Open Access

Downregulation of miR‑29b targets DNMT3b to suppress cellular apoptosis and enhance proliferation in pancreatic cancer

Retraction in: /10.3892/mmr.2023.13049

  • Authors:
    • Li‑Hua Wang
    • Ju Huang
    • Cheng‑Rong Wu
    • Liu‑Ye Huang
    • Jun Cui
    • Zhi‑Zhi Xing
    • Chun‑Yu Zhao
  • View Affiliations

  • Published online on: November 23, 2017     https://doi.org/10.3892/mmr.2017.8145
  • Pages: 2113-2120
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As one of the most aggressive types of tumor, pancreatic cancer is a principal cause of tumor‑associated mortality. Negative associations between microRNA‑29 (miR‑29) and DNA methyltransferases (DNMT) 3a and 3b have been demonstrated to be associated with the carcinogenesis of a number of types of cancer; however, this has not been completely elucidated in pancreatic cancer. In the present study, pancreatic cancer tissues (n=15) and corresponding paracancerous tissues (n=15) were obtained and the results of reverse transcription‑quantitative polymerase chain reaction analysis indicated decreased expression of miR‑29b and enhanced mRNA expression of DNMT3b in pancreatic cancer tissues, compared with the corresponding paracancerous tissues. Increased protein expression of DNMT3b was demonstrated by western blotting and immunohistochemistry. In addition, the negative association between miR‑29b and DNMT3b was noted in pancreatic cancer tissues, and luciferase reporter assays confirmed that miR‑29b was able to directly target DNMT3b in vitro. Notably, miR‑29b overexpression was able to decrease cell viability and to promote the apoptosis by targeting DNMT3b, and the knockdown of DNMT3b exhibited consistent results in vitro and in vivo. The results of the present study suggested that miR‑29b, as a tumor suppressor, may be a novel target for the development of treatments for pancreatic cancer.

Introduction

As one of the most malignant types of human tumor, pancreatic cancer has been regarded as the fourth leading cause of cancer-induced mortality, due to the difficulty of diagnosis at the early stages and resistance to current treatment strategies, including radiotherapy and chemotherapy (e.g., gemcitabine and erlotinib). Although improvements have been made in the treatment and understanding of carcinogenesis, the overall survival of patients with pancreatic cancer has not notably altered and the 5-year survival rate remains <5% (14). Therefore, research is required to identify novel therapeutic strategies and potential targets for pancreatic cancer treatment.

Epigenetic modifications have been demonstrated to serve a role in carcinogenesis (5). The predominant epigenetic modification in mammals is DNA methylation. DNA methyltransferase (DNMT) enzymes 1, 3a and 3b catalyze the addition of a methyl group to the 5′ position of cytosine on DNA to regulate gene expression. It has been hypothesized that DNMT1 is primarily associated with maintenance of an established DNA methylation pattern and methylates newly-biosynthesized DNA, while DNMT3a and DNMT3b exhibit efficient de novo methylation activity (6). The expression levels of DNMTs have been demonstrated to be increased in a number of malignancies, including colon cancer (7), prostate cancer (8), breast cancer (9), leukemia (10) and pancreatic cancer (11), which contributes to the hyper-methylation of promoter CpG-rich regions of tumor suppressor genes. Wnt inhibitory factor-1 (WIF-1), as a tumor suppressor, may antagonize Wnt/β-catenin signaling; however, it was demonstrated to be silenced by overexpressed DNMT3a and DNMT3b-induced promoter hypermethylation in non-small cell lung cancer (12). In addition, patients with increased expression of DNMT3b exhibited a decreased rate of complete remission, and shorter disease-free and overall survival in cytogenetically normal-acute myeloid leukemia (CN-AML); therefore, DNMT3b may be a prognostic factor for CN-AML (13). However, the regulatory mechanisms of DNMTs in pancreatic cancer require further elucidation.

MicroRNAs (miRNAs) are endogenous small (19–25 nucleotides) non-coding RNAs, which negatively regulate gene expression by degrading or suppressing mRNA targets at the post-transcriptional level by recognizing complementary target sites in the 3′-untranslated region (UTR) (14). miRNAs have been demonstrated to be associated with numerous cellular functions, including the immune response, carcinogenesis and resistance to chemotherapy or radiotherapy, and are frequently aberrantly expressed in various types of tumor (15). A number of miRNAs are able to target epigenetic regulators, including DNMTs. miRNA (miR)-148a/152 has been reported to target DNMT1 in pancreatic cancer, gastric cancer and hepatic carcinoma (16). The miR-29 family was observed to target DNMT3a and DNMT3b in multiple myeloma (MM) (17), AML (18) and lung cancer (19). Amodio et al (17) reported that the overexpression of synthetic miR-29b mimics was able to decrease global DNA methylation by targeting DNMT3a and DNMT3b in MM cells, and to markedly increase the growth inhibitory and cell cycle arresting effects of the demethylating agent 5-azacitidine. However, little is known about the expression of miR-29b and the association between miR-29b and DNMT3b in pancreatic cancer tissues.

In the present study, the cell line PANC-1 was selected due to its wide applications in numerous areas of research into pancreatic cancer, including cytotoxicity (20), confocal imaging analysis (21), cellular communication and in vivo analysis (22). In the present study, it was observed that the expression of miR-29b was decreased and the mRNA expression of DNMT3b was increased in pancreatic cancer. It was noted that there existed a negative association between miR-29b and DNMT3b in pancreatic cancer tissues. In vitro, the overexpression of miR-29b inhibited the expression of DNMT3b by directly targeting the 3′-UTR of DNMT3b, and decreased the cell viability and promoted apoptosis. In addition, the knockdown of DNMT3b exhibited similar results and led to limited tumor growth in vivo, which demonstrated the potential of miR-29b as a candidate epi-therapeutic target in pancreatic cancer.

Materials and methods

Cell culture, tissue collection and reagents

The pancreatic cancer PANC-1 cell line was purchased from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) and cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (both from Thermo Fisher Scientific, Inc., Waltham, MA, USA), ampicillin and streptomycin at 37°C with 5% CO2. Pancreatic cancer tissues (n=15) and corresponding paracancerous tissues (n=15) were obtained from Yantai Yuhuangding Hospital (Yantai, China). All samples were collected between August 2015 and March 2016. A total of 6 female and 9 male patients were included (age range, 42–60 years). All diagnoses with primary pancreatic cancer were confirmed using hematoxylin and eosin staining assessed by experienced pathologists. No patients underwent preoperative chemotherapy and/or radiotherapy. Patients diagnosed with autoimmune or other malignant diseases, and pregnant or lactating individuals, were excluded from the experimental group. The present study was approved by the Ethics Committee of Yantai Yuhuangding Hospital, and informed written consent was obtained from all patients.

miR-29b mimics and inhibitors were purchased from Guangzhou RiboBio Co., Ltd. (Guangzhou, China). The sequences of the oligonucleotides used were as follows: miR-29b mimic, 5′-UAGCACCAUUUGAAAUCAGUGUU-3′, 5′-UAGCACCAUUUGAAAUCAGUGUU-3′ and 5′-AUCGUGGUAAACUUUAGUCACUU-3′; miR-29b inhibitor, 5′-AACACUGAUUUCAAAUGGUGCUA-3′. Reporter plasmids containing the full-length 3′-UTR (wild-type or mutant) of DNMT3b mRNA, and the overexpressed plasmid pcDNA3.1-DNMT3b were obtained from Shanghai GenePharma Co., Ltd. (Shanghai, China). Small interfering RNA (siRNA) targeting DNMT3b (5′-UUGUUGUUGGCAACAUCUGAA-3′) or control (5′-CAGAUGUUGCCAACAACAAGA-3′) was purchased from Guangzhou RiboBio Co., Ltd. Anti-DNMT3b (cat. no. 67259S) and GAPDH (cat. no. 2118S) antibodies were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA).

Immunohistochemistry

The expression of DNMT3b was analyzed immunohistochemically using 2-µm-thick, formalin-fixed and paraffin-embedded specimen sections. Slides were incubated in three washes of xylene for 5 min each, followed by two washes of 100% ethanol for 10 min, 95% ethanol for 10 min and ddH2O for 5 min. Antigen retrieval was performed by boiling in pH 9.0, 10 mM Tris/1 mM EDTA, blocking with 3% hydrogen peroxide for 10 min at room temperature and washing. The slides were incubated with anti-DNMT3b antibody (1:100 dilution) at 4°C overnight. The EnVision Detection System kit (Dako; Agilent Technologies, Inc., Santa Clara, CA, USA) was used to visualize the 3,3′-diaminobenzidine chromogen (room temperature for 20 min), followed by nuclear staining using hematoxylin solution (0.2%) at room temperature for 5 min. Neutral gum was used to cover the slides and they were dried at room temperature. Staining was visualized under an Olympus optical microscope (Olympus Corporation, Tokyo, Japan). Staining intensity and extent were graded to evaluate DNMT3b expression. Staining intensity was graded as following: Negative (score 0); weak (score 1); and strong (score 3). Staining extent was graded as following: Negative (score 0), ≤25% (the percentage of high-staining cells in the field); score 1, 25–50%; and score 2, ≤50% (score 3). The total score >4 was regarded as a high expression of DNMT3b.

Cell transfection

According to the manufacturer's instructions, 3×105 PANC-1 cells were seeded on a 6-well plate and transfected with miR-29b mimics or inhibitors, siRNA-DNMT3b, or pcDNA3.1-DNMT3B and its negative control at a concentration of 100 nM using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), and cultured for 24, 48 and 72 h at 37°C.

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA from tissues or PANC-1 cells was extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). First Stand cDNA was synthesized using Bestar™ qPCR RT kit (DBI-2220; DBI Bioscience, Shanghai, China). The mixture was maintained at 37°C for 15 min. qPCR was performed on cDNA using Bestar qPCR master mix SYBR Green (DBI-2043; DBI Bioscience). Bio-Rad iQ5 was used to detect the mRNA expression of genes. Thermocycling conditions were as follows: Pre-denaturation at 95°C for 2 min, denaturation at 95°C for 20 sec and annealing at 58°C for 20 sec for 40 cycles. The relative expression was calculated according to the 2−ΔΔCq method (23). The expression levels of DNMT3b were normalized to the gene expression of GAPDH. The primers are presented in Table I.

Table I.

Sequences of primers used in the present study.

Table I.

Sequences of primers used in the present study.

GenePrimer
GAPDHF: TGTTCGTCATGGGTGTGAA
GAPDHR: ATGGCATGGACTGTGGTCAT
DNMT3bF: GTCATCCGACACCTCTTCGC
DNMT3bR: ACCTCCTGGGTCCTGGCTCT
U6 snRNAF: CTCGCTTCGGCAGCACA
U6 snRNAR: AACGCTTCACGAATTTGCGT
URP CTCAACTGGTGTCGTGGA
hsa-miR-29bR: CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGAACACTG
hsa-miR-29bF: ACACTCCAGCTGGGTAGCACCATTTG

[i] F, forward; R, reverse; DNMT3b, DNA methyltransferase 3b; snRNA, small nuclear RNA; URP, universal reverse primer; miR, microRNA.

Cell Counting Kit-8 (CCK-8) assay

After transfection for 24, 48 and 72 h, PANC-1 cells were harvested and washed with PBS, and the CCK-8 reagent (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) mixed with DMEM was used for the cell viability assay. The absorbance was measured at 450 nm using a microplate reader.

Hoechst staining assay

PANC-1 cells transfected with miR-29b mimics or inhibitors, or siRNA-DNMT3b, were cultured at 37°C for 24 h and stained with the addition of 0.1 µg/ml Hoechst 33342 (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) to the culture medium. Fluorescence microscopy (Olympus IX71; Olympus Corporation) with a filter for Hoechst 33342 (365 nm) was used to detect the alterations in nuclear morphology.

Flow cytometry assay

For the apoptosis analysis, the cells were fixed in cold 70% ethanol at −20°C for 2 h. Cells were subsequently treated with 10 mg/ml RNase and stained with 5 µl (250 µg/ml) Annexin V mixed with 5 µl (1 µg/ml) propidium iodide (PI; eBioscience; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions, and quantified by flow cytometry using a FACSCalibur instrument (BD Biosciences, Franklin Lakes, NJ, USA). The data were analyzed using FlowJo software version 10 (FlowJo LLC, Ashland, OR, USA). Cells positive for Annexin V and PI were considered as apoptotic cells.

Western blotting

Cells for western blotting were collected and total protein was isolated from the cell samples using radioimmunoprecipitation assay lysis buffer (Thermo Fisher Scientific, Inc.). Western blot analysis was performed as previously described (18).

Prediction of miRNA and dual luciferase reporter assay

The gene DNMT3B was predicted to be targeted by miR-29b using miRWalk (www.umm.uni-heidelberg.de/apps/zmf/mirwalk). Comparative analysis was performed using three independent prediction programs (miRecords, mirecords.biolead.org; miRGator, genome.ewha.ac.kr/miRGator/miRGator.html; miRGen, www.diana.pcbi.upenn.edu/miRGen.html) to confirm the accuracy of the prediction. The 3′-UTR of DNMT3B contained the potential target sites of miR-29b, which were conserved among mammals.

The construction of a 3′-UTR-luciferase vector was performed. The genomic DNA was extracted from PANC-1 cells using GenElute Mammalian Genomic DNA Miniprep kit (Sigma-Aldrich; Merck KGaA). A fragment of the DNMT3B mRNA-3′-UTR was amplified using PrimeSTAR HS DNA Polymerase (Takara Biotechnology Co., Ltd., Dalian, China) with the following primers: XhoI forward, 5′-CCGCTCGAGAGGGACAGACATACATT-3′; NotI reverse, 5′-ATAAGAATGCGGCCGCCCCATATTTGTTACGTC-3′. The thermocycling conditions were as follows: Denaturation at 94°C for 30 sec, annealing at 58°C for 30 sec and elongation at 72°C for 45 sec for a total of 20 cycles. The PCR products were purified using a QIAquick Gel Extraction kit (Qiagen Sciences, Inc., Frederick, MD, USA). The purified PCR product was digested by NotI (Takara Biotechnology Co., Ltd.) and XhoI (Takara Biotechnology Co., Ltd.) and cloned into psiCHECK-2 Luciferase vector (Promega Corporation, Madison, WI, USA) downstream of the firefly luciferase gene to construct the 3′UTR luciferase vector of DNMT3B using T4 DNA ligase (Takara Biotechnology Co., Ltd.). According to the manufacturer's instructions, the luciferase reporter assay was performed using the Dual-Luciferase® Reporter Assay system (cat. no. E1910; Promega Corporation). The luciferase activity was detected 48 h following transfection using a Turner 20/20 luminometer (Turner BioSystems, Sunnyvale, CA, USA). The firefly luciferase activity in each sample was normalized to Renilla luciferase.

Tumor model

In order to investigate the tumor suppressive role of siRNA-DNMT3b in vivo, 18 male BALB/c−nu/nu T cell-deficient mice (age, 5–6 weeks; weight, 20±2 g) were purchased from Changzhou Cavens Laboratory Animal Co., Ltd. (Changzhou, China) and were divided into 3 groups (n=6 mice/group). Mice were maintained in specific pathogen-free conditions under 12-h light/dark cycles, at a temperature of 20–22°C and provided with sterilized water and food ad libitum. A total of 3×106 PANC-1 cells were subcutaneously injected into the rear flank of nude mice (6 mice/group). The siRNA-DNMT3b or negative control siRNA (10 nmol) were purchased from Guangzhou RiboBio Co., Ltd. and were delivered via intratumoral injection 6 times, 3 days apart, after the volume of the tumors had reached 1 cm3. The investigation conformed to the Guide for the Care and Use of Laboratory Animals published by the National Institutes of Health (publication no. 85–23, revised 1996; Bethesda, MD, USA). The study protocol was approved by the Ethics Committee of Yantai Yuhuangding Hospital.

Statistical analysis

The statistical analyses were performed using SPSS software (version 16.0; SPSS Inc., Chicago, IL, USA) and the Prism statistical software package (version 5.0; GraphPad Software Inc., La Jolla, CA, USA). Unpaired t-tests or Mann-Whitney U tests were used to compare the two groups, and multiple group comparisons were analyzed with one-way analysis of variance. The post hoc test employed was Tukey's range test. Pearson's correlation coefficient was used to analyze the correlation between the expression of DNMT3b and miR-29b. All of the experiments were performed ≥3 times. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-29b is negatively correlated with DNMT3b in pancreatic cancer tissues

In the present study, pancreatic cancer tissues (n=15) and corresponding paracancerous tissues (n=15) were subjected to RT-qPCR analysis. The results demonstrated that the expression of miR-29b was significantly decreased in pancreatic cancer tissues compared with the corresponding non-neoplastic tissues (Fig. 1A) and that the mRNA expression level of DNMT3b was upregulated during carcinogenesis (Fig. 1B).

The correlation between the expression of miR-29b and DNMT3b in pancreatic cancer tissues was analyzed, and it was observed that an increased level of DNMT3b was associated with a decreased level of miR-29b, and vice versa (Fig. 1C), indicating that DNMT3b was negatively associated with miR-29b in pancreatic cancer and that the increased protein expression of DNMT3b in pancreatic cancer may contribute to the decreased expression of miR-29b.

In order to further investigate the association between miR-29b and DNMT3b, the protein level of DNMT3b was determined. Immunohistochemistry of DNMT3b demonstrated consistent results, in that pancreatic cancer tissues exhibited increased expression of DNMT3b compared with corresponding non-neoplastic tissues (Fig. 1D). Western blot analysis was performed and it was confirmed that the protein level of DNMT3b was increased in pancreatic cancer tissues (Fig. 1E).

miR-29b may directly target and inhibit the expression of DNMT3b

The negative association between miR-29b and DNMT3b suggested that miR-29b may directly target the DNMT3b genes. Therefore, the pancreatic cancer PANC-1 cell line was induced to express miR-29b mimics or inhibitors in vitro, (Fig. 2) and PCR was used to validate the transfection efficiency (Fig. 2C). The present results demonstrated that the mRNA and protein levels of DNMT3b were significantly decreased by the miR-29b mimics and increased by the miR-29b inhibitors (Fig. 2A and B). The predicted genes that may be targeted by miR-29b were screened using miRWalk. Comparative analysis was performed using three independent prediction programs to confirm the accuracy of the prediction. It was observed that the 3′-UTR of DNMT3b contained miR-29b potential target sites (Fig. 2D). In order to confirm the association, a luciferase reporter assay was conducted using a vector containing the full-length 3′-UTR (wild-type or mutant) of DNMT3b mRNA in the PANC-1 cell line, and the results demonstrated that miR-29b mimics were able to significantly decrease the luciferase activity of wild-type DNMT3b-3′UTR, but not the mutant DNMT3b-3′UTR (Fig. 2E). The results of the present study demonstrated that DNMT3b may be directly targeted by miR-29b in pancreatic cancer.

Overexpression of miR-29b impairs the cell vitality and promotes the apoptosis

Since the enhanced expressions of DNMT3 were previously reported to promote carcinogenesis, which may be impaired by miR-29b, the present study investigated the potential therapeutic effect of miR-29b in pancreatic cancer. PANC-1 cells were transfected with miR-29b mimics or inhibitors for 24, 48 and 72 h, and cell viability was measured using a CCK-8 assay. The results indicated that the overexpression of miR-29b mimics decreased the viability of PANC-1 cells at 72 h (Fig. 3A). In addition, the apoptosis of PANC-1 cells was analyzed, and it was observed that an increased rate of apoptosis was induced by the overexpression of miR-29b mimics; conversely, the miR-29b inhibitors or negative control protected the cells from apoptosis (Fig. 3B and C). The results of Hoechst staining indicated that miR-29b mimics were able to markedly increase apoptosis, suggesting that miR-29b may be a tumor suppressor in pancreatic cancer (Fig. 3D).

DNMT3b is able to reverse miR-29b-induced apoptosis and decreased cell vitality

Firstly, the mRNA expression of DNMT3b in cells transfected with siRNA or DNMT3b overexpressed plasmid were detected using qRT-PCR (Fig. 4A). The negative association between miR-29b and DNMT3b in pancreatic cancer indicated that downregulation of DNMT3b produced results consistent with the overexpression of miR-29b. DNMT3b was overexpressed in PANC-1 cells transfected with miR-29b mimics and it was observed that the overexpression of DNMT3b inhibited the decrease in cell viability of the PANC-1 cells (Fig. 4B) and the increased apoptosis induced by miR-29b (Fig. 4C and D). Conversely, knockdown of DNMT3b reversed this effect (Fig. 4A-C). Hoechst staining indicated that DNMT3b was able to reverse the miR-29b-induced decrease in viability and increase in apoptosis (Fig. 4E).

Knockdown of DNMT3b inhibits tumor progression

The role of siRNA-DNMT3b in pancreatic cancer was investigated in vitro and in vivo. PANC-1 cells were transfected with siRNA-DNMT3b or negative control for 24, 48 and 72 h, and cell viability was measured using a CCK-8 assay. It was observed that knockdown of DNMT3b using siRNA significantly decreased the viability of PANC-1 cells at 48 and 72 h (Fig. 5A), which was consistent with the effects of miR-29b overexpression detailed above. The apoptosis of PANC-1 cells was observed to be increased by overexpression of siRNA-DNMT3b, and not the negative control (Fig. 5B and C). Hoechst staining indicated that siRNA-DNMT3b was able to markedly increase the apoptosis of PANC-1 cells (Fig. 5D). The results in vivo additionally indicated that the knockdown of DNMT3b inhibited tumor growth with decreased tumor volume compared with NC siRNA (Fig. 5E and F). The results of the present study demonstrated that overexpression of miR-29b mimics or the knockdown of DNMT3b may be of benefit in the treatment of pancreatic cancer.

Discussion

Pancreatic cancer, particularly pancreatic ductal adenocarcinoma (PDAC), is the fourth most common cause of cancer-associated mortality in the western world. The majority of cases are discovered and diagnosed at advanced stages. Local or regional recurrence rates for patients who have undergone surgical treatment may be as high as 60% (24). Resistance to numerous chemotherapy drug or radiotherapy for the treatment of advanced pancreatic cancer is a concern for patients, with these treatments exhibiting limited benefit for disease progression and survival (25). The present study investigated the role of miR-29b in pancreatic cancer and observed that DNMT3b, which has been reported to be associated with carcinogenesis, may be directly targeted by miR-29b and impair its function. Additionally, miR-29b was able to inhibit the viability of pancreatic cancer cells and promote apoptosis via DNMT3b, suggesting that miR-29b may act as a tumor suppressor in pancreatic cancer.

As the most widely studied epigenetic modification in mammals, DNMTs catalyze the addition of a methyl group at the 5-position of DNA cytosine and is important for fundamental processes, including embryonic development or differentiation, and immune balance; however, the aberrant expression or dysfunction of DNMTs has been identified to be involved in a number of pathologies, including carcinoma (6). The mRNA or protein expression of DNMT1, DNMT3a and DNMT3b is reportedly elevated in different types of malignancy, including pancreatic cancer (8), prostate cancer (26), breast cancer (9) and lymphoma (27). Patra et al (8) utilized immunohistochemical analyses, and demonstrated increased expression of DNMT1 in prostate cancer cell lines and cancer tissues, compared with a benign prostate epithelial cell line and benign prostatic hyperplasia tissues. In pancreatic cancer, Zhang et al (28) reported that the mRNA expression of the three DNMTs in patients with pancreatic cancer increased with the progression of carcinoma, and that patients with increased expression of DNMT3a and DNMT3b, and not DNMT1, exhibited increased tumor size and shorter overall survival times compared with those with lower levels of expression. The protein expression of DNMTs in pancreatic cancer has additionally been studied. Gao et al (11) observed co-expression of DNMT3a and DNMT3b in 23.9 and 77.3% of PDAC tissues, respectively, and demonstrated that the positive DNMT1 expression was correlated with poor overall survival. The results of the present study confirmed that the expression of DNMT3b was upregulated in pancreatic cancer, which was consistent with previous studies.

miRNAs are endogenous small non-coding RNAs (~22 nucleotides in length) with the capacity to regulate gene expression post-transcriptionally by binding to the 3′-UTR of target mRNAs. The miRNA-29 family, including miR-29a, miR-29b and miR-29c, was recently reported to be aberrantly expressed in a number of types of cancer (29). miR-29b is known for its role as a tumor suppressor (30). miR-29 may exert demethylation effects by directly targeting DNMT3a, DNMT3b, methylcytosine dioxygenase TET1 and thymine DNA glycosylase, which contributes to a decreased global level of DNA methylation in a number of tumor suppressors, including WIF-1, p15INK4b and estrogen receptor, and inhibits the progression of acute myeloid leukemia (12,18). The results of the present study indicated that miR-29b was downregulated in pancreatic cancer and, as a tumor suppressor, directly targeted DNMT3b; overexpression of miR-29b was able to markedly inhibit the cell viability of the pancreatic cancer PANC-1 cell line and promoted apoptosis, whereas DNMT3b was able to protect the tumor cells from miR-29b-induced apoptosis. In addition, it was observed that the knockdown of DNMT3b restricted tumor growth in vivo.

However, miR-29b has been demonstrated in previous studies to be a tumor promoter. Wang et al (25) reported that metastatic breast cancer cells and tissues exhibited increased miR-29b expression compared with low-metastasis breast cancer cells and tissues. Overexpression of miR-29b expression was demonstrated to promote cell migration, invasion and apoptotic resistance, through direct repression of phosphatase and tensin homolog expression in breast cancer cells, indicating that the different function of miR-29b may be associated with its targets in distinct types of cancer and cell types (25).

In conclusion, the present study investigated the role of miR-29b in pancreatic cancer, and demonstrated that it was downregulated in pancreatic cancer tissues and enhanced the expression of DNMT3b by targeting its 3′-UTR. Overexpression of miR-29b mimics markedly inhibited cell viability and promoted apoptosis in the pancreatic cancer PANC-1 cell line via the inhibition of DNMT3b. The results of the present study indicated that miR-29b in pancreatic cancer may be a tumor suppressor, to be utilized as a potential therapeutic agent by targeting DNMT3b.

Acknowledgements

The present study was supported by a grant from the Natural Science Foundation of Shandong (grant no. ZR2015HL076). The authors of the present study would like to acknowledge Guangzhou RiboBio Co., Ltd. (Guangzhou, China) for assistance with the design and synthesis of the small interfering RNA.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Demir IE, Friess H and Ceyhan GO: Neural plasticity in pancreatitis and pancreatic cancer. Nat Rev Gastroenterol Hepatol. 12:649–659. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Falasca M, Kim M and Casari I: Pancreatic cancer: Current research and future directions. Biochim Biophys Acta. 1865:123–132. 2016.PubMed/NCBI

5 

Subramaniam D, Thombre R, Dhar A and Anant S: DNA methyltransferases: A novel target for prevention and therapy. Front Oncol. 4:802014. View Article : Google Scholar : PubMed/NCBI

6 

Hamidi T, Singh AK and Chen T: Genetic alterations of DNA methylation machinery in human diseases. Epigenomics. 7:247–265. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Eads CA, Danenberg KD, Kawakami K, Saltz LB, Danenberg PV and Laird PW: CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltransferase overexpression. Cancer Res. 59:2302–2306. 1999.PubMed/NCBI

8 

Patra SK, Patra A, Zhao H and Dahiya R: DNA methyltransferase and demethylase in human prostate cancer. Mol Carcinog. 33:163–171. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Girault I, Tozlu S, Lidereau R and Biéche I: Expression analysis of DNA methyltransferases 1, 3A and 3B in sporadic breast carcinomas. Clin Cancer Res. 9:4415–4422. 2003.PubMed/NCBI

10 

Buchi F, Spinelli E, Masala E, Gozzini A, Sanna A, Bosi A, Ferrari G and Santini V: Proteomic analysis identifies differentially expressed proteins in AML1/ETO acute myeloid leukemia cells treated with DNMT inhibitors azacitidine and decitabine. Leuk Res. 36:607–618. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Gao J, Wang L, Xu J, Zheng J, Man X, Wu H, Jin J, Wang K, Xiao H, Li S and Li Z: Aberrant DNA methyltransferase expression in pancreatic ductal adenocarcinoma development and progression. J Exp Clin Cancer Res. 32:862013. View Article : Google Scholar : PubMed/NCBI

12 

Tan M, Wu J and Cai Y: Suppression of Wnt signaling by the miR-29 family is mediated by demethylation of WIF-1 in non-small-cell lung cancer. Biochem Biophys Res Commun. 438:673–679. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Niederwieser C, Kohlschmidt J, Volinia S, Whitman SP, Metzeler KH, Eisfeld AK, Maharry K, Yan P, Frankhouser D, Becker H, et al: Prognostic and biologic significance of DNMT3B expression in older patients with cytogenetically normal primary acute myeloid leukemia. Leukemia. 29:567–575. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Lee YS and Dutta A: MicroRNAs in cancer. Annu Rev Pathol. 4:199–227. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Melo SA and Esteller M: Dysregulation of microRNAs in cancer: Playing with fire. FEBS Lett. 585:2087–2099. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Amodio N, Rossi M, Raimondi L, Pitari MR, Botta C, Tagliaferri P and Tassone P: miR-29s: A family of epi-miRNAs with therapeutic implications in hematologic malignancies. Oncotarget. 6:12837–12861. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Amodio N, Leotta M, Bellizzi D, Di Martino MT, D'Aquila P, Lionetti M, Fabiani F, Leone E, Gullà AM, Passarino G, et al: DNA-demethylating and anti-tumor activity of synthetic miR-29b mimics in multiple myeloma. Oncotarget. 3:1246–1258. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Garzon R, Liu S, Fabbri M, Liu Z, Heaphy CE, Callegari E, Schwind S, Pang J, Yu J, Muthusamy N, et al: MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1. Blood. 113:6411–6418. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, Liu S, Alder H, Costinean S, Fernandez-Cymering C, et al: MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci USA. 104:pp. 15805–15810. 2007; View Article : Google Scholar : PubMed/NCBI

20 

Guzman EA, Xu Q, Pitts TP, Mitsuhashi KO, Baker C, Linley PA, Oestreicher J, Tendyke K, Winder PL, Suh EM and Wright AE: Leiodermatolide, a novel marine natural product, has potent cytotoxic and antimitotic activity against cancer cells, appears to affect microtubule dynamics, and exhibits antitumor activity. Int J Cancer. 139:2116–2126. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Arundhathi A, Chuang WH, Chen JK, Wang SE, Shyr YM, Chen JY, Liao WN, Chen HW, Teng YM, Pai CC and Wang CH: Prorenin receptor acts as a potential molecular target for pancreatic ductal adenocarcinoma diagnosis. Oncotarget. Jul 13–2016.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

22 

Su MJ, Aldawsari H and Amiji M: Pancreatic cancer cell exosome-mediated macrophage reprogramming and the role of microRNAs 155 and 125b2 transfection using nanoparticle delivery systems. Sci Rep. 6:301102016. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Liang D, Shi S, Xu J, Zhang B, Qin Y, Ji S, Xu W, Liu J, Liu L, Liu C, et al: New insights into perineural invasion of pancreatic cancer: More than pain. Biochim Biophys Acta. 1865:111–122. 2016.PubMed/NCBI

25 

Wang C, Bian Z, Wei D and Zhang JG: miR-29b regulates migration of human breast cancer cells. Mol Cell Biochem. 352:197–207. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Kobayashi Y, Absher DM, Gulzar ZG, Young SR, McKenney JK, Peehl DM, Brooks JD, Myers RM and Sherlock G: DNA methylation profiling reveals novel biomarkers and important roles for DNA methyltransferases in prostate cancer. Genome Res. 21:1017–1027. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Robaina MC, Mazzoccoli L, Arruda VO, Reis FR1, Apa AG, de Rezende LM and Klumb CE: Deregulation of DNMT1, DNMT3B and miR-29 s in Burkitt lymphoma suggests novel contribution for disease pathogenesis. Exp Mol Pathol. 98:200–207. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Zhang JJ, Zhu Y, Wu JL, Liang WB, Zhu R, Xu ZK, Du Q and Miao Y: Association of increased DNA methyltransferase expression with carcinogenesis and poor prognosis in pancreatic ductal adenocarcinoma. Clin Transl Oncol. 14:116–124. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Liu H, Wang B, Lin J and Zhao L: microRNA-29b: An emerging player in human cancer. Asian Pac J Cancer Prev. 15:9059–9064. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Yan B, Guo Q, Fu FJ, Wang Z, Yin Z, Wei YB and Yang JR: The role of miR-29b in cancer: Regulation, function, and signaling. Onco Targets Ther. 8:539–548. 2015.PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang LH, Huang J, Wu CR, Huang LY, Cui J, Xing ZZ and Zhao CY: Downregulation of miR‑29b targets DNMT3b to suppress cellular apoptosis and enhance proliferation in pancreatic cancer Retraction in /10.3892/mmr.2023.13049. Mol Med Rep 17: 2113-2120, 2018
APA
Wang, L., Huang, J., Wu, C., Huang, L., Cui, J., Xing, Z., & Zhao, C. (2018). Downregulation of miR‑29b targets DNMT3b to suppress cellular apoptosis and enhance proliferation in pancreatic cancer Retraction in /10.3892/mmr.2023.13049. Molecular Medicine Reports, 17, 2113-2120. https://doi.org/10.3892/mmr.2017.8145
MLA
Wang, L., Huang, J., Wu, C., Huang, L., Cui, J., Xing, Z., Zhao, C."Downregulation of miR‑29b targets DNMT3b to suppress cellular apoptosis and enhance proliferation in pancreatic cancer Retraction in /10.3892/mmr.2023.13049". Molecular Medicine Reports 17.2 (2018): 2113-2120.
Chicago
Wang, L., Huang, J., Wu, C., Huang, L., Cui, J., Xing, Z., Zhao, C."Downregulation of miR‑29b targets DNMT3b to suppress cellular apoptosis and enhance proliferation in pancreatic cancer Retraction in /10.3892/mmr.2023.13049". Molecular Medicine Reports 17, no. 2 (2018): 2113-2120. https://doi.org/10.3892/mmr.2017.8145