Doxorubicin (DOX)

阿霉素 ( DOX )
  • 文章类型: Journal Article
    化学抗性仍然是导致癌症复发的有效乳腺癌治疗的重大挑战。CRISPR指导的基因编辑成为通过重新编程肿瘤微环境来减少化学耐药性的强大工具。先前的研究表明,中草药提取物具有克服肿瘤化疗耐药性的巨大潜力。然而,由于其较差的肿瘤靶向性和体内耐久性,治疗效果通常是有限的。在这里,我们开发了一种肿瘤微环境响应性纳米平台(H-MnO2(ISLDOX)-PTPN2@HA,M(ID)PH)用于纳米草药和CRISPR共递送以降低化学抗性。通过异甘草素(ISL)与多柔比星(DOX)的治疗实现了协同肿瘤抑制作用,通过基于CRISPR的基因编辑增强,靶向蛋白酪氨酸磷酸酶非受体2型(PTPN2)以启动长期免疫疗法。用M(I+D)PH纳米颗粒处理后观察到有效的PTPN2消耗,这导致肿瘤组织中浸润淋巴细胞的募集和促炎细胞因子的增加。总的来说,我们的纳米颗粒平台为实现协同化疗和免疫疗法提供了多种技术,它为恶性肿瘤提供了有效的治疗选择。
    Chemoresistance remains a significant challenge for effective breast cancer treatment which leads to cancer recurrence. CRISPR-directed gene editing becomes a powerful tool to reduce chemoresistance by reprogramming the tumor microenvironment. Previous research has revealed that Chinese herbal extracts have significant potential to overcome tumor chemoresistance. However, the therapeutic efficacy is often limited due to their poor tumor targeting and in vivo durability. Here we have developed a tumor microenvironment responsive nanoplatform (H-MnO2(ISL + DOX)-PTPN2@HA, M(I + D)PH) for nano-herb and CRISPR codelivery to reduce chemoresistance. Synergistic tumor inhibitory effects were achieved by the treatment of isoliquiritigenin (ISL) with doxorubicin (DOX), which were enhanced by CRISPR-based gene editing to target protein tyrosine phosphatase non-receptor type 2 (PTPN2) to initiate long-term immunotherapy. Efficient PTPN2 depletion was observed after treatment with M(I + D)PH nanoparticles, which resulted in the recruitment of intratumoral infiltrating lymphocytes and an increase of proinflammatory cytokines in the tumor tissue. Overall, our nanoparticle platform provides a diverse technique for accomplishing synergistic chemotherapy and immunotherapy, which offers an effective treatment alternative for malignant neoplasms.
    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
    表皮生长因子受体(EGFR)在肿瘤进展和生存中的作用通常未得到充分发挥。其表达和/或失调与乳腺癌的疾病进展和不良患者预后以及耐药性相关。EGFR通常在乳腺癌中过度表达,尤其是三阴性乳腺癌(TNBC)。目前缺乏分子靶标。我们检查了EGFR抑制剂(EGFRi)与多柔比星(Dox)在雌激素阳性(ER)MCF-7和MDA-MB-231TNBC细胞系中的协同潜力。MDA-MB-231和MCF-7暴露于EGFRi产生的IC50为6.03µM和3.96µM,分别。Dox诱导MDA-MB-231(IC509.67μM)和MCF-7(IC501.4μM)细胞毒性。EGFRi-Dox的组合显着降低了MCF-7(0.46µM)和MBA-MB231(0.01µM)中的IC50。使用Bliss独立性模型证实了两种细胞系中的协同药物相互作用。在0.1-10µMEGFRi和Dox单一处理的MCF-7中发生促凋亡Caspase-3/7激活,而1μMDox对MDA-MB-231产生更有效的作用。EGFRi和Dox单独或组合下调MCF-7和MDA-MB-231中的EGFR基因表达(p<0.001)。这项研究证明了EGFRi与Dox产生协同相互作用的潜力,导致生长抑制增强,凋亡诱导,以及两种细胞系中EGFR的下调。
    The role of the epidermal growth factor receptor (EGFR) in tumor progression and survival is often underplayed. Its expression and/or dysregulation is associated with disease advancement and poor patient outcome as well as drug resistance in breast cancer. EGFR is often overexpressed in breast cancer and particularly triple-negative breast cancer (TNBC), which currently lacks molecular targets. We examined the synergistic potential of an EGFR inhibitor (EGFRi) in combination with doxorubicin (Dox) in estrogen-positive (ER+) MCF-7 and MDA-MB-231 TNBC cell lines. The exposure of MDA-MB-231 and MCF-7 to EGFRi produced an IC50s of 6.03 µM and 3.96 µM, respectively. Dox induced MDA-MB-231 (IC50 9.67 µM) and MCF-7 (IC50 1.4 µM) cytotoxicity. Combinations of EGFRi-Dox significantly reduced the IC50 in MCF-7 (0.46 µM) and MBA-MB 231 (0.01 µM). Synergistic drug interactions in both cell lines were confirmed using the Bliss independence model. Pro-apoptotic Caspase-3/7 activation occurred in MCF-7 at 0.1-10 µM of EGFRi and Dox single treatments, whilst 1 μM Dox yielded a more potent effect on MDA-MB-231. EGFRi and Dox individually and in combination downregulated the EGFR gene expression in MCF-7 and MDA-MB-231 (p < 0.001). This study demonstrates EGFRi\'s potential for eliciting synergistic interactions with Dox, causing enhanced growth inhibition, apoptosis induction, and downregulation of EGFR in both cell lines.
    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
    阿霉素(Dox)可以诱导心脏毒性,从而限制了这种强效药物的效用。在这里,该研究确定了N6-甲基腺苷(m6A)脱甲基酶脂肪量和肥胖相关蛋白(FTO)在Dox诱导的心力衰竭(HF)期间的焦亡和炎症中的作用机制.
    从HF患者收集血清样品用于检测FTO和toll样受体4(TLR4)的表达。选择Dox处理的H9C2心肌细胞进行体外HF建模,然后测量FTO和TLR4的表达。检测心肌细胞的活力,凋亡,NOD-的空间分布,含LRR和Pyrin结构域的蛋白3(NLRP3),和乳酸脱氢酶的水平,炎症因子,氧化应激标志物,和焦亡相关蛋白。检测mRNA的m6A水平。RNA免疫沉淀(RIP)和mRNA稳定性测量用于确定mRNA和蛋白质表达,和RNAm6A斑点印迹和甲基化RIP测定以检测m6A甲基化水平。免疫印迹法检测p-NF-κBp65和p-IκB-α的表达。
    在HF患者的血清中,FTO升高而TLR4降低。Dox处理降低了FTO表达并增加了H9C2细胞中的m6A甲基化水平和TLR4表达。FTO的过表达和TLR4的敲除减少细胞凋亡,细胞毒性,炎症,焦亡,氧化应激,NLRP3共同本地化,和Dox诱导的H9C2细胞中的荧光强度。机械上,FTO通过TLR4的m6A去甲基化导致YTHDF1与TLR4mRNA的结合活性降低,从而降低了TLR4,p-NF-κBp65和p-IκB-α的表达。TLR4敲低抵消FTO敲低对Dox诱导的H9C2细胞的影响。
    FTO通过阻断TLR4/NF-κB途径减轻Dox诱导的HF。
    UNASSIGNED: Doxorubicin (Dox) can induce cardiotoxicity, thereby restricting the utility of this potent drug. Herein, the study ascertained the mechanism of the N6-methyladenosine (m6A) demethylase fat mass and obesity-associated protein (FTO) in pyroptosis and inflammation during Dox-induced heart failure (HF).
    UNASSIGNED: Serum samples were collected from HF patients for detection of the expression of FTO and toll-like receptor 4 (TLR4). Dox-treated H9C2 cardiomyocytes were chosen for in vitro HF modeling, followed by measurement of FTO and TLR4 expression. Cardiomyocytes were detected for viability, apoptosis, spatial distribution of NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3), and the levels of lactic dehydrogenase, inflammatory factors, oxidative stress markers, and pyroptosis-related proteins. The m6A levels of mRNA were examined. RNA immunoprecipitation (RIP) and mRNA stability measurement were used to determine mRNA and protein expression, and RNA m6A dot blot and methylated-RIP assay were performed to detect m6A methylation levels. The expression of p-NF-κB p65 and p-IκB-α was measured by western blotting.
    UNASSIGNED: In the serum of HF patients, FTO was elevated while TLR4 was decreased. Dox treatment reduced FTO expression and increased m6A methylation levels and TLR4 expression in H9C2 cells. Overexpression of FTO and knockdown of TLR4 reduced apoptosis, cytotoxicity, inflammation, pyroptosis, oxidative stress, NLRP3 co-localization, and fluorescence intensity in Dox-induced H9C2 cells. Mechanistically, FTO resulted in reduced binding activity of YTHDF1 to TLR4 mRNA via m6A demethylation of TLR4, thus declining TLR4, p-NF-κB p65, and p-IκB-α expression. TLR4 knockdown counteracted the effects of FTO knockdown on Dox-induced H9C2 cells.
    UNASSIGNED: FTO alleviated Dox-induced HF by blocking the TLR4/NF-κB 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
    多柔比星(DOX)是一种广泛使用的化学治疗剂,可引起严重和频繁的心脏毒性,限制了其临床应用。尽管对DOX引起的心脏毒性进行了广泛的研究,仍然缺乏有效的治疗方法。有必要了解DOX诱导的心脏毒性的分子机制,并寻找新的治疗靶标而不牺牲其抗癌作用。线粒体被认为是由DOX引起的心脏毒性的主要目标。在DOX诱导的心脏毒性中,经常报道以线粒体裂变增加和线粒体融合抑制为特征的线粒体动力学失衡。这可能导致过量的ROS产生,能量代谢紊乱,细胞凋亡,和其他各种问题。此外,线粒体动力学障碍与肿瘤发生有关。令人惊讶的是,最近的研究表明,靶向线粒体动力学蛋白如DRP1和MFN2不仅可以防御DOX诱导的心脏毒性,而且可以增强或不损害其抗癌作用。在这里,我们总结了DOX引起的心脏损伤的线粒体动力学紊乱。此外,我们概述了目前针对线粒体动力学相关蛋白的药理学和非药理学干预措施,以减轻DOX引起的心脏损伤.
    Doxorubicin (DOX) is an extensively used chemotherapeutic agent that can cause severe and frequent cardiotoxicity, which limits its clinical application. Although there have been extensive researches on the cardiotoxicity caused by DOX, there is still a lack of effective treatment. It is necessary to understand the molecular mechanism of DOX-induced cardiotoxicity and search for new therapeutic targets which do not sacrifice their anticancer effects. Mitochondria are considered to be the main target of cardiotoxicity caused by DOX. The imbalance of mitochondrial dynamics characterized by increased mitochondrial fission and inhibited mitochondrial fusion is often reported in DOX-induced cardiotoxicity, which can result in excessive ROS production, energy metabolism disorders, cell apoptosis, and various other problems. Also, mitochondrial dynamics disorder is related to tumorigenesis. Surprisingly, recent studies show that targeting mitochondrial dynamics proteins such as DRP1 and MFN2 can not only defend against DOX-induced cardiotoxicity but also enhance or not impair the anticancer effect. Herein, we summarize mitochondrial dynamics disorder in DOX-induced cardiac injury. Furthermore, we provide an overview of current pharmacological and non-pharmacological interventions targeting proteins involved in mitochondrial dynamics to alleviate cardiac damage caused by DOX.
    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
    背景:骨肉瘤(OS)是最常见的原发性恶性骨肿瘤。然而,单药化疗对OS的疗效有限,并且经常遇到肿瘤耐药性。因此,我们设计并构建了光热疗法(PTT)联合化疗的综合治疗策略,并使用了表面包裹的血小板-骨肉瘤杂化膜(OPM),该杂化膜可延长循环时间并实现OS特异性靶向.
    结果:OPM用作壳结构,封装多个载药纳米核(BPQDs-DOX)并控制阿霉素(DOX)的释放速率。此外,近红外光照射加速DOX的释放,从而延长循环时间并实现光刺激响应释放。OPM封装系统提高了BPQD的稳定性,提高了光热转换效率,并增强PTT功效。体外和离体实验表明,BPQDs-DOX@OPM有效地将药物递送到肿瘤部位,具有延长的循环时间和特异性靶向,与单药化疗相比,具有优异的抗肿瘤活性。此外,这些实验证实了BPQDs-DOX@OPM的良好生物安全性。
    结论:与单药化疗相比,使用BPQDs-DOX@OPM的联合治疗提供了延长的循环时间,靶向给药,增强抗肿瘤活性,和高度的生物安全性,从而为OS的临床治疗提供了一种新的方法。
    BACKGROUND: Osteosarcoma (OS) is the most prevalent primary malignant bone tumor. However, single-agent chemotherapy exhibits limited efficacy against OS and often encounters tumor resistance. Therefore, we designed and constructed an integrated treatment strategy of photothermal therapy (PTT) combined with chemotherapy and used a surface-encapsulated platelet-osteosarcoma hybrid membrane (OPM) that enhances circulation time and enables OS-specific targeting.
    RESULTS: The OPM functions as a shell structure, encapsulating multiple drug-loaded nanocores (BPQDs-DOX) and controlling the release rate of doxorubicin (DOX). Moreover, near-infrared light irradiation accelerates the release of DOX, thereby extending circulation time and enabling photostimulation-responsive release. The OPM encapsulation system improves the stability of BPQDs, enhances their photothermal conversion efficiency, and augments PTT efficacy. In vitro and ex vivo experiments demonstrate that BPQDs-DOX@OPM effectively delivers drugs to tumor sites with prolonged circulation time and specific targeting, resulting in superior anti-tumor activity compared to single-agent chemotherapy. Furthermore, these experiments confirm the favorable biosafety profile of BPQDs-DOX@OPM.
    CONCLUSIONS: Compared to single-agent chemotherapy, the combined therapy using BPQDs-DOX@OPM offers prolonged circulation time, targeted drug delivery, enhanced anti-tumor activity, and high biosafety, thereby introducing a novel approach for the clinical treatment of OS.
    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
    乳腺癌化疗耐药的分子机制尚不清楚。与化学抗性相关的基因的鉴定对于更好地理解驱动抗性的分子过程至关重要。
    本研究利用阿霉素(或多柔比星)耐药MCF-7(MCF-7/ADR)及其亲本MCF-7细胞系的共表达网络分析,探讨乳腺癌耐药机制。使用GEO2R网络工具从从基因表达综合(GEO)数据库获得的两个微阵列数据集(GSE24460和GSE76540)中提取与阿霉素抗性相关的基因。选择共表达网络中具有最高程度和/或介数的候选差异表达基因(DEGs)用于进一步分析。使用qRT-PCR实验验证了主要DEGs的表达。
    我们在MCF-7/ADR中确定了与其亲本MCF-7细胞系相比的十二个DEG,包括10个上调的DEG和2个下调的DEG。功能富集提示IGF2BPs和上皮-间质转化途径的RNA结合在乳腺癌耐药中的关键作用。
    我们的研究结果表明,MMP1,VIM,CNN3,LDHB,NEFH,PLS3、AKAP12、TCEAL2和ABCB1基因在多柔比星耐药中发挥重要作用,可以通过化学合成方法开发新型疗法。
    UNASSIGNED: The molecular mechanism of chemotherapy resistance in breast cancer is not well understood. The identification of genes associated with chemoresistance is critical for a better understanding of the molecular processes driving resistance.
    UNASSIGNED: This study used a co-expression network analysis of Adriamycin (or doxorubicin)-resistant MCF-7 (MCF-7/ADR) and its parent MCF-7 cell lines to explore the mechanisms of drug resistance in breast cancer. Genes associated with doxorubicin resistance were extracted from two microarray datasets (GSE24460 and GSE76540) obtained from the Gene Expression Omnibus (GEO) database using the GEO2R web tool. The candidate differentially expressed genes (DEGs) with the highest degree and/or betweenness in the co-expression network were selected for further analysis. The expression of major DEGs was validated experimentally using qRT-PCR.
    UNASSIGNED: We identified twelve DEGs in MCF-7/ADR compared with its parent MCF-7 cell line, including 10 upregulated and 2 downregulated DEGs. Functional enrichment suggests a key role for RNA binding by IGF2BPs and epithelial-to-mesenchymal transition pathways in drug resistance in breast cancer.
    UNASSIGNED: Our findings suggested that MMP1, VIM, CNN3, LDHB, NEFH, PLS3, AKAP12, TCEAL2, and ABCB1 genes play an important role in doxorubicin resistance and could be targeted for developing novel therapies by chemical synthesis approaches.
    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
    阿霉素(DOX),一种广泛用于癌症化疗的药物,通过多种细胞内相互作用诱导细胞死亡,产生诱导细胞凋亡的活性氧和DNA加合构型,拓扑异构酶II抑制,和希斯通驱逐。尽管它在实体瘤中具有广泛的治疗效果,DOX通常诱导耐药性和心脏毒性。由于低的细胞旁通透性和P-糖蛋白(P-gp)介导的外排,它显示出有限的肠吸收。我们回顾了各种肠胃外DOX制剂,如脂质体,聚合物胶束,聚合物纳米颗粒,和聚合物-药物缀合物,在临床使用或试验中增加其治疗效果。为了提高DOX在静脉和口腔癌治疗中的生物利用度,研究提出了一种对pH或氧化还原敏感的受体靶向系统,用于克服DOX耐药性并提高治疗效果,而不会引起DOX诱导的毒性。具有粘膜粘附性和通过紧密连接调节和P-gp抑制增加的肠通透性的DOX的多功能制剂也已在临床前阶段用作口服生物可利用的DOX。从静脉内制剂开发口服制剂的趋势日益明显,粘膜粘附技术的应用,渗透增强技术,功能性赋形剂的药代动力学调节可能有助于口服DOX的进一步发展。
    Doxorubicin (DOX), a widely used drug in cancer chemotherapy, induces cell death via multiple intracellular interactions, generating reactive oxygen species and DNA-adducted configurations that induce apoptosis, topoisomerase II inhibition, and histone eviction. Despite its wide therapeutic efficacy in solid tumors, DOX often induces drug resistance and cardiotoxicity. It shows limited intestinal absorption because of low paracellular permeability and P-glycoprotein (P-gp)-mediated efflux. We reviewed various parenteral DOX formulations, such as liposomes, polymeric micelles, polymeric nanoparticles, and polymer-drug conjugates, under clinical use or trials to increase its therapeutic efficacy. To improve the bioavailability of DOX in intravenous and oral cancer treatment, studies have proposed a pH- or redox-sensitive and receptor-targeted system for overcoming DOX resistance and increasing therapeutic efficacy without causing DOX-induced toxicity. Multifunctional formulations of DOX with mucoadhesiveness and increased intestinal permeability through tight-junction modulation and P-gp inhibition have also been used as orally bioavailable DOX in the preclinical stage. The increasing trends of developing oral formulations from intravenous formulations, the application of mucoadhesive technology, permeation-enhancing technology, and pharmacokinetic modulation with functional excipients might facilitate the further development of oral DOX.
    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
    我们旨在探讨依帕格列净在阿霉素(Dox)引起的心脏毒性中的功能和可能机制。首先,通过腹腔连续注射Dox建立心脏毒性大鼠模型。然后,将empagliflozin(30mg/kg)灌入大鼠体内。接下来,超声心动图用于检查大鼠的心脏功能,和H&E染色观察心肌组织的病理改变。此外,采用生化检测和酶联免疫吸附法检测肌酸激酶同工酶(CK-MB),N末端脑钠肽前体(NT-proBNP),三磷酸腺苷(ATP),二磷酸腺苷(ADP),和大鼠血清中的一磷酸腺苷(AMP)水平和超氧化物歧化酶(SOD),丙二醛(MDA),和心肌组织中的过氧化氢酶(CAT),分别。此外,Westernblot检测AMPK/SIRT-1/PGC-1α信号通路相关蛋白在心肌组织中的表达。持续腹腔注射Dox可显著升高左心室收缩末期内径(LVESD)和左心室舒张末期内径(LVEDD),缩短分数(FS)和射血分数(EF)降低,显著上调大鼠血清CK-MB和NT-proBNP水平,从而损害心脏功能。Empagliflozin治疗可以通过下调LVEDD和LVESD来改善心肌组织病理学损伤,减轻心功能和组织损伤,上调EF和FS,抑制血清CK-MB和NT-proBNP水平。此外,empagliflozin改善了Dox诱导的过度氧化应激和能量代谢失调。此外,依帕列净激活了Dox引起的心脏毒性大鼠的AMPK/SIRT-1/PGC-1α信号通路。总之,除了改善Dox引起的心脏组织和功能损伤,empagliflozin还可以改善过度的氧化应激和能量代谢。值得注意的是,依帕列净可能通过激活AMPK/SIRT-1/PGC-1α通路发挥心肌保护作用。
    We aimed to probe the functions and possible mechanisms of empagliflozin in doxorubicin (Dox)-caused cardiotoxicity. First, a cardiotoxicity rat model was built by continuously injecting Dox intraperitoneally. Then, empagliflozin (30 mg/kg) was gavaged into the rats. Next, echocardiography was utilized for checking the cardiac function of rats, and H&E staining for observing pathological alterations of the myocardial tissues. Besides, biochemical assays and Enzyme-linked Immunosorbent Assay were adopted to detect the creatine kinase isoenzyme (CK-MB), N-terminal pro-brain natriuretic peptide (NT-proBNP), adenosine triphosphate (ATP), adenosine diphosphate (ADP), and adenosine monophosphate (AMP) levels in rat serum and superoxide dismutase (SOD), malondialdehyde acid (MDA), and catalase (CAT) in myocardial tissue, respectively. Furthermore, the expression of AMPK/SIRT-1/PGC-1α signaling pathway-related proteins in the myocardial tissues was tested by Western blot. Continuous intraperitoneal injection of Dox greatly elevated left ventricular end-systolic diameter (LVESD) and left ventricular end-diastolic diameter (LVEDD), reduced fractional shortening (FS) and ejection fraction (EF), and notably up-regulated CK-MB and NT-proBNP level in rats\' serum, thus impairing cardiac function. Empagliflozin treatment could ameliorate myocardial histopathological damage and alleviate cardiac function and tissue damage by down-regulating LVEDD and LVESD, up-regulating EF and FS, and inhibiting CK-MB and NT-proBNP level in serum. Additionally, empagliflozin improved Dox-induced excessive oxidative stress and dysregulation of energy metabolism. Furthermore, empagliflozin activated the AMPK/SIRT-1/PGC-1α signaling pathway in Dox-caused cardiotoxicity rats. In conclusion, in addition to bettering the cardiac tissue and function injury caused by Dox, empagliflozin also improves excessive oxidative stress and energy metabolism. Notably, empagliflozin may exert cardioprotective effects through activating the AMPK/SIRT-1/PGC-1α 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
    未经证实:氧化应激和细胞凋亡在多柔比星(DOX)诱导的心肌损伤的发病机制中起关键作用。热休克蛋白衍生肽(HSP-17)是在DOX处理的小鼠心脏组织中低表达的肽。具有较高的生物活性和种间序列一致性,并预测具有心肌保护作用。
    未经批准:首先,我们在H9c2细胞培养基中添加1µMDOX,持续24小时,以构建心肌细胞毒性模型。然后我们通过测量细胞活力和乳酸脱氢酶(LDH)水平来检测HSP-17对DOX诱导的H9c2心肌细胞损伤的影响。此外,活性氧(ROS)和四乙基苯并咪唑基碳花青碘化物试剂盒用于评估HSP-17肽对DOX诱导的心肌细胞氧化应激损伤的影响,细胞凋亡相关蛋白检测和流式细胞术检测细胞凋亡水平。此外,蛋白质印迹法检测PI3K/Akt途径的蛋白质表达水平[磷酸化Akt(p-Akt)和磷酸化PI3K(p-PI3K)]。
    UNASSIGNED:我们发现HSP-17肽可以增加细胞活力,保护线粒体潜能,降低LDH水平,减少ROS和心肌细胞凋亡。此外,我们还观察到HSP-17上调p-Akt的表达水平,和LY294002,一种典型的PI3K/Akt抑制剂,发现消除了HSP-17的保护作用。
    未经批准:总而言之,这项研究表明,HSP-17肽通过PI3K/Akt通路激活保护H9c2细胞免受氧化应激和凋亡,为治疗DOX诱导的心肌损伤提供了新的思路。
    UNASSIGNED: Oxidative stress and cell apoptosis play pivotal roles in the pathogenesis of doxorubicin (DOX)-induced myocardial injury. Heat shock protein-derived peptide (HSP-17) is a peptide which is low-expressed in DOX treated mouse heart tissue. It has high bioactivity and interspecies sequence consistency, and is predicted to have myocardial protective effect.
    UNASSIGNED: Firstly, we added 1 µM DOX to H9c2 cell culture medium for 24 hours to construct the myocardial cytotoxicity model. Then we detected the effect of HSP-17 on DOX induced H9c2 cardiomyocyte injury by measuring cell viability and lactate dehydrogenase (LDH) level. In addition, reactive oxygen species (ROS) and tetraethylbenzimidazolylcarbocyanine iodide kits are used to evaluate the effect of the HSP-17 peptide on DOX-induced oxidative stress injury to cardiomyocytes, and the detection of apoptosis related proteins and flow cytometry were applied to detect the level of apoptosis. Furthermore, the protein expression levels [phosphorylated Akt (p-Akt) and phosphorylated PI3K (p-PI3K)] of the PI3K/Akt pathway were also detected by western blotting.
    UNASSIGNED: We found that the HSP-17 peptide can increase cell viability, protect mitochondrial potential, reduce LDH levels, and reduce ROS and cardiomyocyte apoptosis. In addition, we also observed that HSP-17 upregulated the expression level of p-Akt, and LY294002, a typical inhibitor of PI3K/Akt, was found to eliminate the protective roles of HSP-17.
    UNASSIGNED: In conclusion, this study demonstrated that the HSP-17 peptide protected H9c2 cells against oxidative stress and apoptosis via PI3K/Akt pathway activation, which provides a new idea for the treatment of DOX-induced myocardial injury.
    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
    自从它在1960年代被发现以来,多柔比星(DOX)不断引发最广泛的抗人类癌症活性。然而,直接由DOX及其代谢物引起的心脏毒性是在化疗中持续使用DOX引起关注的重大问题。虽然DOX诱导的心脏毒性的确切机制尚未完全了解,最近的研究表明氧化应激,炎症,和几种形式的细胞死亡是支撑多柔比星诱导的心脏毒性(DIC)病因的关键致病机制。值得注意的是,这些关键的机制事件被认为是由3,4-二羟基苯甲酸或原儿茶酸(PCA)负调控-一种基于植物的植物化学物质,具有经过验证的抗氧化剂,抗炎,和抗凋亡特性。这里,我们回顾了实验结果,详细说明了暴露于DOX下PCA的潜在改善作用。我们还讨论了DIC病理生理学的分子见解,强调了潜在的干预点,即使用PCA作为名副其实的化学保护剂可以改善DOX诱导的心脏毒性以及其他抗癌药物如顺铂引起的毒性.虽然我们承认在化疗期间控制口服PCA可能不足以消除由于DOX治疗引起的所有毒性,我们认为PCA阻断氧化应激的能力,减轻炎症,并废除几种形式的心肌细胞死亡,突显了其在改善DIC方面的巨大前景。
    Since its discovery in the 1960 s, doxorubicin (DOX) has constantly elicited the broadest spectrum of cancerocidal activity against human cancers. However, cardiotoxicity caused by DOX directly as well as its metabolites is a great source of concern over the continuous use of DOX in chemotherapy. While the exact mechanism of DOX-induced cardiotoxicity is yet to be completely understood, recent studies indicate oxidative stress, inflammation, and several forms of cell death as key pathogenic mechanisms that underpin the etiology of doxorubicin-induced cardiotoxicity (DIC). Notably, these key mechanistic events are believed to be negatively regulated by 3,4-dihydroxybenzoic acid or protocatechuic acid (PCA)-a plant-based phytochemical with proven anti-oxidant, anti-inflammatory, and anti-apoptotic properties. Here, we review the experimental findings detailing the potential ameliorative effects of PCA under exposure to DOX. We also discuss molecular insights into the pathophysiology of DIC, highlighting the potential intervention points where the use of PCA as a veritable chemoprotective agent may ameliorate DOX-induced cardiotoxicities as well as toxicities due to other anticancer drugs like cisplatin. While we acknowledge that controlled oral administration of PCA during chemotherapy may be insufficient to eliminate all toxicities due to DOX treatment, we propose that the ability of PCA to block oxidative stress, attenuate inflammation, and abrogate several forms of cardiomyocyte cell death underlines its great promise in the amelioration of DIC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号