Open Access

MicroRNA-133b inhibits cell proliferation and promotes apoptosis by targeting cullin 4B in esophageal squamous cell carcinoma

  • Authors:
    • Hairong Huang
    • Yang Xu
    • Zhong Guo
    • Xi Chen
    • Shaiguang Ji
    • Zhiyun Xu
  • View Affiliations

  • Published online on: February 28, 2018     https://doi.org/10.3892/etm.2018.5906
  • Pages: 3743-3750
  • Copyright: © Huang 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

microRNAs (miRs) serve a role as modulators during carcinogenesis. It has been demonstrated that the expression of miR‑133b is decreased in a variety of tumor tissues and cell lines and serves a suppressive role in the proliferation and apoptosis of different types of tumor cells. However, its effect on esophageal squamous cell carcinoma (ESCC) cells remains unclear. In the current study, the expression of mature miR‑133b was measured using reverse transcription-quantitative polymerase chain reaction and the results indicated that miR‑133b was significantly downregulated in ESCC tissues and various ESCC cell lines. The overexpression of miR‑133b significantly inhibited the proliferation and promoted the apoptosis of KYSE150 and Eca‑109 cells. Furthermore, it was demonstrated that cullin 4B (CUL4B) promotes ESCC cell proliferation and inhibits apoptosis by activating the protein kinase B/glycogen synthase kinase 3β/β‑catenin pathway. Taken together, these results demonstrate that miR‑133b/CUL4B serves a tumor suppressive role during ESCC progression and may therefore be used as a potential target to treat patients with ESCC.

Introduction

Esophageal cancer is the eighth most common cancer worldwide, with ~480,000 new cases and 400,000 associated mortalities per year (1). Histologically, there are two main forms of esophageal cancer: Esophageal adenocarcinoma and esophageal squamous cell carcinoma (ESCC), which represents 90% of all esophageal cancer cases (2). Currently, surgery is the only way to treat patients with esophageal cancer and the overall 5-year survival rate is 14–18%. This is due to the fact that surgery can only be performed in a limited number of patients, as in many cases tumors are inoperable (3). Although genetic and epigenetic alterations underpin the development of ESCC, the molecular mechanisms underlying neoplastic progression remain unclear (4,5). Therefore, improving understanding of the molecular biology of ESCC is critical to facilitate the development of more effective diagnostic and therapeutic strategies for ESCC.

MicroRNAs (miRs) are small non-coding RNAs ~22 nucleotides long that bind to the 3′-untranslated region (3′-UTR) of target mRNA (6). Previous studies have demonstrated that different types of miRs regulate the proliferation, apoptosis, invasion, metastasis and the epithelial-mesenchymal transition of tumor cells (7,8). The aberrant expression of miRs has also been identified in ESCC (9,10). The upregulated expression of miR-21 may promote cell proliferation, migration and resistance to apoptosis via the phosphatase and tension homolog/phosphoinositide 3 kinase/protein kinase b signaling pathway in ESCC (11). miR-let-7 is downregulated in ESCC and is considered to be a tumor suppressor, as it regulates the interleukin-6/signal transducer and activator of transcription 3 signaling pathway during neoplastic progression (12). Additionally, it has been demonstrated that miR-142-3p expression is associated with histological differentiation and may be a potential independent prognostic factor in patients with ESCC following surgery (13). It has been demonstrated that miR-133b decreases the invasiveness of esophageal cancer by inhibiting the expression of fascin actin-bundling protein 1 (FSCN1) (14). However, the molecular mechanisms underlying the function of miR-133b in proliferation and apoptosis of esophageal cancer cells remains unknown.

The results of the current study demonstrated that miR-133b expression was downregulated in ESCC tissues and cell lines and its low expression was associated with the clinicopathological features of patients with ESCC. Overexpression of miR-133b in ESCC cell lines decreased cell proliferation and promoted cell apoptosis. Cullin 4B (CUL4B) has been identified as a direct target of miR-133b and it was demonstrated that miR-133b functions as a tumor suppressor by negatively regulating CUL4B expression. Furthermore, it was demonstrated that CUL4B promotes ESCC cell proliferation and inhibits apoptosis by activating the protein kinase B/glycogen synthase kinase 3β/β-catenin (AKT/GSK3β/β-catenin) pathway. Thus, the results of the present study suggest that miR-133b/CUL4B may be a promising therapeutic target for ESCC.

Materials and methods

Tissue collection

Primary ESCC tissues and adjacent non-tumor tissues samples (>5 cm from the edge of tumor) were obtained from 47 untreated patients (27 males and 20 females; mean age of 57 years) undergoing primary surgical resection at the Department of Cardiothoracic Surgery, Jinling Hospital (Nanjing, China) between January 2014 and August 2015. The clinical staging of tumors was performed using the seventh edition of the American Joint Committee on Cancer Staging Manual (15). Samples were snap-frozen in liquid nitrogen prior to RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The current study was approved by the Human Ethics Committee of Jinling Hospital and written informed consent was obtained from each study participant.

Cell lines and cell culture

Six human ESCC cell lines (TE-1, TE-8, KYSE150, KYSE450, Eca-109 and EC9706) and the normal human esophageal epithelial cell line HEECs (cat. no. BNCC337729) were purchased from the Shanghai Institute of the Chinese Academy of Sciences (Shanghai, China). Another normal human esophageal epithelial cell line Het-1A was purchased from American Type Culture Collection (Manassas, VA, USA). TE-1, TE-8, Eca-109, EC9706 and Het-1A cells were cultured in Roswell Park Memorial Institute medium-1640 (Thermo Fisher Scientific Inc., Waltham, MA, USA), and KYSE150, KYSE450 and HEECs cells were cultured in Dulbecco's modified Eagle's medium (Thermo Fisher Scientific, Inc.). These media were supplemented with 10% fetal bovine serum (Invitrogen; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 µg/ml streptomycin in a humidified atmosphere at 37°C with 5% CO2.

RT-qPCR

Tissues samples and culture cells were treated with TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) for total RNA extraction, following the manufacturer's protocols. For miR-133b quantification, 100 ng total RNA was reverse transcribed using the specific stem-loop RT primer and the TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used. The cDNA were quantified using a TaqMan MicroRNA assay (Applied Biosystems; Thermo Fisher Scientific, Inc.) and normalized to U6 RNA levels. The qPCR reaction conditions were as follows: Initial denaturation at 95°C for 10 min and subsequently 40 cycles of 95°C for 15 sec and 60°C for 1 min. For CUL4B mRNA analysis, cDNA was synthesized from 1 µg total RNA using PrimeScript™ RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). qPCR was performed using a TaqMan RT-qPCR assay (Applied Biosystems; Thermo Fisher Scientific, Inc.) and normalized to GAPDH mRNA levels. The qPCR reaction conditions were as follows: Initial denaturation at 95°C for 5 min and subsequently 40 cycles of 95°C for 15 sec, 60°C for 15 sec and 70°C for 20 sec. Each sample was analyzed in triplicate using an ABI 7500 Fast Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). Relative expression levels were evaluated using the 2−ΔΔCq method (16). The following primers were used for qPCR: miR-133b forward, 5′-TTTGGTCCCCTTCAACCAGCTA-3′ and reverse, 5′-GTGCAGGGTCCGAGGT-3′; and CUL4B forward, 5′-GGGAAAGGAATGGTGAA-3′ and reverse, 5′-TGCATAGAGCCGGTTAG-3′. The primers used for the internal controls were as follows: U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′; and GAPDH forward, 5′-TGACTTCAACAGCGACACCCA-3′ and reverse, 5′-CACCCTGTTGCTGTAGCCAAA-3′.

miR-133b transfection

The mature miR-133b sequence (5′-UUUGGUCCCCUUCAACCAGCUA-3′) was obtained from the miRBase database (http://www.mirbase.org/). Subsequently, 0.5 µg miR-133b cDNA sequence was synthesized and inserted into the pcDNA3.1 vector to generate a human pcDNA-miR-133b plasmid. For transfection, KYSE150 or Eca-109 cells (~5×105/well) were seeded in 6-well plates. After 24 h, the cells were transfected with 3 µg pcDNA-miR-NC control vector or pcDNA-miR-133b plasmids using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Cultures were incubated for 72 h prior to collecting samples for subsequent western blotting and RT-qPCR analysis.

Cell proliferation assay

Cell proliferation was determined by culturing ~5×103 cells/well on 96-well plates and cells were transiently transfected with 2 µg pcDNA-miR-133b/miR-NC or pcDNA-CUL4B/empty plasmids (FulenGen, Guangzhou, China) using Lipofectamine 2000. After 72 h, 10 µl Cell Counting kit-8 (CCK-8) solution (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was added to each well and cells were incubated for 1 h at 37°C. Optical density was then detected at a wavelength of 450 nm using an Epoch Microplate Spectrophotometer (BioTek Instruments, Inc., Winooski, VT, USA). Each sample was analyzed in triplicate and data analysis was performed using the mean of the results.

Cell apoptosis assay

Cell apoptosis was analyzed using an Annexin V-fluorescein isothiocyanate (FITC) Apoptosis Detection kit (Beyotime Institute of Biotechnology, Haimen, China). Trypsinized cells were washed three times with PBS and stained with FITC-labeled anti-Annexin V antibody and propidium iodide in the dark for 10 min at room temperature. Cells were then analyzed using the BD FACSCalibur flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA) and FlowJo 7.6.1 software (FlowJo LLC, Ashland, OR, USA).

Western blot analysis

Transfected cells were washed once in PBS and lysed in radioimmunoprecipitation lysis buffer (Beyotime Institute of Biotechnology, Jiangsu, China) and protein concentration was determined using a BCA protein assay kit. A total of 50 µg/lane protein was separated by 10% SDS-polyacrylamide gel electrophoresis and transferred onto 0.22 µm polyvinylidene difluoride membranes, which was incubated with 5% fat-free skimmed milk in Tris-buffered saline containing 0.05% Tween-20 for 1 h at room temperature. Membranes were incubated overnight at 4°C with rabbit anti-CUL4B (cat. no. ab157103; 1:1,000; Abcam, Cambridge, UK), β-catenin (cat. no. ab32572; 1:1,000; Abcam), GSK-3β (cat. no. 9315; 1:1,000; Cell Signaling Technology, Danvars, MA, USA), phosphorylated (p)-GSK-3βTyr216 (cat. no. ab75745; 1:1,000; Abcam) and β-actin (cat. no. sc-130656; 1:1,000; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) primary antibodies. Following extensive washing with PBS supplemented with 0.1% Triton X-100, membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit secondary antibody (cat. no. ab205718; 1:1,500; Abcam) for 1 h at room temperature. Images were visualized using an enhanced chemiluminescent system (EMD Millipore, Billerica, MA, USA). Relative band intensities were determined by densitometry using Quantity One 4.6.2 software (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Luciferase reporter assays

The 3′-UTR of CUL4B with wild-type or mutant binding sites for miR-133b was amplified and subcloned into the pGL3 vector (Promega Corp., Madison, WI, USA), respectively. Eca-109 and KYSE150 cells were co-transfected with 150 ng miR-133b or miR-negative control plasmids and 50 ng pGL3-CUL4B wild-type or mutant reporter plasmids using Lipofectamine 2000. A total of 48 h following transfection, the Dual-luciferase assay kit (Promega, Madison, WI, USA) was used to determine the luciferase activity. Renilla luciferase activity was normalized to the Firefly luciferase activity. Each experiment was performed in triplicate.

Bioinformatic analysis

The target gene of miR-133b was identified and compared using the online target prediction algorithms miRanda (http://www.microrna.org/), miRWalk (http://www.umm.uni-heidelberg.de/apps/zmf/mirwalk/predictedmirnagene.html) and PicTar (http://www.pictar.org/).

Statistical analysis

All statistical analyses were performed using SPSS software 15.0 (SPSS, Inc., Chicago, IL, USA). Data are expressed as the mean ± standard error of the mean from ≥3 independent experiments. Statistical significance was evaluated using a Student's t-test (two-tailed), one-way analysis of variance followed by a Tukey's post hoc test and the Mann-Whitney test. Pearson's correlation analysis was used to measure the correlation between CUL4B and miR-133b expression. P<0.05 was determined to indicate a statistically significant difference.

Results

miR-133b expression is downregulated in human ESCC tissues and cell lines

The biological function of miR-133b in the pathogenesis of ESCC was investigated by examining miR-133b expression in 47 paired ESCC and adjacent non-tumor tissues by RT-qPCR. miR-133b expression was significantly downregulated in ESCC samples compared with adjacent normal tissues (P<0.01; Fig. 1A). Similarly, miR-133b expression in the six ESCC cell lines TE-1 (P<0.01), TE-8 (P<0.05), KYSE150 (P<0.05), KYSE450 (P<0.05), Eca-109 (P<0.01), and EC9706 (P<0.05), was significantly lower than that in the normal esophageal epithelial cell lines Het-1A and HEEC (Fig. 1B). miR-133b expression was also associated with tumor stage (P<0.05), tumor size (P<0.05) and differentiation status (P<0.05); however, it was not associated with other clinicopathological factors of patients with ESCC (Table I).

Table I.

Association between miR-133b expression and clinicopathological features of patients with ESCC.

Table I.

Association between miR-133b expression and clinicopathological features of patients with ESCC.

miR-133b expression

CharacteristicsPatients (n=47)High (n=24)Low (n=23)P-value
Age (years)
  >602915140.417
  <6018  810
Sex
  Male2714130.213
  Female20  911
Tumor localization
  Upper third12  5  70.625
  Middle third1911  8
  Lower third16  7  9
Tumor stage
  I+II25  9160.041a
  III1914  8
Tumor size (cm)
  <528  8200.034a
  ≥51912  7
Lymph node metastasis
  Negative3215170.102
  Positive15  9  6
Differentiation status
  High2813150.048a
  Low1910  9

a P<0.05. Statistical significance was evaluated using Pearson's χ2 test. miR-133b, microRNA-133b; ESCC, esophageal squamous cell carcinoma.

CUL4B is a correlated target gene of miR-133b in ESCC cells

The target gene of miR-133b in ESCC was identified using the online target prediction algorithms. Based on the Gene Expression Omnibus database analysis (17), it was revealed that human CUL4B, an important ubiquitilation molecule associated with apoptosis, contained the conserved putative miR-133b target site (Fig. 2A). Dual-luciferase reporter analysis indicated that co-expression of miR-133b significantly inhibited luciferase activity in KYSE150 (P<0.01) and Eca-109 (P<0.05) cells containing the CUL4B-3′-UTR reporter plasmid, compared with those containing the mutant plasmid (Fig. 2B and C). Endogenous expression of CUL4B was significantly inhibited following transfection of miR-133b in KYSE150 and Eca-109 cells compared with the control (P<0.05; Fig. 2D). Furthermore, CUL4B expression was significantly upregulated in ESCC tissues compared with adjacent non-tumor tissues (P<0.05; Fig. 2E) and there was a negative correlation between miR-133b and CUL4B mRNA levels in ESCC tissues (Fig. 2F). These results indicate that CUL4B is a direct target of miR-133b and suggest that miR-133b may exert its effect by inhibiting CUL4B expression.

miR-133b inhibits cell proliferation and promotes apoptosis by targeting CUL4B in ESCC

The biological function of miR-133b/CUL4B in ESCC was analyzed by transfecting miR-133b and/or CUL4B into KYSE150 and Eca-109 cells (Fig. 3A). The role of miR-133b/CUL4B on the proliferation of ESCC cells was investigated using a CCK-8 assay and the results indicated that overexpression of miR-133b significantly decreased the proliferation of KYSE150 and Eca-109 cells compared with cells transfected with the negative control (P<0.05; Fig. 3B). However, reintroduction of CUL4B into miR-133b-transfected KYSE150 or Eca-109 cells significantly reversed the effects of miR-133b on ESCCC cell proliferation (P<0.05; Fig. 3B). Additionally, overexpression of miR-133b significantly increased apoptosis in KYSE150 cells, which was significantly reversed when CUL4B was reintroduced into miR-133b-transfected KYSE150 cells (all P<0.05; Fig. 3C and D). In Eca-109 cells, miR-133b overexpression also significantly increased levels of apoptosis and the reintroduction of CUL4B inhibited the effects of apoptosis (P<0.05; Fig. 3E and F).

miR-133b inhibits the AKT/GSK3β/β-catenin pathway by downregulating CUL4B in ESCC

It has been demonstrated that CUL4B activates AKT/GSK3β/β-catenin signaling and may promote proliferation and invasion in malignant neoplasms (18). Therefore, it was hypothesized that miR-133b/CUL4B inhibits ESCC cell proliferation and promotes apoptosis by inhibiting AKT/GSK3β/β-catenin signaling pathway. After transfection of miR-133b or miR-NC plasmid, CUL4B, GSK-3β, p-GSK3βTyr216 and β-catenin levels were measured using western blot analysis. The results indicated that the expression of GSK3β (P<0.01) and p-GSK3β (P<0.05) were significantly increased in KYSE150 cells transfected with miR-133b compared with those transfected with the negative control (Fig. 4A and B). By contrast, the expression of the CUL4B (P<0.01) and downstream proliferation/apoptosis-associated target protein β-catenin (P<0.05) were significantly decreased. Similarly, levels of GSK3β and p-GSK3β were significantly increased (P<0.05), but the levels of CUL4B and β-catenin were significantly decreased (P<0.05) in Eca-109 cells transfected with miR-133b compared with cells transfected with negative control (Fig. 4C and D). Taken together, these results indicate that miR-133b/CUL4B may affect ESCC cell proliferation and apoptosis by regulating the AKT/GSK3β/β-catenin pathway.

Discussion

miR-133b is a muscle-specific molecular marker, which serves a role in skeletal muscle development, myoblast differentiation and myogenic-associated disease (19,20). It has been demonstrated that miR-133b serves a suppressive role during tumor growth, invasion, metastasis and apoptosis. miR-133b inhibits gastric cancer cell metastasis in vitro and in vivo by directly suppressing the expression of zinc finger protein Gli1 (21). miR-133b promotes the apoptosis and inhibits the proliferation of osteosarcoma cells by directly targeting B cell lymphoma-2 like protein (22). Downregulation of miR-133b in colorectal cancer tissues compared with adjacent non-tumorous tissues is associated with the poor survival of patients (23). Furthermore, it has been demonstrated that miR-133b inhibits the invasiveness of esophageal types of cancer by inhibiting FSCN1 expression (14). However, the molecular mechanisms of miR-133b in ESCC cell apoptosis and proliferation remain unknown.

The results of the present study demonstrated that miR-133b significantly decreased tumor cell proliferation and promoted apoptosis in vitro. These results indicate that miR-133b may be used as a novel method of treating patients with ESCC. miR-133b levels were also highly associated with tumor stage and differentiation status; differentiation-associated miR-133b levels may be used to predict tumor progression in patients with ESCC that have undergone surgery; however, further studies are required to validate this.

It was also demonstrated that miR-133b/CUL4B served a role in ESCC cell growth and apoptosis. CUL4B is a member of the cullin family and forms a complex that functions as an E3 ubiquitin ligase and catalyzes the polyubiquitination of specific protein substrates in the cell (24,25). Previous studies have demonstrated that CUL4B expression is significantly upregulated in various types of human cancer, promoting cell proliferation, invasion and tumorigenesis (2628). For example, CUL4B promotes the proliferation and inhibits the apoptosis of osteosarcoma and glioblastoma cells (26,27). In addition, CUL4B is a novel prognostic marker correlating with colon cancer pathogenesis and progression (28). The mechanism underlying CUL4B function in cancer progression remains unclear; however, CUL4B may serve a role in epigenetic changes, including heterochromatin formation, histone modification, parental imprinting or X-chromosome inactivation (29,30). CUL4B also promotes cell cycle progression and tumorigenesis via degradation of numerous cyclin-dependent kinase inhibitors or p53 protein (31,32). In addition, CUL4B serves an important role in stabilizing β-catenin against proteasomal degradation in multiple signaling pathways. The results of the current study indicate that CUL4B activates the AKT/GSK3β/β-catenin pathway and may affect ESCC cell proliferation and apoptosis by regulating this pathway. These results provide important insights into the CUL4B pathway and shed light on the functional importance of proliferation and apoptosis in ESCC.

In conclusion, the current study demonstrated that miR-133b is downregulated in ESCC, and its expression is associated with advanced tumor stage and the differentiation status of patients with ESCC. Additionally, it was determined that miR-133b serves a crucial role in inducing proliferation and apoptosis by directly targeting CUL4B and may therefore be a novel therapeutic target to treat patients with ESCC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contribution

HH designed methods and experiments, performed the laboratory experiments and analyzed the data. YX and ZG co-designed the cell proliferation and apoptosis experiments and collaborated on associated data collection and their interpretation. XC and SJ collaborated on bioinformatics analysis and statistical analysis. ZX co-designed experiments, discussed analyses, interpretation, presentation and wrote the paper. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The current study was performed in accordance with the Declaration of Helsinki and approved by the Human Ethics Committee of Jinling Hospital. Written informed consent was obtained from each study participant.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zhou K, Zhang SS, Yan Y and Zhao S: Overexpression of transient receptor potential vanilloid 2 is associated with poor prognosis in patients with esophageal squamous cell carcinoma. Med Oncol. 31:172014. View Article : Google Scholar : PubMed/NCBI

2 

Napier KJ, Scheerer M and Misra S: Esophageal cancer: A review of epidemiology, pathogenesis, staging workup and treatment modalities. World J Gastrointest Oncol. 6:112–120. 2014. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Toh Y, Egashira A and Yamamoto M: Epigenetic alterations and their clinical implications in esophageal squamous cell carcinoma. Gen Thorac Cardiovasc Surg. 61:262–269. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Song Y, Li L, Ou Y, Gao Z, Li E, Li X, Zhang W, Wang J, Xu L, Zhou Y, et al: Identification of genomic alterations in oesophageal squamous cell cancer. Nature. 509:91–95. 2014. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Lin S and Gregory RI: MicroRNA biogenesis pathways in cancer. Nat Rev Cancer. 15:321–333. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Gu J, Wang Y and Wu X: MicroRNA in the pathogenesis and prognosis of esophageal cancer. Curr Pharm Des. 19:1292–1300. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Feber A, Xi L, Luketich JD, Pennathur A, Landreneau RJ, Wu M, Swanson SJ, Godfrey TE and Litle VR: MicroRNA expression profiles of esophageal cancer. J Thorac Cardiovasc Surg. 135:255–260. 2008. View Article : Google Scholar : PubMed/NCBI

10 

He B, Yin B, Wang B, Xia Z, Chen C and Tang J: MicroRNAs in esophageal cancer (Review). Mol Med Rep. 6:459–465. 2012.PubMed/NCBI

11 

Li P, Mao WM, Zheng ZG, Dong ZM and Ling ZQ: Down-regulation of PTEN expression modulated by dysregulated miR-21 contributes to the progression of esophageal cancer. Dig Dis Sci. 58:3483–3493. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Sugimura K, Miyata H, Tanaka K, Hamano R, Takahashi T, Kurokawa Y, Yamasaki M, Nakajima K, Takiguchi S, Mori M and Doki Y: Let-7 expression is a significant determinant of response to chemotherapy through the regulation of IL-6/STAT3 pathway in esophageal squamous cell carcinoma. Clin Cancer Res. 18:5144–5153. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Lin RJ, Xiao DW, Liao LD, Chen T, Xie ZF, Huang WZ, Wang WS, Jiang TF, Wu BL, Li EM and Xu LY: MiR-142-3p as a potential prognostic biomarker for esophageal squamous cell carcinoma. J Surg Oncol. 105:175–182. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Kano M, Seki N, Kikkawa N, Fujimura L, Hoshino I, Akutsu Y, Chiyomaru T, Enokida H, Nakagawa M and Matsubara H: miR-145, miR-133a and miR-133b: Tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma. Int J Cancer. 127:2804–2814. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Edge SB and Compton CC: The American joint committee on cancer: The 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 17:1471–1474. 2010. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Clough E and Barrett T: The gene expression omnibus database. Methods Mol Biol. 1418:93–110. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Qian Y, Yuan J, Hu H, Yang Q, Li J, Zhang S, Jiang B, Shao C and Gong Y: The CUL4B/AKT/β-catenin axis restricts the accumulation of myeloid-derived suppressor cells to prohibit the establishment of a tumor-permissive microenvironment. Cancer Res. 75:5070–5083. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Williams AH, Liu N, van Rooij E and Olson EN: MicroRNA control of muscle development and disease. Curr Opin Cell Biol. 21:461–469. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Kirby TJ and McCarthy JJ: MicroRNAs in skeletal muscle biology and exercise adaptation. Free Radic Biol Med. 64:95–105. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Zhao Y, Huang J, Zhang L, Qu Y, Li J, Yu B, Yan M, Yu Y, Liu B and Zhu Z: MiR-133b is frequently decreased in gastric cancer and its overexpression reduces the metastatic potential of gastric cancer cells. BMC Cancer. 14:342014. View Article : Google Scholar : PubMed/NCBI

22 

Zhao H, Li M, Li L, Yang X, Lan G and Zhang Y: MiR-133b is down-regulated in human osteosarcoma and inhibits osteosarcoma cells proliferation, migration and invasion, and promotes apoptosis. PLoS One. 8:e835712013. View Article : Google Scholar : PubMed/NCBI

23 

Hu G, Chen D, Li X, Yang K, Wang H and Wu W: miR-133b regulates the MET proto-oncogene and inhibits the growth of colorectal cancer cells in vitro and in vivo. Cancer Biol Ther. 10:190–197. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Higa LA, Wu M, Ye T, Kobayashi R, Sun H and Zhang H: CUL4-DDB1 ubiquitin ligase interacts with multiple WD40-repeat proteins and regulates histone methylation. Nat Cell Biol. 8:1277–1283. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Kerzendorfer C, Hart L, Colnaghi R, Carpenter G, Alcantara D, Outwin E, Carr AM and O'Driscoll M: CUL4B-deficiency in humans: Understanding the clinical consequences of impaired Cullin 4-RING E3 ubiquitin ligase function. Mech Ageing Dev. 132:366–373. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Dong J, Wang XQ, Yao JJ, Li G and Li XG: Decreased CUL4B expression inhibits malignant proliferation of glioma in vitro and in vivo. Eur Rev Med Pharmacol Sci. 19:1013–1021. 2015.PubMed/NCBI

27 

Chen Z, Shen BL, Fu QG, Wang F, Tang YX, Hou CL and Chen L: CUL4B promotes proliferation and inhibits apoptosis of human osteosarcoma cells. Oncol Rep. 32:2047–2053. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Jiang T, Tang HM, Wu ZH, Chen J, Lu S, Zhou CZ, Yan DW and Peng ZH: Cullin 4B is a novel prognostic marker that correlates with colon cancer progression and pathogenesis. Med Oncol. 30:5342013. View Article : Google Scholar : PubMed/NCBI

29 

Jia S, Kobayashi R and Grewal SI: Ubiquitin ligase component Cul4 associates with Clr4 histone methyltransferase to assemble heterochromatin. Nat Cell Biol. 7:1007–1013. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Dumbliauskas E, Lechner E, Jaciubek M, Berr A, Pazhouhandeh M, Alioua M, Cognat V, Brukhin V, Koncz C, Grossniklaus U, et al: The Arabidopsis CUL4-DDB1 complex interacts with MSI1 and is required to maintain MEDEA parental imprinting. EMBO J. 30:731–743. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Higa LA, Yang X, Zheng J, Banks D, Wu M, Ghosh P, Sun H and Zhang H: Involvement of CUL4 ubiquitin E3 ligases in regulating CDK inhibitors Dacapo/p27Kip1 and cyclin E degradation. Cell Cycle. 5:71–77. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Nishitani H, Shiomi Y, Iida H, Michishita M, Takami T and Tsurimoto T: CDK inhibitor p21 is degraded by a proliferating cell nuclear antigen-coupled Cul4-DDB1Cdt2 pathway during S phase and after UV irradiation. J Biol Chem. 283:29045–29052. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 15 Issue 4

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
Huang H, Xu Y, Guo Z, Chen X, Ji S and Xu Z: MicroRNA-133b inhibits cell proliferation and promotes apoptosis by targeting cullin 4B in esophageal squamous cell carcinoma. Exp Ther Med 15: 3743-3750, 2018
APA
Huang, H., Xu, Y., Guo, Z., Chen, X., Ji, S., & Xu, Z. (2018). MicroRNA-133b inhibits cell proliferation and promotes apoptosis by targeting cullin 4B in esophageal squamous cell carcinoma. Experimental and Therapeutic Medicine, 15, 3743-3750. https://doi.org/10.3892/etm.2018.5906
MLA
Huang, H., Xu, Y., Guo, Z., Chen, X., Ji, S., Xu, Z."MicroRNA-133b inhibits cell proliferation and promotes apoptosis by targeting cullin 4B in esophageal squamous cell carcinoma". Experimental and Therapeutic Medicine 15.4 (2018): 3743-3750.
Chicago
Huang, H., Xu, Y., Guo, Z., Chen, X., Ji, S., Xu, Z."MicroRNA-133b inhibits cell proliferation and promotes apoptosis by targeting cullin 4B in esophageal squamous cell carcinoma". Experimental and Therapeutic Medicine 15, no. 4 (2018): 3743-3750. https://doi.org/10.3892/etm.2018.5906