Elsevier

Gene

Volume 604, 10 March 2017, Pages 1-8
Gene

Research paper
DNA methylation contributes to deregulation of 12 cancer-associated microRNAs and breast cancer progression

https://doi.org/10.1016/j.gene.2016.12.018Get rights and content

Highlights

  • Hypermethylation of MIR-125b-1, -127, -132, -193a, and -34b was revealed in BC

  • Hypomethylation of MIR-191 was shown in primary breast tumors

  • MIR-124-1, 125b-1, 127, 132, 193a, 34b methylation correlated with down-regulation

  • Levels of miR-127-5p, -124-3p, -375 and DAPK1, BCL2, RASSF1(A) correlated negatively

  • Hypermethylation of MIR-127 and -125b-1 was associated with breast cancer progression

Abstract

The methylation of promoter CpG islands and the interaction between microRNAs (miRNAs) and messenger RNAs (mRNAs) of target genes are considered two crucial mechanisms for gene and pathway deregulation in malignant tumors. The aim of this study was to analyze the role of promoter methylation in altering the expression of 13 miRNAs that are associated with breast cancer (BC): miR-124, -125b, -127, -132, -137, -148a, -191, -193a, -203, -212, -34b, -375, -9. The role of methylation in the deregulation of these miRNAs has not been previously assessed in the representative set of BC samples. We used a set of 58 paired (tumor/normal) breast tissue samples and methylation-specific PCR to demonstrate significant aberrations in the methylation patterns of 9 miRNA genes. In particular, we observed hypermethylation of MIR-127, -132, and -193a, and hypomethylation of MIR-191 for the first time. Using quantitative PCR, we established a strong correlation between promoter methylation and expression levels for 12 miRNA genes (all except MIR-212); this finding demonstrates the functional importance of altered methylation patterns. We also performed a correlation analysis between expression levels of the 13 miRNAs and 5 cancer-associated genes, namely RASSF1(A), CHL1, APAF1, DAPK1, and BCL2, which were predicted as targets for these miRNAs, to investigate the impact of these miRNAs on these genes with key cellular functions in BC. Significant negative correlation was revealed for the following miRNA-mRNA pairs: miR-127-5p and DAPK1, miR-375 and RASSF1(A), and miR-124-3p and BCL2. Additionally, we also found a strong association between hypermethylation of MIR-127 and MIR-125b-1 and BC progression, particularly metastasis. Thus, our findings provide evidence for the significant role of methylation in the deregulation of 12 miRNA genes in BC, identify putative novel functional miRNA-mRNA pairs, and suggest MIR-127 and MIR-125b-1 hypermethylation to be potential biomarkers of BC metastasis.

Introduction

DNA methylation of regulatory sequences at the genomic level and interaction between microRNAs (miRNAs) and the messenger RNAs (mRNAs) of target genes at the posttranscriptional level contribute to the dynamic regulation of gene activity. Aberrations in these mechanisms can lead to impaired functioning of cell signaling pathways, such as that observed in malignant tumors (Lopez-Serra and Esteller, 2012, Hill et al., 2014). The proportion of aberrantly methylated miRNA gene promoters in cancer cells is several fold higher than that of protein-coding genes, indicating that DNA methylation plays an important role in miRNA deregulation in cancer (Vrba et al., 2013). The last decade saw a sharp increase in the interest in identifying targets of methylation and the importance of miRNAs in various tumors, leading to progress in personalized treatment for multiple cancers, including breast cancer (BC) (Stefansson and Esteller, 2013).

BC is the most common cancer among women in all parts of the world, and it accounts for 1/10 of all malignancies detected in both men and women (Dey, 2014). A total of 1.6 million new BC cases are registered annually, and the incidence of BC is increasing worldwide. BC is also the primary cause of cancer-related death among women globally (Dey, 2014).

The aim of this study was to assess the role of methylation of promoter CpG islands in deregulation of 13 cancer-associated miRNAs (miR-124, -125b, -127, -132, -137, -148a, -191, -193a, -203, -212, -34b, -375, and -9) in BC cells. Previously, it was shown that these miRNAs are involved in pathogenesis of BC. MiR-124, -125b, -127, -132, -137, -203, and -212 were reported to act as tumor-suppressors that could prohibit proliferation, invasion, migration, and metastasis (Lv et al., 2011, Feliciano et al., 2013, Wang et al., 2014, Zhang et al., 2014, Eedunuri et al., 2015, Taipaleenmaki et al., 2015, Yang et al., 2015a). The miR-212/132 cluster was suggested to enhance synergistic anti-metastatic properties by suppressing the oncogene SOX4 (Hanieh, 2015). Most miRNAs listed above were shown to be down-regulated in BC; however, up-regulation was observed for miR-191-5p and miR-375 (Saito and Saito, 2012, Mar-Aguilar et al., 2013). It was also found that miR-375, which is overexpressed in BC cells, may have oncogenic functions and could be a key driver for the proliferation of ERα-positive BC cell lines (de Souza Rocha Simonini et al., 2010). Moreover, miR-375 was suggested to be a predictor of local relapse in early-stage BC and metastasis (Madhavan et al., 2016, Zehentmayr et al., 2016).

All the selected miRNA genes harbor CpG islands in their promoter regions. Previously, the role of methylation in the deregulation of miR-9, -148a, and -124 was investigated in representative sets of primary breast tumors (Hsu et al., 2009, Ben Gacem et al., 2014, Sandhu et al., 2014). However, the methylation status alteration of MIR-127, -132, -193a, -212, and -191 has not been studied in BC, although the importance of methylation of these genes in pathogenesis of tumors has been demonstrated in other cancers (Heller et al., 2012, Formosa et al., 2013). Moreover, crucial alterations have been reported in the expression of most of these miRNAs in BC (Mar-Aguilar et al., 2013, Wang et al., 2014, Damavandi et al., 2016). Previous studies on methylation aberrations in the rest of the miRNA genes (MIR-203, -34b, -125b-1, -137, and -375) were limited to cancer cell lines or very small subsets of primary human specimens, and the link between miRNA expression and gene methylation in a representative set of primary BC samples have not been addressed directly (Vogt et al., 2011, Zhang et al., 2011a, Zhang et al., 2011b, Vrba et al., 2013). Here, the methylation status of promoter CpG islands of 13 miRNA genes was assessed by methylation-specific PCR (MSP) in 58 primary BC samples paired with histologically normal samples. The relative expression of these miRNAs was assayed by quantitative PCR (qPCR) in a subset of 38 paired BC samples. The data obtained allowed us to analyze the correlation between methylation and expression of the selected miRNAs and to speculate on the role of DNA methylation in altering the expression of these miRNAs in BC.

To analyze the functional role of the 13 selected miRNAs, we tested their ability to deregulate five cancer-associated genes, which were predicted to be targets for the majority of these miRNAs using bioinformatics. We chose the well-known tumor-suppressor gene, RASSF1(isoform A), which is involved in cell cycle arrest and apoptosis induction; the pro-apoptotic tumor-suppressors APAF1 and DAPK1; anti-apoptotic BCL2; and CHL1, which is involved in cell adhesion, cell migration, invasion, and cancer progression (http://www.genecards.org). All five genes harbor CpG islands in their promoter regions; methylation studies of these genes in solid tumors, including BC, have been reported by us previously (Dreijerink et al., 2001, Zabarovsky et al., 2011a, Zabarovsky et al., 2011b, Braga et al., 2015, Pronina et al., 2016). Here, alterations in the expression of these five genes were assayed using qPCR in a subset of 41 paired BC samples that allowed us to analyze the correlations between the expression levels of the 13 miRNAs and the 5 protein-coding genes and to speculate on possible miRNA-mRNA interacting pairs in BC.

In addition, we analyzed the association between methylation alterations of the 13 miRNA genes and BC progression and identified miRNA genes which hypermethylation was associated with BC progression.

Section snippets

Tissue samples

A total of 58 paired tumor/normal samples of ВС were obtained from the N.N. Blokhin Cancer Research Center (Moscow, Russia). Most of the samples collected were invasive ductal breast carcinomas. The details of the samples are presented in Supplementary Table 1. This set of 58 samples was used in the DNA methylation studies, while subsets of 41 and 38 samples were used for the qPCR expression analyses. The samples were collected in accordance with the guidelines issued by the Ethics Committee of

Expression and methylation profiles of 13 cancer-associated miRNAs in BC

Aberrant expression is the first indicator that a protein-coding gene may function as an oncogene or a tumor-suppressor gene. This is applicable for miRNA genes as well.

In the present study, a high frequency of down-regulation was observed for 5 of the 13 examined miRNAs: miR-124-3p level was decreased in 68% of BC samples, miR-125b-5p in 76%, miR-132-3p in 50%, miR-193a-5p in 79%, and miR-34b-3p in 47% (p  0.05 in all cases, Wilcoxon test; Fig. 1, Supplementary Table 3). Increased expression

Discussion

In this work, we investigated the role of methylation of CpG islands of the promoter in alterations of expression of 13 cancer-associated miRNA genes in BC, possible influence of these miRNAs on deregulation of 5 cancer-associated protein-coding genes (predicted as targets for the miRNAs), and the association of hypermethylation of examined miRNA genes with BC progression and metastasis.

As pointed out in a review (Lehmann, 2014), a number of studies on aberrant methylation of miRNA genes in BC

Conflict of interest

The authors declare no conflicts of interest.

Acknowledgments

We are sincerely grateful to Prof. Michael Lerman (Affina Biotechnologies, USA) for his generous support and useful comments. This work was financially supported by the Russian Science Foundation (RSCF) grant 14-15-00654. The authors thank the Blokhin Cancer Research Center for tissue samples and the Engelhardt Institute of Molecular Biology for the assistance in bioinformatic analyses.

References (75)

  • E.A. Braga et al.

    Methylation in the regulation of the expression of chromosome 3 and microRNA genes in clear-cell renal cell carcinomas

    Russ. J. Genet.

    (2015)
  • H.Y. Chen et al.

    miR-103/107 promote metastasis of colorectal cancer by targeting the metastasis suppressors DAPK and KLF4

    Cancer Res.

    (2012)
  • J. Chen et al.

    miR-127 regulates cell proliferation and senescence by targeting BCL6

    PLoS One

    (2013)
  • Y. Chu et al.

    MicroRNA-590 promotes cervical cancer cell growth and invasion by targeting CHL1

    J. Cell. Biochem.

    (2014)
  • Z. Damavandi et al.

    Aberrant expression of breast development-related MICRORNAS, miR-22, miR-132, and miR-212, in breast tumor tissues

    J. Breast Cancer

    (2016)
  • P. de Souza Rocha Simonini et al.

    Epigenetically deregulated microRNA-375 is involved in a positive feedback loop with estrogen receptor alpha in breast cancer cells

    Cancer Res.

    (2010)
  • D. Deng et al.

    MicroRNA-124-3p regulates cell proliferation, invasion, apoptosis, and bioenergetics by targeting PIM1 in astrocytoma

    Cancer Sci.

    (2016)
  • S. Dey

    Preventing breast cancer in LMICs via screening and/or early detection: the real and the surreal

    World J. Clin. Oncol.

    (2014)
  • K. Dreijerink et al.

    The candidate tumor suppressor gene, RASSF1A, from human chromosome 3p21.3 is involved in kidney tumorigenesis

    Proc. Natl. Acad. Sci. U. S. A.

    (2001)
  • J. Duan et al.

    MicroRNA-34a inhibits cell proliferation and induces cell apoptosis of glioma cells via targeting of Bcl-2

    Mol. Med. Rep.

    (2016)
  • V.K. Eedunuri et al.

    miR-137 targets p160 steroid receptor coactivators SRC1, SRC2, and SRC3 and inhibits cell proliferation

    Mol. Endocrinol.

    (2015)
  • A. Feliciano et al.

    miR-125b acts as a tumor suppressor in breast tumorigenesis via its novel direct targets ENPEP, CK2-alpha, CCNJ, and MEGF9

    PLoS One

    (2013)
  • T. Feng et al.

    MiR-124 inhibits cell proliferation in breast cancer through downregulation of CDK4

    Tumour Biol.

    (2015)
  • M. Ferracin et al.

    miR-125b targets erythropoietin and its receptor and their expression correlates with metastatic potential and ERBB2/HER2 expression

    Mol. Cancer

    (2013)
  • A. Formosa et al.

    DNA methylation silences miR-132 in prostate cancer

    Oncogene

    (2013)
  • Z.B. Han et al.

    MicroRNA-124 suppresses breast cancer cell growth and motility by targeting CD151

    Cell. Physiol. Biochem.

    (2013)
  • H. Hanieh

    Aryl hydrocarbon receptor-microRNA-212/132 axis in human breast cancer suppresses metastasis by targeting SOX4

    Mol. Cancer

    (2015)
  • Y. He et al.

    Hypomethylation of the hsa-miR-191 locus causes high expression of hsa-mir-191 and promotes the epithelial-to-mesenchymal transition in hepatocellular carcinoma

    Neoplasia

    (2011)
  • G. Heller et al.

    Genome-wide miRNA expression profiling identifies miR-9-3 and miR-193a as targets for DNA methylation in non-small cell lung cancers

    Clin. Cancer Res.

    (2012)
  • J.G. Herman et al.

    Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands

    Proc. Natl. Acad. Sci. U. S. A.

    (1996)
  • V.K. Hill et al.

    Stability of the CpG island methylator phenotype during glioma progression and identification of methylated loci in secondary glioblastomas

    BMC Cancer

    (2014)
  • S. Hong et al.

    SHOX2 is a direct miR-375 target and a novel epithelial-to-mesenchymal transition inducer in breast cancer cells

    Neoplasia

    (2014)
  • P.Y. Hsu et al.

    Xenoestrogen-induced epigenetic repression of microRNA-9-3 in breast epithelial cells

    Cancer Res.

    (2009)
  • M. Incoronato et al.

    Epigenetic regulation of miR-212 expression in lung cancer

    PLoS One

    (2011)
  • C.F. Jiang et al.

    Estrogen regulates miRNA expression: implication of estrogen receptor and miR-124/AKT2 in tumor growth and angiogenesis

    Oncotarget

    (2016)
  • G.S. Krasnov et al.

    RPN1, a new reference gene for quantitative data normalization in lung and kidney cancer

    Mol. Biol.

    (2011)
  • U. Lehmann

    Aberrant DNA methylation of microRNA genes in human breast cancer - a critical appraisal

    Cell Tissue Res.

    (2014)
  • Cited by (64)

    • Bone mesenchymal stem cell-derived extracellular vesicles inhibit DAPK1-mediated inflammation by delivering miR-191 to macrophages

      2022, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      These results indicated that BMSC-derived EVs can effectively inhibit the LPS-induced inflammatory activation of THP-1 macrophages, suggesting that the inhibitory effect of BMSCs on macrophage inflammatory activation may be achieved by the delivery of EVs and that BMSC-derived EVs might inhibit the inflammation in THP-1 cells. According to literature reports, DAPK1 regulates inflammation through multiple signaling pathways, such as p38 and mTOR, during the LPS-induced activation of macrophages [25,26,30,31]. We therefore measured the expression of DAPK1 in THP-1 macrophages after treatment with LPS and EVs.

    • The role of microRNAs in regulating cadmium-induced apoptosis by targeting Bcl-2 in IEC-6 cells

      2021, Toxicology and Applied Pharmacology
      Citation Excerpt :

      The results showed that 7 miRNAs, including miR-124-3p and miR-370-3p were upregulated with a dose-effect relationship in the Cd-treated groups, consistent with the sequencing results. Both miR-124-3p and miR-370-3p are believed to promote apoptosis, which is significantly negatively correlated with Bcl-2 (Pronina et al., 2017; Huang et al., 2021). MiR-124-3p has been shown to directly target Bcl2L11 by dual luciferase reporting assay (Cai et al., 2018), but the targeting relationship between miR-370-3p and Bcl-2 has not been confirmed.

    • MicroRNA: A signature for cancer progression

      2021, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      Furthermore, hypermethylation-associated silencing of these miRNAs reduced tumor growth and inhibited metastasis formation in vivo [77]. Hyper-methylation of miR-124, miR-125, miR-126, miR-127, miR-148a, miR-193a, miR-132, miR-133a-3p, miR-137 and miR-34b/c has been found in breast cancer cells [78–82]. In addition, let-7a, miR-9, miR-10b, miR-152, miR-200, miR-195/497 family, and miRNAs at the DLK1-DIO3 imprinted locus harbor similar epigenetic marks [83–85].

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text