study aimed to investigate the neuroprotective potential of auranofin nanoparticles in streptozotocin-induced AD rats. Auranofin-containing polylactic-co-glycolic acid nanoparticles were formulated by the multiple emulsion solvent evaporation method. Characterization was done by determining entrapment efficiency, particle size distribution, surface charge, and morphology. An in vivo study was performed by administering streptozotocin (3 mg/kg/i.c.v., days 1 and 3), auranofin (5 and 10 mg/kg), auranofin nanoparticles (2.5 and 5 mg/kg), and donepezil (2 mg/kg) for 14 days orally. Behavioral deficits were evaluated using the open field test, Morris water maze, objective recognition test, change in oxidative stress levels, and AD markers in the brain. Following the decapitation of the rats, the brains were excised to isolate the hippocampus. Subsequent analyses included the quantification of biochemical and neuroinflammatory markers, as well as the assessment of neurotransmitter levels. The characterization of auranofin nanoparticles showed an entrapment efficiency of 98%, an average particle size of 101.5 ± 10.3 nm, a surface charge of 27.5 ± 5.10 mV, and a polydispersity index of 0.438 ± 0.12. In vivo, administration of auranofin and auranofin nanoparticles significantly reversed streptozotocin-induced cognitive deficits, biochemical alteration, neuroinflammatory markers, and neurotransmitter levels. The present finding suggests that auranofin nanoparticles have more significant neuroprotective potential than auranofin alone. The therapeutic efficacy may be attributed to its antioxidant and anti-inflammatory properties, as well as its positive neuromodulatory effects. Therefore, our findings suggest that it could be a promising candidate for Alzheimer\'s disease therapy.
    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
    需要新的治疗方法来治疗尤因肉瘤肿瘤。我们先前确定尤文肉瘤细胞系对抑制蛋白质翻译的药物敏感。然而,在肿瘤中抑制蛋白质合成的翻译和治疗方法是有限的。在这项工作中,我们发现了活性氧,由多种化疗和其他药物产生,抑制蛋白质合成并降低支持尤文肉瘤细胞肿瘤发生的关键蛋白质的水平。特别是,我们发现过氧化氢和金诺芬,硫氧还蛋白还原酶的抑制剂和氧化应激和活性氧的调节剂,激活蛋白质翻译4E-BP1的阻遏物,并降低尤因和其他肉瘤细胞系中致癌蛋白RRM2和PLK1的水平。这些结果提供了对ROS诱导药物如何通过抑制蛋白质翻译靶向癌细胞的机制的新见解,并确定了ROS与DNA复制(RRM2)和细胞周期调节(PLK1)途径之间的机制联系。
    Novel therapeutic approaches are needed for the treatment of Ewing sarcoma tumors. We previously identified that Ewing sarcoma cell lines are sensitive to drugs that inhibit protein translation. However, translational and therapeutic approaches to inhibit protein synthesis in tumors are limited. In this work, we identified that reactive oxygen species, which are generated by a wide range of chemotherapy and other drugs, inhibit protein synthesis and reduce the level of critical proteins that support tumorigenesis in Ewing sarcoma cells. In particular, we identified that both hydrogen peroxide and auranofin, an inhibitor of thioredoxin reductase and regulator of oxidative stress and reactive oxygen species, activate the repressor of protein translation 4E-BP1 and reduce the levels of the oncogenic proteins RRM2 and PLK1 in Ewing and other sarcoma cell lines. These results provide novel insight into the mechanism of how ROS-inducing drugs target cancer cells via inhibition of protein translation and identify a mechanistic link between ROS and the DNA replication (RRM2) and cell cycle regulatory (PLK1) pathways.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号