Low expression of miR-182 caused by DNA hypermethylation accelerates acute lymphocyte leukemia development by targeting PBX3 and BCL2: miR-182 promoter methylation is a predictive marker for hypomethylation agents + BCL2 inhibitor venetoclax.

IF 4.8 2区 医学 Q1 GENETICS & HEREDITY
Danyang Li, Yigang Yuan, Chen Meng, Zihan Lin, Min Zhao, Liuzhi Shi, Min Li, Daijiao Ye, Yue Cai, Xiaofei He, Haige Ye, Shujuan Zhou, Haixia Zhou, Shenmeng Gao
{"title":"Low expression of miR-182 caused by DNA hypermethylation accelerates acute lymphocyte leukemia development by targeting PBX3 and BCL2: miR-182 promoter methylation is a predictive marker for hypomethylation agents + BCL2 inhibitor venetoclax.","authors":"Danyang Li, Yigang Yuan, Chen Meng, Zihan Lin, Min Zhao, Liuzhi Shi, Min Li, Daijiao Ye, Yue Cai, Xiaofei He, Haige Ye, Shujuan Zhou, Haixia Zhou, Shenmeng Gao","doi":"10.1186/s13148-024-01658-2","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>miR-182 promoter hypermethylation frequently occurs in various tumors, including acute myeloid leukemia, and leads to low expression of miR-182. However, whether adult acute lymphocyte leukemia (ALL) cells have high miR-182 promoter methylation has not been determined.</p><p><strong>Methods: </strong>To assess the methylation status of the miR-182 promoter, methylation and unmethylation-specific PCR analysis, bisulfite-sequencing analysis, and MethylTarget™ assays were performed to measure the frequency of methylation at the miR-182 promoter. Bone marrow cells were isolated from miR-182 knockout (182KO) and 182 wild type (182WT) mice to construct BCR-ABL (P190) and Notch-induced murine B-ALL and T-ALL models, respectively. Primary ALL samples were performed to investigate synergistic effects of the hypomethylation agents (HMAs) and the BCL2 inhibitor venetoclax (Ven) in vitro.</p><p><strong>Results: </strong>miR-182 (miR-182-5P) expression was substantially lower in ALL blasts than in normal controls (NCs) because of DNA hypermethylation at the miR-182 promoter in ALL blasts but not in normal controls (NCs). Knockout of miR-182 (182KO) markedly accelerated ALL development, facilitated the infiltration, and shortened the OS in a BCR-ABL (P190)-induced murine B-ALL model. Furthermore, the 182KO ALL cell population was enriched with more leukemia-initiating cells (CD43<sup>+</sup>B220<sup>+</sup> cells, LICs) and presented higher leukemogenic activity than the 182WT ALL population. Furthermore, depletion of miR-182 reduced the OS in a Notch-induced murine T-ALL model, suggesting that miR-182 knockout accelerates ALL development. Mechanistically, overexpression of miR-182 inhibited proliferation and induced apoptosis by directly targeting PBX3 and BCL2, two well-known oncogenes, that are key targets of miR-182. Most importantly, DAC in combination with Ven had synergistic effects on ALL cells with miR-182 promoter hypermethylation, but not on ALL cells with miR-182 promoter hypomethylation.</p><p><strong>Conclusions: </strong>Collectively, we identified miR-182 as a tumor suppressor gene in ALL cells and low expression of miR-182 because of hypermethylation facilitates the malignant phenotype of ALL cells. DAC + Ven cotreatment might has been applied in the clinical try for ALL patients with miR-182 promoter hypermethylation. Furthermore, the methylation frequency at the miR-182 promoter should be a potential biomarker for DAC + Ven treatment in ALL patients.</p>","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":4.8000,"publicationDate":"2024-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10964616/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Epigenetics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s13148-024-01658-2","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"GENETICS & HEREDITY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: miR-182 promoter hypermethylation frequently occurs in various tumors, including acute myeloid leukemia, and leads to low expression of miR-182. However, whether adult acute lymphocyte leukemia (ALL) cells have high miR-182 promoter methylation has not been determined.

Methods: To assess the methylation status of the miR-182 promoter, methylation and unmethylation-specific PCR analysis, bisulfite-sequencing analysis, and MethylTarget™ assays were performed to measure the frequency of methylation at the miR-182 promoter. Bone marrow cells were isolated from miR-182 knockout (182KO) and 182 wild type (182WT) mice to construct BCR-ABL (P190) and Notch-induced murine B-ALL and T-ALL models, respectively. Primary ALL samples were performed to investigate synergistic effects of the hypomethylation agents (HMAs) and the BCL2 inhibitor venetoclax (Ven) in vitro.

Results: miR-182 (miR-182-5P) expression was substantially lower in ALL blasts than in normal controls (NCs) because of DNA hypermethylation at the miR-182 promoter in ALL blasts but not in normal controls (NCs). Knockout of miR-182 (182KO) markedly accelerated ALL development, facilitated the infiltration, and shortened the OS in a BCR-ABL (P190)-induced murine B-ALL model. Furthermore, the 182KO ALL cell population was enriched with more leukemia-initiating cells (CD43+B220+ cells, LICs) and presented higher leukemogenic activity than the 182WT ALL population. Furthermore, depletion of miR-182 reduced the OS in a Notch-induced murine T-ALL model, suggesting that miR-182 knockout accelerates ALL development. Mechanistically, overexpression of miR-182 inhibited proliferation and induced apoptosis by directly targeting PBX3 and BCL2, two well-known oncogenes, that are key targets of miR-182. Most importantly, DAC in combination with Ven had synergistic effects on ALL cells with miR-182 promoter hypermethylation, but not on ALL cells with miR-182 promoter hypomethylation.

Conclusions: Collectively, we identified miR-182 as a tumor suppressor gene in ALL cells and low expression of miR-182 because of hypermethylation facilitates the malignant phenotype of ALL cells. DAC + Ven cotreatment might has been applied in the clinical try for ALL patients with miR-182 promoter hypermethylation. Furthermore, the methylation frequency at the miR-182 promoter should be a potential biomarker for DAC + Ven treatment in ALL patients.

DNA高甲基化导致的miR-182低表达通过靶向PBX3和BCL2加速急性淋巴细胞白血病的发展:miR-182启动子甲基化是低甲基化药物+BCL2抑制剂venetoclax的预测标记。
背景:miR-182启动子高甲基化经常发生在包括急性髓性白血病在内的各种肿瘤中,并导致miR-182的低表达。然而,成人急性淋巴细胞白血病(ALL)细胞是否存在高miR-182启动子甲基化尚未确定:为了评估 miR-182 启动子的甲基化状态,研究人员进行了甲基化和非甲基化特异性 PCR 分析、亚硫酸氢盐测序分析和 MethylTarget™ 检测,以测量 miR-182 启动子的甲基化频率。从 miR-182 基因敲除(182KO)和 182 野生型(182WT)小鼠身上分离骨髓细胞,分别构建 BCR-ABL (P190) 和 Notch 诱导的小鼠 B-ALL 和 T-ALL 模型。结果:由于ALL blasts中miR-182启动子的DNA超甲基化,ALL blasts中miR-182(miR-182-5P)的表达大大低于正常对照组(NCs),而正常对照组(NCs)则没有。在BCR-ABL(P190)诱导的小鼠B-ALL模型中,敲除miR-182(182KO)明显加速了ALL的发展、促进了浸润并缩短了OS。此外,与 182WT ALL 细胞群相比,182KO ALL 细胞群富含更多的白血病启动细胞(CD43+B220+ 细胞,LICs),并具有更高的致白血病活性。此外,在Notch诱导的小鼠T-ALL模型中,去掉miR-182会降低OS,这表明miR-182敲除会加速ALL的发展。从机理上讲,miR-182的过表达通过直接靶向PBX3和BCL2抑制增殖并诱导凋亡,而PBX3和BCL2是两个著名的致癌基因,是miR-182的关键靶点。最重要的是,DAC联合Ven对miR-182启动子高甲基化的ALL细胞有协同作用,但对miR-182启动子低甲基化的ALL细胞没有协同作用:总之,我们发现miR-182是ALL细胞中的肿瘤抑制基因,而miR-182因高甲基化而低表达会促进ALL细胞的恶性表型。DAC+Ven联合治疗可用于miR-182启动子高甲基化ALL患者的临床试验。此外,miR-182启动子的甲基化频率应成为DAC+Ven治疗ALL患者的潜在生物标志物。
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
自引率
5.30%
发文量
150
期刊介绍: Clinical Epigenetics, the official journal of the Clinical Epigenetics Society, is an open access, peer-reviewed journal that encompasses all aspects of epigenetic principles and mechanisms in relation to human disease, diagnosis and therapy. Clinical trials and research in disease model organisms are particularly welcome.
×
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