Identification of genes underlying different methylation profiles in refractory anemia with excess blast and refractory cytopenia with multilineage dysplasia in myelodysplastic syndrome.

The Korean Journal of Hematology Pub Date : 2012-09-01 Epub Date: 2012-09-25 DOI:10.5045/kjh.2012.47.3.186
Suee Lee, Hyuk-Chan Kwon, Sung-Hyun Kim, Sung Yong Oh, Ji Hyun Lee, Yeon-Su Lee, Daekwan Seo, Jin-Yeong Han, Hyo-Jin Kim
{"title":"Identification of genes underlying different methylation profiles in refractory anemia with excess blast and refractory cytopenia with multilineage dysplasia in myelodysplastic syndrome.","authors":"Suee Lee,&nbsp;Hyuk-Chan Kwon,&nbsp;Sung-Hyun Kim,&nbsp;Sung Yong Oh,&nbsp;Ji Hyun Lee,&nbsp;Yeon-Su Lee,&nbsp;Daekwan Seo,&nbsp;Jin-Yeong Han,&nbsp;Hyo-Jin Kim","doi":"10.5045/kjh.2012.47.3.186","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Myelodysplastic syndrome (MDS) is a preleukemic condition that transforms into acute myeloid leukemia. However, the genetic events underlying this transformation remain poorly understood. Aberrant DNA methylation may play a causative role in the disease and its prognosis. Thus, we compared the DNA methylation profiles in refractory anemia with excess blast (RAEB) to those in refractory cytopenia with multilineage dysplasia (RCMD).</p><p><strong>Methods: </strong>Bone marrow samples were collected from 20 patients with primary MDS (9 with RAEB and 11 with RCMD), and peripheral blood samples were collected from 4 healthy controls. These samples were assessed using a commercial whole genome-wide methylation assay. Methylation-specific polymerase chain reaction (PCR) was used to detect the methylation of candidate gene promoters in RAEB and RCMD.</p><p><strong>Results: </strong>Microarray data revealed significant hypermethylation in 69 genes within RAEB but not RCMD. Candidate genes were mapped to 5 different networks, and network 1 had the highest score due to its involvement in gene expression, cancer, and cell cycle. Five genes (GSTM5, BIK, CENPH, RERG, and ANGPTL2) were associated with malignant disease progression. Among them, the methylated promoter pairs of GSTM5 (55.5% and 20%), BIK (20% and 0%), and ANGPTL2 (44.4% and 10%) were observed more frequently in RAEB.</p><p><strong>Conclusion: </strong>DNA methylation of GSTM5, BIK, and ANGPTL2 may induce epigenetic silencing and contribute to the increasing blasts and resulting MDS progression; however, the functions of these genes were not determined. Further study focusing on epigenetic silencing using various detection modalities is required.</p>","PeriodicalId":23001,"journal":{"name":"The Korean Journal of Hematology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2012-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.5045/kjh.2012.47.3.186","citationCount":"8","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"The Korean Journal of Hematology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.5045/kjh.2012.47.3.186","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2012/9/25 0:00:00","PubModel":"Epub","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 8

Abstract

Background: Myelodysplastic syndrome (MDS) is a preleukemic condition that transforms into acute myeloid leukemia. However, the genetic events underlying this transformation remain poorly understood. Aberrant DNA methylation may play a causative role in the disease and its prognosis. Thus, we compared the DNA methylation profiles in refractory anemia with excess blast (RAEB) to those in refractory cytopenia with multilineage dysplasia (RCMD).

Methods: Bone marrow samples were collected from 20 patients with primary MDS (9 with RAEB and 11 with RCMD), and peripheral blood samples were collected from 4 healthy controls. These samples were assessed using a commercial whole genome-wide methylation assay. Methylation-specific polymerase chain reaction (PCR) was used to detect the methylation of candidate gene promoters in RAEB and RCMD.

Results: Microarray data revealed significant hypermethylation in 69 genes within RAEB but not RCMD. Candidate genes were mapped to 5 different networks, and network 1 had the highest score due to its involvement in gene expression, cancer, and cell cycle. Five genes (GSTM5, BIK, CENPH, RERG, and ANGPTL2) were associated with malignant disease progression. Among them, the methylated promoter pairs of GSTM5 (55.5% and 20%), BIK (20% and 0%), and ANGPTL2 (44.4% and 10%) were observed more frequently in RAEB.

Conclusion: DNA methylation of GSTM5, BIK, and ANGPTL2 may induce epigenetic silencing and contribute to the increasing blasts and resulting MDS progression; however, the functions of these genes were not determined. Further study focusing on epigenetic silencing using various detection modalities is required.

Abstract Image

Abstract Image

Abstract Image

鉴定骨髓增生异常综合征中难治性贫血伴细胞过多和难治性细胞减少伴多系发育不良的不同甲基化谱的基因。
背景:骨髓增生异常综合征(MDS)是一种转化为急性髓系白血病的白血病前期疾病。然而,这种转变背后的遗传事件仍然知之甚少。异常DNA甲基化可能在疾病及其预后中起致病作用。因此,我们比较了难治性贫血伴细胞过多(RAEB)和难治性细胞减少伴多系发育不良(RCMD)的DNA甲基化谱。方法:采集20例原发性MDS患者(9例合并RAEB, 11例合并RCMD)骨髓标本,4例健康对照者外周血标本。这些样本使用商业全基因组甲基化测定法进行评估。采用甲基化特异性聚合酶链反应(PCR)检测RAEB和RCMD候选基因启动子的甲基化。结果:微阵列数据显示RAEB中有69个基因显着高甲基化,而RCMD中没有。候选基因被映射到5个不同的网络中,网络1因其参与基因表达、癌症和细胞周期而得分最高。5个基因(GSTM5、BIK、CENPH、RERG和ANGPTL2)与恶性疾病进展相关。其中,GSTM5(55.5%和20%)、BIK(20%和0%)和ANGPTL2(44.4%和10%)的甲基化启动子对在RAEB中出现频率更高。结论:GSTM5、BIK和ANGPTL2的DNA甲基化可能诱导表观遗传沉默,并有助于细胞增加和MDS的进展;然而,这些基因的功能尚未确定。需要使用各种检测方式对表观遗传沉默进行进一步的研究。
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
0.00%
发文量
0
×
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