DNMT1-microRNA126 epigenetic circuit contributes to esophageal … · 2014. 12. 13. · Another 30...

34
1 DNMT1-microRNA126 epigenetic circuit contributes to esophageal squamous cell carcinoma growth via ADAM9-EGFR-AKT signaling Ronghua Liu a, b, # , Jie Gu c, # , Pei Jiang a, b , Yijie Zheng a, b , Xiaoming Liu a, b , Xuechao Jiang a, b , Enyu Huang a, b , Shudao Xiong a, d ,Fengkai Xu c , Guangwei Liu a, b , Di Ge c, *, Yiwei Chu a, b, * a Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai,200032, People's Republic of China. b Biotherapy Research Center, Fudan University, Shanghai 200032, China. c Department of Thoracic Surgery, The Affiliated Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China. d Department of Hematology/Oncology, The Second Hospital of Anhui Medical University, Hefei, Anhui Province, People's Republic of China. # These authors contributed equally to this paper. *Correspondence: Yiwei Chu, M.D, Ph.D, Department of Immunology and Key Laboratory of Molecular, Medicine of Ministry of Education, Shanghai Medical College, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, P.R.China, Tel. /fax: +86 21 54237324, E-mail address: [email protected]; or Di Ge, M.D, Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R.China, Tel. /fax: +86 21 64041990-2559, E-mail address. [email protected]. Research. on June 12, 2021. © 2014 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

Transcript of DNMT1-microRNA126 epigenetic circuit contributes to esophageal … · 2014. 12. 13. · Another 30...

  • 1

    DNMT1-microRNA126 epigenetic circuit contributes to esophageal

    squamous cell carcinoma growth via ADAM9-EGFR-AKT signaling

    Ronghua Liu a, b, #, Jie Gu c, #, Pei Jiang a, b, Yijie Zheng a, b, Xiaoming Liu a, b, Xuechao

    Jiang a, b , Enyu Huang a, b , Shudao Xiong a, d ,Fengkai Xu c, Guangwei Liu a, b, Di Gec,*,

    Yiwei Chua, b, *

    a Department of Immunology, Key Laboratory of Medical Molecular

    Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University,

    Shanghai,200032, People's Republic of China.

    b Biotherapy Research Center, Fudan University, Shanghai 200032, China.

    c Department of Thoracic Surgery, The Affiliated Zhongshan Hospital of Fudan University,

    Shanghai, 200032, People's Republic of China.

    d Department of Hematology/Oncology, The Second Hospital of Anhui Medical University,

    Hefei, Anhui Province, People's Republic of China.

    # These authors contributed equally to this paper.

    *Correspondence:

    Yiwei Chu, M.D, Ph.D, Department of Immunology and Key Laboratory of Molecular,

    Medicine of Ministry of Education, Shanghai Medical College, Fudan University, No.138,

    Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, P.R.China, Tel. /fax: +86 21 54237324,

    E-mail address: [email protected]; or Di Ge, M.D, Department of Thoracic Surgery,

    Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R.China,

    Tel. /fax: +86 21 64041990-2559, E-mail address. [email protected].

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 2

    RUNNING TITLE: DNMT1-microRNA126 epigenetic circuit regulates ESCC growth

    Key Words: epigenetics; microRNA; DNA methylation; prognosis

    Number of words in text: 4870;

    Supplementary Tables: 6; Supplementary Figures: 6.

    Disclosure of Potential Conflicts of Interest

    The authors declare no conflicts of interest.

    Translational Relevance

    Esophageal squamous cell carcinoma growth (ESCC) is one of the most aggressive

    malignancies of the gastrointestinal tract. Previous studies have described complicated

    genetic and epigenetic alternations involved in ESCC progression, but the underlying

    mechanisms have not been fully elucidated. Recent evidences show that microRNAs

    (miRNAs) are involved in and are controlled by epigenetic regulation, and thereby form a

    reciprocal regulatory circuit in many systems. In this study, using NGS-based miRNA

    profiling, we found the downregulation of miR-126 in ESCC and proposed a

    “DNMT1-miR-126 epigenetic circuit” involved in ESCC progression. Moreover, miR-126

    acted as a novel tumor suppressor that effectively suppresses ESCC cell proliferation and

    migration by repressing ADAM9-EGFR-AKT signaling transduction and as an

    independent prognostic indicator for ESCC patients. Taking together, our funding of

    “DNMT1-miR-126 epigenetic circuit” suggests another insight into pathological

    mechanisms underlying ESCC initiation and progression, implying its clinical significance

    in developing targets for ESCC prediction and therapy.

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 3

    Abstract

    Purpose: MicroRNAs (miRNAs) are involved in and are controlled by epigenetic

    regulation, and thereby form a reciprocal regulatory circuit. Using next-generation

    sequencing (NGS)-based miRNA profiling, this study aimed to discover esophageal

    squamous cell carcinoma (ESCC)-specific miRNAs and miRNA-related epigenetic

    modulations.

    Experimental Design: NGS-based miRNA profiles were generated for 4 pairs of ESCC

    tissues and adjacent normal tissues. In situ hybridization was used to assess miRNA

    expression and its correlation with prognosis. miRNA-related DNA methylations were

    identified using bisulfite genomic sequencing, and the role of DNA methyltransferase 1

    (DNMT1) was investigated using RNA interference. miRNA targets were screened by

    mRNA sequencing, and functional validation was performed in vitro and in vivo.

    Results: NGS-based miRNA profiling identified 78 differentially expressed miRNAs in

    ESCC. Among them, microRNA126-3p (miR-126) was significantly downregulated, and its

    downregulation correlated with poor ESCC prognosis. Downregulation of miR-126 was

    due to promoter hypermethylation of its host gene, Egfl7. DNMT1 was aberrantly

    upregulated in ESCC and responsible for the hypermethylation of Egfl7. Intriguingly,

    DNMT1 was suppressed by overexpression of miR-126, indicating the existence a

    regulatory feedback circuit. ADAM9 was identified as a key target of miR-126. Ectopic

    expression of miR-126 or silencing of ADAM9 reduced ESCC cell proliferation and

    migration by inhibiting epidermal growth factor receptor-AKT signaling.

    Conclusions: Our results indicate that miR-126 is a potential prognostic indicator for

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 4

    ESCC and suggest that a novel “DNMT1-miR-126 epigenetic circuit” is involved in ESCC

    progression. Consequently, miR-126-based epigenetic modulations may provide a basic

    rationale for new approaches to anti-tumor therapeutics.

    Introduction

    Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive

    malignancies of the gastrointestinal tract(1). Despite improvements in diagnostic

    approaches and the introduction of adjuvant chemotherapy, the 5-year survival rate in

    patients with esophageal cancer remains less than 30%(2). Previous studies have

    described complicated genetic and epigenetic alternations involved in ESCC progression

    (3, 4), but the underlying mechanisms have not been fully elucidated. Therefore, in order

    to develop better approaches for ESCC diagnosis and therapy, a comprehensive

    understanding of the molecular variables that affect ESCC pathways is needed.

    MicroRNAs (miRNAs) have emerged as key players in human tumors; their

    dysregulation affects cell proliferation(5), differentiation(6), invasion(7), and the

    epithelial-mesenchymal transition (EMT)(8). miRNA expression profiles have been

    developed for use as prognostic/predictive biomarkers and been used to identify the

    molecular signatures and underlying functional mechanisms of various cancers. Previous

    miRNA profiling studies were conducted using miRNA expression microarray platforms (9,

    10). However, in recent decades, the emergence of next-generation sequencing (NGS)

    platforms has revolutionized genomic studies. In comparison with microarray, NGS-based

    sequencing has advantages for the analysis of widely expressed miRNAs, the

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 5

    identification of miRNA sequence variations, and the discovery of novel miRNAs(11, 12).

    Although, microarray-based miRNA profiles of ESCC have been described (13), to our

    knowledge, no studies have used an NGS-based platform to identify ESCC-specific

    miRNAs. Furthermore, the pathological mechanisms underlying their dysregulation

    remain incompletely understood.

    Epigenetic modulations, including DNA methylation and histone acetylation, play a

    critical role in various diseases by regulating gene expression without changing the DNA

    sequence(14). Recent evidence indicates that miRNAs integrate into the epigenetic

    machinery via the transcriptional regulation of epigenetic factors, creating an

    “epigenetic-miRNA feedback loop” (15, 16). In this study, we propose that a

    “DNMT1-miR-126 epigenetic circuit” is involved in ESCC progression. MiR-126 was

    silenced by promoter hypermethylation of its host gene in ESCC tissues, while DNMT1

    was aberrantly overexpressed. Silencing of DNMT1 increased miR-126 expression by

    reducing the hypermethylation, while miR-126 suppressed DNMT1 expression via a

    negative feedback process. MiR-126 was also identified as a novel tumor suppressor and

    a potential prognostic indicator in ESCC. Overexpression of miR-126 repressed ESCC

    cell proliferation and migration by inhibiting the activation of the ADAM9/EGFR/AKT

    pathway. Understanding the epigenetic-miRNA network in ESCC progression and

    modulating miRNA-related epigenetic dynamics hold promise for ESCC targeted therapy.

    Materials and Methods

    Samples and cells

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 6

    ESCC specimens embedded in paraffin were obtained from 185 patients who underwent

    esophagectomy at Zhongshan Hospital (Shanghai, People's Republic of China) in 2007

    with informed consent, and clinicopathological characteristics of patients were listed in the

    supplementary table S1. Charlson comorbidity index (CCI) was used for the analysis of

    comorbidities(17). Each paraffin blocks was well formalin-fixed, paraffin-embedded and

    without necrosis and hemorrhage. Follow-up was conducted every six mouths, and ended

    in December 2012, with a median follow-up of 32 months. The clinical endpoint was the

    survival state of patient at the last follow-up. Disease-specific survival was defined as the

    interval between surgery and death of ESCC. The data were censored for patients who

    died of causes other than ESCC or survived at the last follow-up. Another 30 pairs of

    frozen human primary human ESCC tissues (ECs) and matched adjacent non-cancerous

    tissues (NMs) were obtained from 30 patients at Zhongshan Hospital in 2012. Tumor

    stage was determined according to 7th edition of the Union for International Cancer

    Control-American Joint Committee on Cancer tumor, node, metastasis staging system

    (18). Approval for the study was obtained from the Research Ethics Committee of

    Zhongshan Hospital.

    ESCC cell line ECa-109 was obtained from the Institute of Biochemistry and Cell

    Biology of the Chinese Academy of Science (TCHu 69, Shanghai, China), and ESCC cell

    line KYSE-510 (JCRB1436, National Institute for Biomedical Innovation, Japan) and

    normal human esophageal epithelial cell line HEEpiC (HEEC) (#2720, ScienCell

    Research Laboratories, USA) was supplied by Dr. Lu (19), which were authenticated

    using short tandem repeat (STR) profiling and used in the 6 month after receipt in this

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 7

    research. All the cells were cultured in RPMI-1640 supplemented with 10% FBS, 2 μM

    glutamine, 100 IU/mL penicillin, and 100 μg/mL streptomycin sulfates.

    NGS-based miRNA profiling

    Total RNA from 4 pairs of ECs and matched NMs were isolated using a mirVana miRNA

    Isolation Kit (Ambion, Austin, TX, USA) according to the manufacturer’s instructions. After

    PAGE purification, small RNA molecules were amplified and sequenced on an Illumina

    platform according to the manufacturer’s instructions (20).

    Tissue microarray and in situ hybridization (ISH)

    Tissue microarrays were constructed as described in a previous study(21). Digoxin

    (DIG)-labeled oligonucleotide probes complementary to hsa-miR-126 were designed as

    previously described and purchased from ExonBIO (Guangzhou, China)(22). The probe

    sequence was 5′-UCGUACCGUGAGUAAUAAUGCG-3′. The slides were hybridized with

    DIG-conjugated hsa-miR-126 probes overnight at 42°C and stained using an Enhanced

    Sensitive ISH Detection Kit II (AP) with phosphatase-conjugated anti-DIG antibody

    (BOSTER, China) according to the manufacturer’s instructions.

    Statistical analysis

    Data are presented as the mean ± SEM of replicate experiments (n > 3). Results were

    analyzed using SPSS 16.0 software (SPSS, Chicago, IL, USA) and PRISM 5.0

    (GraphPad Software Inc., San Diego, CA, USA). Student’s unpaired t-test or unpaired

    t-test with Welch's correction was used to analyze intergroup differences for 2 groups,

    ANOVA was used to analyze more than 2 groups, and Pearson’s correlation coefficient

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 8

    was used to analyze the correlation between groups. The cumulative survival time was

    calculated using the Kaplan-Meier method and analyzed using the log-rank test.

    Univariate and multivariate analyses were based on the Cox proportional hazards

    regression model. P values < 0.05 were considered statistically significant.

    Results

    NGS-based discovery of miR-126 and validation of its downregulation in ESCC

    To identify ESCC-specific miRNA profiles, NGS-based miRNA profiling was performed on

    4 pairs of frozen ECs and adjacent NMs. When ECs and NMs were compared, 78

    miRNAs were differentially expressed, of which 44 were downregulated and 34 were

    upregulated in ECs (P < 0.05 with fold-change > 3) (Fig. 1A and Supplementary Table

    S2). Among the differentially regulated miRNAs, the downregulation of miR-126-3p in ECs

    exhibited the strongest statistical significance. For validation, qPCR was used to assess

    the expression of miR-126 in 30 pairs of frozen ECs and matched NMs, as well as in

    ESCC cell lines (ECa-109 and KYSE-510) and HEECs, normal human esophageal

    epithelial cells. The results showed that miR-126 expression was also downregulated in

    ESCC tissues (P < 0.001) (Fig. 1B) and cell lines (P < 0.001) (Fig. 1C), when compared

    with expression in NMs and HEECs, respectively.

    Downregulation of miR-126 correlates with poor prognosis in ESCC patients

    To address the clinical significance of miR-126 in ESCC, ISH was performed in 185 ESCC

    specimens. Patients with negative miR-126 expression exhibited lower 5-year

    disease-specific survival rate than that with positive miR-126 expression (P = 0.014,

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 9

    Figure 1D). In multivariate analysis, CCI, tumor stage and miR-126 expression were

    identified as independent prognostic factors in patients’ disease-specific survival

    (Supplementary Table S3). These results suggest that downregulation of miR-126 is an

    independent indicator of prognosis in ESCC.

    Hypermethylation of the Egfl7 promoter induces miR-126 downregulation in EC

    Given that the miR-126 gene is located in an intron of Egfl7 and its expression is

    reportedly upregulated in cancer cells treated with inhibitors of DNA methylation(23), we

    analyzed the sequence of Egfl7 and found that a large CpG island was present in the

    promoter region (Fig. 2A). To understand the correlation between methylation of the Egfl7

    promoter and the downregulation of miR-126, we analyzed CpG island methylation in ECs

    and matched NMs. The results showed that Egfl7 promoter methylation was higher in ECs

    than in NMs (Fig. 2B). Samples with higher methylation exhibited lower miR-126

    expression (Fig. 2C). To determine the effect of methylation of the Egfl7 promoter on the

    expression of miR-126, genomic DNA was extracted from ECa-109 and KYSE-510 cells

    treated with 5-azadC or vehicle. After sodium-bisulfite modification, MSP was carried out

    using the specific primers for the unmethylated (U) or methylated (M) Egf7 promoter

    sequence. As a Result, compared with vehicle treated cells in which the Egf7 promoter

    was hypermethylated, cells treated with the demethylating agent 5-aza-dC to induce

    demethylation of the Egfl7 promoter showed more unmethylated sequences and an

    increase in miR-126 (Fig. 2D). These results indicate that hypermethylation of the Egfl7

    promoter directly downregulates miR-126 in ESCC.

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 10

    DNMT1-miR-126 regulatory circuit contributes to Egfl7 promoter hypermethylation

    DNA methyltransferases (DNMTs) are critical for gene-specific methylation and the

    consequent transcriptional silencing of cancer-related genes(24). In this study, we found

    that DNMT1 expression was dramatically increased in ESCC tumor tissues and cell lines

    (Fig. 3A, Supplementary Fig. S1A). Furthermore, correlation analysis showed that the

    expression of DNMT1 inversely correlated with that of endogenous miR-126 (Fig. 3A). To

    investigate the role of DNMT1 in the regulation of miR-126 expression, we silenced

    DNMT1 expression in cells using siRNA. DNMT1 knockdown reduced the

    hypermethylation of the miR-126 host gene (Fig. 3B), leading to miR-126 upregulation

    (Fig. 3C). Interestingly, overexpression of miR-126 suppressed DNMT1 expression in

    both KYSE-510 and ECa-109 cells (Fig. 3D). Collectively, overexpression of DNMT1

    resulted in the hypermethylation and downregulation of miR-126, while overexpression of

    miR-126 suppressed DNMT1 expression. Our findings indicate the involvement of a novel

    DNMT1-miR-126 regulatory loop in ESCC.

    Overexpression of miR-126 suppresses ESCC cell proliferation and migration

    Given the low expression of miR-126 in ESCC, we further evaluated the biological role

    played by miR-126 in ESCC cell lines. miR-126 mimics were transfected into ECa-109

    and KYSE-510 cells to upregulate miR-126 expression (Supplementary Fig. S2A).

    Overexpression of miR-126 markedly suppressed cell proliferation (Fig. 4A) and the rate

    of colony formation (Fig. 4B) in ECa-109 and KYSE-510 cells when compared to the

    negative control. Further analysis showed that miR-126 mainly promoted cell apoptosis

    (Supplementary Fig. S2B), but without a marked effect on the cell cycle (Supplementary

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 11

    Fig. S2C). In addition, overexpression of miR-126 inhibited EC cell migration in vitro (Fig.

    4C, Supplemetary Fig. S3A).

    To confirm the suppressive role of miR-126 in ESCC, we next investigated the effect

    of miR-126 on ESCC tumor growth in vivo. ECa-109 and KYSE-510 cells transfected with

    either miR-126 or miR-NC (control group) were implanted subcutaneously into the

    bilateral posterior flank of nude mice. Compared to tumor growth in the control group,

    tumor growth in the miR-126-transfected group was markedly suppressed, as reflected by

    the smaller tumor sizes (Fig. 4D, Supplemetary Fig. S3B). Collectively, these results

    indicate that miR-126 functions as a tumor suppressor by inhibiting ESCC cell growth and

    migration.

    The ADAM9/EGFR/AKT signaling directly mediates the suppressive role of miR-126

    To investigate the targets and downstream signaling pathway of miR-126, we compared

    mRNA profiles of ECa-109 cells transfected with miR-126 (126-EC cells) or miR-NC

    (NC-EC cells) mimics using NGS-based mRNA sequencing. In 126-EC cells, 31 genes

    were significantly downregulated relative to the levels in NC-EC cells (Supplementary

    Table S4), including ADAM9. Consistently, in bioinformatics analysis, ADAM9, a putative

    target gene of miR-126, was identified with 4 prediction algorithms (PicTar, TargetScan,

    miRanda, and miRDB) (Supplementary Table S4). Therefore, ADAM9 was selected for

    further validation and functional analyses. Results from qPCR, western blot, and

    immunohistochemistry (IHC) showed that ADAM9 was overexpressed in ECs and ESCC

    cell lines and that its mRNA levels inversely correlated with miR-126 levels in ECs (Fig.

    5A-B, Supplementary Fig. S4A). This inverse correlation strongly suggested the

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 12

    involvement of ADAM9 in the miR-126-related network. Bioinformatics analysis showed

    that the binding sites in the 3′-UTR of ADAM9 and miR-126 are highly conserved (Fig. 5C).

    We constructed the ADAM9-3′-UTR reporter plasmid and performed luciferase reporter

    assays in cells transfected with the 3′-UTR plasmid. As expected, miR-126 repressed

    luciferase activity in cells expressing ADAM9-3′-UTR, indicating a direct interaction

    between miR-126 and ADAM9 in ESCC cells (Fig. 5C). Furthermore, a decrease in

    ADAM9 was detected in miR-126-overexpressing ESCC cells (P < 0.05) (Fig. 5D). To

    address the direct involvement of ADAM9 in miR-126-regulated tumor suppression,

    ECa-109, KYSE-510, and KYSE-510 cells were transfected with siRNAs against ADAM9

    mRNA; si-1 (si-ADAM9) was selected for further study because of its efficient interference

    (Supplementary Fig. S4B). EC cell proliferation was inhibited in

    ADAM9-siRNA-transfected ECa-109 and KYSE-510 cells and the cell migration rate was

    lower than in si-Ctrl-transfected cells (Fig. 6A). Moreover, after silencing DNMT1, the

    expression of ADAM9 in protein level was also reduced, with decreasing cell viability and

    migration (Fig. 5D, Supplementary Fig. S1B–C). These data indicate that ADAM9 is the

    target of miR-126 and is regulated by the “DNMT1-miR-126 circuit” in ESCC.

    ADAM9 activates EGFR by cleaving the transmembrane pro-heparin-binding

    epidermal growth factor-like growth factor (pro-HB-EGF) precursor to yield HB-EGF, a

    soluble EGFR ligand. HB-EGF binds and activates intracellular signaling cascades

    downstream of EGFR, such as the AKT pathway, and thereby promotes the growth of

    normal and neoplastic cells (25, 26). In our study, we found that the EGFR/AKT pathway

    was involved in the suppressive action of miR-126/ADAM9 in ESCC. Both overexpression

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 13

    of miR-126 and blockage of ADAM9 reduced levels of p-EGFR and p-AKT (Fig. 6B), cell

    proliferation, and cell migration decreased (Fig. 6C). These results indicate that the

    ADAM9/EGFR/AKT pathway directly mediates the suppressive action of miR-126 in

    ESCC (Fig. 6D).

    Discussion

    The ”epigenetic-miRNA loop,” wherein miRNAs regulate or are regulated by epigenetic

    factors, was proposed by Pagano et al(27). They suggested that epigenetic-miRNA loops

    contributed to hematopoiesis by orchestrating gene expression in hematopoietic cells. In

    this study, we identified the DNMT1-miR-126 epigenetic loop in ESCC and investigated its

    contribution to ESCC development via modulation of the ADAM9/EGFR/AKT pathway.

    Dysregulation of miRNAs in diseases is frequently due to epigenetic changes in the

    miRNA genome(28, 29). Previous studies of miRNA genomic sequences have indicated

    that promoter hypermethylation of miRNAs occurs frequently as a result of the aberrant

    expression of DNMTs, histone deacetylases, and methyl-CpG-binding proteins (29-31). In

    the present study, we found that miR-126 was downregulated in ECs and that its

    downregulation was induced by promoter hypermethylation of its host gene.

    Dysregulation of miR-126 has also been observed in other tumors, such as colon

    cancer(32) and lung cancer (33), but little is known about its function in ESCC. This is the

    first report to show that miR-126 is downregulated and acts as a new tumor suppressor in

    ESCC. Moreover, we found that downregulation of miR-126 is a potential prognostic

    indicator for ESCC. However, the results was different from the prior report that

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 14

    miRNA-126 failed to show a relation to outcome of patients with esophageal

    adenocarcinoma (34). Such kind of conflict might due to the distinct biology and clinical

    features of esophageal squamous carcinoma in our study and esophageal

    adenocarcinoma in the previous study. The differences in races (Asian vs. American)

    might also contribute to it.

    Although evidence from Zhang et al. indicated that the downregulation of miR-126 in

    breast cancer positively correlated with hypermethylation of the Egfl7 T2 promoter (35),

    the underlying molecular mechanism was not elucidated. Notably, our results demonstrate

    that DNMT1 is overexpressed in primary ECs and cell lines and that DNMT1 responsible

    for the hypermethylation of the Egfl7 promoter, leading to miR-126 downregulation.

    DNMT1, a member of the DNMT family (DNMT1, DNMT3A, and DNMT3B), is

    overexpressed in tumors and is essential for maintaining promoter hypermethylation of

    tumor suppressive genes, leading to their silencing (36). In breast cancer, overexpressed

    DNMT1 induces miR-148a/152 silencing by maintaining the hypermethylation of their

    promoters and contributes to tumor transformation and angiogenesis (37). In ESCC, we

    demonstrate that overexpressed DNMT1 was responsible for the promoter

    hypermethylation of the miR-126 host gene, thereby downregulating miR-126 and

    inhibiting ESCC growth and cell migration. Interestingly, in a feedback mechanism,

    DNMT1 was suppressed by miR-126 overexpression in ESCC cells, indicating the

    presence of a novel “DNMT1-miR-126 epigenetic circuit.” We also showed that DNMT1

    was the direct target of miR-126 (Supplementary Fig. S5). Regulation of DNMT1 by

    miR-126 has also been described in CD4+ T cells from systemic lupus erythematosus

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 15

    patients (37). Similar to the “DNMT1-miR-126 epigenetic circuit” in ESCC, a negative

    feedback regulatory loop between miR-148/152 and DNMT1 has been reported in breast

    cancer (38). Thus, epigenetic-miRNA loops are widely present in various tumors. Given

    that both miRNA and epigenetic modulations strongly regulate the expression of various

    disease-associated genes(14), controlling the dynamic balance of epigenetic-miRNA

    loops might be of great clinical significance.

    The expression of ADAM9 has been reported to correlate with cell adherence and

    migration (25, 33, 39, 40). This study found that ADAM9 functioned as a direct target of

    miR-126 and contributed to miR-126 repressing cell migration in ESCC. Consistently, the

    targeting of miR-126 to ADAM9 and the role of ADAM9 in cell migration was also reported

    in a previous study by S. Hamada et al (39). A similar approach with our study using

    restoration of miR-126 and silencing of ADAM9 showed inhibition of the invasive growth.

    In order to systematically investigate the target of miR-126 in ESCC, RNA sequencing

    was applied in the present study to examine the differential gene expression in

    miR-126-overexpressed ESCC EC-109 cells vs. control cells. ADAM9 was showed to be a

    functional target of miR-126, while other potential targets are under exploration. Therefore,

    apart from pancreatic cancer in S. Hamada’s study, this study also identified the

    interaction between miR-126 and ADAM9 in ESCC. In addition, we investigated the

    down-stream pathway and found that ADAM9 was critical for promoting cell proliferation in

    ESCC by targeting EGFR-AKT signaling. ADAM9 cleaved the pro-HB-EGF precursor to

    yield soluble HB-EGF, which binds to EGFR, thereby activating downstream signal

    transduction (26, 41). The EGFR/AKT pathway is important for cell growth and migration

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 16

    (42, 43). Our results showed that blocking ADAM9 inhibits EGFR/AKT activation by

    suppressing phosphorylation, but has no effect on ERK phosphorylation. We also found

    that the expression of phosphoinositide-3-kinase, regulatory subunit 2 (beta) was

    downregulated upon miR-126 overexpression (Supplementary Table S4 and

    Supplementary Fig. S6A). However, its expression in ECs and in NMs did not differ (P >

    0.05), and its inverse correlation with miR-126 expression was poor (R=-0.1791,

    P=0.5255) (Supplementary Fig. S6B-C). Other growth-related targets of miR-126

    identified in other tumors, such as Crk(44) and IRS(45), were not downregulated in

    miR-126-overexpressing ECa-109 cells (Supplementary Fig. S6A), implying they are not

    key targets of miR-126 in ESCC growth.

    With accumulating evidences demonstrating the importance of miRNAs in cancer

    biology, miRNA based cancer therapy emerges as a hot issue and also a big challenge in

    the current miRNA research. To be excited, MRX34, as the first microRNA-based therapy

    for cancer, is entering the phase 1 trial(46). In the present study, our primary data showed

    that the exogenous miR-126 mimics suppressed ESCC cell growth, which implies its

    potential therapeutic significance in ESCC using a miRNAs’ “replacement” strategy.

    However, miR-126 based therapy for ESCC in practice still needs to be further explored,

    considering some challenges, especially technical limitations, such as how to improve its

    accumulation in the target tissues and balance the safety and efficiency. In general, there

    is a long way ahead to realize the translation of fundamental research of miR-126 to

    clinical applications in ESCC.

    In summary, this study proposes a “DNMT1-miR-126 epigenetic circuit” in ESCC

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 17

    and sheds light on its importance for ESCC progression via regulation of

    ADAM9-EGFR-AKT signaling. Altering critical gene expression and signal transduction by

    modulating miR-126 related epigenetic dynamics might be of clinical significance in ESCC

    prediction and therapy.

    Grant Support

    This work was supported by the National Science Foundation of China (81273215,

    81372313, 81401876 and 81272259), the Major Research Plan of the National Natural

    Science Foundation of China (91229110), the National Basic Research Program of China

    (973 Programs, 2011CB910404) and the Specialized Research Fund for the Doctoral

    Program of Higher Education (20120071110046).

    References

    1. Holmes RS, Vaughan TL. Epidemiology and pathogenesis of esophageal cancer.

    Semin Radiat Oncol. 2007;17:2-9.

    2. Yamasaki M, Miyata H, Miyazaki Y, Takahashi T, Kurokawa Y, Nakajima K, et al.

    Evaluation of the Nodal Status in the 7th Edition of the UICC-TNM Classification for

    Esophageal Squamous Cell Carcinoma: Proposed Modifications for Improved Survival

    Stratification : Impact of Lymph Node Metastases on Overall Survival after

    Esophagectomy. Ann Surg Oncol. 2014;21:2850-6

    3. Zhang W, Zhu H, Liu X, Wang Q, Zhang X, He J, et al. Epidermal Growth Factor

    Receptor Is a Prognosis Predictor in Patients With Esophageal Squamous Cell

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 18

    Carcinoma. Ann Thorac Surg. 2014;98:513-9.

    4. Wang G, Zhang W, Jiang W, Luan L. Overexpression of her-2 associated with the

    progression of esophageal cancer patients. Hepatogastroenterology. 2013;60:1972-8.

    5. Xiong S, Zheng Y, Jiang P, Liu R, Liu X, Chu Y. MicroRNA-7 inhibits the growth of

    human non-small cell lung cancer A549 cells through targeting BCL-2. Int J Biol Sci.

    2011;7:805-14.

    6. Yu X, Li H, Jiang X, Guo L, Jiang W, Lu SH. MiR-203 inhibits the proliferation and

    self-renewal of esophageal cancer stem-like cells by suppressing stem renewal factor

    Bmi-1. Stem Cells Dev. 2013;23:576-85.

    7. Matsushima K, Isomoto H, Yamaguchi N, Inoue N, Machida H, Nakayama T, et al.

    MiRNA-205 modulates cellular invasion and migration via regulating zinc finger E-box

    binding homeobox 2 expression in esophageal squamous cell carcinoma cells. J Transl

    Med. 2011;9:30.

    8. Ohashi S, Natsuizaka M, Naganuma S, Kagawa S, Kimura S, Itoh H, et al. A

    NOTCH3-mediated squamous cell differentiation program limits expansion of

    EMT-competent cells that express the ZEB transcription factors. Cancer Res.

    2011;71:6836-47.

    9. Di Leva G, Croce CM. miRNA profiling of cancer. Curr Opin Genet Dev. 2013;23:3-11.

    10. Zheng Y, Xiong S, Jiang P, Liu R, Liu X, Qian J, et al. Glucocorticoids inhibit

    lipopolysaccharide-mediated inflammatory response by downregulating microRNA-155: a

    novel anti-inflammation mechanism. Free Radic Biol Med. 2012;52:1307-17.

    11. Fehniger TA, Wylie T, Germino E, Leong JW, Magrini VJ, Koul S, et al.

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 19

    Next-generation sequencing identifies the natural killer cell microRNA transcriptome.

    Genome Res. 2010;20:1590-604.

    12. Tam S, de Borja R, Tsao MS, McPherson JD. Robust global microRNA expression

    profiling using next-generation sequencing technologies. Lab Invest. 2014;94:350-8.

    13. Kong KL, Kwong DL, Chan TH, Law SY, Chen L, Li Y, et al. MicroRNA-375 inhibits

    tumour growth and metastasis in oesophageal squamous cell carcinoma through

    repressing insulin-like growth factor 1 receptor. Gut. 2012;61:33-42.

    14. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nature

    reviews Genetics. 2002;3:415-28.

    15. Sun X, He Y, Huang C, Ma TT, Li J. The epigenetic feedback loop between DNA

    methylation and microRNAs in fibrotic disease with an emphasis on DNA

    methyltransferases. Cellular signalling. 2013;25:1870-6.

    16. Iorio MV, Piovan C, Croce CM. Interplay between microRNAs and the epigenetic

    machinery: an intricate network. Biochimica et biophysica acta. 2010;1799:694-701.

    17. Tougeron D, Di Fiore F, Thureau S, Berbera N, Iwanicki-Caron I, Hamidou H, et al.

    Safety and outcome of definitive chemoradiotherapy in elderly patients with oesophageal

    cancer. Br J Cancer. 2008;99:1586-92.

    18. Talsma K, van Hagen P, Grotenhuis BA, Steyerberg EW, Tilanus HW, van Lanschot

    JJ, et al. Comparison of the 6th and 7th Editions of the UICC-AJCC TNM Classification for

    Esophageal Cancer. Ann Surg Oncol. 2012;19:2142-8.

    19. Lu CL, Guo J, Gu J, Ge D, Hou YY, Lin ZW, et al. CXCR4 heterogeneous expression

    in esophageal squamous cell cancer and stronger metastatic potential with

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 20

    CXCR4-positive cancer cells. Dis Esophagus. 2014;27:294-302.

    20. Hou J, Lin L, Zhou W, Wang Z, Ding G, Dong Q, et al. Identification of miRNomes in

    human liver and hepatocellular carcinoma reveals miR-199a/b-3p as therapeutic target for

    hepatocellular carcinoma. Cancer Cell. 2011;19:232-43.

    21. Gu J, Ding JY, Lu CL, Lin ZW, Chu YW, Zhao GY, et al. Overexpression of CD88

    predicts poor prognosis in non-small-cell lung cancer. Lung Cancer. 2013;81:259-65.

    22. Li J, Li X, Li Y, Yang H, Wang L, Qin Y, et al. Cell-specific detection of miR-375

    downregulation for predicting the prognosis of esophageal squamous cell carcinoma by

    miRNA in situ hybridization. PLoS One. 2013;8:e53582.

    23. Felli N, Felicetti F, Lustri AM, Errico MC, Bottero L, Cannistraci A, et al.

    miR-126&126* restored expressions play a tumor suppressor role by directly regulating

    ADAM9 and MMP7 in melanoma. PLoS One. 2013;8:e56824.

    24. Di Ruscio A, Ebralidze AK, Benoukraf T, Amabile G, Goff LA, Terragni J, et al.

    DNMT1-interacting RNAs block gene-specific DNA methylation. Nature. 2013;503:371-6.

    25. Fry JL, Toker A. Secreted and membrane-bound isoforms of protease ADAM9 have

    opposing effects on breast cancer cell migration. Cancer Res. 2010;70:8187-98.

    26. Itabashi H, Maesawa C, Oikawa H, Kotani K, Sakurai E, Kato K, et al. Angiotensin II

    and epidermal growth factor receptor cross-talk mediated by a disintegrin and

    metalloprotease accelerates tumor cell proliferation of hepatocellular carcinoma cell lines.

    Hepatol Res. 2008;38:601-13.

    27. Pagano F, De Marinis E, Grignani F, Nervi C. Epigenetic role of miRNAs in normal

    and leukemic hematopoiesis. Epigenomics. 2013;5:539-52.

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 21

    28. Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell.

    2007;128:669-81.

    29. Weber B, Stresemann C, Brueckner B, Lyko F. Methylation of human microRNA

    genes in normal and neoplastic cells. Cell Cycle. 2007;6:1001-5.

    30. Meng X, Wu J, Pan C, Wang H, Ying X, Zhou Y, et al. Genetic and epigenetic

    down-regulation of microRNA-212 promotes colorectal tumor metastasis via dysregulation

    of MnSOD. Gastroenterology. 2013;145:426-36 e1-6.

    31. Roy SS, Gonugunta VK, Bandyopadhyay A, Rao MK, Goodall GJ, Sun LZ, et al.

    Significance of PELP1/HDAC2/miR-200 regulatory network in EMT and metastasis of

    breast cancer. Oncogene. 2013;33:3707-16.

    32. Liu Y, Zhou Y, Feng X, An P, Quan X, Wang H, et al. MicroRNA-126 functions as a

    tumor suppressor in colorectal cancer cells by targeting CXCR4 via the AKT and ERK1/2

    signaling pathways. Int J Oncol. 2014;44:203-10.

    33. Crawford M, Brawner E, Batte K, Yu L, Hunter MG, Otterson GA, et al. MicroRNA-126

    inhibits invasion in non-small cell lung carcinoma cell lines. Biochem Biophys Res

    Commun. 2008;373:607-12.

    34. Hu Y, Correa AM, Hoque A, Guan B, Ye F, Huang J, et al. Prognostic significance of

    differentially expressed miRNAs in esophageal cancer. Int J Cancer. 2011;128:132-43.

    35. Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, et al. miR-126 and miR-126* repress

    recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast

    cancer metastasis. Nat Cell Biol. 2013;15:284-94.

    36. Jin B, Robertson KD. DNA methyltransferases, DNA damage repair, and cancer. Adv

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 22

    Exp Med Biol. 2013;754:3-29.

    37. Xu Q, Jiang Y, Yin Y, Li Q, He J, Jing Y, et al. A regulatory circuit of miR-148a/152 and

    DNMT1 in modulating cell transformation and tumor angiogenesis through IGF-IR and

    IRS1. Journal of molecular cell biology. 2013;5:3-13.

    38. Zhao S, Wang Y, Liang Y, Zhao M, Long H, Ding S, et al. MicroRNA-126 regulates

    DNA methylation in CD4+ T cells and contributes to systemic lupus erythematosus by

    targeting DNA methyltransferase 1. Arthritis Rheum. 2011;63:1376-86.

    39. Hamada S, Satoh K, Fujibuchi W, Hirota M, Kanno A, Unno J, et al. MiR-126 acts as a

    tumor suppressor in pancreatic cancer cells via the regulation of ADAM9. Mol Cancer Res.

    2012;10:3-10.

    40. Shintani Y, Higashiyama S, Ohta M, Hirabayashi H, Yamamoto S, Yoshimasu T, et al.

    Overexpression of ADAM9 in non-small cell lung cancer correlates with brain metastasis.

    Cancer Res. 2004;64:4190-6.

    41. Bakken AM, Protack CD, Roztocil E, Nicholl SM, Davies MG. Cell migration in

    response to the amino-terminal fragment of urokinase requires epidermal growth factor

    receptor activation through an ADAM-mediated mechanism. J Vasc Surg.

    2009;49:1296-303.

    42. de Graauw M, Cao L, Winkel L, van Miltenburg MH, le Devedec SE, Klop M, et al.

    Annexin A2 depletion delays EGFR endocytic trafficking via cofilin activation and

    enhances EGFR signaling and metastasis formation. Oncogene. 2013;33:2610-9.

    43. Li Y, Huang X, Zhang J, Ma K. Synergistic inhibition of cell migration by tetraspanin

    CD82 and gangliosides occurs via the EGFR or cMet-activated Pl3K/Akt signalling

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 23

    pathway. Int J Biochem Cell Biol. 2013;45:2349-58.

    44. Liu LY, Wang W, Zhao LY, Guo B, Yang J, Zhao XG, et al. Mir-126 inhibits growth of

    SGC-7901 cells by synergistically targeting the oncogenes PI3KR2 and Crk, and the

    tumor suppressor PLK2. Int J Oncol. 2014;45:1257-65.

    45. Zhou Y, Feng X, Liu YL, Ye SC, Wang H, Tan WK, et al. Down-Regulation of miR-126

    Is Associated with Colorectal Cancer Cells Proliferation, Migration and Invasion by

    Targeting IRS-1 via the AKT and ERK1/2 Signaling Pathways. PLoS One. 2013;8:e81203.

    46. Bouchie A. First microRNA mimic enters clinic. Nature biotechnology. 2013;31:577.

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 24

    Figure legends

    Figure 1. NGS-based discovery of miR-126 and validation of its downregulation in

    ESCC. (A) Heatmap and sample clustering analysis of the miRNAs differentially

    expressed in ECs and NMs, and the Pearson correlation coefficient are shown. The

    expression of miR-126 detected by RNA-Seq was confirmed by qPCR. (B) Levels of

    miRNA-126 in 30 matched ECs and NMs were detected using qPCR and normalized to

    U6 levels. The percentage of ESCC samples with upregulated, downregulated, and

    unchanged miR-126 expression was analyzed. The qPCR results are shown as the mean

    ± SD (***P < 0.001). (C) Relative levels of miR-126 were detected in ECa-109, KYSE-510,

    and HEECs by qPCR and normalized to U6 levels. The mean ± SD was determined from

    3 replicates (***P < 0.001). (D) ISH was used to detect the presence of miR-126 in ECs.

    Kaplan-Meier analysis showed that negative miR-126 expression was associated with

    poor disease-specific survival in ECs (P = 0.014, log-rank test).

    Figure 2. Hypermethylation of the Egfl7 promoter induces miR-126 downregulation

    in ESCC. (A) The location of the miR-126 gene (MIR 126) in Egfl7 was analyzed. The

    CpG island consisting of 12 CpG sites was used for bisulfite genomic sequencing (BSP)

    and methylation-specific PCR (MSP) analyses. (B) BSP was used to determine the

    methylation ratio at the Egfl7 promoter in ECs (n = 4) and matched NMs (n = 4). Black

    circles and open circles represent methylated (M) and unmethylated (U) CpG

    dinucleotides respectively. Data shown are the mean ± SD (**P < 0.01). (C) The

    correlation between miR-126 levels (black bars) and Egfl7 promoter methylation levels

    (red lines) was analyzed in the 4 pairs of esophageal samples. (D) Egfl7 promoter

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 25

    methylation levels and miR-126 expression were determined by MSP and qPCR

    respectively both in ECa-109 and KYSE-510 cells treated with 5-aza-dC (5 µM) or vehicle.

    MSP was carried out using the specific primers for the unmethylated (U) or methylated (M)

    Egf7 promoter sequence. The mean ± SD was determined from 3 replicates (***P <

    0.001).

    Figure 3. DNMT1-miR-126 regulatory circuit contributes to Egfl7 promoter

    hypermethylation. (A) DNMT1 RNA levels in 30 matched ECs and their NMs were

    detected using qPCR and normalized to GAPDH expression. Data shown are the mean ±

    SD values (**P < 0.01). DNMT1 RNA levels in cancer tissues negatively correlated with

    miR-126 levels (P = 0.0074; R = -0.4787). DNMT1 protein levels were measured in 6

    pair-matched ESCC samples and in cell lines. (B) ECa-109 and KYSE-510 cells were

    transfected with control RNA (si-Ctrl, 100 nM) or DNMT1 siRNA (si-DNMT1, 100 nM) as

    indicated. DNA from each sample was extracted, and Egfl7 promoter methylation levels

    were analyzed using MSP. M: methylated; U: unmethylated. (C) miR-126 expression was

    determined using qPCR and normalized to U6 expression. ADAM9 and DNMT1

    expression was detected and normalized to GAPDH. The mean ± SD was determined

    from 3 replicates (**P < 0.01). (D) DNMT1 expression in ECa-109 and KYSE-510 cells

    transfected with miR-126 and miR-NC mimics was determined by western blotting.

    Figure 4. Overexpression of miR-126 suppresses ESCC cell proliferation and

    migration. (A) ECa-109 and KYSE-510 cells were transfected with miR-126 (100 nM) or

    miR-NC (100 nM) mimics for 24 h, and cell viability and proliferation were assessed with

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 26

    the CCK-8 assay at 24, 48, 72, and 96 h. (B) Cell colonies were counted after transfection

    for 12 days. The colony formation ratio was calculated as: (visible colony number/original

    number of cells) × 100. (C) The total number of migrated cells was calculated at 48 h after

    transfection. (D) ECa-109 or KYSE-510 cells transfected with miR-126 or miR-NC mimics

    were injected subcutaneously into either side of the posterior flank of the same nude

    mouse, respectively (n = 4). Photographs show mice and tumors 4 weeks after tumor cell

    implantation. Tumor sizes are shown as the mean ± SD. Values represent the mean ± SD

    (*P < 0.05; **P < 0.01; ***P < 0.001).

    Figure 5. ADAM9 is the target of miR-126 in ESCC. (A) ADAM9 RNA levels in 30

    matched esophageal cancer tissues and their adjacent normal tissues were determined

    using qPCR and normalized to GAPDH expression. Data shown are the mean ± SD

    values (***P < 0.001). ADAM9 RNA levels in cancer tissues negatively correlated with

    endogenous miR-126 levels (P = 0.0019, R = -0.5446, n = 30). (B) ADAM9 protein levels

    were measured in 6 matched ESCC samples and 3 cell lines by western blotting. IHC

    straining for ADAM9 was performed in ECs and matched NMs. Magnification: 200×; 400×.

    (C) Alignment of miR-126 with the ADAM9 3′-UTR. MiR-126 (100 nM) or miR-NC (100 nM)

    mimics were co-transfected with the ADAM9-3′-UTR reporter plasmid, along with a control

    Renilla luciferase pRL-TK vector, into ECa-109 or KYSE-510 cells. The relative luciferase

    activity in each group was analyzed 48 h later and expressed relative to the activity in the

    miR-NC-transfected group. Data are representative of 3 independent experiments. Values

    represent the mean ± SD (***P < 0.001). (D) ADAM9 expression in ECa-109 and

    KYSE-510 cells transfected with miR-126 mimics or siRNA against DNMT1 (si-D) was

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • 27

    determined in by western blotting.

    Figure 6. The ADAM9/EGFR/AKT signaling directly mediates the suppressive role of

    miR-126 in ESCC. (A) ECa-109 and KYSE-510 cells were transfected with control siRNA

    (si-Ctrl, 100 nM) or ADAM9 siRNA (si-ADAM9, 100 nM) as indicated, and cell viability was

    assessed at different time points. Cell migration ability was determined with the transwell

    assay, and the migrated cell number was calculated. (B) ADAM9, pEGFR, EGFR, pAKT,

    AKT, pERK, and ERK protein levels were determined with western blotting and compared

    to GAPDH levels. (C) ECa-109 cells were treated with an EGFR inhibitor (AG1478, 10

    μM), and pEGFR, EGFR, pAKT, and AKT protein levels in ECa-109 cells were analyzed

    with western blotting. Cell viability and cell migration were determined at different time

    points after treatment with AG1478 (10 μM). The data are representative of 3 independent

    experiments. (D) Diagram summarizing the findings. Values represent the mean ± SD (*P

    < 0.05; **P < 0.01; ***P < 0.001).

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/

  • Published OnlineFirst December 15, 2014.Clin Cancer Res Ronghua Liu, Jie Gu, Pei Jiang, et al. ADAM9-EGFR-AKT signalingesophageal squamous cell carcinoma growth via DNMT1-microRNA126 epigenetic circuit contributes to

    Updated version

    10.1158/1078-0432.CCR-14-1740doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2014/12/16/1078-0432.CCR-14-1740.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/early/2014/12/13/1078-0432.CCR-14-1740To request permission to re-use all or part of this article, use this link

    Research. on June 12, 2021. © 2014 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 15, 2014; DOI: 10.1158/1078-0432.CCR-14-1740

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-14-1740http://clincancerres.aacrjournals.org/content/suppl/2014/12/16/1078-0432.CCR-14-1740.DC1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2014/12/13/1078-0432.CCR-14-1740http://clincancerres.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4Figure 5Figure 6