Open Access

Downregulation of δ opioid receptor by RNA interference enhances the sensitivity of BEL/FU drug‑resistant human hepatocellular carcinoma cells to 5‑FU Corrigendum in /10.3892/mmr.2018.9415

Retraction in: /10.3892/mmr.2023.12974

Downregulation of δ opioid receptor by RNA interference enhances the sensitivity of BEL/FU drug‑resistant human hepatocellular carcinoma cells to 5‑FU

Corrigendum in: /10.3892/mmr.2018.9415 Retraction in /10.3892/mmr.2023.12974

  • Authors:
    • Bo Tang
    • Zhigao Hu
    • Yang Li
    • Shengguang Yuan
    • Zhenran Wang
    • Shuiping Yu
    • Songqing He
  • View Affiliations

  • Published online on: November 6, 2015     https://doi.org/10.3892/mmr.2015.4511
  • Pages: 59-66
  • Copyright: © Tang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

δ opioid receptor (DOR) was the first opioid receptor of the G protein‑coupled receptor family to be cloned. Our previous studies demonstrated that DOR is involved in regulating the development and progression of human hepatocellular carcinoma (HCC), and is involved in the regulation of the processes of invasion and metastasis of HCC cells. However, whether DOR is involved in the development and progression of drug resistance in HCC has not been reported and requires further elucidation. The aim of the present study was to investigate the expression levels of DOR in the drug‑resistant HCC BEL‑7402/5‑fluorouracil (BEL/FU) cell line, and its effects on drug resistance, in order to preliminarily elucidate the effects of DOR in HCC drug resistance. The results of the present study demonstrated that DOR was expressed at high levels in the BEL/FU cells, and the expression levels were higher, compared with those in normal liver cells. When the expression of DOR was silenced, the proliferation of the drug‑resistant HCC cells were unaffected. However, when the cells were co‑treated with a therapeutic dose of 5‑FU, the proliferation rate of the BEL/FU cells was significantly inhibited, a large number of cells underwent apoptosis, cell cycle progression was arrested and changes in the expression levels of drug‑resistant proteins were observed. Overall, the expression of DOR was upregulated in the drug‑resistant HCC cells, and its functional status was closely associated with drug resistance in HCC. Therefore, DOR may become a recognized target molecule with important roles in the clinical treatment of drug‑resistant HCC.

Introduction

Hepatocellular carcinoma (HCC) is the fifth most common type of malignant tumor worldwide, and 500,000 patients succumb to mortality from HCC each year (14). Among the numerous therapeutic strategies used to treat HCC, chemotherapy remains indispensable. However, HCC readily develops multiple drug resistance to chemotherapeutic drugs, which frequently results in unsatisfactory chemotherapeutic treatment of HCC (5,6). There are several mechanisms underlying the generation of multiple drug resistance in HCC, among which the increased expression of the P-glycoprotein (P-gp) transport protein, or its encoding gene multidrug resistance 1 (MDR1), in HCC cells is key (7,8). P-gp is an important member of the ATP-binding cassette transporter family, and is expressed on the cell membrane where it forms a specific pore channel. The pore channel is opened following activation with ATP, and mediates the transport of several substrate molecules, including chemotherapeutic drugs, across extracellular and intracellular membranes (9). Previous studies have demonstrated that the expression of P-gp is significantly increased in multiple-drug-resistant HCC cells (10,11). The increased expression of P-gp facilitates the efflux of chemotherapeutic drugs out of cells, resulting in the increased drug resistance of HCC. By contrast, when the expression of P-gp is reduced or its function is inhibited, multiple drug resistance in drug-resistant HCC cells is reduced (12,13). Therefore, the expression levels of P-gp may be used as an indicator to measure multiple drug resistance in HCC. Our previous study demonstrated that δ opioid receptor (DOR) was expressed extensively in human HCC cells, and its functional status directly affects the proliferation, apoptosis, invasion and migration of HCC cells (14). In addition, high expression levels of DOR were detected in the multiple-drug-resistant HCC BEL7402/5-fluouracil (BEL/FU) cell line. The effects of DOR on the proliferative ability and drug resistance of multiple-drug-resistant HCC cells remains to be elucidated. In the present study, the BEL/FU cell line was used as the study subject, and DOR was downregulated using RNA interference, in order to determine the effects of DOR on the proliferative ability of the BEL/FU cells. In addition, the expression levels of P-gp and MDR1 were detected, to elucidate the effects of DOR on the proliferative ability and drug resistance of multiple-drug-resistant HCC cells. The present study may provide suitable targets to improve liver cancer chemotherapy drug resistance sensitivity.

Materials and methods

Cell culture

BEL and Chang liver cells were purchased from American Type Culture Collection (Danvers, MA, USA) and cultured in RPMI 1640 culture medium (Gibco, Thermo Fisher Scientific, Inc., Waltham, MA USA) supplemented with 10% fetal bovine serum (FBS; Gibco). To obtain 5-FU-drug-resistant BEL cells, the cells were cultured in complete RPMI-1640 culture medium supplemented with 1.0×10−7 mol/l 5-FU (Sigma-Aldrich, St. Louis, MO, USA) for 6 months. Once the drug resistance assessment was successful, the cells were cultured in RPMI-1640 supplemented with 10% FBS, at 37°C in an atmosphere containing 5% CO2. The cells were passaged every 3–4 days.

Small interfering RNA (siRNA) transfection

DOR-specific siRNA was designed and synthesized by Shanghai GenePharma Co., Ltd. (Shanghai, China). The siRNA consisted of a 21-bp duplex oligonucleotide with a sense strand corresponding to the human DOR mRNA sequence: 5′-GCCAAGCUGAUCAACAUCUTT-3′. BEL/FU cells were inoculated into 6-well plates at a density of 5×105 cells/well in the absence of antibiotics. After 24 h, the cells reached 70% confluence and transfection was performed. Briefly, the culture medium was replaced with antibiotic-free medium 24 h prior to transfection. Aliquots (4 µl) of the siRNAs of the si-control, siDOR, and siDOR + 5-Fu groups were thoroughly mixed with serum-free RPMI medium (250 µl) and incubated at room temperature for 5 min. Lipofectamine® 2000 (10 µl; Invitrogen; Thermo Fisher Scientific, Inc.) was mixed thoroughly with serum-free RPMI medium (250 µl) and incubated at room temperature for 5 min. Subsequently, the prepared siRNA and Lipofectamine® 2000 solutions were mixed thoroughly and incubated at room temperature for 20 min. The siRNA and 500 µl Lipofectamine® 2000 mixture was then added to the 6-well plates and cultured in a CO2 incubator following thorough mixing. The culture medium was replaced with fresh RPMI 1640 medium following 4–6 h of transfection, in order to continue the culture. The cells were collected after 24, 48 and 72 h.

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

Total RNA was extracted from the cells in each group using TRIzol® (Invitrogen). RNA was reverse transcribed to cDNA using the PrimeScript 1st Strand cDNA Synthesis kit (Takara Biotechnology Co., Ltd., Dalian, China) according to the manufacturer's instructions. RT-qPCR was performed using an RT-PCR reagent kit (Takara Biotechnology Co., Ltd.), according to the manufacturer's instructions. β-actin was used as an internal control. The primers for DOR, MDR1 and β-actin were synthesized by Invitrogen, and the primer sequences are presented in Table I. The qPCR reaction contained a total volume of 50 µl (2 µl template, 2 µl primer 1 (10 µM), 2 µl primer 2 (10 µM), 25 µl PCR MasterMix, 19 µl ddH2O). An SYBR® Green-based RT-qPCR assay (Beyotime Institute of Biotechnology, Shanghai, China)was used to determine the mRNA expression levels using an ABI PRISM 7900HT Sequence Detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.), and the cycling conditions were as follows: 94°C for 2 min, followed by 30 cycles of dena-turation at 94°C for 30 sec, annealing at 64°C for 30 sec, and extension at 72°C for 30 sec. β-actin was used as an internal control to normalize gene expression levels. The PCR products were subsequently subjected to 1.0% agarose gel electrophoresis, and the results were scanned and analyzed using a gel documentation system (Syngene, Cambridge, UK).

Table I

Primer sequences of the DOR, MDR1 and β-actin genes.

Table I

Primer sequences of the DOR, MDR1 and β-actin genes.

PrimerForwardReverse
DOR 5′-ACCAAGATCTGCGTGTTCCT-3′ 5′-CGATGACGAAGATGTGGATG-3′
MDR1 5′-CCCATCATTGCAATAGCAGG-3′ 5′-GTTCAAACTTCTGCTCCTGA-3′
β-actin 5′-AAGGAAGGCTGGAAGAGTGC-3′ 5′-CTGGGACGACATGGAGAAAA-3′

[i] DOR, δ opioid receptor; MDR1, multidrug resistance 1.

Detection of cell proliferation using an MTT assay

BEL/FU cells in the logarithmic phase were dissociated using 0.25% trypsin (BD Biosciences, Franklin Lakes, NJ, USA), inoculated into 96-well plates at a density of 1×105 cells/well, and conventionally cultured for 24 h. The proliferative ability of the cells was detected using an MTT assay (BD Biosciences) following DOR gene silencing and/or 5-FU treatment. The specific procedure was performed as previously described (15). Briefly, the plates were incubated in a 5% CO2 incubator for 48 h, and 20 µl MTT (5 mg/ml) was then added and incubated for a further 4 h at 37°C. Following incubation, the excess liquid was discarded and the cells were incubated with 150 µl dimethyl sulfoxide (Sigma-Aldrich) at room temperature for 10 min on a shaker. The OD570 value was measured using a microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA), and the cell growth inhibition rate (GIR) was detected and calculated as follows: GIR = (cell number of the control group − cell number of the treatment group)/(cell number of the control group) ×100%.

Detection of cell apoptosis using flow cytometry

Following DOR gene silencing and/or 5-FU treatment, the BEL/FU cells in each experimental group were collected using the trypsin method (0.25%; 37°C; 1–3 min), and the cell density was adjusted to 1×106 cells/ml. The cells were then precipitated by centrifugation (4°C; 1,000 × g; 5 min), washed twice with phosphate-buffered saline (PBS), and stained with 10 µl Annexin V-fluorescein isothiocyanate and 5 µl propidium iodide (PI) staining solution (Bioseal Biotechnology Co., Ltd., Beijing, China). The solution was incubated at 37°C in the dark for 15 min, and the samples were immediately subjected to flow cytometric analysis (BD Bioscience).

Analysis of cell cycle distribution using flow cytometry

Following DOR gene silencing and/or 5-FU treatment, the BEL/FU cells in each experimental group were collected using the trypsin method, and the cell density was adjusted to 1×106 cells/ml. The cells were then precipitated by centrifugation, washed twice with PBS, and fixed in 70% cold ethanol at 4°C overnight. The cells were then washed twice with PBS, and were subsequently stained with 500 µl DNAStain solution (10 µg/ml RNase, 50 µg/ml PI and 1% Triton X-100; Bioseal Biotechnology Co., Ltd.) at 37°C in the dark for 30 min. The stained cells were analyzed using a flow cytometer (BD Biosciences).

Western blot analysis

The BEL/FU cells in each experimental group were collected using the trypsin method, and were resuspended at a density of 1×106 cells/ml in pre-cooled cell lysis buffer (Beyotime Institute of Biotechnology), containing 8 M urea, 4% CHAPS (w/v), 65 mM DTT, 1 mM EDTA, 0.5 mM EGTA, 1 mM PMSF, 40 mM Tris-HCl (pH 7.4) and 1x protease inhibitor cocktail tablet. The protein samples were collected and protein concentrations were determined usinga bicinchoninic acid assay. Subsequently, the protein samples (50 µg) from each group were subjected to 10% SDS-PAGE, and were transferred to polyvinylidene fluoride membranes (Bio-Rad Laboratories, Inc.) using a semi-dry method. The membranes were then blocked in 5% skim milk overnight. Subsequently, the membranes were washed with Tris-buffered saline containing 0.05% Tween (TBST) and incubated with the following primary antibodies at 37°C for 1 h: Mouse monoclonal anti-human MDR1 antibody to cyclin D1 and CDK4 (cat. no. sc-55510; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), rabbit polyclonal anti-human DOR antibody (cat. no. sc-7492; Santa Cruz Biotechnology, Inc.), goat polyclonal anti-human P-gp antibody (cat. no. sc-241605; Santa Cruz Biotechnology, Inc.), goat polyclonal anti-human β-actin antibody (cat. no. sc-1616; Santa Cruz Biotechnology, Inc.). The membranes were washed again with TBST and incubated with rabbit horseradish peroxidase (HRP)-conjugated anti-mouse IgG (cat. no. sc-358917, Santa Cruz Biotechnology, Inc.) and bovine HRP-conjugated anti-goat IgG (cat. no. sc-2378, Santa cruz Biotechnology, Inc.) secondary antibodies at 37°C for 1 h. The blots were then incubated with enhanced chemiluminescence (Beyotime Institute of Biotechnology) working solution at room temperature for 1 min and exposed to X-ray films (Canon, Inc., Tokyo, Japan). The protein bands were scanned and the optical density values were analyzed.

Statistical analysis

All data were analyzed using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA). All data are presented as the mean ± standard deviation. The comparison between two groups was performed using Student's t-test, and comparisons between multiple groups were performed by one-way analysis of variance. P<0.05 was considered to indicate a statistically significant difference.

Results

mRNA and protein expression levels of DOR in BEL/FU cells

Total RNA was extracted from Chang liver cells and BEL/FU cells, to perform RT-PCR analyses. The mRNA expression levels of DOR were detected in the BEL/FU cells, and the expression levels were significantly higher, compared with those in the Chang liver cells (P<0.05; Fig. 1A and B). In addition, total protein was extracted from the Chang liver cells and BEL/FU cells, and the protein expression levels of DOR were detected using western blot analysis. The protein expression levels of DOR was also at a higher level in the BEL/FU cells, and were significantly higher, compared with those in the Chang liver cells (P<0.05; Fig. 1C and D).

Effects of DOR gene silencing on the proliferation of BEL/FU cells

An MTT assay was performed to determine the effects of DOR gene silencing on the proliferative ability of the BEL/FU cells. Compared with the untransfected group and the negative control group transfected with control oligonucleotides, no differences were observed in the proliferative ability of the BEL/FU cells in the 5-FU treatment group and the DOR siRNA transfection group (P>0.05). However, following DOR siRNA transfection and 5-FU treatment, the proliferative ability of the BEL/FU cells was significantly reduced (P<0.05; Fig. 2).

Effects of DOR gene silencing on the apoptosis of BEL/FU cells

To investigate whether DOR was associated with the apoptosis of BEL/FU cells, the expression of DOR was silenced by RNA interference, and the apoptotic rates of the cells were detected using flow cytometry. Compared with the negative control group transfected with control oligonucleotides, the rate of apoptosis of the BEL/FU cells in the 5-FU treatment group and in the DOR siRNA transfection group did not exhibit significant changes (P>0.05). However, following both DOR siRNA transfection and 5-FU treatment, the rate of apoptosis of the BEL/FU cells was significantly increased (P<0.01; Fig. 3).

Effects of DOR gene silencing on the cell cycle distribution of BEL/FU cells

To investigate whether the inhibition of BEL/FU cell proliferation by DOR gene silencing was associated with cell cycle progression, cell cycle distribution was analyzed using flow cytometry. Compared with the negative control group transfected with control oligonucleotides, the cell cycle distribution of the BEL/FU cells showed no significantly differences following transfection with DOR siRNA (P>0.05). However, following both DOR siRNA transfection and 5-FU treatment, the BEL/FU cells were significantly arrested at the G0/G1 phase (P<0.05; Fig. 4A and B).

As DOR gene silencing and 5-FU treatment resulted in G0/G1 cell cycle arrest in the BEL/FU cells, the present study examined the expression levels of the cell cycle-associated proteins, cyclin D1 and cyclin-dependent kinase (CDK)4, which regulate the G0/G1 phase in BEL/FU cells following DOR gene silencing (16). Compared with the negative control group transfected with control oligonucleotides and the group transfected with DOR siRNA alone, the protein expression levels of cyclin D1 and CDK4 in the cells subjected to DOR siRNA and 5-FU treatment were significantly decreased (P<0.05; Fig. 4C and D).

DOR gene silencing enhances the sensitivity of BEL/FU cells to 5-FU

The GIR of the cells in the DOR siRNA transfection and 5-FU treatment group was significantly higher, compared with the GIRs in the negative control group transfected with control oligonucleotides and in the group transfected with DOR siRNA alone (P<0.05; Fig. 5).

The generation of multiple-drug resistance in HCC is closely associated with the P-gp transport protein In order to further investigate the specific mechanisms underlying the increased sensitivity of BEL/FU cells to 5-FU following DOR gene silencing, the expression levels P-gp/MDR1 were detected. Following DOR gene silencing, the gene expression levels of MDR1 decreased, and the protein expression levels of P-gp decreased accordingly (P<0.05; Fig. 6).

Discussion

The present study aimed to evaluate the function of DOR in the treatment of multiple drug-resistant HCC, and to determine its value in clinical application. The results demonstrated that the gene and protein expression levels of DOR were significantly increased in the multiple drug-resistant HCC BEL/FU cells. These results indicated that DOR was important in the development of multiple drug resistance in HCC; and may, therefore, be a potential biomarker and therapeutic target for the treatment of multiple drug resistance in HCC. Our previous study demonstrated that DOR was expressed in HCC tissues and normal human liver tissues, and its expression levels were significantly higher in the HCC tissues, compared with those in the normal liver tissues (14). A previous study demonstrated that DOR may be used as a promising marker for HCC diagnosis to increase the efficiency of HCC imaging detection (17). In addition, DOR overexpression may increase cholestasis, and the malignant progression and invasion of cholangiocarcinoma, with silencing of the expression of DOR being important in the treatment of late stage cholangiocarcinoma (18). The present study hypothesized that DOR promotes the progression of multiple drug resistance in HCC and promotes the proliferation of multiple drug-resistant HCC cells. The results of the present study revealed significant differences in the expression levels of DOR between normal liver cells and multiple drug-resistant HCC cells.

To further elucidate the mechanism underlying the action of DOR in the present study, the gene expression of DOR was silenced in multiple drug-resistant HCC cells using RNA interference technology. RNA interference has been extensively used for the analysis of mammalian gene functions and may become a potential method for gene therapy (19,20). In the present study, DOR-specific siRNA was effectively transfected into multiple drug-resistant HCC cells, to rapidly inhibit the gene expression of DOR. A previous study indicated that the activation of DOR stimulates the proliferation of human glioblastoma T98 G cells (21). However, other studies have reported that the activation of DOR inhibits the proliferation of breast cancer cells (22) and colorectal cancer cells (23). The results of the present study suggested that DOR was expressed at high levels in multiple drug-resistant HCC cells, suggesting that DOR may promote the proliferation of multiple drug-resistant HCC cells and the progression of multiple drug resistance in HCC. However, silencing of the expression of DOR alone did not inhibit the proliferation of multiple drug-resistant HCC cells or induce apoptosis in these cells. In addition, no significant differences in the cell cycle progression of the HCC cells were observed. As a conventionally used chemotherapeutic drug, 5-FU is used extensively for the clinical treatment of HCC. In the present study, the administration of a therapeutic dose of 5-FU did not produce cytotoxic effects in the BEL/FU cells; however, DOR gene silencing combined with 5-FU treatment significantly induced apoptosis in the BEL/FU cells, and the cell cycle was arrested at the G0/G1 phase. These results indicated that DOR gene silencing required combination with chemotherapeutic drug treatment in order to exert inhibitory effects on the BEL/FU cells.

However, how DOR gene silencing increased the sensitivity of multiple drug-resistant HCC cells to 5-FU remained to be fully elucidated. To address this, the gene expression levels of MDR1 were detected. The results demonstrated that the gene expression of MDR1 was downregulated following DOR gene silencing, indicating that the expression of these two genes exhibited a certain correlation; however, the specific association between these two genes in HCC cells remains to be fully elucidated. It has previously been shown that downregulation or upregulation of the BMI-1 gene in HCC cells downregulates or upregulates the gene expression of MDR1, respectively (24). The DOR gene and the MDR1 gene encode a transmembrane protein; however, they do not belong to the same family. As no significant correlation was observed between the expression levels of these two genes, their association cannot be confirmed. Therefore, the specific association between these two genes and whether they have interactive effects requires further investigation. The MDR1 gene is a drug resistance gene, and P-gp is its encoded membrane protein, which is expressed at high levels in drug-resistant HCC cells (25,26) and enhanced the efflux of chemotherapeutic drugs from cells. In the present study, following silencing of the expression of DOR, the protein expression levels of P-gp also decreased. Therefore, it was hypothesized that downregulation of the P-gp protein attenuated the efflux ability of the cells against 5-FU, causing a rapid increase in the intracellular concentrations of 5-FU and eventually causing BEL/FU cells to regain sensitivity to chemotherapeutic drugs.

In conclusion, the present study demonstrated that DOR is expressed in high levels in multiple drug-resistant HCC cells, and that DOR gene silencing inhibited the development of multiple drug resistance in HCC. Therefore, DOR gene silencing may be considered as a novel method for the treatment of multiple drug resistant HCC.

Acknowledgments

The present study was supported by the Project of Science and Technology of Guangxi University (grant no. 2013ZD046), the National Natural Science Foundation of China (grant no. 81360367), the Specific Project of Traditional Chinese Medicine and Technology of the Department of Health, Guangxi (grant no. GZPT13-45), the Project of Establishment of Key Laboratory for Molecular Medicine of Liver Injury and Repair, Guangxi (grant no. SYS2013009), and the Self-raising Project of the Department of Health, Guangxi (grant no Z2013464).

References

1 

Okuda K: Hepatocellular carcinoma. J Hepatol. 32(1 Suppl): 225–237. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Wörns MA and Galle PR: Future perspectives in hepatocellular carcinoma. Dig Liver Dis. 42(Suppl 3): S302–S309. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Rampone B, Schiavone B, Martino A, Viviano C and Confuorto G: Current management strategy of hepatocellular carcinoma. World J Gastroenterol. 15:3210–3216. 2009. View Article : Google Scholar : PubMed/NCBI

4 

El-Serag HB and Rudolph KL: Hepatocellular carcinoma: Epidemiology and molecular carcinogenesis. Gastroenterology. 132:2557–2576. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Poupon R, Fartoux L and Rosmorduc O: Therapeutic advances in hepatocellular carcinoma. Bull Acad Natl Med. 192:23–32. 2008.In French.

6 

Marin JJ, Romero MR and Briz O: Molecular bases of liver cancer refractoriness to pharmacological treatment. Curr Med Chem. 17:709–740. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Li G, Chen X, Wang Q, Xu Z, Zhang W and Ye L: The roles of four multi-drug resistance proteins in hepatocellular carcinoma multidrug resistance. J Huazhong Univ Sci Technolog Med Sci. 27:173–175. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Chow EK, Fan LL, Chen X and Bishop JM: Oncogene-specific formation of chemoresistant murine hepatic cancer stem cells. Hepatology. 56:1331–1341. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Takara K, Sakaeda T and Okumura K: An update on overcoming MDR1-mediated multidrug resistance in cancer chemotherapy. Curr Pharm Des. 12:273–286. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Zhai BJ, Shao ZY, Zhao CL, Hu K and Wu F: Development and characterization of multidrug resistant human hepatocarcinoma cell line in nude mice. World J Gastroenterol. 12:6614–6619. 2006.PubMed/NCBI

11 

Yan F, Wang XM, Pan C and Ma QM: Down-regulation of extracellular signal-regulated kinase 1/2 activity in P-glycoprotein-mediated multidrug resistant hepatocellular carcinoma cells. World J Gastroenterol. 15:1443–1451. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Li B, Ye T, Zhao L, Li DH, Gou XH, Zhao LY, Han L, Chen L, Yan LN and Gong JP: Effects of multidrug resistance, antisense RNA on the chemosensitivity of hepatocellular carcinoma cells. Hepatobiliary Pancreat Dis Int. 5:552–559. 2006.PubMed/NCBI

13 

Warmann S, Göhring G, Teichmann B, Geerlings H, Pietsch T and Fuchs J: P-glycoprotein modulation improves in vitro chemosensitivity in malignant pediatric liver tumors. Anticancer Res. 23:4607–4611. 2003.

14 

Tang B, Li Y, Yuan S, Tomlinson S and He S: Upregulation of the δ opioid receptor in liver cancer promotes liver cancer progression both in vitro and in vivo. Int J Oncol. 43:1281–1290. 2013.PubMed/NCBI

15 

Zhang B, Zhang X, Tang B, Zheng P and Zhang Y: Investigation of elemene-induced reversal of tamoxifen resistance in MCF-7 cells through oestrogen receptor α (ERα) re-expression. Breast Cancer Res Treat. 136:399–406. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Wang C, Lisanti MP and Liao DJ: Reviewing once more the c-myc and Ras collaboration: Converging at the cyclin D1-CDK4 complex and challenging basic concepts of cancer biology. Cell Cycle. 10:57–67. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Collier TL, Schiller PW and Waterhouse RN: Radiosynthesis and in vivo evaluation of the pseudopeptide delta-opioid antagonist [(125)I] ITIPP(psi). Nucl Med Biol. 28:375–381. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Nicoll J, Axiotis CA and Bergasa NV: The delta opioid receptor 1 is expressed by proliferating bile ductules in rats with cholestasis: Implications for the study of liver regeneration and malignant transformation of biliary epithelium. Med Hypotheses. 65:1099–1105. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Hannon GJ: RNA interference. Nature. 418:244–251. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Ren YJ and Zhang Y: An update on RNA interference-mediated gene silencing in cancer therapy. Expert Opin Biol Ther. 14:1581–1592. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Lazarczyk M, Matyja E and Lipkowski AW: A comparative study of morphine stimulation and biphalin inhibition of human glioblastoma T98G cell proliferation in vitro. Peptides. 31:1606–1612. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Hatzoglou A, Bakogeorgou E and Castanas E: The antiprolif-erative effect of opioid receptor agonists on the T47D human breast cancer cell line, is partially mediated through opioid receptors. Eur J Pharmacol. 296:199–207. 1996. View Article : Google Scholar : PubMed/NCBI

23 

Kuniyasu H, Luo Y, Fujii K, Sasahira T, Moriwaka Y, Tatsumoto N, Sasaki T, Yamashita Y and Ohmori H: CD10 enhances metastasis of colorectal cancer by abrogating the anti-tumoural effect of methionine-enkephalin in the liver. Gut. 59:348–356. 2010. View Article : Google Scholar

24 

Effendi K, Mori T, Komuta M, Masugi Y, Du W and Sakamoto M: Bmi-1 gene is upregulated in early-stage hepatocellular carcinoma and correlates with ATP-binding cassette transporter B1 expression. Cancer Sci. 101:666–672. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Fantappiè O, Solazzo M, Lasagna N, Platini F, Tessitore L and Mazzanti R: P-glycoprotein mediates celecoxib-induced apoptosis in multiple drug-resistant cell lines. Cancer Res. 67:4915–4923. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Ling X, He Y, Zhang G, Zhou Y and Yan B: Increased P-glycoprotein expression in mitochondria is related to acquired multidrug resistance in human hepatoma cells depleted of mitochondrial DNA. Int J Oncol. 40:109–118. 2012.

Related Articles

Journal Cover

January-2016
Volume 13 Issue 1

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
Tang B, Hu Z, Li Y, Yuan S, Wang Z, Yu S and He S: Downregulation of δ opioid receptor by RNA interference enhances the sensitivity of BEL/FU drug‑resistant human hepatocellular carcinoma cells to 5‑FU Corrigendum in /10.3892/mmr.2018.9415 Retraction in /10.3892/mmr.2023.12974. Mol Med Rep 13: 59-66, 2016
APA
Tang, B., Hu, Z., Li, Y., Yuan, S., Wang, Z., Yu, S., & He, S. (2016). Downregulation of δ opioid receptor by RNA interference enhances the sensitivity of BEL/FU drug‑resistant human hepatocellular carcinoma cells to 5‑FU Corrigendum in /10.3892/mmr.2018.9415 Retraction in /10.3892/mmr.2023.12974. Molecular Medicine Reports, 13, 59-66. https://doi.org/10.3892/mmr.2015.4511
MLA
Tang, B., Hu, Z., Li, Y., Yuan, S., Wang, Z., Yu, S., He, S."Downregulation of δ opioid receptor by RNA interference enhances the sensitivity of BEL/FU drug‑resistant human hepatocellular carcinoma cells to 5‑FU Corrigendum in /10.3892/mmr.2018.9415 Retraction in /10.3892/mmr.2023.12974". Molecular Medicine Reports 13.1 (2016): 59-66.
Chicago
Tang, B., Hu, Z., Li, Y., Yuan, S., Wang, Z., Yu, S., He, S."Downregulation of δ opioid receptor by RNA interference enhances the sensitivity of BEL/FU drug‑resistant human hepatocellular carcinoma cells to 5‑FU Corrigendum in /10.3892/mmr.2018.9415 Retraction in /10.3892/mmr.2023.12974". Molecular Medicine Reports 13, no. 1 (2016): 59-66. https://doi.org/10.3892/mmr.2015.4511