The sensitivity of acute myeloid leukemia cells to cytarabine is increased by suppressing the expression of Heme oxygenase-1 and hypoxia-inducible factor 1-alpha.

IF 5.3 2区 医学 Q1 ONCOLOGY
Mohammad Sadeghi, Asma Moslehi, Hadiseh Kheiry, Fariba Karoon Kiani, Asieh Zarei, Atefeh Khodakarami, Vahid Karpisheh, Ali Masjedi, Badrossadat Rahnama, Mohammad Hojjat-Farsangi, Mortaza Raeisi, Mehdi Yousefi, Ali Akbar Movasaghpour Akbari, Farhad Jadidi-Niaragh
{"title":"The sensitivity of acute myeloid leukemia cells to cytarabine is increased by suppressing the expression of Heme oxygenase-1 and hypoxia-inducible factor 1-alpha.","authors":"Mohammad Sadeghi, Asma Moslehi, Hadiseh Kheiry, Fariba Karoon Kiani, Asieh Zarei, Atefeh Khodakarami, Vahid Karpisheh, Ali Masjedi, Badrossadat Rahnama, Mohammad Hojjat-Farsangi, Mortaza Raeisi, Mehdi Yousefi, Ali Akbar Movasaghpour Akbari, Farhad Jadidi-Niaragh","doi":"10.1186/s12935-024-03393-3","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Acute myeloid leukemia (AML), a malignancy Often resistant to common chemotherapy regimens (Cytarabine (Ara-c) + Daunorubicin (DNR)), is accompanied by frequent relapses. Many factors are involved in causing chemoresistance. Heme Oxygenase-1 (HO-1) and Hypoxia-Inducible Factor 1-alpha (HIF-1α) are two of the most well-known genes, reported to be overexpressed in AML and promote resistance against chemotherapy according to several studies. The main chemotherapy agent used for AML treatment is Ara-c. We hypothesized that simultaneous targeting of HO-1 and HIF-1α could sensitize AML cells to Ara-c.</p><p><strong>Method: </strong>In this study, we used our recently developed, Trans-Activator of Transcription (TAT) - Chitosan-Carboxymethyl Dextran (CCMD) - Poly Ethylene Glycol (PEG) - Nanoparticles (NPs), to deliver Ara-c along with siRNA molecules against the HO-1 and HIF-1α genes to AML primary cells (ex vivo) and cell lines including THP-1, KG-1, and HL-60 (in vitro). Subsequently, the effect of the single or combinational treatment on the growth, proliferation, apoptosis, and Reactive Oxygen Species (ROS) formation was evaluated.</p><p><strong>Results: </strong>The designed NPs had a high potential in transfecting cells with siRNAs and drug. The results demonstrated that treatment of cells with Ara-c elevated the generation of ROS in the cells while decreasing the proliferation potential. Following the silencing of HO-1, the rate of apoptosis and ROS generation in response to Ara-c increased significantly. While proliferation and growth inhibition were considerably evident in HIF-1α-siRNA-transfected-AML cells compared to cells treated with free Ara-c. We found that the co-inhibition of genes could further sensitize AML cells to Ara-c treatment.</p><p><strong>Conclusions: </strong>As far as we are aware, this study is the first to simultaneously inhibit the HO-1 and HIF-1α genes in AML using NPs. It can be concluded that HO-1 causes chemoresistance by protecting cells from ROS damage. Whereas, HIF-1α mostly exerts prolific and direct anti-apoptotic effects. These findings imply that simultaneous inhibition of HO-1 and HIF-1α can overcome Ara-c resistance and help improve the prognosis of AML patients.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3000,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11197338/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Cell International","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s12935-024-03393-3","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Acute myeloid leukemia (AML), a malignancy Often resistant to common chemotherapy regimens (Cytarabine (Ara-c) + Daunorubicin (DNR)), is accompanied by frequent relapses. Many factors are involved in causing chemoresistance. Heme Oxygenase-1 (HO-1) and Hypoxia-Inducible Factor 1-alpha (HIF-1α) are two of the most well-known genes, reported to be overexpressed in AML and promote resistance against chemotherapy according to several studies. The main chemotherapy agent used for AML treatment is Ara-c. We hypothesized that simultaneous targeting of HO-1 and HIF-1α could sensitize AML cells to Ara-c.

Method: In this study, we used our recently developed, Trans-Activator of Transcription (TAT) - Chitosan-Carboxymethyl Dextran (CCMD) - Poly Ethylene Glycol (PEG) - Nanoparticles (NPs), to deliver Ara-c along with siRNA molecules against the HO-1 and HIF-1α genes to AML primary cells (ex vivo) and cell lines including THP-1, KG-1, and HL-60 (in vitro). Subsequently, the effect of the single or combinational treatment on the growth, proliferation, apoptosis, and Reactive Oxygen Species (ROS) formation was evaluated.

Results: The designed NPs had a high potential in transfecting cells with siRNAs and drug. The results demonstrated that treatment of cells with Ara-c elevated the generation of ROS in the cells while decreasing the proliferation potential. Following the silencing of HO-1, the rate of apoptosis and ROS generation in response to Ara-c increased significantly. While proliferation and growth inhibition were considerably evident in HIF-1α-siRNA-transfected-AML cells compared to cells treated with free Ara-c. We found that the co-inhibition of genes could further sensitize AML cells to Ara-c treatment.

Conclusions: As far as we are aware, this study is the first to simultaneously inhibit the HO-1 and HIF-1α genes in AML using NPs. It can be concluded that HO-1 causes chemoresistance by protecting cells from ROS damage. Whereas, HIF-1α mostly exerts prolific and direct anti-apoptotic effects. These findings imply that simultaneous inhibition of HO-1 and HIF-1α can overcome Ara-c resistance and help improve the prognosis of AML patients.

通过抑制血红素加氧酶-1 和缺氧诱导因子 1-α 的表达,可提高急性髓性白血病细胞对阿糖胞苷的敏感性。
背景:急性髓性白血病(AML急性髓性白血病(AML)是一种常对普通化疗方案(阿糖胞苷(Ara-c)+多柔比星(DNR))产生耐药性的恶性肿瘤,经常复发。导致化疗耐药性的因素有很多。血红素加氧酶-1(HO-1)和缺氧诱导因子 1-α(HIF-1α)是其中两个最著名的基因,多项研究表明,它们在急性髓细胞性白血病中过度表达,并促进化疗耐药性的产生。治疗急性髓细胞白血病的主要化疗药物是 Ara-c。我们假设,同时靶向 HO-1 和 HIF-1α 可使 AML 细胞对 Ara-c 敏感:在这项研究中,我们使用最近开发的转录激活剂(TAT)-壳聚糖-羧甲基葡聚糖(CCMD)-聚乙二醇(PEG)-纳米颗粒(NPs),将Ara-c与针对HO-1和HIF-1α基因的siRNA分子一起递送到AML原代细胞(体内外)和细胞系(体外),包括THP-1、KG-1和HL-60。随后,评估了单一或组合处理对细胞生长、增殖、凋亡和活性氧(ROS)形成的影响:结果:所设计的 NPs 在用 siRNAs 和药物转染细胞方面具有很高的潜力。结果表明,用 Ara-c 处理细胞会增加细胞中 ROS 的生成,同时降低细胞的增殖潜力。沉默HO-1后,细胞对Ara-c的凋亡率和ROS生成率显著增加。与用游离 Ara-c 处理的细胞相比,HIF-1α-siRNA 转染的AML 细胞的增殖和生长抑制更为明显。我们发现,基因的联合抑制可使 AML 细胞对 Ara-c 处理进一步敏感:据我们所知,这项研究是首次利用 NPs 同时抑制 AML 中的 HO-1 和 HIF-1α 基因。可以得出结论:HO-1通过保护细胞免受ROS损伤而导致化疗抵抗。而 HIF-1α 则主要发挥增殖和直接抗凋亡作用。这些发现意味着,同时抑制HO-1和HIF-1α可以克服Ara-c耐药性,有助于改善急性髓细胞白血病患者的预后。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
10.90
自引率
1.70%
发文量
360
审稿时长
1 months
期刊介绍: Cancer Cell International publishes articles on all aspects of cancer cell biology, originating largely from, but not limited to, work using cell culture techniques. The journal focuses on novel cancer studies reporting data from biological experiments performed on cells grown in vitro, in two- or three-dimensional systems, and/or in vivo (animal experiments). These types of experiments have provided crucial data in many fields, from cell proliferation and transformation, to epithelial-mesenchymal interaction, to apoptosis, and host immune response to tumors. Cancer Cell International also considers articles that focus on novel technologies or novel pathways in molecular analysis and on epidemiological studies that may affect patient care, as well as articles reporting translational cancer research studies where in vitro discoveries are bridged to the clinic. As such, the journal is interested in laboratory and animal studies reporting on novel biomarkers of tumor progression and response to therapy and on their applicability to human cancers.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信