4.7 Article

Genome-wide methylation sequencing identifies progression-related epigenetic drivers in myelodysplastic syndromes

期刊

CELL DEATH & DISEASE
卷 11, 期 11, 页码 -

出版社

SPRINGERNATURE
DOI: 10.1038/s41419-020-03213-2

关键词

-

资金

  1. National Natural Science Foundation of China [81900166, 81900163, 81970118, 81970156]
  2. Medical Innovation Team of Jiangsu Province [CXTDB2017002]
  3. Natural Science Foundation of Jiangsu Province for Youths [BK20180280]
  4. Zhenjiang Clinical Research Center of Hematology [SS2018009]
  5. Social Development Foundation of Zhenjiang [SH2017040, SH2018044, SH2019065, SH2019067, SH2020055]
  6. Scientific Research Project of The Fifth 169 Project of Zhenjiang [21]
  7. Youth Medical Talents Project of Ke Jiao Qiang Wei Project of Jiangsu Province [QNRC2016450]
  8. Medical Field of Zhenjiang Jin Shan Ying Cai Project
  9. Scientific Research Foundation of Affiliated People's Hospital of Jiangsu University for PhD [KFB202002]

向作者/读者索取更多资源

The potential mechanism of myelodysplastic syndromes (MDS) progressing to acute myeloid leukemia (AML) remains poorly elucidated. It has been proved that epigenetic alterations play crucial roles in the pathogenesis of cancer progression including MDS. However, fewer studies explored the whole-genome methylation alterations during MDS progression. Reduced representation bisulfite sequencing was conducted in four paired MDS/secondary AML (MDS/sAML) patients and intended to explore the underlying methylation-associated epigenetic drivers in MDS progression. In four paired MDS/sAML patients, cases at sAML stage exhibited significantly increased methylation level as compared with the matched MDS stage. A total of 1090 differentially methylated fragments (DMFs) (441 hypermethylated and 649 hypomethylated) were identified involving in MDS pathogenesis, whereas 103 DMFs (96 hypermethylated and 7 hypomethylated) were involved in MDS progression. Targeted bisulfite sequencing further identified that aberrant GFRA1, IRX1, NPY, and ZNF300 methylation were frequent events in an additional group of de novo MDS and AML patients, of which only ZNF300 methylation was associated with ZNF300 expression. Subsequently, ZNF300 hypermethylation in larger cohorts of de novo MDS and AML patients was confirmed by real-time quantitative methylation-specific PCR. It was illustrated that ZNF300 methylation could act as a potential biomarker for the diagnosis and prognosis in MDS and AML patients. Functional experiments demonstrated the anti-proliferative and pro-apoptotic role of ZNF300 overexpression in MDS-derived AML cell-line SKM-1. Collectively, genome-wide DNA hypermethylation were frequent events during MDS progression. Among these changes, ZNF300 methylation, a regulator of ZNF300 expression, acted as an epigenetic driver in MDS progression. These findings provided a theoretical basis for the usage of demethylation drugs in MDS patients against disease progression.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据