DOX-induced cardiotoxicity

  • 文章类型: Journal Article
    二甲双胍(Met)通过潜在的作用机制假设对阿霉素(Dox)诱导的心脏毒性的保护作用,具有未知的可靠性和可信度。
    本研究旨在研究Met对Dox诱导的心脏毒性的保护作用及其潜在的作用机制,以及检查他们的可靠性和可信度。
    在PubMed中进行了全面搜索,Embase,WebofScience,科学直接,Scopus,和CNKI数据库从成立到2023年12月31日。评估Met对Dox诱导的心脏毒性的功效的动物实验包括在该研究中。主要疗效结果是心肌损伤的标志物。使用连续变量的标准化平均差来测量效应大小。使用Stata18统计软件包中的随机效应模型汇总数据。
    21项研究包括用Dox治疗的203-208只动物和用Dox和Met治疗的271-276只动物。质量评估显示出高质量的分数。汇总结果有利于基于血清乳酸脱氢酶(LDH)的Met治疗,肌酸激酶-心肌带(CK-MB),心肌肌钙蛋白I(cTnI),和天冬氨酸转氨酶水平。使用留一法的敏感性分析显示出稳定的结果。漏斗地块,Egger\'stest,和Begg的测试证实了潜在的出版偏见。基于大量证据,对氧化应激假说进行了广泛的研究。
    Met对于防止Dox引起的心脏毒性是有效且安全的,从而使其成为临床研究的合适药物。作用机制的氧化应激假说已经建立,具有最高的可靠性和可信度。
    UNASSIGNED: The protective effects of metformin (Met) against doxorubicin (Dox)-induced cardiotoxicity via potential hypotheses of mechanisms of action with unknown reliability and credibility.
    UNASSIGNED: This study aimed to investigate the protective effects of Met against Dox-induced cardiotoxicity and the underlying mechanisms of action, as well as examine their reliability and credibility.
    UNASSIGNED: A comprehensive search was conducted within the PubMed, Embase, Web of Science, Science Direct, Scopus, and CNKI databases from inception to 31 December 2023. Animal experiments evaluating the efficacy of Met against Dox-induced cardiotoxicity were included in this study. The primary efficacy outcomes were markers of myocardial injury. Effect size was measured using the standardized mean difference for continuous variables. Data were pooled using a random-effects model in the Stata 18 statistical software package.
    UNASSIGNED: Twenty-one studies involving 203-208 animals treated with Dox and 271-276 animals treated with Dox and Met were included in this analysis. Quality assessment revealed high-quality scores. Pooled results favored Met treatment based on the serum lactate dehydrogenase (LDH), creatine kinase-myocardial band (CK-MB), cardiac troponin I (cTnI), and aspartate aminotransferase levels. Sensitivity analysis using the leave-one-out method demonstrated stable results. Funnel plots, Egger\'s test, and Begg\'s test confirmed potential publication bias. The oxidative stress hypothesis has been investigated extensively based on abundant evidence.
    UNASSIGNED: Met is effective and safe for protecting against Dox-induced cardiotoxicity, thus making it an appropriate drug for clinical investigation. The oxidative stress hypothesis of mechanism of action is well established with highest reliability and credibility.
    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:阿霉素(Dox)是各种癌症的有效化疗药物,但其临床应用受到严重心脏毒性的限制。Dox治疗可以转录激活心肌细胞中多个心脏毒性相关基因,这种全球基因激活的潜在机制仍然知之甚少。
    结果:这里,我们整合了动物模型的数据,Dox处理后的CUT&Tag和RNA-seq,并且发现在Dox处理后心肌细胞中H3K27ac(与基因激活相关的组蛋白修饰)的水平显著增加。C646,组蛋白乙酰转移酶的抑制剂,逆转Dox诱导的H3K27ac在心肌细胞中的积累,随后阻止了Dox诱导的DNA损伤和细胞凋亡的增加。此外,C646通过恢复射血分数和逆转分数缩短百分比来减轻Dox处理的小鼠的心脏功能障碍。此外,Dox治疗增加了包括Bax在内的多个心脏毒性基因启动子处的H3K27ac沉积,Fas和Bnip3,导致它们的上调。此外,H3K27ac在心脏毒性相关基因的沉积在整个基因组中表现出广泛的特征.基于H3K27ac的沉积和mRNA表达水平,预测了几个可能导致Dox诱导心脏毒性的潜在基因.最后,Dox治疗后H3K27ac调节的心脏毒性基因的上调在物种之间是保守的。
    结论:综合来看,Dox诱导的表观遗传修饰,特别是H3K27ac,作为分子开关,激活强大的心脏毒性相关基因,导致心肌细胞死亡和心脏功能障碍。这些发现为Dox诱导的心脏毒性与表观遗传调控之间的关系提供了新的见解。并确定H3K27ac是预防和治疗Dox引起的心脏毒性的潜在靶标。
    BACKGROUND: Doxorubicin (Dox) is an effective chemotherapeutic drug for various cancers, but its clinical application is limited by severe cardiotoxicity. Dox treatment can transcriptionally activate multiple cardiotoxicity-associated genes in cardiomyocytes, the mechanisms underlying this global gene activation remain poorly understood.
    RESULTS: Herein, we integrated data from animal models, CUT&Tag and RNA-seq after Dox treatment, and discovered that the level of H3K27ac (a histone modification associated with gene activation) significantly increased in cardiomyocytes following Dox treatment. C646, an inhibitor of histone acetyltransferase, reversed Dox-induced H3K27ac accumulation in cardiomyocytes, which subsequently prevented the increase of Dox-induced DNA damage and apoptosis. Furthermore, C646 alleviated cardiac dysfunction in Dox-treated mice by restoring ejection fraction and reversing fractional shortening percentages. Additionally, Dox treatment increased H3K27ac deposition at the promoters of multiple cardiotoxic genes including Bax, Fas and Bnip3, resulting in their up-regulation. Moreover, the deposition of H3K27ac at cardiotoxicity-related genes exhibited a broad feature across the genome. Based on the deposition of H3K27ac and mRNA expression levels, several potential genes that might contribute to Dox-induced cardiotoxicity were predicted. Finally, the up-regulation of H3K27ac-regulated cardiotoxic genes upon Dox treatment is conservative across species.
    CONCLUSIONS: Taken together, Dox-induced epigenetic modification, specifically H3K27ac, acts as a molecular switch for the activation of robust cardiotoxicity-related genes, leading to cardiomyocyte death and cardiac dysfunction. These findings provide new insights into the relationship between Dox-induced cardiotoxicity and epigenetic regulation, and identify H3K27ac as a potential target for the prevention and treatment of Dox-induced cardiotoxicity.
    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)介导的心脏毒性可加剧肿瘤患者的死亡率,但相关的药物治疗措施相对有限。Ferroptosis最近被认为是DOX诱导的心脏毒性的主要机制。Idebenone,一种新型的铁凋亡抑制剂,是一种广泛使用的临床药物。然而,其在DOX诱导的心脏毒性中的作用和病理机制尚不清楚。在这项研究中,我们证明了艾地苯醌对DOX诱导的心脏毒性的影响,并阐明了其潜在的机制。单次腹膜内注射DOX(15mg/kg)以建立DOX诱导的心脏毒性。结果表明,艾地苯醌由于具有调节急性DOX诱导的Fe2和ROS超负荷的能力,可明显减轻DOX诱导的心功能不全。导致铁性凋亡。CESTA和BLI进一步揭示了艾地苯醌的抗铁凋亡作用是由FSP1介导的。有趣的是,在DOX存在下,艾地苯醌增加FSP1蛋白水平,但不影响Fsp1mRNA水平。艾地苯醌可以在K355与FSP1蛋白形成稳定的氢键,这可能影响其与泛素的缔合。结果证实艾地苯醌通过抑制其泛素化降解来稳定FSP1蛋白水平。总之,这项研究证明艾地苯醌通过调节FSP1抑制铁凋亡来减轻DOX诱导的心脏毒性,使其成为接受DOX治疗的患者的潜在临床药物。
    Doxorubicin (DOX)-mediated cardiotoxicity can exacerbate mortality in oncology patients, but related pharmacotherapeutic measures are relatively limited. Ferroptosis was recently identified as a major mechanism of DOX-induced cardiotoxicity. Idebenone, a novel ferroptosis inhibitor, is a well-described clinical drug widely used. However, its role and pathological mechanism in DOX-induced cardiotoxicity are still unclear. In this study, we demonstrated the effects of idebenone on DOX-induced cardiotoxicity and elucidated its underlying mechanism. A single intraperitoneal injection of DOX (15 mg/kg) was administrated to establish DOX-induced cardiotoxicity. The results showed that idebenone significantly attenuated DOX-induced cardiac dysfunction due to its ability to regulate acute DOX-induced Fe2+ and ROS overload, which resulted in ferroptosis. CESTA and BLI further revealed that idebenone\'s anti-ferroptosis effect was mediated by FSP1. Interestingly, idebenone increased FSP1 protein levels but did not affect Fsp1 mRNA levels in the presence of DOX. Idebenone could form stable hydrogen bonds with FSP1 protein at K355, which may influence its association with ubiquitin. The results confirmed that idebenone stabilized FSP1 protein levels by inhibiting its ubiquitination degradation. In conclusion, this study demonstrates idebenone attenuated DOX-induced cardiotoxicity by inhibiting ferroptosis via regulation of FSP1, making it a potential clinical drug for patients receiving DOX 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
    黄芪桂枝五物汤(HQGZWWD)在治疗多种心血管疾病方面显示出广阔的潜力。本研究旨在阐明HQGZWWD治疗多柔比星(DOX)诱导的心肌损伤的分子基础和治疗作用。利用HQGZWWD的HPLC指纹图谱对活性成分进行分析。建立了DOX诱导的大鼠心肌损伤模型,使用超声心动图评价HQGZWWD的治疗效果,心肌酶水平,苏木精和伊红染色。网络药理学用于筛选治疗目标,进行蛋白质印迹和免疫组织化学以评估细胞焦亡水平。使用测定试剂盒测量氧化应激水平,使用透射电子显微镜检查线粒体损伤。建立了DOX诱导细胞损伤的体外模型,使用含有HQGZWWD和N-乙酰半胱氨酸(NAC)的血清进行治疗。使用测定试剂盒和DCFH-DA检测氧化应激水平,而细胞焦亡水平通过WB评估,免疫荧光,和ELISA测定。HQGZWWD改善DOX诱导的心肌损伤。网络药理学将IL-1β和IL-18确定为关键靶标。HQGZWWD下调炎症因子IL-1β和IL-18的蛋白水平,抑制GSDMD-NT的表达,同时抑制了Caspase-1,ASC,NLRP3和Caspase-11。此外,HQGZWWD抑制氧化应激,并且使用NAC作为氧化应激抑制剂导致H9C2细胞中GSDMD-NT蛋白的显着抑制。这些发现强调了HQGZWWD通过抑制氧化应激和抑制规范和非规范的焦变途径的心肌保护作用。
    Huangqi Guizhi Wuwu Decoction (HQGZWWD) has shown promising potential in treating various cardiovascular diseases. This study aimed to elucidate the molecular basis and therapeutic role of HQGZWWD in the treatment of doxorubicin (DOX)-induced myocardial injury. The HPLC fingerprint of HQGZWWD was used to analyze the active components. A DOX-induced myocardial damage rat model was developed, and the therapeutic effects of HQGZWWD were evaluated using echocardiography, myocardial enzyme levels, and hematoxylin and eosin staining. Network pharmacology was used to screen treatment targets, and western blotting and immunohistochemistry were performed to assess cellular pyroptosis levels. Oxidative stress levels were measured using assay kits, and mitochondrial damage was examined using transmission electron microscopy. An in vitro model of DOX-induced cell damage was established, and treatment was administered using serum containing HQGZWWD and N-acetylcysteine (NAC). Oxidative stress levels were detected using assay kits and DCFH-DA, whereas cellular pyroptosis levels were assessed through WB, immunofluorescence, and ELISA assays. HQGZWWD ameliorated DOX-induced myocardial injury. Network pharmacology identified IL-1β and IL-18 as crucial targets. HQGZWWD downregulated the protein levels of the inflammatory factors IL-1β and IL-18, inhibited the expression of GSDMD-NT, and simultaneously suppressed the synthesis of Caspase-1, ASC, NLRP3, and Caspase-11. Additionally, HQGZWWD inhibited oxidative stress, and the use of NAC as an oxidative stress inhibitor resulted in significant inhibition of the GSDMD-NT protein in H9C2 cells. These findings highlight the myocardial protective effects of HQGZWWD by inhibiting oxidative stress and suppressing both canonical and non-canonical pyroptotic pathways.
    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:阿霉素(DOX)诱导的心脏毒性(DIC)是其临床应用的主要障碍。探索减轻DIC的替代治疗分子或药物是必不可少的。WGX50,一种来自花椒的有机提取物,具有抗炎和抗氧化的生物活性,然而,其在DIC中的作用和机制尚不清楚。
    方法:我们在体外和体内建立了DOX诱导的心脏毒性模型。超声心动图和组织学分析用于确定小鼠心脏损伤的严重程度。心肌损伤标志物cTnT,CK-MB,ANP,BNP,和铁凋亡相关指标Fe2+,MDA,和GPX4使用ELISA测量,RT-qPCR,和蛋白质印迹分析。用透射电镜观察线粒体的形态。线粒体膜电位的水平,线粒体ROS,使用JC-1、MitoSOX™检测脂质ROS,和C11-BODIPY581/591探头。
    结果:我们的发现表明WGX50通过抑制线粒体ROS和铁凋亡来保护DOX诱导的心脏毒性。在体内,WGX50可有效缓解阿霉素引起的心功能不全,心脏损伤,纤维化,线粒体损伤,和氧化还原失衡。体外,WGX50通过降低线粒体膜电位水平和增加线粒体ATP产生来保持线粒体功能。此外,WGX50减少铁积累和线粒体ROS,增加GPX4表达,并调节脂质代谢以抑制DOX诱导的铁凋亡。
    结论:综合来看,WGX50通过线粒体ROS和铁凋亡途径保护DOX诱导的心脏毒性,这为WGX50作为心脏保护的有希望的候选药物提供了新的见解。
    Doxorubicin (DOX)-induced cardiotoxicity (DIC) is a major impediment to its clinical application. It is indispensable to explore alternative treatment molecules or drugs for mitigating DIC. WGX50, an organic extract derived from Zanthoxylum bungeanum Maxim, has anti-inflammatory and antioxidant biological activity, however, its function and mechanism in DIC remain unclear.
    We established DOX-induced cardiotoxicity models both in vitro and in vivo. Echocardiography and histological analyses were used to determine the severity of cardiac injury in mice. The myocardial damage markers cTnT, CK-MB, ANP, BNP, and ferroptosis associated indicators Fe2+, MDA, and GPX4 were measured using ELISA, RT-qPCR, and western blot assays. The morphology of mitochondria was investigated with a transmission electron microscope. The levels of mitochondrial membrane potential, mitochondrial ROS, and lipid ROS were detected using JC-1, MitoSOX™, and C11-BODIPY 581/591 probes.
    Our findings demonstrate that WGX50 protects DOX-induced cardiotoxicity via restraining mitochondrial ROS and ferroptosis. In vivo, WGX50 effectively relieves doxorubicin-induced cardiac dysfunction, cardiac injury, fibrosis, mitochondrial damage, and redox imbalance. In vitro, WGX50 preserves mitochondrial function by reducing the level of mitochondrial membrane potential and increasing mitochondrial ATP production. Furthermore, WGX50 reduces iron accumulation and mitochondrial ROS, increases GPX4 expression, and regulates lipid metabolism to inhibit DOX-induced ferroptosis.
    Taken together, WGX50 protects DOX-induced cardiotoxicity via mitochondrial ROS and the ferroptosis pathway, which provides novel insights for WGX50 as a promising drug candidate for cardioprotection.
    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
    通过磁性操作对细胞进行图案化以创建三维(3D)细胞培养模型是一种有前途的技术,这是快速的,简单,和成本效益。这项研究介绍了一种新的生物图案化方法,该方法基于使用藻酸盐组成的生物墨水对细胞进行磁性操作,细胞,和磁性纳米粒子。Plackett-Burman和Box-Behnken实验设计模型用于优化其中NIH-3T3细胞用作模型细胞系的生物墨水制剂。通过7天培养时间通过光学显微镜确认图案化能力。然后,使用H9c2心肌细胞形成生物图案的3D心脏结构。细胞和细胞外成分,F-肌动蛋白和I型胶原蛋白,和心脏特异性生物标志物,成功观察到生物图案化的3D心脏结构的肌钙蛋白T和MYH6。此外,针对开发的3D模型研究了多柔比星(DOX)诱导的心脏毒性,对于生物图案化的3D心脏结构,IC50值计算为8.1μM,与传统的二维细胞培养相比,它对DOX暴露具有更高的抗性。特此,开发的生物图案化方法被证明是制造3D心脏模型的简单而快速的方法,特别是药物筛选应用。
    Impact statement Contactless manipulation and cell patterning techniques provide rapid and cost-effective three-dimensional (3D) cell culture model formation for tissue engineering applications. The present study introduces a new methodology that comprised alginate-based bioink to pattern cells via contactless magnetic manipulation to fabricate 3D cardiac structures. The developed cardiac model was evaluated in terms of Doxorubicin-induced cardiotoxicity and biopatterned 3D cardiac structures were found more resistant to drug exposure compared to two-dimensional control.
    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
    聚古洛糖醛酸(PG),一种来自藻酸盐的多糖,具有优异的生物活性。我们通过酸水解制备了具有10.41kDa分子量(Mw)和59平均聚合度(DP)的高纯度PG,三个pH等级,Q-Sepharose柱洗脱,和SephadexG-25柱脱盐。然后,我们在体外和体内评估了PG对阿霉素诱导的心脏毒性(DIC)的保护作用。无毒的PG增强了细胞活力,减少的细胞焦亡形态,LDH和IL-1β释放减少,和下调ASC寡聚化的表达,NLRP3、cl-CASP1和GSDMD,在阿霉素刺激的HL-1细胞和C57BL/6J小鼠中,PG保护心肌细胞免受NLRP3炎性体介导的焦亡。可能的潜在机制可能是PG下调阿霉素诱导的Peli1,其缺乏可以抑制阿霉素诱导的NLRP3炎性体介导的焦亡。这些结果表明,来自藻酸盐的多糖PG可以预防DIC,并且可能是预防DIC的潜在治疗剂或生物活性材料。
    Polyguluronic acid (PG), a polysaccharide from alginate, possesses excellent bioactivities. We prepared high-purity PG with 10.41 kDa molecular weight (Mw) and a 59 average degree of polymerization (DP) by acid hydrolysis, three pH grades, Q-Sepharose column elution, and Sephadex G-25 column desalination. Then, we evaluated the PG protective effects on doxorubicin-induced cardiotoxicity (DIC) in vitro and in vivo. The nontoxic PG enhanced cellular viability, reduced cell pyroptosis morphology, diminished the LDH and IL-1β release, and downregulated expressions of ASC oligomerization, NLRP3, cl-CASP1, and GSDMD, by which PG protected the cardiomyocytes from NLRP3 inflammasome-mediated pyroptosis in doxorubicin-stimulated HL-1 cells and C57BL/6J mice. The probable underlying mechanism may be that PG downregulated doxorubicin -induced Peli1, the deficiency of which could inhibit doxorubicin-induced NLRP3 inflammasome-mediated pyroptosis. These results suggested that polysaccharide PG from alginate could prevent DIC and may be a potential therapeutic agent or bioactive material for preventing 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
    多柔比星(DOX)是一种高效的化疗药物,但长期使用会引起心脏毒性和耐药性。越来越多的证据表明p53直接参与DOX的毒性和耐药性。DOX抗性的主要原因之一是p53的突变或失活。此外,因为由DOX引起的p53的非特异性激活可以杀死非癌细胞,p53是降低毒性的流行靶标。然而,通过p53抑制减少DOX诱导的心脏毒性(DIC)通常与p53再激活的抗肿瘤优势不一致.因此,为了提高DOX的有效性,由于p53基因复杂的调控网络和多态性,迫切需要探索p53靶向的抗癌策略.在这次审查中,我们总结了p53在DIC和耐药中的作用和潜在机制。此外,我们专注于应用膳食营养素的进步和挑战,天然产品,和其他药理学策略来克服DOX诱导的化学抗性和心脏毒性。最后,我们提出了解决关键问题的潜在治疗策略,以期为增加DOX的临床使用和提高其抗癌益处提供新思路.
    Doxorubicin (DOX) is a highly effective chemotherapeutic drug, but its long-term use can cause cardiotoxicity and drug resistance. Accumulating evidence demonstrates that p53 is directly involved in DOX toxicity and resistance. One of the primary causes for DOX resistance is the mutation or inactivation of p53. Moreover, because the non-specific activation of p53 caused by DOX can kill non-cancerous cells, p53 is a popular target for reducing toxicity. However, the reduction in DOX-induced cardiotoxicity (DIC) via p53 suppression is often at odds with the antitumor advantages of p53 reactivation. Therefore, in order to increase the effectiveness of DOX, there is an urgent need to explore p53-targeted anticancer strategies owing to the complex regulatory network and polymorphisms of the p53 gene. In this review, we summarize the role and potential mechanisms of p53 in DIC and resistance. Furthermore, we focus on the advances and challenges in applying dietary nutrients, natural products, and other pharmacological strategies to overcome DOX-induced chemoresistance and cardiotoxicity. Lastly, we present potential therapeutic strategies to address key issues in order to provide new ideas for increasing the clinical use of DOX and improving its anticancer benefits.
    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
    Ferritinophagy是影响心脏功能的最新分子机制之一。此外,这是阿霉素的途径之一,常用的抗癌药物之一,对心肌产生负面影响,导致心脏功能受损。这种副作用限制了阿霉素的使用。铁螯合剂在阻碍铁细胞吞噬中起重要作用。抗氧化剂还可以通过改善氧化应激来影响铁细胞吞噬。在这项研究中,据推测,褪黑素的抗氧化功能可以促进去铁胺的作用,一种铁螯合剂,在铁氧体吞噬的水平。将42只雄性Wistar大鼠(150-200g)分为7组(n=6),其中包括I组:正常对照,第二组:多柔比星(Dox),第三组:褪黑素(Mel),第四组:去铁胺(Des),组V:Mel+Dox,VI组:Des+Dox,和VII组:Mel+Des+Dox。第三组,V和VII用褪黑激素20mg/kg/天口服预处理7天。第四组,在给予Dox之前,在D4上用250mg/kg/剂量的去铁胺处理VI和VII一次。阿霉素以20mg/kgip单剂量给予。第8天,轻度麻醉大鼠进行心电图分析和超声心动图检查.收集血清样品,然后处死用于组织取样。进行了以下生化评估:NCOA4,IREB2,FTH1,SLC7A11和GPX4的PCR;和血清cTnI的ELISA,血清转铁蛋白,组织GSH,还有丙二醛.此外,心脏损伤的组织病理学评估;caspase-3,Bax,和Bcl2;并通过ECG和ECHO进行生理功能评估。多柔比星诱导的急性显著心脏损伤,铁细胞自噬和凋亡增加,对单一和联合预防性治疗有反应,其中联合治疗大多表现出最佳效果。总之,使用褪黑激素作为抗氧化剂和铁螯合剂,去铁胺,可能会阻碍阿霉素的有害心脏毒性作用。然而,需要进一步的研究来检测高剂量褪黑素和去铁胺对延长治疗期的影响.
    Ferritinophagy is one of the most recent molecular mechanisms affecting cardiac function. In addition, it is one of the pathways by which doxorubicin, one of the anticancer drugs commonly used, negatively impacts the cardiac muscle, leading to cardiac function impairment. This side effect limits the use of doxorubicin. Iron chelators play an important role in hindering ferritinophagy. Antioxidants can also impact ferritinophagy by improving oxidative stress. In this study, it was assumed that the antioxidant function of melatonin could promote the action of deferoxamine, an iron chelator, at the level of ferritinophagy. A total of 42 male Wistar rats (150-200 g) were divided into seven groups (n = 6) which consisted of group I: control normal, group II: doxorubicin (Dox), group III: melatonin (Mel), group IV: deferoxamine (Des), group V: Mel + Dox, group VI: Des + Dox, and group VII: Mel + Des + Dox. Groups III, V and VII were orally pretreated with melatonin 20 mg/kg/day for 7 days. Groups IV, VI and VII were treated with deferoxamine at a 250 mg/kg/dose once on D4 before Dox was given. Doxorubicin was given at a 20 mg/kg ip single dose. On the 8th day, the rats were lightly anaesthetized for electrocardiography analysis and echocardiography. Serum samples were collected and then sacrificed for tissue sampling. The following biochemical assessments were carried out: PCR of NCOA4, IREB2, FTH1, SLC7A11, and GPX4; and ELISA for serum cTnI, serum transferrin, tissue GSH, and malondialdehyde. In addition, histopathological assessment of heart injury; immunostaining of caspase-3, Bax, and Bcl2; and physiological function assessment by ECG and ECHO were carried out. Doxorubicin-induced acute significant cardiac injury with increased ferritinophagy and apoptosis responded to single and combined prophylactic treatment, in which the combined treatment showed mostly the best results. In conclusion, using melatonin as an antioxidant with an iron chelator, deferoxamine, could hinder the hazardous cardiotoxic effect of doxorubicin. However, further studies are needed to detect the impact of higher doses of melatonin and deferoxamine with a prolonged treatment period.
    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
    Doxorubicin (DOX) is widely used in the treatment of various cancers, increasing the great risk of adverse cardiovascular events, while the clinical intervention effect is not ideal. Histamine has been documented to participate in pathophysiological processes of cardiovascular diseases and inflammation-associated carcinogenesis. However, the potential roles of histamine in antitumor-related cardiotoxicity have not been fully elucidated. In this study, cardiomyocytes (hiPSC-CMs, HL-1 cells) and mice were treated with DOX to establish DOX-induced cardiotoxicity (DIC) models. Histidine decarboxylase knockout mice (HDC-/-) mice and histamine 1 receptor (H1R) antagonist were used to explore the effect of histamine/H1R signaling on DIC. Our results demonstrated that histamine deficiency or pharmaceutical inhibition of H1R accelerated myocardial ferroptosis, which is responsible for the aggravated DIC both in vivo and in vitro, while the supplementation of exogenous histamine reversed these changes. Our data revealed that the dysfunction of histamine/H1R signaling repressed the activation of transducer and activator of transcription 3 (STAT3), accompanying with decreased expression of solute carrier family7member11 (SLC7A11), a major modulator of ferroptosis. Conclusively, the disruption of histamine/H1R axis triggered ferroptosis and exacerbated DIC possibly by modulating STAT3-SLC7A11 pathway. Our findings point to a potential therapeutic target for DIC and provide more consideration on the usage of antihistamine drugs.
    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