MicroRNA‑216b reduces growth, migration and invasion of pancreatic ductal adenocarcinoma cells by directly targeting ρ‑associated coiled‑coil containing protein kinase 1

Retraction in: /10.3892/ol.2022.13363

  • Authors:
    • Yang‑An Liu
    • Yue Zhang
    • Zhi Zheng
    • Kai Li
    • Xin‑Hua Wu
    • Qiu‑Guo Du
    • Xiao Ye
    • Lili Wang
    • Ling Zhu
  • View Affiliations

  • Published online on: February 23, 2018     https://doi.org/10.3892/ol.2018.8109
  • Pages: 6745-6751
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Developments in cancer therapy have greatly improved the survival time for patients with pancreatic ductal adenocarcinoma (PDAC); however, the prognosis of patients with PDAC remains poor. Understanding the expression patterns and functions of microRNAs may provide strategies for the diagnosis and treatment of patients with PDAC. The present study aimed to explore the expression and functions of microRNA‑216b (miR‑216b) in PDAC. The expression of miR‑216b in PDAC tissues and cell lines was quantified with reverse transcription‑quantitative polymerase chain reaction. An miR‑216b mimic was introduced into PDAC cells to induce the effects of miR‑21b overexpression. The effects of miR‑216b overexpression on growth, migration and invasion of PDAC cells were evaluated by cell proliferation assay, migration and invasion assays, respectively. The molecular mechanism underlying the suppressive effects of miR‑216b on PDAC was also examined; a direct target gene of miR‑216b, ρ‑associated coiled‑coil containing protein kinase 1 (ROCK1), was downregulated by ROCK1 short interfering RNA to investigate the effects on growth, migration and invasion of PDAC cells. The present study revealed that miR‑216b was significantly downregulated in PDAC tissues and cell lines. Overexpression of miR‑216b inhibited growth, migration and invasion of PDAC cells in vitro. ROCK1 was identified as a direct target gene of miR‑216b in pancreatic cancer and the downregulation of ROCK1 resembled the effects of miR‑216b overexpression in PDAC cells. Taken together, miR‑216b acted as a tumor suppressor in PDAC and may represent a novel therapeutic target in PDAC.

Introduction

Pancreatic cancer is the fourth leading cause for cancer-associated mortality in the United States (1). It is projected to be the second-leading cause of cancer-associated mortalities by 2030 (2). Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, which accounts for ~90% for all pancreatic tumors (3). A range of risk factors is associated with PDAC progression, including advanced age, smoking, obesity and chronic pancreatitis (46). PDAC is an aggressive and destructive disease, characterized by rapid progression, early metastasis and a limited response to radiotherapy and chemotherapy (7). Radical excision is central to the successful therapy of patients with PDAC (8); however, only 10–15% of patients are diagnosed at the early stages when surgical resection can be performed (9). At present, developments in cancer therapy have improved the survival time for patients with PDAC; however, the prognosis of patients with PDAC remains poor, with a five-year overall survival rate of <5% and a median survival time of 6 months (10). Therefore, the identification of novel therapeutic targets is urgently required.

MicroRNAs (miRNAs) are a conserved group of single stranded, 17–25-nucleotide, non-coding RNA molecules that are endogenously expressed in mammals and other groups of organisms (11). They downregulate protein expression by directly binding to the 3′-untranslational region (3′UTR) of target genes, therefore inhibiting translation or inducing mRNA degradation (12). A single type of miRNA may regulate a large number of genes, often targeting multiple components of complex intracellular networks (13). The abnormal expression of an individual miRNA may have a marked influence on cellular physiology, potentially leading to disease, including cancer (14). Accumulating reports have demonstrated that miRNAs are involved in carcinogenesis and disease progression of a number of types of human cancer, and that they regulate diverse biological processes, including cell proliferation, the cell cycle, differentiation, apoptosis, migration, invasion, metastasis and sensitivity to chemotherapy and radiation (15,16). It is reported that numerous miRNAs are deregulated in cancer tissue when compared with normal tissue (1719), acting either as tumor suppressors or oncogenes depending on the functions of their target genes (20,21). Therefore, further investigation of the expression and molecular mechanisms of miRNAs in PDAC is likely to provide therapeutic targets for patients with PDAC.

The present study aimed to determine the expression level of miR-216b, its association with PDAC progression and the underlying molecular mechanisms. The results demonstrated that miR-216b was significantly downregulated in PDAC tissues and cell lines. In addition, it was revealed that miR-216b acted as a tumor suppressor in PDAC, which was mediated by directly targeting ρ-associated coiled-coil containing protein kinase 1 (ROCK1).

Materials and methods

Tissue samples, cell culture and transfection

A total of 22 paired PDAC and normal adjacent tissues (NATs) were obtained from patients with PDAC (male, 14; female, 8; age range, 36–68 years) who had undergone pancreaticoduodenectomy at the Wuhan Central Hospital of Tongji Medical College (Wuhan, China) between July 2013 and February 2015. The tissue samples were immediately snap-frozen in liquid nitrogen tanks and stored at −80°C. The present study was approved by the Ethics Committee of Wuhan Central Hospital of Tongji Medical College. Written informed consent was obtained from all patients prior to enrollment in the study.

Panc-1, Bxpc-3, Sw1990 and Aspc-1 human PDAC cells, HPDE6c7 human normal pancreatic cells and HEK293T human embryonic kidney cells were purchased from American Type Culture Collection (Manassas, VA, USA). All cells were cultured in RPMI-1640 medium (Bxpc-3, Aspc-1) or Dulbecco's modified Eagle's medium (Panc-1, Sw1990, HPDE6c7, HEK293T) supplemented with 10% fetal bovine serum (FBS), 2 mM glutamine, 100 IU/ml penicillin and 100 ug/ml streptomycin (all Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in a humidified atmosphere of 5% CO2 and 95% air at 37°C.

Small RNAs, including an miR-216b mimic, negative control (NC) miRNA, ROCK1 short interfering (si)RNA and an siRNA control, were purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China). The miR-216b mimics sequence was 5′-AAAUCUCUGCAGGCAAAUGUGA-3′ and the NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. The ROCK1 siRNA sequence was 5′-GGGUAACUCAUCUGGUAAATT-3′ and the siRNA control sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. The RNAs were introduced into cells at a final concentration of 50 nM using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the patient tissues, and HPDE6c7, Panc-1, Bxpc-3, Sw1990 and Aspc-1 cells, using TRIzol® regent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The concentration of total RNA was determined using an ND-1000 spectrophotometer (NanoDrop Technologies; Thermo Fisher Scientific, Inc., Wilmington, DE, USA) to measure absorbance at 260 and 280 nm. A TaqMan miRNA assay kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used to evaluate the miR-216b expression level, using U6 as an internal control. The cycling conditions were as follows: 50°C for 2 min, 95°C for 10 min; 40 cycles of denaturation at 95°C for 15 sec; and annealing/extension at 60°C for 60 sec. To determine the ROCK1 mRNA expression level, total RNA was reverse transcribed into cDNA using the PrimeScript RT reagent kit (Takara Biotechnology, Inc., Dalian, China) according to the manufacturer's protocol. SYBR Green Premix Ex Taq (Takara Biotechnology, Inc.) was used to determine ROCK1 mRNA expression relative to GADPH. The cycling conditions were as follows: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. The primers were designed as follows: miR-216b, 5′-GCCGCGCTAAAGTGCTTATAGTG-3′ (forward) and 5′-CACCAGGGTCCGAGGT-3′ (reverse); U6, 5′-CTCGCTTCGGCAGCACA-3′ (forward) and 5′-AACGCTTCACGAATTTGCGT-3′ (reverse); ROCK1, 5′-AGGAAGGCGGACATATTGATCCCT-3′ (forward) and 5′-AGACGATAGTTGGGTCCCGGC-3′ (reverse); and GAPDH, 5′-ACAACTTTGGTATCGTGGAAGG-3′ (forward) and 5′-GCCATCACGCCACAGTTTC-3′ (reverse). Each sample was analyzed in triplicate, and analyzed by the 2−ΔΔCq method (22).

Cell proliferation assay

Cell proliferation was examined using a Cell Counting kit 8 assay (CCK8; Dojindo Molecular Technologies, Inc., Kumamoto, Japan). At 24 h after transfection, Panc-1 and Sw1990 cells were collected and seeded into 96-well plates at 3×103 cells per well, in triplicate. Following 1, 2, 3 or 4 days in the previously described growth conditions, 10 µl CCK8 assay solution was added to each well. The cells were incubated with CCK8 for a further 2 h at 37°C, and optical density values were evaluated at 450 nm.

Transwell migration and invasion assay

Cell migration and invasion were quantified using Panc-1 and Sw1990 cells in Transwell chambers (24-well insert; 8 µm pore size; Corning Incorporated, Corning, NY, USA). For the migration assay, 1×105 cells in 200 µl serum-free medium of the appropriate type were seeded in the top Transwell chamber. For the Transwell invasion assay, 1×105 cells in 200 µl serum-free medium were plated in the top Transwell chamber onto a Matrigel-coated (40 ug/well; BD Biosciences, San Jose, CA, USA) membrane. For both assays, 500 µl culture medium of the appropriate type supplemented with 20% FBS was added to the lower chamber as a chemoattractant. Following a 24-h incubation in the previously described growth conditions, cells on the top surface of the Transwell chamber were removed using a cotton swab. Cells on the lower surface of the membrane were fixed with 100% methanol for 30 min and stained with 0.05% crystal violet for 30 min at room temperature, and five random fields were selected for quantification under an inverted microscope (×200 magnification; IX83; Olympus Corporation, Tokyo, Japan).

Western blot analysis

Transfected Panc-1 and Sw1990 cells were washed with PBS, lysed with radioimmunoprecipitation lysis buffer (Sigma-Aldrich; Merck Millipore, Darmstadt, Germany) supplemented with a protease inhibitor mixture (Roche Diagnostics, Basel, Switzerland), and protein concentrations were determined using a bicinchoninic acid protein assay (Thermo Fisher Scientific, Inc.). Equal amounts of protein (20 µg) were subjected to 10% SDS-PAGE, transferred to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA) and blocked with 5% fat-free milk in TBST buffer (0.1% Tween-20) at room temperature for 2 h. Subsequently, the membranes were probed with primary antibodies at 4°C overnight against ROCK1 (1;1,000 dilution; sc-365628) and GADPH (1;1,000 dilution; sc-69778; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), washed three times with TBST and incubated with goat anti-mouse horseradish peroxidase-conjugated secondary antibody (sc-2005; 1:5,000 dilution; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at room temperature for 2 h. Following three washes with TBST, immunoreactive bands were visualized using ECL Plus reagents (Pierce; Thermo Fisher Scientific, Inc.). This experiment was repeated three times.

Identification of miR216b target genes

TargetScan (http://www.targetscan.org/), miRanda (http://www.microrna.org) and MicroCosm (http://www.ebi.ac.uk/enright-srv/microcosm/htdocs/targets/v5/), were used to predict the potential target genes of miR-216b.

Dual-luciferase report assay

The pMIR-ROCK1-3′UTR wild type (Wt) and mutant (Mut) plasmids (sites 1 and 2) were synthesized by Shanghai GenePharma Co., Ltd. An miR-216b mimic or NC was co-transfected with pMIR-ROCK1-3′UTR Wt or Mut into HEK293T cells using Lipofectamine 2000 according to the manufacturer's protocol. At 48 h after transfection, luciferase activity was determined using the Dual-Luciferase Reporter Assay System (Promega Corporation, Madison, WI, USA) according to the manufacturer's protocol. Firefly luciferase activity was used for normalization to Renilla luciferase activity. The experiment was performed at least in triplicate.

Statistical analysis

Statistical analysis was performed with Student's t-tests or one-way analysis of variance (ANOVA) plus multiple comparisons using SPSS version 17.0 software (SPSS Inc., Chicago, IL, USA). SNK was used as a post hoc test following ANOVA. Data were expressed as the mean with standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-216b is underexpressed in PDAC tissues and cell lines

RT-qPCR was performed to determine the expression of miR-216b in PDAC tissues and matched NATs. The results showed that the expression levels of miR-216b in PDAC tissues were significantly lower than matched NATs (Fig. 1A).

The present study then used RT-qPCR to evaluate the expression levels of miR-216b in PDAC cell lines and HPDE6c7 normal pancreatic cell line. It was revealed that the expression levels of miR-216b were significantly decreased in all PDAC cell lines compared with in HPDE6c7 cells (Fig. 1B).

miR-216b inhibits the proliferation, migration and invasion of PDAC cells

To investigate the effects of miR-216 on PDAC cells, miR-216b or NC was introduced into PDAC cells. The expression levels of miR-216b in Panc-1 and Sw1990 cells were lower than Bxpc-3 and Aspc-1 cells. Therefore, Panc-1 and Sw1990 were selected to be transfected with miR-216b mimics or NC miRNA. Following transfection, the transfection efficiency was determined with RT-qPCR. It was revealed that the expression levels of miR-216b were significantly increased by the miR-216b mimic in Panc-1 and Sw1990 compared with the NC groups (Fig. 2A).

Subsequently, the effects of miR-216b overexpression on PDAC cell proliferation, migration and invasion were evaluated. The cell proliferation assay demonstrated that overexpression of miR-216b significantly inhibited the proliferation of Panc-1 and Sw1990 PDAC cells (Fig. 2B). In addition, cell migration and invasion abilities were significantly decreased by the miR-216b mimic in Panc-1 and Sw1990 cells (Fig. 2C). These results indicated that overexpression of miR-216b acted as a tumor suppressor in PDAC.

ROCK1 is a direct target of miR-216b in PDAC

To explore the molecular mechanism underlying the suppressive role of miR-216b in PDAC progression, miRNA prediction software and databases were used to predict the potential target genes of miR-216b. The analysis indicated that the 3′UTR of ROCK1 contained two predicted binding sites for miR-216b (Fig. 3A). A Dual-Luciferase reporter assay was then performed to confirm that miR-216b was directly targeting ROCK1. The results revealed that the luciferase activity of pMIR-ROCK1-3′UTR site1 Wt and pMIR-ROCK1-3′UTR site2 Wt was significantly lower compared with the NC groups. The luciferase activity of pMIR-ROCK1-3′UTR site1 Mut and pMIR-ROCK1-3′UTR site2 Mut was rescued in HEK293T cells (Fig. 3B). Furthermore, the mRNA and protein expression levels of ROCK1 in Panc-1 and Sw1990 cells under the regulation of miR-216b were investigated. As presented in Fig. 3C and D, ROCK1 was significantly downregulated in Panc-1 and Sw1990 cells at the mRNA and protein levels. Thus, ROCK1 was confirmed as a direct target gene of miR-216b in PDAC.

ROCK1 is associated with the miR-216b-induced suppression of PDAC

siRNA was used to knockdown the ROCK1 expression level in PDAC cells. Panc-1 and Sw1990 cells were transfected with ROCK1 siRNA or an siRNA control. At 72 h after transfection, the transfection efficiency was determined by western blot analysis. As presented in Fig. 4, ROCK1 was significantly downregulated in ROCK1 siRNA-transfected Panc-1 and Sw1990 cells compared with cells transfected with the siRNA control. The effects of ROCK1 knockdown on the growth, migration and invasion of PDAC cells were investigated. The results demonstrated that ROCK1 significantly suppressed growth, migration and invasion of Panc-1 and Sw1990 cells, which was similar to the functions induced by miR-216b overexpression in PDAC cells, implying that ROCK1 is likely to be a direct functional target of miR-216b in PDAC.

Discussion

Understanding the expression and functions of miRNAs may provide strategies for the diagnosis and treatment of patients with PDAC (23). The abnormal expression of miRNAs in PDAC was previously reported (2426); however, few studies have investigated the role of specific miRNAs in carcinogenesis and progression of PDAC. The present study has presented the first data characterizing the expression, biological functions and molecular mechanisms underlying miR-216b in PDAC. The present study revealed that miR-216b was significantly downregulated in PDAC tissues and cell lines, and that overexpressing miR-216b inhibited the cell proliferation, migration and invasion of PDAC cells. ROCK1 was identified as a direct functional target of miR-216b in PDAC. Above all, miR-216b acted as a tumor suppressor in PDAC and should be further investigated as a target for therapy for patients with PDAC.

miR-216b, a member of the miR-216 family, is located at chromosome 2p16.1 (27). miR-216b has been previously investigated in gastric adenocarcinoma (28), heaptocellular carcinoma (29), breast cancer (30), colorectal cancer (31) and nasopharyngeal carcinoma (32,33). Wang et al (28) reported that the expression levels of miR-216b were decreased in gastric adenocarcinoma tissue samples and cell lines; the expression levels of miR-216b were positively associated with the clinical outcome of patients with gastric adenocarcinoma. In hepatocellular carcinoma, miR-216b expression level was reduced in the tumor compared with adjacent liver tissues, and the low expression level of miR-216b was associated with tumor volume, hepatitis B (HBV) infection, HBV DNA quantity and vascular invasion (29). Another study indicated that miR-216b expression levels were decreased in nasopharyngeal carcinoma, and the decreased expression level of miR-216b was associated with advanced clinical stage and lymph node metastasis (32,33). Overall, miR-216b was downregulated in these types of cancer and this may be implicated in their progression.

In functional studies, miR-216b was demonstrated to act as a tumor suppressor in in various types of malignant tumor. In gastric adenocarcinoma, upregulation of miR-216b inhibited cell proliferation and cell cycle progress via directly targeting histone deacetylases (28). A previous study revealed that the ectopic expression of miR-216b decreased the proliferation, migration and invasion of hepatocellular carcinoma cells by targeting insulin like growth factor 2 mRNA binding protein 2 (29). Zheng et al (30) demonstrated that miR-216b was downregulated in breast cancer, and targeted P2X purinoceptor 7 to attenuate cell growth and enhance apoptosis. Deng et al (32,33) indicated that miR-216b suppressed nasopharyngeal carcinoma cell proliferation and invasion via blockade of K-Ras and protein kinase Cα.

Identification of cancer-specific miR-216b, as well as their target genes, is important for elucidating the functions of miRNAs in carcinogenesis and progression of various types of cancer, and may provide promising therapeutic targets. In the present study, ROCK1 was identified as a direct target gene of miR-216b in PDAC. To investigate the molecular mechanism underlying miR-216b in PDAC, bioinformatic analysis was performed to predict the potential target genes of miR-126b. It demonstrated that ROCK1 contained two predicted binding sites for miR-216b. A dual-luciferase reporter assay was performed to explore whether miR-216b directly targeted ROCK1. The results indicated that miR-216b inhibited the luciferase activity of ROCK1-3′UTR Wt1 and ROCK1-3′UTR Wt2. Furthermore, RT-qPCR and western blotting demonstrated that miR-216b decreased ROCK1 expression at the mRNA and protein expression levels. Finally, the effect of ROCK1 knockdown in PDAC cells was similar with the functions induced by miR-216b overexpression, rendering ROCK1 as a direct functional target of miR-216b in PDAC. Collectively, it is reasonable to assume that alterations to miR-216b expression level may modulate the growth and metastasis of PDAC cells by directly targeting ROCK1.

ROCK1, located at chromosome 18 (18q11.1) (34), is frequently upregulated in human cancer (35). In a study on pancreatic cancer, the expression level of ROCK1 was increased in tumor tissue, and may have contributed to pancreatic cancer invasion and metastasis (36). A number of previous studies have demonstrated that ROCK1 acts as an oncogene and may be regulated by miRNAs in various types of human cancer (3741). For example, in hepatocellular carcinoma, miR-145 targeted ROCK1 to inhibit cell growth and metastasis (37). Cai et al (38) revealed that miR-144 suppressed cell migration and proliferation in rectal cancer by directly targeting ROCK1. ROCK1 was also targeted by miRNAs in other types of human cancer, including miR-144 in osteosarcoma (39), miR-124 in gastric cancer (40) and miR-135a in prostate cancer (41). The present study demonstrated that miR-216b inhibited the growth and metastasis of PDAC cells via the downregulation of ROCK1. To the best of our knowledge, this is the first study that demonstrated that ROCK1 may be regulated by miRNAs in PDAC. Further studies should investigate it as a therapeutic target for inhibiting the growth and metastasis of PDAC.

In conclusion, the present study revealed that miR-216b was significantly downregulated in PDAC tissues and cell lines. Overexpression of miR-216b inhibited proliferation, migration and invasion of PDAC cells. The tumor suppressive functions of miR-216b were mediated by the downregulation of its downstream target gene ROCK1. These results suggested that miR-216b may be a potential therapeutic target for the treatment of PDAC.

Acknowledgements

The present study was supported by The Hospital Fund of Wuhan Central Hospital (grant no. YQ14B03) and the fund of Health and Family Planning Commission of Wuhan Municipality(grant no. WX16D37).

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 

Humeau M, Vignolle-Vidoni A, Sicard F, Martins F, Bournet B, Buscail L, Torrisani J and Cordelier P: Salivary MicroRNA in pancreatic cancer patients. PLoS One. 10:e01309962015. View Article : Google Scholar : PubMed/NCBI

3 

Subramani R, Gangwani L, Nandy SB, Arumugam A, Chattopadhyay M and Lakshmanaswamy R: Emerging roles of microRNAs in pancreatic cancer diagnosis, therapy and prognosis (Review). Int J Oncol. 47:1203–1210. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Fuchs CS, Colditz GA, Stampfer MJ, Giovannucci EL, Hunter DJ, Rimm EB, Willett WC and Speizer FE: A prospective study of cigarette smoking and the risk of pancreatic cancer. Arch Intern Med. 156:2255–2260. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Gapstur SM, Gann PH, Lowe W, Liu K, Colangelo L and Dyer A: Abnormal glucose metabolism and pancreatic cancer mortality. JAMA. 283:2552–2558. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Michaud DS, Giovannucci E, Willett WC, Colditz GA, Stampfer MJ and Fuchs CS: Physical activity, obesity, height, and the risk of pancreatic cancer. JAMA. 286:921–929. 2001. View Article : Google Scholar : PubMed/NCBI

7 

He D, Miao H, Xu Y, Xiong L, Wang Y, Xiang H, Zhang H and Zhang Z: MiR-371-5p facilitates pancreatic cancer cell proliferation and decreases patient survival. PLoS One. 9:e1129302014. View Article : Google Scholar : PubMed/NCBI

8 

Wagner M, Redaelli C, Lietz M, Seiler CA, Friess H and Büchler MW: Curative resection is the single most important factor determining outcome in patients with pancreatic adenocarcinoma. Br J Surg. 91:586–594. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Stathis A and Moore MJ: Advanced pancreatic carcinoma: Current treatment and future challenges. Nat Rev Clin Oncol. 7:163–172. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Siegel R, Ward E, Brawley O and Jemal A: Cancer statistics, 2011: The impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 61:212–236. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Boudreau RL, Jiang P, Gilmore BL, et al: Transcriptome-wide discovery of microRNA binding sites in human brain. Neuron. 81:294–305. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Mendell JT and Olson EN: MicroRNAs in stress signaling and human disease. Cell. 148:1172–1187. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Zhao L, Chen X and Cao Y: New role of microRNA: Carcinogenesis and clinical application in cancer. Acta Biochim Biophys Sin (Shanghai). 43:831–839. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Hagan JP and Croce CM: MicroRNAs in carcinogenesis. Cytogenet Genome Res. 118:252–259. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Ganci F, Vico C, Korita E, Sacconi A, Gallo E, Mori F, Cambria A, Russo E, Anile M, Vitolo D, et al: MicroRNA expression profiling of thymic epithelial tumors. Lung Cancer. 85:197–204. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Thorns C, Schurmann C, Gebauer N, et al: Global microRNA profiling of pancreatic neuroendocrine neoplasias. Anticancer Res. 34:2249–2254. 2014.PubMed/NCBI

19 

Devor EJ, Hovey AM, Goodheart MJ, Ramachandran S and Leslie KK: microRNA expression profiling of endometrial endometrioid adenocarcinomas and serous adenocarcinomas reveals profiles containing shared, unique and differentiating groups of microRNAs. Oncol Rep. 26:995–1002. 2011.PubMed/NCBI

20 

Fabbri M, Ivan M, Cimmino A, Negrini M and Calin GA: Regulatory mechanisms of microRNAs involvement in cancer. Expert Opin Biol Ther. 7:1009–1019. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Zhang B, Pan X, Cobb GP and Anderson TA: microRNAs as oncogenes and tumor suppressors. Dev Biol. 302:1–12. 2007. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Liu H, Xu XF, Zhao Y, Tang MC, Zhou YQ, Lu J and Gao FH: MicroRNA-191 promotes pancreatic cancer progression by targeting USP10. Tumour Biol. 35:12157–12163. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Zhu Z, Xu Y, Du J, Tan J and Jiao H: Expression of microRNA-218 in human pancreatic ductal adenocarcinoma and its correlation with tumor progression and patient survival. J Surg Oncol. 109:89–94. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Zhao C, Zhang J, Zhang S, Yu D, Chen Y, Liu Q, Shi M, Ni C and Zhu M: Diagnostic and biological significance of microRNA-192 in pancreatic ductal adenocarcinoma. Oncol Rep. 30:276–284. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Sadakari Y, Ohtsuka T, Ohuchida K, Tsutsumi K, Takahata S, Nakamura M, Mizumoto K and Tanaka M: MicroRNA expression analyses in preoperative pancreatic juice samples of pancreatic ductal adenocarcinoma. JOP. 11:587–592. 2010.PubMed/NCBI

27 

Shao JY, Huang XM, Yu XJ, Huang LX, Wu QL, Xia JC, Wang HY, Feng QS, Ren ZF, Ernberg I, et al: Loss of heterozygosity and its correlation with clinical outcome and Epstein-Barr virus infection in nasopharyngeal carcinoma. Anticancer Res. 21:3021–3029. 2001.PubMed/NCBI

28 

Wang Y, Xu P, Yao J, Yang R, Shi Z, Zhu X, Feng X and Gao S: MicroRNA-216b is down-regulated in human gastric adenocarcinoma and inhibits proliferation and cell cycle progression by targeting oncogene HDAC8. Target Oncol. 11:197–207. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Liu FY, Zhou SJ, Deng YL, Zhang ZY, Zhang EL, Wu ZB, Huang ZY and Chen XP: MiR-216b is involved in pathogenesis and progression of hepatocellular carcinoma through HBx-miR-216b-IGF2BP2 signaling pathway. Cell Death Dis. 6:e16702015. View Article : Google Scholar : PubMed/NCBI

30 

Zheng L, Zhang X, Yang F, Zhu J, Zhou P, Yu F, Hou L, Xiao L, He Q and Wang B: Regulation of the P2×7R by microRNA-216b in human breast cancer. Biochem Biophys Res Commun. 452:197–204. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Kim SY, Lee YH and Bae YS: MiR-186, miR-216b, miR-337-3p, and miR-760 cooperatively induce cellular senescence by targeting α subunit of protein kinase CKII in human colorectal cancer cells. Biochem Biophys Res Commun. 429:173–179. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Deng M, Tang H, Zhou Y, Zhou M, Xiong W, Zheng Y, Ye Q, Zeng X, Liao Q, Guo X, et al: miR-216b suppresses tumor growth and invasion by targeting KRAS in nasopharyngeal carcinoma. J Cell Sci. 124:2997–3005. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Deng M, Liu JF, Gu YX, Zheng GP and He ZM: miR-216b suppresses cell proliferation and invasion by targeting PKCα in nasopharyngeal carcinoma cells. Zhonghua Zhong Liu Za Zhi. 35:645–650. 2013.(In Chinese). PubMed/NCBI

34 

Lock FE, Ryan KR, Poulter NS, Parsons M and Hotchin NA: Differential regulation of adhesion complex turnover by ROCK1 and ROCK2. PLoS One. 7:e314232012. View Article : Google Scholar : PubMed/NCBI

35 

Zhou X, Wei M and Wang W: MicroRNA-340 suppresses osteosarcoma tumor growth and metastasis by directly targeting ROCK1. Biochem Biophys Res Commun. 437:653–658. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Kaneko K, Satoh K, Masamune A, Satoh A and Shimosegawa T: Expression of ROCK-1 in human pancreatic cancer: Its down-regulation by morpholino oligo antisense can reduce the migration of pancreatic cancer cells in vitro. Pancreas. 24:251–257. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Ding W, Tan H, Zhao C, Li X, Li Z, Jiang C, Zhang Y and Wang L: MiR-145 suppresses cell proliferation and motility by inhibiting ROCK1 in hepatocellular carcinoma. Tumour Biol. 37:6255–6260. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Cai SD, Chen JS, Xi ZW, Zhang LJ, Niu ML and Gao ZY: MicroRNA-144 inhibits migration and proliferation in rectal cancer by downregulating ROCK1. Mol Med Rep. 12:7396–7402. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Huang J, Shi Y, Li H, Yang M and Liu G: MicroRNA-144 acts as a tumor suppressor by targeting Rho-associated coiled-coil containing protein kinase 1 in osteosarcoma cells. Mol Med Rep. 12:4554–4559. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Hu CB, Li QL, Hu JF, Zhang Q, Xie JP and Deng L: miR-124 inhibits growth and invasion of gastric cancer by targeting ROCK1. Asian Pac J Cancer Prev. 15:6543–6546. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Kroiss A, Vincent S, Decaussin-Petrucci M, Meugnier E, Viallet J, Ruffion A, Chalmel F, Samarut J and Allioli N: Androgen-regulated microRNA-135a decreases prostate cancer cell migration and invasion through downregulating ROCK1 and ROCK2. Oncogene. 34:2846–2855. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 15 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
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 Retraction in /10.3892/ol.2022.13363. Oncol Lett 15: 6745-6751, 2018
APA
Liu, Y., Zhang, Y., Zheng, Z., Li, K., Wu, X., Du, Q. ... Zhu, L. (2018). MicroRNA‑216b reduces growth, migration and invasion of pancreatic ductal adenocarcinoma cells by directly targeting ρ‑associated coiled‑coil containing protein kinase 1 Retraction in /10.3892/ol.2022.13363. Oncology Letters, 15, 6745-6751. https://doi.org/10.3892/ol.2018.8109
MLA
Liu, Y., Zhang, Y., Zheng, Z., Li, K., Wu, X., Du, Q., Ye, X., Wang, L., 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 Retraction in /10.3892/ol.2022.13363". Oncology Letters 15.5 (2018): 6745-6751.
Chicago
Liu, Y., Zhang, Y., Zheng, Z., Li, K., Wu, X., Du, Q., Ye, X., Wang, L., 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 Retraction in /10.3892/ol.2022.13363". Oncology Letters 15, no. 5 (2018): 6745-6751. https://doi.org/10.3892/ol.2018.8109