miR‑212 regulated by HIF‑1α promotes the progression of pancreatic cancer

  • Authors:
    • Hui Yue
    • Lin Liu
    • Zhenguo Song
  • View Affiliations

  • Published online on: January 29, 2019     https://doi.org/10.3892/etm.2019.7213
  • Pages: 2359-2365
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA‑212 (miR‑212) is dysregulated in numerous tissues and cancer types and serves a role in the progression of human cancer. However, the function and mechanism of miR‑212 in the development of pancreatic ductal adenocarcinoma (PDAC) remain unknown, particularly in a hypoxic microenvironment. In the present study, miR‑212 expression was observed to be significantly upregulated in PDAC tissues compared with normal tissues. Clinical data analysis indicated that miR‑212 was positively associated with a large tumor size, Tumor‑Node‑Metastasis stage, lymph node metastasis and vessel invasion, and influenced the overall survival time. Notably, there was a positive association between the expression of hypoxia‑inducible factor-1α (HIF‑1α) and miR‑212 in vivo and in vitro in hypoxic conditions. Mechanistically, HIF‑1α bound directly to a hypoxia response element in the miR‑212 promoter region and activated miR‑212 expression in PDAC cells. Collectively, these results demonstrated that HIF‑1α positively regulated miR‑212 expression and resulted in PDAC progression.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-associated mortality in the USA and likely to rise to the second by the year 2030 (1). The 5-year overall survival rate is only 8.0% and the median survival time of patients is <6 months (2). Despite rapid improvements in PDAC diagnosis and therapy, the overall survival time of patients with pancreatic cancer has not changed substantially (3). Patients with PDAC mostly succumb due to recurrence and metastasis to distant organs (4). Furthermore, there remains a lack of underlying biomarkers and therapeutic targets for PDAC diagnosis and therapy. Therefore, it is necessary to identify novel biomarkers and therapeutic targets in order to improve the efficacy of treatment for patients with PDAC.

MicroRNAs (miRNAs/miRs) are small noncoding RNAs, usually 18–25 nucleotides in length, which regulate protein translation by binding to the 3′-untranslated region of their target mRNAs (5). miRNAs serve roles in numerous physiological and pathological processes including apoptosis, the cell cycle, cellular growth, proliferation, differentiation, metabolism and aging (6). Accumulating evidence reveals that miRNA dysfunction influences the metastasis, chemo-radioresistance and overall survival time of patients with cancer, including patients with PDAC (710). Previous reports have revealed miRNA involvement in PDAC progression, including roles of miR-216b (11), miR-10a-5p (10), miR-374b-5p (12), miR-1271 (13), miR-7-5p (14), miR-4295 (15) and miR-185 (16). However, the molecular mechanisms underlying the involvement of miRNAs in PDAC progression remain unclear.

miR-212, a cancer-associated miRNA located on chromosome 17p13.3, participates in the progression of different types of human cancer (17). It has been demonstrated that miR-212, as an oncogene, is upregulated in breast cancer (18) and pancreatic cancer (19); and as a suppressor gene, is downregulated in hepatocellular carcinoma (20), prostate cancer (21), non-small cell lung cancer (22) and nasopharyngeal carcinoma (23). Furthermore, aforementioned studies revealed that dysregulated miR-212 has an influence on the progression of a number of solid malignancies. However, to the best of our knowledge, the function and regulatory mechanism of miR-212 in PDAC has not yet been examined.

The present study aimed to investigate the function and expression mechanism of miR-212 in PDAC. The results revealed that miR-212 was overexpressed in PDAC specimens and associated with certain clinicopathological features, particularly the overall survival time of patients. miR-212 overexpression in PDAC was revealed to be mediated by hypoxia-inducible factor-1α (HIF-1α), which directly bound to the miR-212 promoter and upregulated its activity. The results of the present study indicate that miR-212 is a novel direct target of HIF-1α and may function as a novel biomarker for the diagnosis and therapy of PDAC.

Materials and methods

Cell culture and hypoxic treatment

Human PDAC cell lines MiaPaca2 and AsPc1, and the 293 cell line, were obtained from the Cell Bank of Type Culture Collection of Chinese Academy of Sciences (Shanghai, China). These cells were cultured at 37°C in a humidified atmosphere of 5% CO2 using Dulbecco's modified Eagle's medium with 10% fetal bovine serum (both Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). For hypoxic treatment, cells were placed in a modulator incubator chamber (Thermo Fisher Scientific, Inc.) with 93.5% N2, 5% CO2 and 1.5% O2 for 6 and 12 h. Oxygen at 21% was used during normoxia condition.

With an approval from the Ethics Committee of the Tianjin Cancer Institute and Hospital (Tianjin, China), PDAC and adjacent normal pancreatic tissue specimens were obtained from patients (age, 51–75 years; male, 12; female, 29) who underwent surgical resection subsequent to the histopathological diagnosis of PDAC (n=41). Patients were included if they were diagnosed with PDAC via pathological diagnosis following radical resection and if they were willing to participate. Patients were excluded if they did not undergo radical resection. Written informed consent was obtained from individuals or their guardians. Samples were collected between January and December 2015 at Tianjin Cancer Institute and Hospital with a follow-up period of 2 years to analyze over survival.

miR-212 structure search

miRbase (www.mirbase.org) was searched using ‘miR-212’ and MI0000288 was selected to identify precursor and mature miR-212 structures. The GC box and the TATA box located at the transcription initiation point of miR-212 were identified using the University of California Santa Cruz genome browser (genome.ucsc.edu) by selecting genomes and searching ‘miR-212’ and selecting ‘MIR212 (ENST00000586026.1)’ for further studies. Five hypoxia response elements (HREs) were identified (5′-A/GCGTG-3′) to potentially to bind HIF-1α at the miR-212 promoter.

Small interfering RNA (siRNA), plasmid constructs and transient transfection

siRNAs against HIF-1α (siHIF-1α) were designed and synthesized by Shanghai GenePharma Co., Ltd. (Shanghai, China), including the following duplexes: siHIF-1α-1, forward, 5′-CCAGCAGACUCAAAUACAATT-3′ and reverse, 5′-UUGUAUUUGAGUCUGCUGGTT-3′; siHIF-1α-2, forward, 5′-GCCUCUUUGACAAACUUAATT-3′ and reverse, 5′-UAAGUUUGUCAAAGAGGCTT-3′; siHIF-1α-3, forward, 5′-CAGGCCACAUUCACGUAUATT-3′ and reverse, 5′-UAUACGUGAAUGUGGCCUGTT-3′; and siNC, forward, 5′-UUCUCCGAACGUGUCACGUTT-3′ and reverse, 5′-ACGUGACACGUUCGGAGAATT-3′. Following the examination transfection efficacy, siHIF-1α-3 was selected for subsequent experiments. The human HIF-1α cDNA was cloned into the pcDNA3.1 plasmid expression vector (Thermo Fisher Scientific, Inc.) for overexpression experiments and the empty vector was used as a control. siRNAs (50 nM) and plasmids (2 µg) were prepared and transfected into the PDAC cell lines using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) (18,19). At 48 h of transfection, mRNA and protein levels were tested by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting.

RT-qPCR

Total RNA was isolated from the transfected cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and used for first-strand cDNA synthesis (42°C for 20 min; 95°C for 5 min; and 5°C for 5 min) using the First-Strand Synthesis System for RT-PCR (Takara Biotechnology Co., Ltd., Dalian, China). Each sample was processed in triplicate using a SYBR-Green PCR kit (cat no RR820A; Takara Biotechnology Co., Ltd.) with the following thermocycling conditions: 95°C for 30 sec followed by 40 cycles of 95°C for 5 sec and 60°C for 30 sec. Relative mRNA expression levels were quantified using the 2ΔΔCq method (24). U6 RNA and β-actin were used as reference genes. The following primers were obtained from Shanghai GenePharma Co., Ltd. (Shanghai, China): has-miR-212 sense, 5′-ACACTCCAGCTGGGTAACAGTCTCCAGTC-3′; HIF-1α sense, 5′-AGAAACCACCTATGACCTGC-3′ and antisense, 5′-GTCGTGCTGAATAATACCACTC-3′; U6 sense, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ and antisense, 5′-CGCTTCACGAATTTGCGTGTCAT-3′; β-actin sense, 5′-CTGGAACGGTGAAGGTGACA-3′ and antisense, 5′-AAGGGACTTCCTGTAACAATGCA-3′.

Western blot analysis

Whole-cell extracts were prepared by lysing cells with radioimmunoprecipitation assay lysis buffer in a proteinase inhibitor cocktail (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). Protein concentrations were quantified using a bicinchoninic acid protein assay kit (Pierce; Thermo Fisher Scientific, Inc.). Protein lysates (20 µg/lane) were separated on 10% SDS-PAGE gels and transferred to polyvinylidene fluoride membrane. Membranes were blocked with PBS plus 0.1% Tween-20 with 5% non-fat milk at room temperature for 2 h. Target proteins were detected by probing with antibodies against HIF-1α (1:1,000; cat. no. ab16066; Abcam, Cambridge, UK) and β-actin (1:1,000; cat. no. ab8226; Abcam) overnight at 4°C. Following, membranes were incubated with anti-mouse IgG second antibody (1:1,000; cat. no. ab190475; Abcam) at room temperature for 2 h. Specific proteins were visualized using an enhanced chemiluminescence detection reagent (cat. no. #32106; Pierce; Thermo Fisher Scientific, Inc.).

Chromatin immunoprecipitation (ChIP) assay

A chromatin immunoprecipitation assay was performed using a commercial kit (cat no. #17-295; Upstate Biotechnology, Inc., Lake Placid, NY, USA) according to the manufacturer's protocol. MiaPaca2 and 293 cells were treated under hypoxia conditions (93.5% N2, 5% CO2 and 1.5% O2) in the modulator incubator chamber at 37°C for 6 and 12 h. Oxygen at 21% was used during normoxia. ChIP assays were performed to confirm HREs that bind to HIF-1α. The following groups were analyzed: Negative control (NC), positive control (PC), normoxia (N) and hypoxia (H).

Dual-luciferase reporter assay

Genomic DNA fragments of the miR-212 gene, spanning from +1 to −2,000 relative to the transcription initiation sites were generated by PCR and inserted into pGL3-Basic vectors (pGL3-miR-212; cat. no. E1751; Promega Corporation, Madison, WI, USA). A mutation (mut) HRE of miR-212 was created for dual-luciferase reporter assays in addition to wild type fragments. All constructs were sequenced to confirm their identity. Plasmids (2 µg) were transfected into MiaPaca2 and 293 cells using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.). At 48 h of transfection, cells were prepared for Dual-Luciferase Reporter assays. Luciferase activity was measured using the Dual-Luciferase Reporter assay system (Promega Corporation) as previously described (25). Renilla activity was used for normalization.

Statistical analysis

Differences between groups were compared using a Student's t-test or one-way analysis of variance followed by a least significant difference post hoc test. Categorical data were analyzed using either Fisher's exact test or the χ2 test, as appropriate. Each experiment was conducted independently at least three times, and the values are presented as the mean ± standard error of the mean unless otherwise stated. Statistical analyses were performed using SPSS software (version 21.0; IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-212 and HIF-1α mRNA are overexpressed in PDAC and associated with the clinicopathological features and prognosis of patients with PDAC

The expression level of miR-212 and HIF-1α mRNA was detected by RT-qPCR using paired specimens from patients with PDAC. The data indicated that miR-212 and HIF-1α mRNA expression levels were significantly upregulated in PDAC samples compared with adjacent normal pancreatic tissue samples (P<0.05; Fig. 1A). There was a positive association identified between miR-212 and HIF-1α at the mRNA level. Subsequently, miR-212 expression level in PDAC samples was assessed by RT-qPCR and it was revealed that there was a significant association between miR-212 expression level and tumor size, lymph node metastasis and vessel invasion among patients with PDAC (all P<0.05; Table I). miR-212 was quartered according to the range of miR-212 expression, <25% was considered as negative expression (−), 26–50% was low expression (+), 51–75% was termed medium expression (++) and >76% identified high expression (+++). Patients with PDAC with a high expression level of miR-212 and HIF-1α mRNA had a significantly worse overall survival time compared with patients with a low expression level (P=0.022 and P=0.028, respectively; Fig. 1B and C), suggesting that miR-212 and HIF-1α may serve a role in the survival of patients with PDAC. The results revealed that miR-212 and HIF-1α are overexpressed in PDAC samples and their expression was associated with clinicopathological features in PDAC, particularly the overall survival time of patients.

Table I.

Association between miR-212 expression and clinicopathological features in patients with pancreatic ductal adenocarcinoma.

Table I.

Association between miR-212 expression and clinicopathological features in patients with pancreatic ductal adenocarcinoma.

miR-212

Variables−/+++/+++χ2 testP-value
Tumor size (cm) 10.0410.004
  T162
  T2627
Histological grade 0.2770.702
  G1/G21022
  G327
pTNM stage 2.3050.165
  I/II1017
  III/IV212
Lymph node metastasis
  N0765.5550.029
  N1523
Vessel invasion
  Negative654.6400.041
  Positive624
HIF-1α 16.443<0.001
  −/+82
  ++/+++427

[i] P-values were calculated using a χ2 test. miR, microRNA; HIF-1α, hypoxia-inducible factor-1α; pTNM, pathological Tumor-Node-Metastasis; (−), negative expression (<25%); (+), low expression (26–50%); (++) medium expression (51–75%); (+++) high expression (>76%)

Structure of miR-212

The present study used the miRBase database to identify the precursor and mature miR-212 structures (Fig. 2A). The GC box and the TATA box located at the transcription initiation point of miR-212 were identified using the University of California Santa Cruz genome browser (Fig. 2B) and a total of five HREs were identified (5′-A/GCGTG-3′) that were potentially to bind with HIF-1α at the miR-212 promoter (Fig. 2B). Therefore, HIF-1α may bind to the HRE of miR-212 to initialize the transcription of miR-212.

miR-212 and HIF-1α mRNA levels were upregulated under in vitro hypoxic conditions

To further confirm the positive association between miR-212 and HIF-1α mRNA expression, the pancreatic cancer cell lines (MiaPaca2 and AsPc1) were used for in vitro experiments in a hypoxic microenvironment. MiaPaca2 and AsPc1 cell lines were maintained in hypoxic conditions for different durations (6 and 12 h) and RT-qPCR was used to assess the mRNA expression of miR-212 and HIF-1α. The results indicated that the expression levels of miR-212 and HIF-1α mRNA were significantly upregulated following hypoxia stimulation at 6 h compared with 0 h and at 12 h compared with 6 h (P<0.05; Fig. 3A and B). Additionally, HIF-1α protein expression levels were markedly increased in hypoxia conditions at different time points in MiaPaca2 and AsPc1 cells (Fig. 3C and D). Furthermore, it was revealed that miR-212 and HIF-1α mRNA expression levels were positively associated in MiaPaca2 and AsPc1 cell lines in hypoxic milieu 12 h.

HIF-1α influences the expression of miR-212

In order to confirm the influence of HIF-1α on miR-212 expression, siHIF-1α and HIF-1α plasmid were transfected into MiaPaca2 and AsPc1 cell lines. The results indicated that miR-212 expression was significantly downregulated in MiaPaca2 and AsPc1 cell lines transfected with siHIF-1α compared with control cells (P<0.05; Fig. 4A and B). By contrast, miR-212 expression was significantly upregulated in MiaPaca2 and AsPc1 cell lines transfected with the HIF-1α overexpression plasmid compared with control cells (P<0.05; Fig. 4A and B). Fig. 4C and D presents the protein expression levels of HIF-1α subsequent to transfection with siHIF-1α and HIF-1α plasmid in MiaPaca2 and AsPc1 cell lines, respectively. The data implied that HIF-1α influenced the expression of miR-212.

HIF-1α directly regulates the expression of miR-212 in PDAC by binding to the HRE at the miR-212 gene promoter

The above results indicated that HIF-1α may serve a role in modulating miR-212 expression; therefore, the present study further investigated whether HIF-1α directly targeted miR-212. To demonstrate the binding of HIF-1α to the miR-212 promoter in pancreatic cancer cells, a chromatin immunoprecipitation assay was performed in MiaPaca2 cells at 1.5% O2 or 21% O2. In chromatin fractions pulled-down by an anti-HIF-1α antibody, only the HRE of the miR-212 promoter located at −481 bp was detected (Fig. 5A). The binding affinity markedly increased under hypoxic conditions compared with normal conditions.

To determine whether HIF-1α binding to the miR-212 promoter activated its transcription, miR-212 luciferase promoter vectors were constructed and transfected with or without a HIF-1α expression vector (pCDH-HIF-1α) into MiaPaca2 and 293 cell lines. Luciferase analysis demonstrated that HIF-1α overexpression significantly increased miR-212 promoter activity in HRE −481 bp in MiaPaca2 and 293 cell lines compared with the EV group (P<0.01; Fig. 5B and C). Additionally, the miR-212 promoter activity was not induced in the pGL3-HRE mut group. Altogether, these results suggested that HIF-1α not only directly bound to the miR-212 promoter but also upregulated the activity of miR-212 in PDACs.

Discussion

The present study investigated the association between the expression levels of miR-212 and HIF-1α in PDAC. The results revealed that miR-212 and HIF-1α mRNA expression levels were higher in PDAC samples compared with in adjacent normal pancreatic tissues. miR-212 has been demonstrated to be upregulated and downregulated in different types of solid tumors, as an oncogene and suppressor gene, respectively (17). Using clinical data analysis, associations were identified between the expression levels of miR-212 and tumor size, lymph node metastasis and vessel invasion among patients with PDAC. Notably, patients with PDAC with high miR-212 expression had significantly worse overall survival time compared with patients with low expression. The aforementioned data demonstrated that miR-212 was overexpressed in PDAC samples, and, as an oncogene, influenced the overall survival of patients with PDAC.

PDAC is characterized by stroma fibrosis, named desmoplasia, which hampers the efficacy of therapeutic treatments for PDAC (26). Desmoplasia causes hypovascularity and poor blood supply in pancreatic cancer tissues resulting in a hypoxic milieu (27). In a hypoxic microenvironment, PDAC cells constitutively express HIF-1α (28). HIF-1α as a nuclear factor may directly bind to the HREs of target genes and regulate their transcription (29). Clinical data indicated that the expression of miR-212 was correlated with the expression of HIF-1α mRNA in PDAC tissues. The expression levels of miR-212 and HIF-1α mRNA were markedly increased in hypoxic compared with normoxic conditions in vitro. In order to demonstrate the effect of HIF-1α on miR-212 expression, a HIF-1α overexpression plasmid and siHIF-1α were transfected into PDAC cell lines. The results demonstrated that miR-212 expression was upregulated subsequent to HIF-1α overexpression. By contrast, miR-212 expression was downregulated following transfection with siHIF-1α. These results revealed that there was a positive association between the expression of miR-212 and HIF-1α mRNA.

To confirm that HIF-1α regulated miR-212 expression, chromatin immunoprecipitation and dual-luciferase assays were performed. The results revealed that HIF-1α was able to directly bind to the HRE (−481 bp) of the miR-212 promoter and the binding affinity increased under hypoxic conditions. Notably, the overexpression of HIF-1α increased miR-212 promoter activity in PDAC and 293 cell lines.

In conclusion, the present study confirmed that miR-212 was upregulated in PDAC samples, compared with normal adjacent pancreatic tissues, and was associated with a poor clinical prognosis. Furthermore, a positive association between miR-212 and HIF-1α mRNA expression was identified in PDAC tissues and in vitro experiments. The results revealed that HIF-1α regulated miR-212 expression by binding to the HRE. Therefore, miR-212 may function as a novel biomarker for diagnosis, and the inhibition of miR-212 expression may be an effective therapeutic method for the treatment of patients with PDAC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Tianjin Science and Technology Commission Joint Program (grant no. 15JCYBJC49600).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

ZS was responsible for conception, design, study supervision, and manuscript writing, review and revision. ZS, HY and LL were responsible for the development of methodology, and acquisition, analysis and interpretation of data.

Ethics approval and consent to participate

Ethics approval was obtained from the Ethics Committee of the Tianjin Cancer Institute and Hospital (Tianjin, China). Written informed consent was obtained from individuals or their guardians.

Patient consent for publication

Patient consent for publication was obtained.

Competing interests

The authors declare that they have no competing interests.

References

1 

Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM and Matrisian LM: Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 74:2913–2921. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Adamska A, Domenichini A and Falasca M: Pancreatic ductal adenocarcinoma: Current and evolving therapies. Int J Mol Sci. 18:E13382017. View Article : Google Scholar : PubMed/NCBI

4 

Lewis AR, Pihlak R and McNamara MG: The importance of quality-of-life management in patients with advanced pancreatic ductal adenocarcinoma. Curr Probl Cancer. 42:26–39. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Uddin A and Chakraborty S: Role of miRNAs in lung cancer. J Cell Physiol. 20–Apr;2018.(Epub ahead of print). View Article : Google Scholar

6 

Hwang HW and Mendell JT: MicroRNAs in cell proliferation, cell death, and tumorigenesis. Br J Cancer. 94:776–780. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Wang Y, Bao W, Liu Y, Wang S, Xu S, Li X, Li Y and Wu S: miR-98-5p contributes to cisplatin resistance in epithelial ovarian cancer by suppressing miR-152 biogenesis via targeting Dicer1. Cell Death Dis. 9:4472018. View Article : Google Scholar : PubMed/NCBI

8 

Idichi T, Seki N, Kurahara H, Fukuhisa H, Toda H, Shimonosono M, Okato A, Arai T, Kita Y, Mataki Y, et al: Molecular pathogenesis of pancreatic ductal adenocarcinoma: Impact of passenger strand of pre-miR-148a on gene regulation. Cancer Sci. 109:2013–2026. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Liu B, Yang H, Taher L, Denz A, Grützmann R, Pilarsky C and Weber GF: Identification of prognostic biomarkers by combined mRNA and miRNA expression microarray analysis in pancreatic cancer. Transl Oncol. 11:700–714. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Xiong G, Huang H, Feng M, Yang G, Zheng S, You L, Zheng L, Hu Y, Zhang T and Zhao Y: MiR-10a-5p targets TFAP2C to promote gemcitabine resistance in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res. 37:762018. View Article : Google Scholar : PubMed/NCBI

11 

Liu YA, Zhang Y, Zheng Z, Li K, Wu XH, Du QG, Ye X, Wang L and Zhu L: MicroRNA-216b reduces growth, migration and invasion of pancreatic ductal adenocarcinoma cells by directly targeting ρ-associated coiled-coil containing protein kinase 1. Oncol Lett. 15:6745–6751. 2018.PubMed/NCBI

12 

Sun D, Wang X, Sui G, Chen S, Yu M and Zhang P: Downregulation of miR-374b-5p promotes chemotherapeutic resistance in pancreatic cancer by upregulating multiple anti-apoptotic proteins. Int J Oncol. 14–Mar;2018.(Epub ahead of print). View Article : Google Scholar

13 

Xie F, Huang Q, Liu CH, Lin XS, Liu Z, Liu LL, Huang DW and Zhou HC: MiR-1271 negatively regulates AKT/MTOR signaling and promotes apoptosis via targeting PDK1 in pancreatic cancer. Eur Rev Med Pharmacol Sci. 22:678–686. 2018.PubMed/NCBI

14 

Zhu W, Wang Y, Zhang D, Yu X and Leng X: MiR-7-5p functions as a tumor suppressor by targeting SOX18 in pancreatic ductal adenocarcinoma. Biochem Biophys Res Commun. 497:963–970. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Yuan Q, Zhang Y, Li J, Cao G and Yang W: High expression of microRNA-4295 contributes to cell proliferation and invasion of pancreatic ductal adenocarcinoma by the down-regulation of Glypican-5. Biochem Biophys Res Commun. 497:73–79. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Xia D, Li X, Niu Q, Liu X, Xu W, Ma C, Gu H, Liu Z, Shi L, Tian X, Chen X and Zhang Y: MicroRNA-185 suppresses pancreatic cell proliferation by targeting transcriptional coactivator with PDZ-binding motif in pancreatic cancer. Exp Ther Med. 15:657–666. 2018.PubMed/NCBI

17 

Wanet A, Tacheny A, Arnould T and Renard P: miR-212/132 expression and functions: Within and beyond the neuronal compartment. Nucleic Acids Res. 40:4742–4753. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Xie M, Fu Z, Cao J, Liu Y, Wu J, Li Q and Chen Y: MicroRNA-132 and microRNA-212 mediate doxorubicin resistance by down-regulating the PTEN-AKT/NF-κB signaling pathway in breast cancer. Biomed Pharmacother. 102:286–294. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Wu Z, Zhou L, Ding G and Cao L: Overexpressions of miR-212 are associated with poor prognosis of patients with pancreatic ductal adenocarcinoma. Cancer Biomark. 18:35–39. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Jia P, Wei G, Zhou C, Gao Q, Wu Y, Sun X and Li X: Upregulation of MiR-212 inhibits migration and tumorigenicity and inactivates Wnt/β-catenin signaling in human hepatocellular carcinoma. Technol Cancer Res Treat. 17:15330346187652212018. View Article : Google Scholar : PubMed/NCBI

21 

Hu B, Jin X and Wang J: MicroRNA-212 targets mitogen-activated protein kinase 1 to inhibit proliferation and invasion of prostate cancer cells. Oncol Res. 26:1093–1102. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Tang T, Huan L, Zhang S, Zhou H, Gu L, Chen X and Zhang L: MicroRNA-212 functions as a tumor-suppressor in human non-small cell lung cancer by targeting SOX4. Oncol Rep. 38:2243–2250. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Jiang C, Wang H, Zhou L, Jiang T, Xu Y and Xia L: MicroRNA-212 inhibits the metastasis of nasopharyngeal carcinoma by targeting SOX4. Oncol Rep. 38:82–88. 2017. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Zhao T, Jiang W, Wang X, Wang H, Zheng C, Li Y, Sun Y, Huang C, Han ZB, Yang S, et al: ESE3 inhibits pancreatic cancer metastasis by upregulating E-cadherin. Cancer Res. 77:874–885. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Whatcott CJ, Diep CH, Jiang P, Watanabe A, LoBello J, Sima C, Hostetter G, Shepard HM, Von Hoff DD and Han H: Desmoplasia in primary tumors and metastatic lesions of pancreatic cancer. Clin Cancer Res. 21:3561–3568. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Liu H, Ma Q, Xu Q, Lei J, Li X, Wang Z and Wu E: Therapeutic potential of perineural invasion, hypoxia and desmoplasia in pancreatic cancer. Curr Pharm Des. 18:2395–2403. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Akakura N, Kobayashi M, Horiuchi I, Suzuki A, Wang J, Chen J, Niizeki H, Kawamura Ki, Hosokawa M and Asaka M: Constitutive expression of hypoxia-inducible factor-1alpha renders pancreatic cancer cells resistant to apoptosis induced by hypoxia and nutrient deprivation. Cancer Res. 61:6548–6554. 2001.PubMed/NCBI

29 

Dengler VL, Galbraith M and Espinosa JM: Transcriptional regulation by hypoxia inducible factors. Crit Rev Biochem Mol Biol. 49:1–15. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 17 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yue H, Liu L and Song Z: miR‑212 regulated by HIF‑1α promotes the progression of pancreatic cancer. Exp Ther Med 17: 2359-2365, 2019
APA
Yue, H., Liu, L., & Song, Z. (2019). miR‑212 regulated by HIF‑1α promotes the progression of pancreatic cancer. Experimental and Therapeutic Medicine, 17, 2359-2365. https://doi.org/10.3892/etm.2019.7213
MLA
Yue, H., Liu, L., Song, Z."miR‑212 regulated by HIF‑1α promotes the progression of pancreatic cancer". Experimental and Therapeutic Medicine 17.3 (2019): 2359-2365.
Chicago
Yue, H., Liu, L., Song, Z."miR‑212 regulated by HIF‑1α promotes the progression of pancreatic cancer". Experimental and Therapeutic Medicine 17, no. 3 (2019): 2359-2365. https://doi.org/10.3892/etm.2019.7213