Low-dose hypomethylating agents cooperate with ferroptosis inducers to enhance ferroptosis by regulating the DNA methylation-mediated MAGEA6-AMPK-SLC7A11-GPX4 signaling pathway in acute myeloid leukemia.

IF 9.4 1区 医学 Q1 HEMATOLOGY Experimental Hematology & Oncology Pub Date : 2024-02-20 DOI:10.1186/s40164-024-00489-4
Shuya Feng, Yigang Yuan, Zihan Lin, Min Li, Daijiao Ye, Liuzhi Shi, Danyang Li, Min Zhao, Chen Meng, Xiaofei He, Shanshan Wu, Fang Xiong, Siyu Ye, Junjun Yang, Haifeng Zhuang, Lili Hong, Shenmeng Gao
{"title":"Low-dose hypomethylating agents cooperate with ferroptosis inducers to enhance ferroptosis by regulating the DNA methylation-mediated MAGEA6-AMPK-SLC7A11-GPX4 signaling pathway in acute myeloid leukemia.","authors":"Shuya Feng, Yigang Yuan, Zihan Lin, Min Li, Daijiao Ye, Liuzhi Shi, Danyang Li, Min Zhao, Chen Meng, Xiaofei He, Shanshan Wu, Fang Xiong, Siyu Ye, Junjun Yang, Haifeng Zhuang, Lili Hong, Shenmeng Gao","doi":"10.1186/s40164-024-00489-4","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Ferroptosis is a new form of nonapoptotic and iron-dependent type of cell death. Glutathione peroxidase-4 (GPX4) plays an essential role in anti-ferroptosis by reducing lipid peroxidation. Although acute myeloid leukemia (AML) cells, especially relapsed and refractory (R/R)-AML, present high GPX4 levels and enzyme activities, pharmacological inhibition of GPX4 alone has limited application in AML. Thus, whether inhibition of GPX4 combined with other therapeutic reagents has effective application in AML is largely unknown.</p><p><strong>Methods: </strong>Lipid reactive oxygen species (ROS), malondialdehyde (MDA), and glutathione (GSH) assays were used to assess ferroptosis in AML cells treated with the hypomethylating agent (HMA) decitabine (DAC), ferroptosis-inducer (FIN) RAS-selective lethal 3 (RSL3), or their combination. Combination index (CI) analysis was used to assess the synergistic activity of DAC + RSL3 against AML cells. Finally, we evaluated the synergistic activity of DAC + RSL3 in murine AML and a human R/R-AML-xenografted NSG model in vivo.</p><p><strong>Results: </strong>We first assessed GPX4 expression and found that GPX4 levels were higher in AML cells, especially those with MLL rearrangements, than in NCs. Knockdown of GPX4 by shRNA and indirect inhibition of GPX4 enzyme activity by RSL3 robustly induced ferroptosis in AML cells. To reduce the dose of RSL3 and avoid side effects, low doses of DAC (0.5 µM) and RSL3 (0.05 µM) synergistically facilitate ferroptosis by inhibiting the AMP-activated protein kinase (AMPK)-SLC7A11-GPX4 axis. Knockdown of AMPK by shRNA enhanced ferroptosis, and overexpression of SLC7A11 and GPX4 rescued DAC + RSL3-induced anti-leukemogenesis. Mechanistically, DAC increased the expression of MAGEA6 by reducing MAGEA6 promoter hypermethylation. Overexpression of MAGEA6 induced the degradation of AMPK, suggesting that DAC inhibits the AMPK-SLC7A11-GPX4 axis by increasing MAGEA6 expression. In addition, DAC + RSL3 synergistically reduced leukemic burden and extended overall survival compared with either DAC or RSL3 treatment in the MLL-AF9-transformed murine model. Finally, DAC + RSL3 synergistically reduced viability in untreated and R/R-AML cells and extended overall survival in two R/R-AML-xenografted NSG mouse models.</p><p><strong>Conclusions: </strong>Our study first identify vulnerability to ferroptosis by regulating MAGEA6-AMPK-SLC7A11-GPX4 signaling pathway. Combined treatment with HMAs and FINs provides a potential therapeutic choice for AML patients, especially for R/R-AML.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":9.4000,"publicationDate":"2024-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10877917/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Experimental Hematology & Oncology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s40164-024-00489-4","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"HEMATOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Ferroptosis is a new form of nonapoptotic and iron-dependent type of cell death. Glutathione peroxidase-4 (GPX4) plays an essential role in anti-ferroptosis by reducing lipid peroxidation. Although acute myeloid leukemia (AML) cells, especially relapsed and refractory (R/R)-AML, present high GPX4 levels and enzyme activities, pharmacological inhibition of GPX4 alone has limited application in AML. Thus, whether inhibition of GPX4 combined with other therapeutic reagents has effective application in AML is largely unknown.

Methods: Lipid reactive oxygen species (ROS), malondialdehyde (MDA), and glutathione (GSH) assays were used to assess ferroptosis in AML cells treated with the hypomethylating agent (HMA) decitabine (DAC), ferroptosis-inducer (FIN) RAS-selective lethal 3 (RSL3), or their combination. Combination index (CI) analysis was used to assess the synergistic activity of DAC + RSL3 against AML cells. Finally, we evaluated the synergistic activity of DAC + RSL3 in murine AML and a human R/R-AML-xenografted NSG model in vivo.

Results: We first assessed GPX4 expression and found that GPX4 levels were higher in AML cells, especially those with MLL rearrangements, than in NCs. Knockdown of GPX4 by shRNA and indirect inhibition of GPX4 enzyme activity by RSL3 robustly induced ferroptosis in AML cells. To reduce the dose of RSL3 and avoid side effects, low doses of DAC (0.5 µM) and RSL3 (0.05 µM) synergistically facilitate ferroptosis by inhibiting the AMP-activated protein kinase (AMPK)-SLC7A11-GPX4 axis. Knockdown of AMPK by shRNA enhanced ferroptosis, and overexpression of SLC7A11 and GPX4 rescued DAC + RSL3-induced anti-leukemogenesis. Mechanistically, DAC increased the expression of MAGEA6 by reducing MAGEA6 promoter hypermethylation. Overexpression of MAGEA6 induced the degradation of AMPK, suggesting that DAC inhibits the AMPK-SLC7A11-GPX4 axis by increasing MAGEA6 expression. In addition, DAC + RSL3 synergistically reduced leukemic burden and extended overall survival compared with either DAC or RSL3 treatment in the MLL-AF9-transformed murine model. Finally, DAC + RSL3 synergistically reduced viability in untreated and R/R-AML cells and extended overall survival in two R/R-AML-xenografted NSG mouse models.

Conclusions: Our study first identify vulnerability to ferroptosis by regulating MAGEA6-AMPK-SLC7A11-GPX4 signaling pathway. Combined treatment with HMAs and FINs provides a potential therapeutic choice for AML patients, especially for R/R-AML.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
在急性髓性白血病中,低剂量低甲基化药物与铁变态反应诱导剂合作,通过调节 DNA 甲基化介导的 MAGEA6-AMPK-SLC7A11-GPX4 信号通路来增强铁变态反应。
背景:铁凋亡是一种新的非凋亡和铁依赖型细胞死亡形式。谷胱甘肽过氧化物酶-4(GPX4)通过减少脂质过氧化在抗铁细胞凋亡中发挥着重要作用。虽然急性髓性白血病(AML)细胞,尤其是复发和难治性(R/R)-AML,存在较高的 GPX4 水平和酶活性,但单纯的 GPX4 药物抑制在 AML 中的应用有限。因此,结合其他治疗试剂抑制 GPX4 是否能有效地应用于急性髓细胞性白血病,在很大程度上还是未知数:方法:采用脂质活性氧(ROS)、丙二醛(MDA)和谷胱甘肽(GSH)检测法评估接受低甲基化剂(HMA)地西他滨(DAC)、铁变态反应诱导剂(FIN)RAS选择性致死3(RSL3)或它们的组合治疗的急性髓细胞的铁变态反应。联合指数(CI)分析用于评估 DAC + RSL3 对 AML 细胞的协同活性。最后,我们评估了 DAC + RSL3 在小鼠 AML 和人 R/R-AML 异种移植 NSG 模型中的协同活性:我们首先评估了GPX4的表达,发现AML细胞,尤其是MLL重排细胞中的GPX4水平高于NC细胞。通过 shRNA 敲除 GPX4 并用 RSL3 间接抑制 GPX4 酶的活性,可在 AML 细胞中强力诱导铁变态反应。为了减少RSL3的剂量并避免副作用,低剂量的DAC(0.5 µM)和RSL3(0.05 µM)通过抑制AMP激活蛋白激酶(AMPK)-SLC7A11-GPX4轴协同促进铁中毒。通过 shRNA 敲除 AMPK 可增强铁凋亡,而过表达 SLC7A11 和 GPX4 可挽救 DAC + RSL3 诱导的抗白血病生成。从机制上讲,DAC通过减少MAGEA6启动子的超甲基化来增加MAGEA6的表达。MAGEA6 的过表达诱导了 AMPK 的降解,这表明 DAC 通过增加 MAGEA6 的表达抑制了 AMPK-SLC7A11-GPX4 轴。此外,在 MLL-AF9 转化的小鼠模型中,与 DAC 或 RSL3 治疗相比,DAC + RSL3 能协同减轻白血病负担并延长总生存期。最后,在两种 R/R-AML 异种移植的 NSG 小鼠模型中,DAC + RSL3 协同降低了未经处理和 R/R-AML 细胞的存活率,并延长了总存活期:我们的研究通过调节 MAGEA6-AMPK-SLC7A11-GPX4 信号通路,首次发现了易受铁变态反应影响的细胞。HMAs和FINs的联合治疗为急性髓细胞白血病患者,尤其是R/R-AML患者提供了一种潜在的治疗选择。
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 去求助
来源期刊
CiteScore
12.60
自引率
7.30%
发文量
97
审稿时长
6 weeks
期刊介绍: Experimental Hematology & Oncology is an open access journal that encompasses all aspects of hematology and oncology with an emphasis on preclinical, basic, patient-oriented and translational research. The journal acts as an international platform for sharing laboratory findings in these areas and makes a deliberate effort to publish clinical trials with 'negative' results and basic science studies with provocative findings. Experimental Hematology & Oncology publishes original work, hypothesis, commentaries and timely reviews. With open access and rapid turnaround time from submission to publication, the journal strives to be a hub for disseminating new knowledge and discussing controversial topics for both basic scientists and busy clinicians in the closely related fields of hematology and oncology.
期刊最新文献
Correction: Efficacy of NKG2D CAR-T cells with IL-15/IL-15Rα signaling for treating Epstein-Barr virus-associated lymphoproliferative disorder Identifying ADGRG1 as a specific marker for tumor-reactive T cells in acute myeloid leukemia. Ubiquitin modification in the regulation of tumor immunotherapy resistance mechanisms and potential therapeutic targets. Polyunsaturated fatty acids promote M2-like TAM deposition via dampening RhoA-YAP1 signaling in the ovarian cancer microenvironment. Genetic factors, risk prediction and AI application of thrombotic diseases.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1