4.6 Article

Alterations in the methylome of the stromal tumour microenvironment signal the presence and severity of prostate cancer

期刊

CLINICAL EPIGENETICS
卷 12, 期 1, 页码 -

出版社

BMC
DOI: 10.1186/s13148-020-00836-2

关键词

Prostate cancer; Tumour microenvironment; Cancer-associated fibroblast; Stroma; Methylation; Field effect; EPIC microarray

资金

  1. Cancer Australia [1044458]
  2. National Health and Medical Research Council [1106870, SJC 1063559, GPR 1002648, 1102752, MGL 1035721]
  3. Cancer Institute of New South Wales [14/ECF/1-23]
  4. Prostate Cancer Foundation of Australia (Movember Young Investigator Grant) [YI0911]
  5. Cancer Council New South Wales [RG 18-09]
  6. Victorian Government through the Victorian Cancer Agency [MCRF15023, MCRF18017]
  7. Australian Government Research Training Program (RTP)
  8. RT Hall Trust
  9. TissuPath Pathology
  10. National Health and Medical Research Council of Australia [1106870, 1102752] Funding Source: NHMRC

向作者/读者索取更多资源

Background Prostate cancer changes the phenotype of cells within the stromal microenvironment, including fibroblasts, which in turn promote tumour progression. Functional changes in prostate cancer-associated fibroblasts (CAFs) coincide with alterations in DNA methylation levels at loci-specific regulatory regions. Yet, it is not clear how these methylation changes compare across CAFs from different patients. Therefore, we examined the consistency and prognostic significance of genome-wide DNA methylation profiles between CAFs from patients with different grades of primary prostate cancer. Results We used Infinium MethylationEPIC BeadChips to evaluate genome-wide DNA methylation profiles from 18 matched CAFs and non-malignant prostate tissue fibroblasts (NPFs) from men with moderate to high grade prostate cancer, as well as five unmatched benign prostate tissue fibroblasts (BPFs) from men with benign prostatic hyperplasia. We identified two sets of differentially methylated regions (DMRs) in patient CAFs. One set of DMRs reproducibly differed between CAFs and fibroblasts from non-malignant tissue (NPFs and BPFs). Indeed, more than 1200 DMRs consistently changed in CAFs from every patient, regardless of tumour grade. The second set of DMRs varied between CAFs according to the severity of the tumour. Notably, hypomethylation of the EDARADD promoter occurred specifically in CAFs from high-grade tumours and correlated with increased transcript abundance and increased EDARADD staining in patient tissue. Across multiple cohorts, tumours with low EDARADD DNA methylation and high EDARADD mRNA expression were consistently associated with adverse clinical features and shorter recurrence free survival. Conclusions We identified a large set of DMRs that are commonly shared across CAFs regardless of tumour grade and outcome, demonstrating highly consistent epigenome changes in the prostate tumour microenvironment. Additionally, we found that CAFs from aggressive prostate cancers have discrete methylation differences compared to CAFs from moderate risk prostate cancer. Together, our data demonstrates that the methylome of the tumour microenvironment reflects both the presence and the severity of the prostate cancer and, therefore, may provide diagnostic and prognostic potential.

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

4.6
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