Doxorubicin

阿霉素
  • 文章类型: Journal Article
    背景:曲贝替丁与聚乙二醇化脂质体阿霉素(PLD)的组合被批准用于治疗铂敏感的复发性卵巢癌患者。然而,在现实世界中,目前关于老年卵巢癌患者的这种治疗的信息有限.
    方法:这项观察性和多中心研究回顾性评估了trabectedin联合PLD在现实世界中治疗被诊断为铂类敏感的复发性卵巢癌的老年患者,根据15家GEICO相关医院的产品特征摘要(SmPC)进行治疗。治疗开始时年龄≥70岁的患者和无铂间隔≥6个月被认为是合格的。
    结果:43例患者的中位年龄为74.0岁。2015年12月31日,2019年在15个西班牙中心。4例患者达到完全缓解(9.3%),14(32.6%)部分反应,13例(30.2%)疾病稳定为最佳放射学反应。在根据CA125血清水平分析生物学总体反应(即,Rustin标准),14人对治疗有反应(32.6%),11人回应并恢复正常(25.6%),3例患者稳定(7.0%),3例进展(7.0%).研究人群的中位无进展生存期(PFS)和总生存期(OS)分别为7.7和19.5个月,分别。最常见的3/4级不良事件是中性粒细胞减少(n=8,18.7%)和虚弱(n=5,11.6%)。
    结论:这项分析表明,曲贝替丁联合PLD治疗老年铂敏感型复发性卵巢癌是一种可行且有效的治疗方法。显示出可接受的安全性,这对这些患者的姑息治疗至关重要。
    BACKGROUND: Trabectedin in combination with pegylated liposomal doxorubicin (PLD) is approved for the treatment of patients with platinum-sensitive relapsed ovarian cancer. Nevertheless, there is currently limited information regarding this treatment in elderly patients with ovarian cancer in a real-world setting.
    METHODS: This observational and multicentric study retrospectively evaluated trabectedin plus PLD in a real-world setting treatment of elderly patients diagnosed with platinum-sensitive relapsed ovarian cancer, treated according to the Summary of Product Characteristics (SmPC) from 15 GEICO-associated hospitals. Patients ≥ 70 years old at the time of treatment initiation and platinum-free intervals ≥ 6 months were considered eligible.
    RESULTS: Forty-three patients with a median age of 74.0 years were treated between January 1st, 2015, and December 31st, 2019 in 15 Spanish centers. Four patients achieved complete response (9.3%), 14 (32.6%) partial response, and 13 (30.2%) stable disease as the best radiological response. In the analysis of biological overall response according to CA125 serum levels (i.e., Rustin criteria), 14 responded to the treatment (32.6%), 11 responded and normalized (25.6%), three patients stabilized (7.0%) and three progressed (7.0%). Median progression-free survival (PFS) and overall survival (OS) in the study population were 7.7 and 19.5 months, respectively. The most common grade 3/4 adverse events were neutropenia (n = 8, 18.7%) and asthenia (n = 5, 11.6%).
    CONCLUSIONS: This analysis demonstrated that trabectedin combined with PLD is a feasible and effective treatment in elderly patients with platinum-sensitive relapsed ovarian cancer, showing an acceptable safety profile, which is crucial in the palliative treatment of these patients.
    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
    严重的心脏毒性限制了阿霉素(DOX)的临床应用。中药穿心莲及其主要活性成分(脱水穿心莲内酯,DA)以其多种心血管保护作用而闻名。然而,DA对Dox诱导的心脏毒性(DIC)的影响尚不清楚.在这项研究中,我们在体内和体外评估了DA对DIC的影响,并揭示了DA对DIC的潜在机制。通过超声心动图和组织学测定系统地评估了DA对DIC的影响。Westernblot和流式细胞术检测心肌细胞凋亡。TEM和免疫荧光进一步用于测定自噬通量。我们的结果表明,DA给药可显着改善心功能并减轻DOX诱导的心肌细胞凋亡。机械上,DA通过抑制Dox诱导的mTOR信号通路激活和增加TFEB向细胞核的转位,恢复自噬通量和溶酶体功能。然而,mTOR的激活或TFEB的敲减通过影响溶酶体功能和自噬通量而显著抑制DA对DIC的保护作用。总之,我们的结果表明,DA可能是一种潜在的抗DIC心脏保护剂.
    The clinical application of doxorubicin (DOX) was limited by the serious cardiotoxicity. The traditional Chinese medicine Andrographis paniculata and its principal active component (Dehydroandrographolide, DA) have been well known for their diverse cardiovascular protective effects. However, the effects of DA on Dox-induced cardiotoxicity (DIC) were still unknown. In this study, we evaluated the effects and revealed the potential mechanisms of DA on DIC both in vivo and in vitro. The effects of DA on DIC were systematically assessed by echocardiography and histological assays. Western blot and flow cytometry were used to measure apoptosis of cardiomyocytes. TEM and immunofluorescence were further used to assay autophagic flux. Our results showed that DA administration significantly improved cardiac function and attenuated DOX-induced cardiomyocyte apoptosis. Mechanically, DA restored autophagic flux and lysosome function via inhibiting Dox-induced mTOR signal pathway activation and increasing the translocation of TFEB to the nucleus. However, activation of mTOR or knockdown of TFEB significantly inhibited the protective effects of DA against DIC by impacting lysosomal functions and autophagic flux. In conclusion, our results revealed that DA might be a potential cardioprotective agent against DIC.
    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
    乳腺癌(BC)是女性最常见的致命癌症。需要探索新的治疗策略以通过克服BC细胞的抗性来增强阿霉素的功效。NUF2是Ndc80着丝粒复合物的组成部分,是介导有丝分裂的关键物质,并影响多种肿瘤的进展。然而,NUF2抗性在BC中的作用和机制尚不清楚。本研究旨在揭示NUF2在BC耐药中的作用。我们在这里发现NUF2在人类BC中高度表达。NUF2消耗来源的外泌体阻断BC细胞的生长。Further,NUF2消融来源的外泌体抑制BC细胞自噬。此外,NUF2消融衍生的外泌体改善了BC细胞的阿霉素抗性。机械上,NUF2消融衍生的外泌体阻断了BC细胞中的PI3K/AKT/mTOR轴。总之,NUF2消融来源的外泌体通过介导PI3K/AKT/mTOR轴阻断BC细胞自噬并改善阿霉素耐药性。
    Breast cancer (BC) is the most common type of fatal cancer in women. New therapeutic strategies need to be explored to enhance the efficacy of doxorubicin by overcoming the resistance of BC cells. NUF2 is a component of the Ndc80 centromere complex and is a key substance in mediating mitosis and affects the progression of multiple tumors. However, the role as well as mechanisms of NUF2 resistance in BC remain unclear. This study aims to reveal the role of NUF2 in drug resistance in BC. We here revealed that NUF2 was highly expressed in human BC. NUF2 depletion-derived exosomes blocked the growth of BC cells. Further, NUF2 ablation-derived exosomes inhibited autophagy in BC cells. Also, NUF2 ablation-derived exosomes improved doxorubicin resistance in BC cells. Mechanically, NUF2 ablation-derived exosomes blocked PI3K/AKT/mTOR axis in BC cells. In summary, NUF2 ablation-derived exosomes blocked the autophagy of BC cells and improved doxorubicin resistance via mediating PI3K/AKT/mTOR axis.
    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
    透明质酸酶通过切割透明质酸增加细胞外液的组织渗透性和扩散,细胞外基质的主要成分。透明质酸酶聚乙二醇化(Hyal-PEG)降低其清除率并增强生物分布。在体外(大鼠胶质母细胞瘤101.8球体的形态学分析)和体内(通过脑内移植肿瘤后大鼠的存活时间和形态学分析)研究了Hyal-PEG和Hyal-PEG与阿霉素的组合的促和抗癌活性。在体外培养基中存在阿霉素和Hyal-PEG的情况下,球状体失去了粘附在基质上的能力并分解为单个细胞。用Hyal-PEG对肿瘤组织进行脑内移植不会加速胶质母细胞瘤的生长。接受单独移植肿瘤和与Hyal-PEG联合移植的动物的平均存活时间为13天和20天,分别。在一只移植肿瘤和Hyal-PEG的大鼠中,这个参数增加了53%。接受阿霉素和Hyal-PEG全身治疗的大鼠的存活时间显着增加(p=0.003)。在体外大鼠胶质母细胞瘤101.8模型上证明了治疗剂量的阿霉素与Hyal-PEG的抗肿瘤作用。Hyal-PEG抑制肿瘤细胞的粘附,但没有造成他们的死亡.Hyal-PEG处理的肿瘤的移植不减少动物存活时间。治疗剂量的阿霉素与Hyal-PEG的全身给药增加了患有胶质母细胞瘤的大鼠的存活时间101.8。
    Hyaluronidase increases tissue permeability and diffusion of the extracellular fluid by cleaving hyaluronan, the primary component of the extracellular matrix. Hyaluronidase pegylation (Hyal-PEG) decreases its clearance and enhances biodistribution. The pro- and anticancer activity of Hyal-PEG and a combination of Hyal-PEG with doxorubicin were studied in vitro (morphological analysis of rat glioblastoma 101.8 spheroids) and in vivo (by the survival time of rats after intracerebral transplantation of the tumor and morphological analysis). In the presence of doxorubicin and Hyal-PEG in the culture medium in vitro, spheroids lost their ability to adhere to the substrate and disintegrate into individual cells. Intracerebral transplantation of the tumor tissue with Hyal-PEG did not accelerate glioblastoma growth. The mean survival time for animals receiving transplantation of the tumor alone and in combination with Hyal-PEG was 13 and 20 days, respectively. In one rat with transplanted tumor and Hyal-PEG, this parameter increased by 53%. The survival time of rats receiving systemic therapy with doxorubicin and Hyal-PEG significantly increased (p=0.003). Antitumor effect of therapeutic doses of doxorubicin combined with Hyal-PEG was demonstrated on the model of rat glioblastoma 101.8 in vitro. Hyal-PEG inhibited adhesion of tumor cells, but did not cause their death. Transplantation of Hyal-PEG-treated tumor did not reduce animal survival time. Systemic administration of therapeutic doses of doxorubicin with Hyal-PEG increased survival time of rats with glioblastoma 101.8.
    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
    智能纳米药物递送系统(Cu/ZIF-8@GOx-DOX@HA,以下为CZGDH),由掺杂Cu的沸石咪唑酯骨架8(Cu/ZIF-8,以下为CZ)组成,葡萄糖氧化酶(GOx),多柔比星(DOX),透明质酸(HA)用于肿瘤的靶向给药和协同治疗。CZGDH通过HA的靶向作用特异性进入肿瘤细胞,并表现出酸度触发的生物降解作用,随后释放GOx,DOX,和肿瘤微环境(TME)中的Cu2+。GOx氧化肿瘤细胞中的葡萄糖(Glu)以产生H2O2和葡萄糖酸用于饥饿治疗(ST)。DOX进入肿瘤内细胞核进行化疗(CT)。释放的Cu2+消耗肿瘤细胞中过表达的谷胱甘肽(GSH)以产生Cu+。生成的Cu+和H2O2引发类Fenton反应生成有毒的羟基自由基(·OH),这破坏了肿瘤细胞的氧化还原平衡,并有效地杀死了肿瘤细胞进行化学动力学治疗(CDT)。因此,通过TME激活的级联反应实现了协同多峰肿瘤治疗。纳米药物递送系统具有高的载药率(48.3wt%),三模式协同治疗对肿瘤细胞有很强的杀伤作用(67.45%)。
    An intelligent nanodrug delivery system (Cu/ZIF-8@GOx-DOX@HA, hereafter CZGDH) consisting of Cu-doped zeolite imidazolate framework-8 (Cu/ZIF-8, hereafter CZ), glucose oxidase (GOx), doxorubicin (DOX), and hyaluronic acid (HA) was established for targeted drug delivery and synergistic therapy of tumors. The CZGDH specifically entered tumor cells through the targeting effect of HA and exhibited acidity-triggered biodegradation for subsequent release of GOx, DOX, and Cu2+ in the tumor microenvironment (TME). The GOx oxidized the glucose (Glu) in tumor cells to produce H2O2 and gluconic acid for starvation therapy (ST). The DOX entered the intratumoral cell nucleus for chemotherapy (CT). The released Cu2+ consumed the overexpressed glutathione (GSH) in tumor cells to produce Cu+. The generated Cu+ and H2O2 triggered the Fenton-like reaction to generate toxic hydroxyl radicals (·OH), which disrupted the redox balance of tumor cells and effectively killed tumor cells for chemodynamic therapy (CDT). Therefore, synergistic multimodal tumor treatment via TME-activated cascade reaction was achieved. The nanodrug delivery system has a high drug loading rate (48.3 wt%), and the three-mode synergistic therapy has a strong killing effect on tumor cells (67.45%).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    阿霉素是常用的化疗药物,已用于治疗各种类型的恶性肿瘤,包括软组织和骨肉瘤以及乳腺癌等重要器官的癌症。子房,膀胱,和甲状腺。它也用于治疗白血病和淋巴瘤,然而,这是一个障碍,因为它们突出的副作用包括心脏毒性和肺纤维化,我们的目的是确定CoQ10作为抗氧化剂在阻止阿霉素对组织退行性影响的有害影响方面的作用。要做到这一点,将27只大鼠细分为3组,每组9只;辅酶Q10暴露组,阿霉素暴露组,和CoQ10加多柔比星组。在研究结束时,处死动物,收获带有心脏的肺,并准备载玻片在显微镜下检查。结果表明,阿霉素诱导了异常的细胞结构,从而破坏了肺和心脏的细胞结构,而CoQ10则阻碍了这些破坏作用并几乎恢复正常的组织结构。因此,CoQ10将维持肺和心脏的正常组织。
    Doxorubicin is the common chemotherapeutic agent that has been harnessed for the treatment of various types of malignancy including the treatment of soft tissue and osteosarcoma and cancers of the vital organs like breast, ovary, bladder, and thyroid. It is also used to treat leukaemia and lymphoma, however, this is an obstacle because of their prominent side effects including cardiotoxicity and lung fibrosis, we do aim to determine the role of CoQ10 as an antioxidant on the impeding the deleterious impacts of doxorubicin on tissue degenerative effects. To do so, 27 rats were subdivided into 3 groups of 9 each; CoQ10 exposed group, Doxorubicin exposed group, and CoQ10 plus Doxorubicin group. At the end of the study, the animals were sacrificed and lungs with hearts were harvested, and slides were prepared for examination under a microscope. The results indicated that doxorubicin induced abnormal cellular structure resulting in damaging cellular structures of the lung and heart while CoQ10 impeded these damaging effects and nearly restoring normal tissue structure. As a result, CoQ10 will maintain normal tissue of the lung and heart.
    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
    由困扰癌症幸存者的慢性压力触发的交感激活是肿瘤发生的新兴调节剂。肾上腺素能阻断以前与改善三阴性乳腺癌(TNBC)对阿霉素(DOX)的反应有关,然而,确切的潜在机制仍然不清楚。化疗期间癌症干细胞(CSC)的恢复力促进抵抗和复发。缺氧诱导因子-1α(HIF-1α)和β-catenin是富含CSC的相互交织的转录因子,有证据表明它们的表达可能受到系统性肾上腺素能信号的调节。在这里,我们旨在探讨使用卡维地洛(CAR)阻断肾上腺素受体对DOX的影响及其调节CSC克服化疗耐药的潜力.为了实现这一目标,使用肾上腺素预孵育的MDA-MB-231细胞进行体外研究,并使用慢性束缚应激促进的实体瘤小鼠模型进行体内研究。结果表明,肾上腺素增加了TNBC的增殖,并诱导了一个让人联想到CSC的表型转换,如增强的乳腺球形成所证明的。这些结果与醛脱氢酶-1(ALDH-1)和Nanog表达水平以及HIF-1α和β-catenin激增平行。在体内,与未应激的小鼠相比,在慢性应激下观察到更大的肿瘤体积.使用CAR的肾上腺素能阻滞,然而,通过增强凋亡增强DOX对停止TNBC细胞增殖和肿瘤生长的影响。CAR还抑制了HIF-1α和β-连环蛋白肿瘤水平,随后抑制了ALDH-1和SOX2。我们的研究揭示了HIF-1α通过β-catenin途径连接应激诱导的交感神经激活促进CSC富集的核心作用。它还强调了对CAR逆转TNBCDOX化学耐药能力的新见解。
    Sympathetic activation triggered by chronic stress afflicting cancer survivors is an emerging modulator of tumorigenesis. Adrenergic blockade was previously associated with improving response to doxorubicin (DOX) in triple-negative breast cancer (TNBC), yet the precise underlying mechanisms remain obscure. The resilience of cancer stem cells (CSCs) during chemotherapy fosters resistance and relapse. Hypoxia-inducible factor-1α (HIF-1α) and β-catenin are intertwined transcriptional factors that enrich CSCs and evidence suggests that their expression could be modulated by systemic adrenergic signals. Herein, we aimed to explore the impact of adrenoreceptor blockade using carvedilol (CAR) on DOX and its potential to modulate CSCs overcoming chemoresistance. To achieve this aim, in vitro studies were conducted using adrenaline-preincubated MDA-MB-231 cells and in vivo studies using a chronic restraint stress-promoted solid tumor mouse model. Results revealed that adrenaline increased TNBC proliferation and induced a phenotypic switch reminiscent of CSCs, as evidenced by enhanced mammosphere formation. These results paralleled an increase in aldehyde dehydrogenase-1 (ALDH-1) and Nanog expression levels as well as HIF-1α and β-catenin upsurge. In vivo, larger tumor volumes were observed in mice under chronic stress compared to their unstressed counterparts. Adrenergic blockade using CAR, however, enhanced the impact DOX had on halting TNBC cell proliferation and tumor growth via enhanced apoptosis. CAR also curbed HIF-1α and β-catenin tumor levels subsequently suppressing ALDH-1 and SOX2. Our study unveils a central role for HIF-1α linking stress-induced sympathetic activation fueling CSC enrichment via the β-catenin pathway. It also highlights novel insights into CAR\'s capacity in reversing DOX chemoresistance in TNBC.
    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
    多柔比星(DOX)是与对男性生殖健康的不利影响相关的化学治疗剂。小球藻(ChV)是一种有效的天然抗氧化剂,在维持健康和预防氧化应激相关疾病方面具有广阔的应用前景。本研究旨在探讨ChV对DOX致睾丸毒性的保护作用。将25只Wistar大鼠(230±20g)随机分为5组(n=5),包括对照组,假手术组(每天口服生理盐水,每周一次腹腔(IP)),DOX组(3mg/kg;每周一次;IP),ChV组(300mg/kg/天;口服灌胃),和DOX(3mg/kg;每周一次;IP)+ChV(300mg/kg/天;经口灌胃)组。治疗8周后,对大鼠实施安乐死,血清睾酮水平,睾丸组织形态计量学,性腺指数(GSI),凋亡基因表达,氧化应激指数,和精子参数进行了评估。结果表明,DOX导致组织学指标显著下降,睾酮水平,GSI,精子参数,和Bcl-2基因表达和P-53和Bax基因表达增加,和氧化应激标志物(P<0.05)。在DOX+ChV组中施用ChV显著改善了睾酮水平,精子参数,睾丸组织凋亡,抗氧化酶,和睾丸的结构完整性(P<0.05)。研究结果表明,ChV的共同给药可能是一种有前途的治疗剂,可以减少DOX对男性生殖性能的不利影响。
    Doxorubicin (DOX) is a chemotherapy agent associated with adverse effects on male reproductive health. Chlorella vulgaris (ChV) is a potent natural antioxidant with promising applications in maintaining health and preventing oxidative stress-related diseases. The present study aimed to investigate the protective effect of ChV on DOX-induced testicular toxicity. Twenty-five Wistar rats (230 ± 20g) were randomly assigned to five groups (n = 5), including the control group, sham group (received normal saline by oral gavage daily and intraperitoneally (IP) once a week), DOX group (3mg/kg; once a week; IP), ChV group (300mg/kg/day; by oral gavage), and DOX (3mg/kg; once a week; IP) + ChV (300mg/kg/day; by oral gavage) group. After 8 weeks of treatment, the rats were euthanized and serum testosterone level, testes histomorphometry, gonadosomatic index (GSI), apoptotic gene expression, oxidative stress index, and sperm parameters were assessed. The results showed that DOX led to a significant decrease in histological indexes, testosterone level, GSI, sperm parameters, and Bcl-2 gene expression and increased expression of P-53 and Bax genes, and oxidative stress markers (P<0.05). The administration of ChV in the DOX+ChV group significantly improved testosterone levels, sperm parameters, testicular tissue apoptosis, antioxidant enzymes, and structural integrity of the testes (P<0.05). The findings suggest that the co-administration of ChV can be a promising therapeutic agent to reduce the adverse effects of DOX on male reproductive performance.
    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
    BACKGROUND: The anthracycline chemotherapeutic antibiotic doxorubicin (DOX) can induce cumulative cardiotoxicity and lead to cardiac dysfunction. Long non-coding RNAs (lncRNAs) can function as important regulators in DOX-induced myocardial injury.
    OBJECTIVE: This study aims to investigate the functional role and molecular mechanism of lncRNA OXCT1 antisense RNA 1 (OXCT1-AS1) in DOX-induced myocardial cell injury in vitro.
    METHODS: Human cardiomyocytes (AC16) were stimulated with DOX to induce a myocardial cell injury model. OXCT1-AS1, miR-874-3p, and BDH1 expression in AC16 cells were determined by RT-qPCR. AC16 cell viability was measured by XTT assay. Flow cytometry was employed to assess the apoptosis of AC16 cells. Western blotting was used to evaluate protein levels of apoptosis-related markers. Dual-luciferase reporter assay was conducted to verify the binding ability between miR-874-3p and OXCT1-AS1 and between miR-874-3p and BDH1. The value of p<0.05 indicated statistical significance.
    RESULTS: OXCT1-AS1 expression was decreased in DOX-treated AC16 cells. Overexpression of OXCT1-AS1 reversed the reduction of cell viability and promotion of cell apoptosis caused by DOX. OXCT1-AS1 is competitively bound to miR-874-3p to upregulate BDH1. BDH1 overexpression restored AC16 cell viability and suppressed cell apoptosis under DOX stimulation. Knocking down BDH1 reversed OXCT1-AS1-mediated attenuation of AC16 cell apoptosis under DOX treatment.
    CONCLUSIONS: LncRNA OXCT1-AS1 protects human myocardial cells AC16 from DOX-induced apoptosis via the miR-874-3p/BDH1 axis.
    OBJECTIVE: O antibiótico quimioterápico antraciclina doxorrubicina (DOX) pode induzir cardiotoxicidade cumulativa e levar à disfunção cardíaca. RNAs não codificantes longos (lncRNAs) podem funcionar como importantes reguladores na lesão miocárdica induzida por DOX.
    OBJECTIVE: Este estudo tem como objetivo investigar o papel funcional e o mecanismo molecular do RNA antisense lncRNA OXCT1 1 (OXCT1-AS1) na lesão celular miocárdica induzida por DOX in vitro.
    UNASSIGNED: Cardiomiócitos humanos (AC16) foram estimulados com DOX para induzir um modelo de lesão celular miocárdica. A expressão de OXCT1-AS1, miR-874-3p e BDH1 em células AC16 foi determinada por RT-qPCR. A viabilidade das células AC16 foi medida pelo ensaio XTT. A citometria de fluxo foi empregada para avaliar a apoptose de células AC16. Western blotting foi utilizado para avaliar os níveis proteicos de marcadores relacionados à apoptose. O ensaio repórter de luciferase dupla foi conduzido para verificar a capacidade de ligação entre miR-874-3p e OXCT1-AS1 e entre miR-874-3p e BDH1. O valor de p<0,05 indicou significância estatística.
    RESULTS: A expressão de OXCT1-AS1 foi diminuída em células AC16 tratadas com DOX. A superexpressão de OXCT1-AS1 reverteu a redução da viabilidade celular e a promoção da apoptose celular causada pela DOX. OXCT1-AS1 está ligado competitivamente ao miR-874-3p para regular positivamente o BDH1. A superexpressão de BDH1 restaurou a viabilidade das células AC16 e suprimiu a apoptose celular sob estimulação com DOX. A derrubada do BDH1 reverteu a atenuação da apoptose de células AC16 mediada por OXCT1-AS1 sob tratamento com DOX.
    UNASSIGNED: LncRNA OXCT1-AS1 protege células miocárdicas humanas AC16 da apoptose induzida por DOX através do eixo miR-874-3p/BDH1.
    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
    癌症耐药性与代谢适应有关。已显示癌细胞在细胞死亡期间的适应中涉及乙酰化多胺。然而,目前尚缺乏探索乙酰化多胺作为潜在抗癌药物的模拟物。我们对用多柔比星(DOX)处理的人乳腺癌MCF-7细胞进行了细胞内代谢物谱分析,一种众所周知的抗癌药物。采用新颖的内部垂直管凝胶电泳辅助程序,然后进行LC-HRMS分析,以检测乙酰化多胺,例如N1-乙酰亚精胺。我们设计了一种模拟物N1-乙酰亚精胺(MINAS),它是组蛋白脱乙酰酶10(HDAC10)的已知底物。使用分子对接和分子动力学(MD)模拟来评估MINAS对HDAC10的抑制潜力。将MINAS的抑制潜力和ADMET曲线与已知的HDAC10抑制剂TubastatinA.N1-乙酰亚精胺进行了比较,聚胺的乙酰化形式,在用DOX处理的MCF-7细胞中细胞内检测到超过DMSO处理的MCF-7细胞。我们设计并策划了MINAS(PubChemCID162679241)。分子对接和MD模拟表明,MINAS(〜8.2kcal/mol)对TubastatinA(〜8.4kcal/mol)具有强大且可比的抑制潜力。MINAS和TubastatinA在HDAC10上具有相似的结合位点,包括Ser138、Ser140、Tyr183和Cys184。此外,MINAS与TubastatinA相比具有更好的ADMET曲线,具有较高的MRTD值和较低的毒性。总之,数据显示,N1-乙酰亚精胺水平在DOX诱导的乳腺癌细胞死亡过程中升高.此外,MINAS,一种N1-乙酰亚精胺模拟化合物,当与DOX等化疗联合使用时,可以作为潜在的抗癌药物进行研究。
    Cancer drug resistance is associated with metabolic adaptation. Cancer cells have been shown to implicate acetylated polyamines in adaptations during cell death. However, exploring the mimetic of acetylated polyamines as a potential anticancer drug is lacking. We performed intracellular metabolite profiling of human breast cancer MCF-7 cells treated with doxorubicin (DOX), a well known anticancer drug. A novel and in-house vertical tube gel electrophoresis assisted procedure followed by LC-HRMS analysis was employed to detect acetylated polyamines such as N1-acetylspermidine. We designed a mimetic N1-acetylspermidine (MINAS) which is a known substrate of histone deacetylase 10 (HDAC10). Molecular docking and molecular dynamics (MDs) simulations were used to evaluate the inhibitory potential of MINAS against HDAC10. The inhibitory potential and the ADMET profile of MINAS were compared to a known HDAC10 inhibitor Tubastatin A. N1-acetylspermidine, an acetylated form of polyamine, was detected intracellularly in MCF-7 cells treated with DOX over DMSO-treated MCF-7 cells. We designed and curated MINAS (PubChem CID 162679241). Molecular docking and MD simulations suggested the strong and comparable inhibitory potential of MINAS (-8.2 kcal/mol) to Tubastatin A (-8.4 kcal/mol). MINAS and Tubastatin A share similar binding sites on HDAC10, including Ser138, Ser140, Tyr183, and Cys184. Additionally, MINAS has a better ADMET profile compared to Tubastatin A, with a high MRTD value and lower toxicity. In conclusion, the data show that N1-acetylspermidine levels rise during DOX-induced breast cancer cell death. Additionally, MINAS, an N1-acetylspermidine mimetic compound, could be investigated as a potential anticancer drug when combined with chemotherapy like DOX.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号