Pitavastatin

匹伐他汀
  • 文章类型: Journal Article
    背景:尽管有有效的策略,EGFR突变肺癌的耐药性仍是一个挑战.代谢重编程是肿瘤耐药的主要机制之一。一类被称为“他汀类药物”的药物抑制脂质胆固醇代谢,广泛用于心血管疾病患者。先前的研究也证明了其在接受EGFR-TKI治疗的肺癌患者中改善治疗效果的能力。因此,他汀类药物对肺癌靶向耐药的影响仍有待研究。
    方法:长时间接触吉非替尼导致从亲本敏感细胞系(PC9)中出现耐药肺癌细胞系(PC9GR),表现出传统的EGFR突变。CCK-8测定用于评估各种浓度的匹伐他汀对细胞增殖的影响。进行RNA测序以检测差异表达的基因及其相关途径。为了检测蛋白质的表达,进行蛋白质印迹。通过异种移植小鼠模型在体内评估匹伐他汀的抗肿瘤活性。
    结果:低剂量维持诱导PC9吉非替尼耐药菌株。细胞培养和动物相关研究验证了匹伐他汀对肺癌细胞增殖的抑制作用,促进细胞凋亡,抑制了EGFR-TKIs的获得性耐药。KEGG通路分析表明,相对于PC9细胞,在PC9GR细胞中hippo/YAP信号通路被激活,匹伐他汀对YAP的表达有抑制作用。用YAPRNA干扰,pAKT,pBAD和BCL-2表达降低,而BAX表达则增加。因此,YAP下调显著增加吉非替尼耐药肺癌细胞的凋亡并降低其存活率。SC79增加pAKT后,吉非替尼诱导的YAP下调细胞凋亡减少,细胞存活率提高。机械上,匹伐他汀的这些作用与YAP途径有关,从而抑制下游AKT/BAD-BCL-2信号通路。
    结论:本研究为临床应用降脂药物匹伐他汀增强肺癌患者对EGFR-TKI药物的敏感性、缓解耐药提供了分子依据。
    BACKGROUND: Despite effective strategies, resistance in EGFR mutated lung cancer remains a challenge. Metabolic reprogramming is one of the main mechanisms of tumor drug resistance. A class of drugs known as \"statins\" inhibit lipid cholesterol metabolism and are widely used in patients with cardiovascular diseases. Previous studies have also documented its ability to improve the therapeutic impact in lung cancer patients who receive EGFR-TKI therapy. Therefore, the effect of statins on targeted drug resistance to lung cancer remains to be investigated.
    METHODS: Prolonged exposure to gefitinib resulted in the emergence of a resistant lung cancer cell line (PC9GR) from the parental sensitive cell line (PC9), which exhibited a traditional EGFR mutation. The CCK-8 assay was employed to assess the impact of various concentrations of pitavastatin on cellular proliferation. RNA sequencing was conducted to detect differentially expressed genes and their correlated pathways. For the detection of protein expression, Western blot was performed. The antitumor activity of pitavastatin was evaluated in vivo via a xenograft mouse model.
    RESULTS: PC9 gefitinib resistant strains were induced by low-dose maintenance. Cell culture and animal-related studies validated that the application of pitavastatin inhibited the proliferation of lung cancer cells, promoted cell apoptosis, and restrained the acquired resistance to EGFR-TKIs. KEGG pathway analysis showed that the hippo/YAP signaling pathway was activated in PC9GR cells relative to PC9 cells, and the YAP expression was inhibited by pitavastatin administration. With YAP RNA interference, pAKT, pBAD and BCL-2 expression was decreased, while BAX expression as increased. Accordingly, YAP down-regulated significantly increased apoptosis and decreased the survival rate of gefitinib-resistant lung cancer cells. After pAKT was increased by SC79, apoptosis of YAP down-regulated cells induced by gefitinib was decreased, and the cell survival rate was increased. Mechanistically, these effects of pitavastatin are associated with the YAP pathway, thereby inhibiting the downstream AKT/BAD-BCL-2 signaling pathway.
    CONCLUSIONS: Our study provides a molecular basis for the clinical application of the lipid-lowering drug pitavastatin enhances the susceptibility of lung cancer to EGFR-TKI drugs and alleviates drug resistance.
    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
    高血压和高脂血症是两种常见的疾病,需要有效的管理以降低心血管疾病的风险。在通常用于治疗这些疾病的药物中,缬沙坦和匹伐他汀在降低血压和胆固醇水平方面显示出显著的功效,分别。在这项研究中,同步荧光光谱法与化学计量分析工具相结合,特别是浓度残差增广经典最小二乘(CRACLS)和光谱残差增广经典最小二乘(SRACLS),同时测定缬沙坦和匹伐他汀的含量。开发的模型表现出出色的预测性能,缬沙坦和匹伐他汀的预测相对均方根误差(RRMSEP)为2.253和2.1381,分别。因此,这些模型已成功应用于分析合成样品和商业配方以及血浆样品,具有很高的准确度和精密度。此外,还评估了所确定样品的绿色和蓝色轮廓,以评估其对环境的影响和分析实用性。结果显示了优异的绿色和蓝色评分,AGREE评分为0.7,BAGI评分为75,表明该方法是测定缬沙坦和匹伐他汀的可靠和灵敏的方法,在药物质量控制中具有潜在的应用。生物分析研究,和治疗药物监测。
    Hypertension and hyperlipidemia are two common conditions that require effective management to reduce the risk of cardiovascular diseases. Among the medications commonly used for the treatment of these conditions, valsartan and pitavastatin have shown significant efficacy in lowering blood pressure and cholesterol levels, respectively. In this study, synchronous spectrofluorimetry coupled to chemometric analysis tools, specifically concentration residual augmented classical least squares (CRACLS) and spectral residual augmented classical least squares (SRACLS), was employed for the determination of valsartan and pitavastatin simultaneously. The developed models exhibited excellent predictive performance with relative root mean square error of prediction (RRMSEP) of 2.253 and 2.1381 for valsartan and pitavastatin, respectively. Hence, these models were successfully applied to the analysis of synthetic samples and commercial formulations as well as plasma samples with high accuracy and precision. Besides, the greenness and blueness profiles of the determined samples were also evaluated to assess their environmental impact and analytical practicability. The results demonstrated excellent greenness and blueness scores with AGREE score of 0.7 and BAGI score of 75 posing the proposed method as reliable and sensitive approach for the determination of valsartan and pitavastatin with potential applications in pharmaceutical quality control, bioanalytical studies, and therapeutic drug monitoring.
    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
    他汀类药物可降低慢性肾脏病(CKD)患者心血管事件的风险。尽管糖尿病(DM)是他汀类药物治疗的副作用,一些研究表明,匹伐他汀不会引起DM。本研究调查了匹伐他汀对红细胞膜脂肪酸(FA)含量的影响,影响DM和心血管疾病的发生。此外,评估匹伐他汀治疗后脂联素和糖化血红蛋白(HbA1c)水平的变化.
    共纳入45名患者,其中28人完成了研究。超过24周,16例患者接受2mg匹伐他汀,12例患者接受10mg阿托伐他汀。如果需要额外的脂质对照,则在12周后调整剂量。匹伐他汀和阿托伐他汀组分别有10例和9例DM患者,分别。红细胞膜FAs和脂联素水平测定采用气相色谱法和酶联免疫吸附试验,分别。
    在两组中,饱和FAs,棕榈酸,反式油酸,总胆固醇,低密度脂蛋白胆固醇水平显著低于基线水平.匹伐他汀组红细胞膜中花生四烯酸(AA)含量明显增加,但脂联素水平未受影响。用匹伐他汀治疗的患者HbA1c水平降低。没有与他汀类药物治疗相关的不良反应。
    匹伐他汀治疗CKD患者可能通过改变红细胞膜AA水平来改善糖代谢。此外,匹伐他汀对CKD和DM患者的血糖控制无不良影响.
    UNASSIGNED: Statins reduce the risk of cardiovascular events in patients with chronic kidney disease (CKD). Although diabetes mellitus (DM) is a reported side effect of statin treatment, some studies have indicated that pitavastatin does not cause DM. The present study investigated the effect of pitavastatin on the fatty acid (FA) content of erythrocyte membranes, which affects the occurrence of DM and cardiovascular diseases. In addition, changes in adiponectin and glycated hemoglobin (HbA1c) levels were evaluated after pitavastatin treatment.
    UNASSIGNED: A total of 45 patients were enrolled, 28 of whom completed the study. Over 24 weeks, 16 patients received 2 mg pitavastatin and 12 patients received 10 mg atorvastatin. Dosages were adjusted after 12 weeks if additional lipid control was required. There were 10 and nine patients with DM in the pitavastatin and atorvastatin groups, respectively. Erythrocyte membrane FAs and adiponectin levels were measured using gas chromatography and enzyme-linked immunosorbent assay, respectively.
    UNASSIGNED: In both groups, saturated FAs, palmitic acid, trans-oleic acid, total cholesterol, and low-density lipoprotein cholesterol levels were significantly lower than those at baseline. The arachidonic acid (AA) content in the erythrocyte membrane increased significantly in the pitavastatin group, but adiponectin levels were unaffected. HbA1c levels decreased in patients treated with pitavastatin. No adverse effects were associated with statin treatment.
    UNASSIGNED: Pitavastatin treatment in patients with CKD may improve glucose metabolism by altering erythrocyte membrane AA levels. In addition, pitavastatin did not adversely affect glucose control in patients with CKD and DM.
    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
    鉴于由于固有的抗癌性机制而导致的针对各种癌症类型的当前治疗方式的有限功效,发现有效的抗癌剂提出了巨大的挑战。癌症免疫化疗是乳腺癌治疗和克服癌症耐药性的替代策略。人吲哚胺2,3-双加氧酶(hIDO1)和人色氨酸2,3-双加氧酶2(hTDO2)在色氨酸代谢中起关键作用,导致犬尿氨酸和其他生物活性代谢产物的产生。此过程促进烟酰胺二核苷酸(NAD)的从头合成,促进抗癌性。这项研究使用FDA批准的药物的药物再利用策略确定了一种新的双重hIDO1/hTDO2抑制剂。在这里,我们描述了基于配体的药效团模型的发展,该模型基于12种已报道hIDO1/hTDO2抑制活性的化合物的训练集。我们进行了药效团搜索,然后对2568种FDA批准的针对这两种酶的药物进行了高通量虚拟筛选。导致十次点击,其中四个具有双重抑制活性的高潜力。为了进一步进行计算机模拟和体外生物学研究,抗高胆固醇血症药物匹伐他汀被认为是本研究的首选药物。分子动力学(MD)模拟表明,匹伐他汀与hIDO1和hTDO2受体形成稳定的复合物,为其潜在的治疗功效提供结构基础。在纳摩尔(nM)浓度下,它对两种检查的酶都表现出显着的体外酶抑制活性。此外,匹伐他汀对BT-549,MCF-7和HepG2细胞系表现出有效的细胞毒性活性(IC50=16.82,9.52和1.84µM,分别)。其抗癌活性主要是由于通过HepG2癌细胞的细胞周期分析发现的G1/S期停滞的诱导。最终,用匹伐他汀治疗HepG2癌细胞影响caspase-3的显著激活,伴随着细胞凋亡生物标志物如IDO的下调,TDO,STAT3、P21、P27、IL-6和AhR。
    Discovering effective anti-cancer agents poses a formidable challenge given the limited efficacy of current therapeutic modalities against various cancer types due to intrinsic resistance mechanisms. Cancer immunochemotherapy is an alternative strategy for breast cancer treatment and overcoming cancer resistance. Human Indoleamine 2,3-dioxygenase (hIDO1) and human Tryptophan 2,3-dioxygenase 2 (hTDO2) play pivotal roles in tryptophan metabolism, leading to the generation of kynurenine and other bioactive metabolites. This process facilitates the de novo synthesis of Nicotinamide Dinucleotide (NAD), promoting cancer resistance. This study identified a new dual hIDO1/hTDO2 inhibitor using a drug repurposing strategy of FDA-approved drugs. Herein, we delineate the development of a ligand-based pharmacophore model based on a training set of 12 compounds with reported hIDO1/hTDO2 inhibitory activity. We conducted a pharmacophore search followed by high-throughput virtual screening of 2568 FDA-approved drugs against both enzymes, resulting in ten hits, four of them with high potential of dual inhibitory activity. For further in silico and in vitro biological investigation, the anti-hypercholesterolemic drug Pitavastatin deemed the drug of choice in this study. Molecular dynamics (MD) simulations demonstrated that Pitavastatin forms stable complexes with both hIDO1 and hTDO2 receptors, providing a structural basis for its potential therapeutic efficacy. At nanomolar (nM) concentration, it exhibited remarkable in vitro enzyme inhibitory activity against both examined enzymes. Additionally, Pitavastatin demonstrated potent cytotoxic activity against BT-549, MCF-7, and HepG2 cell lines (IC50 = 16.82, 9.52, and 1.84 µM, respectively). Its anticancer activity was primarily due to the induction of G1/S phase arrest as discovered through cell cycle analysis of HepG2 cancer cells. Ultimately, treating HepG2 cancer cells with Pitavastatin affected significant activation of caspase-3 accompanied by down-regulation of cellular apoptotic biomarkers such as IDO, TDO, STAT3, P21, P27, IL-6, and AhR.
    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
    脂质紊乱,主要是高胆固醇血症,是波兰最常见的心血管(CV)危险因素(这甚至适用于3/4的人)。低密度脂蛋白胆固醇(LDL-C)血清水平是确定CV风险并确定降脂治疗(LLT)目标的基本脂质参数。降脂治疗可改善心血管预后,并在一级和二级心血管预防中延长寿命。尽管有有效的降脂药物和有关其有益作用的可靠数据,LDL-C控制水平严重不足.这是相关的,除其他外,医生的惰性和患者对副作用的恐惧。血脂学的发展使药物具有良好的安全性并使治疗个性化。匹伐他汀,第三有效的降脂他汀类药物,其特点是肌肉并发症和新的糖尿病病例的风险较低,因为它的代谢不同。因此,匹伐他汀对于糖尿病高危患者或现有糖尿病患者是非常好的治疗选择,以及有心血管风险的患者。这份专家意见文件试图就使用匹伐他汀治疗脂质紊乱的地方和可能性提出建议。
    Lipid disorders, primarily hypercholesterolemia, are the most common cardiovascular (CV) risk factor in Poland (this applies even 3/4 of people). The low-density lipoprotein cholesterol (LDL-C) serum level is the basic lipid parameter that should be measured to determine CV risk and determines the aim and target of lipid-lowering treatment (LLT). Lipid-lowering treatment improves cardiovascular prognosis and prolongs life in both primary and secondary cardiovascular prevention. Despite the availability of effective lipid-lowering drugs and solid data on their beneficial effects, the level of LDL-C control is highly insufficient. This is related, among other things, to physician inertia and patients\' fear of side effects. The development of lipidology has made drugs available with a good safety profile and enabling personalisation of therapy. Pitavastatin, the third most potent lipid-lowering statin, is characterised by a lower risk of muscle complications and new cases of diabetes due to its being metabolised differently. Thus, pitavastatin is a very good therapeutic option in patients at high risk of diabetes or with existing diabetes, and in patients at cardiovascular risk. This expert opinion paper attempts at recommendation on the place and possibility of using pitavastatin in the treatment of lipid disorders.
    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
    肺癌是全球癌症相关死亡的最常见原因,由多种因素引起,包括高脂肪饮食(HFD)。CD36,一种脂肪酸受体,与代谢相关的疾病密切相关,包括心血管疾病和癌症。然而,CD36在HFD加速非小细胞肺癌(NSCLC)中的作用尚不清楚.在体内,我们饲喂C57BL/6J野生型(WT)和CD36敲除(CD36-/-)小鼠正常食物或HFD在有或没有匹伐他汀的2周前皮下注射LLC1细胞。体外,用游离脂肪酸(FFA)处理A549和NCI-H520细胞以模拟HFD情况以探索潜在机制。我们发现HFD在体内促进LLC1肿瘤生长,FFA增加A549和NCI-H520细胞的细胞增殖和迁移。降脂药匹伐他汀抑制了增强的细胞或肿瘤生长,减少脂质积累。更重要的是,我们发现,NSCLC患者的血浆可溶性CD36(sCD36)水平高于健康患者.与WT小鼠相比,在CD36-/-小鼠中LLC1细胞的增殖被大大抑制,在HFD组中被匹伐他汀进一步抑制。在分子水平上,我们发现CD36抑制,用匹伐他汀或质粒,通过AKT/mTOR途径降低增殖和迁移相关蛋白的表达。一起来看,我们证明匹伐他汀或其他抑制剂抑制CD36表达可能是NSCLC治疗的可行策略.
    Lung cancer is the most common cause of cancer-related deaths worldwide and is caused by multiple factors, including high-fat diet (HFD). CD36, a fatty acid receptor, is closely associated with metabolism-related diseases, including cardiovascular disease and cancer. However, the role of CD36 in HFD-accelerated non-small-cell lung cancer (NSCLC) is unclear. In vivo, we fed C57BL/6J wild-type (WT) and CD36 knockout (CD36-/-) mice normal chow or HFD in the presence or absence of pitavastatin 2 weeks before subcutaneous injection of LLC1 cells. In vitro, A549 and NCI-H520 cells were treated with free fatty acids (FFAs) to mimic HFD situation for exploration the underlying mechanisms. We found that HFD promoted LLC1 tumor growth in vivo and that FFAs increased cell proliferation and migration in A549 and NCI-H520 cells. The enhanced cell or tumor growth was inhibited by the lipid-lowering agent pitavastatin, which reduced lipid accumulation. More importantly, we found that plasma soluble CD36 (sCD36) levels were higher in NSCLC patients than those in healthy ones. Compared to that in WT mice, the proliferation of LLC1 cells in CD36-/- mice was largely suppressed, which was further repressed by pitavastatin in HFD group. At the molecular level, we found that CD36 inhibition, either with pitavastatin or plasmid, reduced proliferation- and migration-related protein expression through the AKT/mTOR pathway. Taken together, we demonstrate that inhibition of CD36 expression by pitavastatin or other inhibitors may be a viable strategy for NSCLC treatment.
    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
    为临床开发选择候选药物,准确及时地预测人体药代动力学(PK)谱,评估药物-药物相互作用(DDI),预测疾病人群中潜在的PK变化是药物发现的关键步骤。当肝转运蛋白参与药物清除(CL)和分布体积(Vss)时,预测人类PK的复杂性显着增加。这里制定了一个战略框架,以匹伐他汀为例。该框架包括构建基于猴子生理的PK(PBPK)模型,模型校准,以获得各种清除参数的体外-体内外推(IVIVIVE)的比例因子(SF),人体模型的开发和验证,以及疾病人群中DDI和PK变化的评估。通过整合体外人体参数和来自Clint的3.45、0.14和1.17的猴子模型的校准SF,活跃,CLINT,被动,和CLINT,胆汁,分别,以及从体外研究中获得的单个转运蛋白转运的相对分数和OATP抑制的优化Ki值,该模型合理地捕获了观察到的匹伐他汀PK曲线,携带遗传多态性的人类受试者的DDI和PK变异,即,AUC在20%以内。最后,当应用基于测量的OATP1B生物标志物的功能减少时,该模型可以充分预测肝功能损害人群的PK变化。本研究提出了早期药物开发的战略框架,能够预测PK概况和评估DDI等场景中的PK变化,遗传多态性,和肝功能损害相关的疾病状态,特别关注OATP底物。
    To select a drug candidate for clinical development, accurately and promptly predicting human pharmacokinetic (PK) profiles, assessing drug-drug interactions (DDIs), and anticipating potential PK variations in disease populations are crucial steps in drug discovery. The complexity of predicting human PK significantly increases when hepatic transporters are involved in drug clearance (CL) and volume of distribution (Vss). A strategic framework is developed here, utilizing pitavastatin as an example. The framework includes the construction of a monkey physiologically-based PK (PBPK) model, model calibration to obtain scaling factors (SF) of in vitro-in vivo extrapolation (IVIVE) for various clearance parameters, human model development and validation, and assessment of DDIs and PK variations in disease populations. Through incorporating in vitro human parameters and calibrated SFs from the monkey model of 3.45, 0.14, and 1.17 for CLint,active, CLint,passive, and CLint,bile, respectively, and together with the relative fraction transported by individual transporters obtained from in vitro studies and the optimized Ki values for OATP inhibition, the model reasonably captured observed pitavastatin PK profiles, DDIs and PK variations in human subjects carrying genetic polymorphisms, i.e., AUC within 20%. Lastly, when applying the functional reduction based on measured OATP1B biomarkers, the model adequately predicted PK changes in the hepatic impairment population. The present study presents a strategic framework for early-stage drug development, enabling the prediction of PK profiles and assessment of PK variations in scenarios like DDIs, genetic polymorphism, and hepatic impairment-related disease states, specifically focusing on OATP substrates.
    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
    匹伐他汀,一种有效的3-羟甲基戊二酰辅酶A还原酶抑制剂,用于治疗高胆固醇血症和混合性血脂异常。匹伐他汀的肝摄取主要由有机阴离子转运多肽1B1(OATP1B1)和溶质载体有机阴离子转运蛋白家族成员1B1(SLCO1B1)基因占据,它是编码OATP1B1的多态基因。SLCO1B1基因多态性显著改变匹伐他汀的药代动力学。本研究旨在建立基于生理的药代动力学(PBPK)模型,以根据SLCO1B1基因多态性预测匹伐他汀的药代动力学。采用PK-Sim®10.0版建立匹伐他汀的全身PBPK模型。我们的药物基因组学数据和总共27个具有不同剂量给药和人口统计学特性的临床药代动力学数据用于开发和验证模型。分别。匹伐他汀的理化性质和处置特征是从先前报道的数据中获得的,或经过优化以捕获不同SLCO1B1二倍体型中的血浆浓度-时间曲线。通过将预测的药代动力学参数和曲线与观察到的数据进行比较来进行模型评估。预测的血浆浓度-时间曲线在视觉上类似于在非基因型群体和不同SLCO1B1二倍体中观察到的曲线。AUC和Cmax的所有倍数误差值包括在观察值的两倍范围内。因此,正确建立了匹伐他汀在不同SLCO1B1复型中的PBPK模型。本研究可用于在不同年龄的个体中个体化匹伐他汀的剂量给药策略。种族,和SLCO1B1二倍体。
    Pitavastatin, a potent 3-hydroxymethylglutaryl coenzyme A reductase inhibitor, is indicated for the treatment of hypercholesterolemia and mixed dyslipidemia. Hepatic uptake of pitavastatin is predominantly occupied by the organic anion transporting polypeptide 1B1 (OATP1B1) and solute carrier organic anion transporter family member 1B1 (SLCO1B1) gene, which is a polymorphic gene that encodes OATP1B1. SLCO1B1 genetic polymorphism significantly alters the pharmacokinetics of pitavastatin. This study aimed to establish the physiologically based pharmacokinetic (PBPK) model to predict pitavastatin pharmacokinetics according to SLCO1B1 genetic polymorphism. PK-Sim® version 10.0 was used to establish the whole-body PBPK model of pitavastatin. Our pharmacogenomic data and a total of 27 clinical pharmacokinetic data with different dose administration and demographic properties were used to develop and validate the model, respectively. Physicochemical properties and disposition characteristics of pitavastatin were acquired from previously reported data or optimized to capture the plasma concentration-time profiles in different SLCO1B1 diplotypes. Model evaluation was performed by comparing the predicted pharmacokinetic parameters and profiles to the observed data. Predicted plasma concentration-time profiles were visually similar to the observed profiles in the non-genotyped populations and different SLCO1B1 diplotypes. All fold error values for AUC and Cmax were included in the two fold range of observed values. Thus, the PBPK model of pitavastatin in different SLCO1B1 diplotypes was properly established. The present study can be useful to individualize the dose administration strategy of pitavastatin in individuals with various ages, races, and SLCO1B1 diplotypes.
    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
    匹伐他汀(PITA)是一种3-羟基-3-甲基戊二酰辅酶A(HMG-CoA)还原酶抑制剂,用于治疗高胆固醇血症,最近的研究集中于其潜在的抗癌作用。本研究旨在阐明PITA单独和联合顺铂对宫颈癌细胞(HeLa)的体外作用。通过3-(4,5-二甲基噻唑-2-基)-2,5-二苯基四唑溴化物(MTT)和中性红摄取(NRU)测定24、48和72小时来评估PITA(5-200μM)的细胞毒性。在流式细胞术(0.1-100μM)中进行细胞凋亡和细胞周期分析。使用标准彗星测定法对PITA(2-200μM)的遗传毒性作用和氧化DNA损伤进行评估,甲酰氨基嘧啶糖基化酶(fpg)修饰的彗星试验,和HeLa细胞中的活性氧(ROS)活化。PITA单独以剂量依赖性方式降低细胞活力(20-200、20-200和5-200μM持续24、48和72小时,分别,在MTT中)。PITA与顺铂的联合治疗导致显著更大的细胞活力抑制。ROS和DNA损伤在100μM持续4小时和20μM持续24小时时显著增加,分别。PITA诱导的细胞凋亡,亚G1细胞比例增加,被监控,而且,通过蛋白质印迹和caspase3/8/9多重分析试剂盒,它增加了活性caspase-9和caspase-3的表达,并上调了裂解的聚二磷酸腺苷核糖聚合酶(PARP)。我们得出结论,PITA可以用于有效的宫颈癌研究,并获得了有希望的发现供进一步研究。
    Pitavastatin (PITA) is a 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase inhibitor to treat hypercholesterolemia and in recent studies is focused that its potential anti-cancer effect. This study was aimed to elucidate the effect of PITA alone and in combination with cisplatin on cervical cancer cells (HeLa) in vitro. Cytotoxicity of PITA (5-200 μM) was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and neutral red uptake (NRU) assays for 24, 48, and 72 h. Cell apoptosis and cell cycle analyses were performed in flow cytometry (0.1-100 μM). The evaluation of genotoxic effects and oxidative DNA damage of PITA (2-200 μM) were performed with standard comet assay, formamidopyrimidine glycosylase (fpg)-modified comet assay, and reactive oxygen species (ROS) activation in HeLa cells. PITA alone reduced cell viability in a dose-dependent manner (20-200, 20-200, and 5-200 μM for 24, 48, and 72 h, respectively, in MTT). The combined treatment of PITA with cisplatin resulted in significantly greater inhibition of cell viability. ROS and DNA damage increased significantly at 100 μM for 4 h and 20 μM for 24 h, respectively. PITA-induced apoptosis, an increased proportion of sub G1 cells, was monitored, and also, it increased the expression of active caspase-9 and caspase-3 and upregulated cleaved poly adenosine diphosphate ribose polymerase (PARP) by western blotting and caspase 3/8/9 multiple assay kit. We conclude that PITA can be used to efficiently cervical cancer studies, and promising findings have been obtained for further 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
    犬口腔黑色素瘤是一种高度恶性的癌症,预后不良。他汀类药物,常用的治疗血脂异常的药物,对黑素瘤细胞具有多效抗癌作用和明显的抗增殖作用。他汀类药物的抗癌作用各不相同;在人类癌症中,与亲水性他汀相比,亲脂性他汀类药物显示出更强的抗癌作用。然而,缺乏各种他汀类药物对犬癌细胞影响差异的数据,因此,治疗犬黑色素瘤的最佳他汀类药物仍然未知。因此,这项研究旨在通过比较亲水性瑞舒伐他汀和亲脂性阿托伐他汀的抗癌作用来阐明最有效的他汀类药物,辛伐他汀,氟伐他汀和匹伐他汀对三种犬口腔黑色素瘤细胞系的影响。细胞融合的时间依赖性测量表明,亲脂性他汀类药物对所有细胞系的抗增殖作用均强于亲水性瑞舒伐他汀。乳酸脱氢酶释放的定量,细胞毒性的指标,结果表明,亲脂性他汀类药物比亲水性瑞舒伐他汀更有效地诱导细胞死亡。亲脂性他汀类药物影响细胞增殖的抑制和细胞死亡的诱导。他汀类药物对犬口腔黑素瘤细胞的抗癌作用在以下IC50值的升序上不同:匹伐他汀<氟伐他汀=辛伐他汀<阿托伐他汀<瑞舒伐他汀。匹伐他汀的所需浓度约为瑞舒伐他汀的1/20。在这项研究中使用的他汀类药物中,匹伐他汀的抗癌作用最高。我们的结果表明,亲脂性匹伐他汀是治疗犬口服黑色素瘤的最佳他汀类药物。
    Canine oral melanoma is a highly malignant cancer with a poor prognosis. Statins, commonly used drugs for treating dyslipidemia, exhibit pleiotropic anticancer effects and marked anti-proliferative effects against melanoma cells. The anticancer effects among statins vary; in human cancers, lipophilic statins have shown stronger anticancer effects compared with hydrophilic statins. However, data on the differences in the effects of various statins on canine cancer cells are lacking, hence the optimal statins for treating canine melanoma remain unknown. Therefore, this study aimed to clarify the most effective statin by comparing the anticancer effects of hydrophilic rosuvastatin and lipophilic atorvastatin, simvastatin, fluvastatin and pitavastatin on three canine oral melanoma cell lines. Time-dependent measurement of cell confluence showed that lipophilic statins had a stronger anti-proliferative effect on all cell lines than hydrophilic rosuvastatin. Quantification of lactate dehydrogenase release, an indicator of cytotoxicity, showed that lipophilic statins more effectively induced cell death than hydrophilic rosuvastatin. Lipophilic statins affected both inhibition of cell proliferation and induction of cell death. The anticancer effects of statins on canine oral melanoma cells differed in the following ascending order of IC50 values: pitavastatin < fluvastatin = simvastatin < atorvastatin < rosuvastatin. The required concentration of pitavastatin was approximately 1/20th that of rosuvastatin. Among the statins used in this study, pitavastatin had the highest anticancer effect. Our results suggest lipophilic pitavastatin as the optimal statin for treating canine oral melanoma.
    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