Let-7c-5p inhibits cell proliferation and induces cell apoptosis by targeting ERCC6 in breast cancer

  • Authors:
    • Xiaonan Fu
    • Xuan Mao
    • Yuxiang Wang
    • Xianfeng Ding
    • Yongfeng Li
  • View Affiliations

  • Published online on: July 19, 2017     https://doi.org/10.3892/or.2017.5839
  • Pages: 1851-1856
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In this study, we found that let-7c-5p expression was clearly downregulated in breast cancer tissues compared with that of corresponding adjacent tissues. Furthermore, overexpression of let-7c-5p in MCF-7 breast cancer cells could significantly inhibit cell proliferation and induce cell apoptosis. The target genes of let-7c-5p were predicted by the way of bioinformatics, and validated by dual luciferase reporter assay and western blotting demonstrating that excision repair cross complementing 6 (ERCC6) gene was a direct target. Collectively, the present study suggested that let-7c-5p acted as a tumor suppressor in breast cancer possibly by negatively regulating ERCC6, which took an important part in nucleotide excision repair and it may provide a new potential strategy for breast cancer therapy.

Introduction

Breast cancer is one of the most common malignant diseases among women both in developed and developing countries, accounting for 29% of new cancer cases and 14% of cancer-associated deaths among American women (1). In China, the incidence of breast cancer is lower than in America, but during the past decade, the incidence has been increasing markedly, especially in rural areas (2). Therefore, more effective methods for breast cancer diagnosis and treatments are urgently needed.

MicroRNAs (miRNAs) are endogenous non-coding RNAs that function as gene regulators mainly by binding to the 3′UTR of their target mRNAs, inducing mRNAs degradation or translation repression (3). Biological processes, including cell proliferation, differentiation, apoptosis and senescence are controlled by miRNAs. Aberrant expression of miRNAs is associated with a variety of cancers, such as lung cancer, gastric cancer, and breast cancer (46). Let-7 was originally discovered in Caenorhabditis elegans, controlling the timing of stem-cell division and differentiation (7). Subsequently, let-7 and its family members have been found playing important roles in tumor suppression. Akao et al reported that let-7 might suppress the growth of human colon cancer cells and reduced oncogene RAS and c-myc expression (8). Esquela-Kerscher et al found that let-7 could inhibit the growth of lung cancer cell lines, as well as the growth of lung cancer cell xenografts in mice (9). Let-7c-5p, a member of let-7 family, is downregulated in prostate cancer, overexpression of let-7c-5p suppresses androgen receptor expression and leads to inhibition of prostate cancer cell proliferation (10). Nwaeburu et al found that forced expression of let-7c-5p by quercetin could activate Numbl expression and resulted in suppression of pancreatic cancer progression (11). Some other studies suggest that let-7c-5p functions as a anti-oncogene in human non-small cell lung cancer, hepatocellular carcinoma and colorectal cancer (1214).

Although let-7c-5p has been studied in several cancers, its function and molecular mechanism in breast cancer remain to be further identified. Here, we measured let-7c-5p expression in breast cancer tissues and corresponding adjacent tissues, and examined the effects of let-7c-5p on human breast cancer cell proliferation and apoptosis. Moreover, for the first time, we found that ERCC6 was a target of let-7c-5p in breast cancer.

Materials and methods

Tissues collection

Nine paraffin-embedded breast cancer and corresponding adjacent tissue samples were collected from Zhejiang Cancer Hospital (Hangzhou, China). This study was approved by the ethics committee of the Zhejiang Sci-Tech University (Hangzhou, China) and Zhejiang Cancer Hospital. All the samples were stored at 4°C until RNA extraction.

RNA extraction and qRT-PCR

Total RNA was extracted from paraffin-embedded tissues using Recover All™ Total Nucleic Acid Isolation (Ambion, Austin, TX, USA) following the manufacturer's instructions. RNA concentrations were measured by the NanoDrop ND-2000 spectrophotometer (NanoDrop Technologies, Wilmington, DE, USA). RNA reverse transcription was performed with 100 ng of total RNA. Expression of let-7c-5p, ERCC6 and internal control GAPDH were detected using SYBR Premix Ex Taq™ II (Takara, Dalian, China). qRT-PCR was performed on the Applied Biosystems 7500 real-time PCR system (Applied Biosystems, Foster City, CA, USA). Relative expression was calculated using the 2−∆∆Ct method, the expression of let-7c-5p and ERCC6 was normalized with GAPDH. Primers for qRT-PCR are listed in Table I.

Table I.

Primers for qRT-PCR.

Table I.

Primers for qRT-PCR.

PrimerSequence (5′-3′)
let-7c-5p GTCGTATCCAGTGCAGGGTCCGAGG
stem-loop TATTCGCACTGGATACGACAACCAT
let-7c-5pF: GAGGTAGTAGGTTGTATGGTTG
R: GCAGGGTCCGAGGTATTC
ERCC6F: CAATAGTCTGCCTCCCCACCCC
R: CAACTTCTCGTTCCTCAACACATC
GAPDHF: TGCCAAATATGATGACATCAAGAA
R: GGAGTGGGTGTCGCTGTTG

[i] F, forward; R, reverse.

Cell culture and transfection

The MCF-7 human breast cancer cell line was obtained from Zhejiang Sci-Tech University, cultured in DMEM-high glucose medium supplemented with 10% fetal bovine serum (Gibco, Grand Island, NY, USA), at 37°C in a 5% CO2 humidified incubator (HF90, Heal Force, Hong Kong). Mimics for miR-NC (5′-CAGUACUUUUGUGUAGUACAA-3′) and let-7c-5p (5′-UGAGGUAGUAGGUUGUAUGGUU-3′) were purchased from GenePharma (Shanghai, China). Reverse transfection was performed in this study using GeneTran™ III High Efficiency Transfection Reagent (Biomiga, Inc., San Diego, CA, USA) according to the manufacturer's recommendations.

MTT assay

MCF-7 cells were seeded in 96-well plates at approximately 4000 cells per well and transfected with miR-NC or let-7c-5p mimics. MTT (Sigma, St. Louis, MO, USA) was used to examine the effects of let-7c-5p on cell proliferation at 24, 48 and 72 h post-transfection. At each time, 20 µl MTT solution was added to each well, and incubated at 37°C for 4 h, the formazan produced by viable cells was dissolved in 150 µl dimethylsulfoxide (DMSO). The cell numbers were evaluated by reading the absorbance at 490 nm (15).

Cell apoptosis assay

Cells were seeded in 6-well plates at approximately 3×105 cells per well and transfected with miR-NC or let-7c-5p mimics. All cells were harvested at 24 h post-transfection, washed with cold PBS for twice, stained with FITC and PI using BD Pharmingen FITC Annexin V™ apoptosis detection kit I (BD Biosciences, San Jose, CA, USA) following the manufacturer's instructions. Flow cytometry analysis was finished on BD Accuri C6 flow cytometer with C6 software.

Dual luciferase reporter assay

The potential target genes of let-7c-5p were predicted using the algorithms of TargetScan (http://www.targetscan.org/vert_71/), PicTar (http://pictar.mdc-berlin.de/) and starBase (http://starbase.sysu.edu.cn/). ERCC6 was taken as a potential target gene of let-7c-5p. There was only one potential complementary site for let-7c-5p in the 3′UTR of ERCC6 mRNA, and the wild-type 3′UTR fragment containing putative binding site was amplified by PCR and cloned into the XbaI and NotI sites downstream of Renilla luciferase vector pRL-TK (Promega, Beijing, China), the recombinant plasmid was named pRL-ERCC6-wt. The putative let-7c-5p binding site was deleted by overlap-extension PCR, and the recombinant plasmid was named pRL-ERCC6-mut. Primers are listed in Table II, constructs were verified by sequencing.

Table II.

Primers for construction of recombinant plasmids.

Table II.

Primers for construction of recombinant plasmids.

PrimerSequence (5′-3′)
Wild-typeF: ACAACATTGCTTCCTA
3′UTRR: TCAATCCAAGTATTTTCTCC
MutantF: ACAACATTGCTTCCTA
3′UTR-1R: AAGTTTTAATTCACATCATGCAAACAA
MutantF: TGTTTGCATGATGTGAATTACAACTT
3′UTR-2R: TCAATCCAAGTATTTTCTCC

[i] F, forward; R, reverse.

For dual luciferase reporter assay, cells were seeded in 24-well plates in triplicate and cotransfected with the firefly luciferase vector pGL3 and recombinant plasmid pRL-ERCC6-wt or pRL-ERCC6-mut, together with mimics for either miR-NC or let-7c-5p at a final concentration of 20 nM. After 48 h, luciferase activities were measured using the dual luciferase reporter assay system (Promega). Renilla luciferase activity was normalized with firefly luciferase activity.

Western blotting

Cells were harvested at 48 h post-transfection and lysed in RIPA buffer on ice. Total proteins were separated by 10% SDS-PAGE, electroblotted onto PVDF membranes. Membranes were blocked with 5% non-fat milk powder for 2 h at room temperature and incubated overnight at 4°C with a polyclonal antibody: anti-ERCC6 antibody (1:2000 dilution; Abcam, Cambridge, MA, USA) or anti-GAPDH antibody (1:2000 dilution; HuaBio, China). After washing three times with TBS-T, membranes were incubated with a secondary antibody anti-rabbit HRP-conjugate (1:2000 dilution; HuaBio) for 2 h at room temperature (16). Antibody detection was performed with ECL Western Blotting Substrate (Solarbio, Beijing, China), and photos were taken using the Tanon 5500 imaging system (Tanon, Shanghai, China).

Statistical analysis

Statistical analysis was performed using SPSS software version 17.0 (SPSS, Inc., Chicago, IL, USA). All data were presented as mean ± standard deviation (SD). Statistical significance was tested by Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Reduced expression of let-7c-5p in breast cancer tissues

As shown in Fig. 1, expression of let-7c-5p was significantly decreased in the nine breast cancer tissues, indicating that let-7c-5p could potentially serve as a biomarker for breast cancer.

Overexpression of let-7c-5p inhibits MCF-7 cell proliferation and induces cell apoptosis

To determine the transfection efficiency, cells were harvested after transfection with 40 or 80 nM let-7c-5p mimics for 24 h. Total RNA was isolated from cells using the traditional TRIzol method. qRT-PCR was performed to measure the expression of let-7c-5p. As shown in Fig. 2A, let-7c-5p expression was enhanced significantly by let-7c-5p mimics.

To examine the effects of let-7c-5p on MCF-7 cell proliferation in vitro, MTT assay was performed at 24, 48 and 72 h post-transfection, the cell growth curve showed that the proliferation ability of transfected cells was reduced remarkably compared to control group cells (Fig. 2B).

Additionally, we explored whether cell apoptosis could be induced by let-7c-5p. After transfection for 24 h, cells were harvested, and cell apoptosis was detected by flow cytometer. As shown in Fig. 2C, early stage apoptotic cells are presented in the lower right quadrant, a higher rate of apoptosis was observed in transfected cells, especially in those transfected with 80 nM let-7c-5p mimics (Fig. 2D).

ERCC6 is a direct target gene of let-7c-5p in breast cancer

TargetScan, PicTar and starBase were used to predict the target genes of let-7c-5p, and the intersection of the prediction results from the three algorithms showed that there were 41 candidate genes with at least one binding site of let-7c-5p (Table III). Of these genes, ITGB3 and MAP4K3 have been verified as targets of let-7c-5p in human non-small cell lung cancer. In this experiment, we aimed to find a new target of let-7c-5p in breast cancer. ERCC6, which has been reported to be associated with cancer risk, was selected as a putative target gene.

Table III.

Potential target genes of let-7c-5p predicted by TargetScan, PicTar and starBase.

Table III.

Potential target genes of let-7c-5p predicted by TargetScan, PicTar and starBase.

Potential target genesPosition
ADRB2 chr5:148207936–148207943
AHCTF1 chr1:247003542–247003549
APBB3 chr5:139937904–139937911
BACH1 chr21:30717135–30717142
CD200R1 chr3:112641981–112641988
CDV3 chr3:133307221–133307228
CLDN12 chr7:90043275–90043282
COIL chr17:55016284–55016291
COL1A1 chr17:48262068–48262074
DMD chrX:31138095–31138102
E2F6 chr2:11585579–11585586
ERCC6 chr10:50666729–50666736
FGD6 chr12:95474757–95474764
FNIP1 chr5:130979161–130979168
FRAS1 chr4:79462429–79462436
GAN chr16:81412340–81412347
GNPTAB chr12:102140912–102140919
GOLT1B chr12:21670278–21670285
HAND1 chr5:153854615–153854622
INTS2 chr17:59943299–59943306
IRS2 chr13:110408191–110408198
ITGB3 chr17:45388633–45388639
KLHDC8B chr3:49213851–49213858
LRIG2 chr1:113666778–113666785
LRIG3 chr12:59266314–59266321
MAP4K3 chr2:39476696–39476703
MED8 chr1:43850492–43850499
MLL2 chr12:49414882–49414889
NAP1L1 chr12:76441001–76441008
NLK chr17:26522778–26522784
PRPF38B chr1:109243302–109243309
RANBP2 chr2:109401057–109401064
RNF20 chr9:104324767–104324773
RRM2 chr2:10269649–10269656
SEMA4C chr2:97526289–97526296
SLC20A1 chr2:113421361–113421367
SLC35D2 chr9:99083376–99083383
TMEM2 chr9:74298636–74298643
WDR37 chr10:1176192–1176199
ZCCHC3 chr20:280315–280321
ZFYVE26 chr14:68214907–68214914

The let-7c-5p binding sites in 3′UTR of ERCC6 mRNA were predicted by TargetScan, as a result, there was only one potential complementary sequence for let-7c-5p at nt 125–132 (Fig. 3A). To validate whether let-7c-5p targets the 3′UTR of ERCC6 mRNA, we used the firefly luciferase vector pGL3 as an internal control plasmid, and co-transfected pGL3 and recombinant plasmid pRL-ERCC6-wt or pRL-ERCC6-mut along with miR-NC or let-7c-5p mimics into MCF-7 cells. Luciferase activities were measured after co-transfection for 48 h, as shown in Fig. 3B, dual luciferase reporter assay demonstrated that the Renilla luciferase activity of pRL-ERCC6-wt, which contained the wild-type 3′UTR fragment of ERCC6, was decreased by approximately 50% by let-7c-5p, compared to the miR-NC group. In addition, the luciferase activity of pRL-ERCC6-mut, in which the predicted binding site has been deleted, was not suppressed significantly. These results showed that ERCC6 was a direct target gene for let-7c-5p in breast cancer.

To better understand the effects of let-7c-5p on ERCC6, we evaluated the ERCC6 mRNA and protein expression level in MCF-7 cells after treatment with let-7c-5p mimics for 48 h. As shown in Fig. 3C and D, protein expression of ERCC6 was clearly inhibited by let-7c-5p, but the mRNA expression of ERCC6 did not change significantly. These findings suggested that let-7c-5p regulated ERCC6 mainly through suppression of ERCC6 protein accumulation, but not affecting the mRNA level.

Discussion

A large number of studies suggest that miRNAs are involved in multiple human cancers, some are oncogenes, like miR-183, which is over-expressed in synovial sarcoma, rhabdomyosarcoma and colon cancer, targets the tumor suppressor gene EGR1 and contributes to cell migration in these tumor types (17). Some other miRNAs may act as tumor suppressor genes, such as miR-100, which is found to reduce colon cancer cell motility and growth by targeting Lgr5 in vitro (18). MiRNA let-7c-5p acts as a cancer suppressor in different ways, such as preventing early cancer progression through repressing HMGA2 expression (19), inhibiting migration and invasion of human non-small cell lung cancer and colorectal cancer (13,14), and inducing cell apoptosis and disrupting cell cycle in human hepatocellular carcinoma cells (12). As for breast cancer, let-7c-5p is downregulated in both tissues and serum of the patients, and the expression of let-7c-5p is affected by postmenopausal status (20). Higher expression level of let-7c-5p is reported to be correlated with better clinical prognosis of patients with estrogen receptor-positive breast cancer (21), thus we hypothesize that let-7c-5p may play a suppressive part in breast cancer.

In this study, we also confirmed that let-7c-5p expression was decreased in breast cancer tissues compared with that of matched adjacent tissues, moreover, we found that overexpression of let-7c-5p could inhibit breast cancer cell proliferation and induce cell apoptosis significantly, these observations authenticated the hypothesis that let-7c-5p acted as a tumor suppressor in breast cancer. Furthermore, we found that let-7c-5p could bind to the 3′UTR of ERCC6 mRNA. However, miRNAs regulate their target mRNAs in different ways, if the miRNAs have perfect or near-perfect complementarity to the 3′UTR of target mRNAs, the target mRNAs will be destroyed by miRNA, in this situation, both the mRNA and protein levels will be affected. On the other hand, the miRNAs have partial complementary sites in the 3′UTR of target mRNAs, the inhibition of protein accumulation will be direct, but the mRNA level may not be affected significantly (22,23). Herein, let-7c-5p has partial complementarity to the 3′UTR of ERCC6 mRNA, as a result, ERCC6 protein expression was suppressed but ERCC6 mRNA expression was not inhibited obviously by let-7c-5p in MCF-7 cells.

ERCC6, also known as CSB, is a chromatin remodeling factor. The protein encoded by ERCC6 gene is a DNA-binding protein, which has important activity in nucleotide excision repair (24). It has been identified that mutations in ERCC6 gene are associated with the rare disease Cockayne syndrome type B (25). In recent years, ERCC6 is also showed to be involved in cancers. Ma et al unraveled some genetic variants of ERCC6 jointly taking part in the lung cancer development among Chinese people (26). Chiu et al and Liu et al also reported that ERCC6 polymorphisms were associated with the increased risk of gastric cancer and oral cancer (27,28).

Chromosomal instability is observed in most cancers including breast cancer. In cancer cells with high levels of chromosomal instability and replication stress, DNA damage occurs more frequently, and accumulation of DNA damage results in cell malfunction and cell apoptosis (29,30). So more DNA repair mechanisms need to be activated to correct the damage and maintain the activity of cancer cells (31,32). In the current study, we find that suppression of ERCC6 protein expression by let-7c-5p is related with reduced growth ability of breast cancer cells and higher rate of apoptosis, indicating ERCC6 may be involved in DNA damage repair in breast cancer and reduced ERCC6 protein level leads to DNA damage accumulation.

Many clinical anticancer drugs target DNA directly (33), such as cisplatin, which interferes with DNA replication and induces DNA damage by binding to DNA and forming DNA-cisplatin adducts. However, cisplatin resistance is a serious problem in cancer treatment, and activation of DNA repair mechanism is one of the main mechanisms involved in cisplatin resistance (34). The expression of excision repair cross-complementation group 1 (ERCC1), which participates in nucleotide excision repair, can be induced by cisplatin treatment, and increased excision of DNA-cisplatin adducts contributes to cisplatin resistance (35). As a DNA-binding protein, ERCC6 is also likely to be associated with anticancer drugs resistance via the nucleotide excision repair. Nevertheless, this potential mechanism of ERCC6 has not been verified, thus more studies on ERCC6 are still needed.

In conclusion, our study suggested that let-7c-5p was a crucial contributor to cell apoptosis and inhibition of cell proliferation in breast cancer, partially through targeting ERCC6. These findings may provide a new potential strategy for breast cancer treatment.

Acknowledgements

This study was supported by the Natural Science Foundation of Zhejiang Province (LY15C050002 and LQ17H160013). This study was also supported by the National Natural Science Foundation of China (81402100), the Foundation of Health and Family Planning Commission of Jiangsu Province (Q201408) and the Social Development Foundation of Zhejiang (SH2016031, SH2014026).

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Filipowicz W, Bhattacharyya SN and Sonenberg N: Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nat Rev Genet. 9:102–114. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Chen LT, Xu SD, Xu H, Zhang JF, Ning JF and Wang SF: MicroRNA-378 is associated with non-small cell lung cancer brain metastasis by promoting cell migration, invasion and tumor angiogenesis. Med Oncol. 29:1673–1680. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Guo B, Liu L, Yao J, Ma R, Chang D, Li Z, Song T and Huang C: miR-338-3p suppresses gastric cancer progression through a PTEN-AKT axis by targeting P-REX2a. Mol Cancer Res. 12:313–321. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Shi Y, Luo X, Li P, Tan J, Wang X, Xiang T and Ren G: miR-7-5p suppresses cell proliferation and induces apoptosis of breast cancer cells mainly by targeting REGγ. Cancer Lett. 358:27–36. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Roush S and Slack FJ: The let-7 family of microRNAs. Trends Cell Biol. 18:505–516. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Akao Y, Nakagawa Y and Naoe T: let-7 microRNA functions as a potential growth suppressor in human colon cancer cells. Biol Pharm Bull. 29:903–906. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Esquela-Kerscher A, Trang P, Wiggins JF, Patrawala L, Cheng A, Ford L, Weidhaas JB, Brown D, Bader AG and Slack FJ: The let-7 microRNA reduces tumor growth in mouse models of lung cancer. Cell Cycle. 7:759–764. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Nadiminty N, Tummala R, Lou W, Zhu Y, Zhang J, Chen X, White RW eVere, Kung HJ, Evans CP and Gao AC: MicroRNA let-7c suppresses androgen receptor expression and activity via regulation of Myc expression in prostate cancer cells. J Biol Chem. 287:1527–1537. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Nwaeburu CC, Bauer N, Zhao Z, Abukiwan A, Gladkich J, Benner A and Herr I: Up-regulation of microRNA let-7c by quercetin inhibits pancreatic cancer progression by activation of Numbl. Oncotarget. 7:58367–58380. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Zhu X, Wu L, Yao J, Jiang H, Wang Q, Yang Z and Wu F: MicroRNA let-7c inhibits cell proliferation and induces cell cycle arrest by targeting CDC25A in human hepatocellular carcinoma. PLoS One. 10:e01242662015. View Article : Google Scholar : PubMed/NCBI

13 

Han HB, Gu J, Zuo HJ, Chen ZG, Zhao W, Li M, Ji DB, Lu YY and Zhang ZQ: Let-7c functions as a metastasis suppressor by targeting MMP11 and PBX3 in colorectal cancer. J Pathol. 226:544–555. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Zhao B, Han H, Chen J, Zhang Z, Li S, Fang F, Zheng Q, Ma Y, Zhang J, Wu N, et al: MicroRNA let-7c inhibits migration and invasion of human non-small cell lung cancer by targeting ITGB3 and MAP4K3. Cancer Lett. 342:43–51. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Mao Y, Wu S, Zhao R and Deng Q: MiR-205 promotes proliferation, migration and invasion of nasopharyngeal carcinoma cells by activation of AKT signalling. J Int Med Res. 44:231–240. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Wu Q, Jin H, Yang Z, Luo G, Lu Y, Li K, Ren G, Su T, Pan Y, Feng B, et al: MiR-150 promotes gastric cancer proliferation by negatively regulating the pro-apoptotic gene EGR2. Biochem Biophys Res Commun. 392:340–345. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Sarver AL, Li L and Subramanian S: MicroRNA miR-183 functions as an oncogene by targeting the transcription factor EGR1 and promoting tumor cell migration. Cancer Res. 70:9570–9580. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Zhou MK, Liu XJ, Zhao ZG and Cheng YM: MicroRNA-100 functions as a tumor suppressor by inhibiting Lgr5 expression in colon cancer cells. Mol Med Rep. 11:2947–2952. 2015.PubMed/NCBI

19 

Park SM, Shell S, Radjabi AR, Schickel R, Feig C, Boyerinas B, Dinulescu DM, Lengyel E and Peter ME: Let-7 prevents early cancer progression by suppressing expression of the embryonic gene HMGA2. Cell Cycle. 6:2585–2590. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Li XX, Gao SY, Wang PY, Zhou X, Li YJ, Yu Y, Yan YF, Zhang HH, Lv CJ, Zhou HH, et al: Reduced expression levels of let-7c in human breast cancer patients. Oncol Lett. 9:1207–1212. 2015.PubMed/NCBI

21 

Sun X, Xu C, Tang SC, Wang J, Wang H, Wang P, Du N, Qin S, Li G, Xu S, et al: Let-7c blocks estrogen-activated Wnt signaling in induction of self-renewal of breast cancer stem cells. Cancer Gene Ther. 23:83–89. 2016. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Pillai RS, Bhattacharyya SN and Filipowicz W: Repression of protein synthesis by miRNAs: How many mechanisms? Trends Cell Biol. 17:118–126. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Selby CP and Sancar A: Human transcription-repair coupling factor CSB/ERCC6 is a DNA-stimulated ATPase but is not a helicase and does not disrupt the ternary transcription complex of stalled RNA polymerase II. J Biol Chem. 272:1885–1890. 1997. View Article : Google Scholar : PubMed/NCBI

25 

Ghai SJ, Shago M, Shroff M and Yoon G: Cockayne syndrome caused by paternally inherited 5 Mb deletion of 10q11.2 and a frameshift mutation of ERCC6. Eur J Med Genet. 54:272–276. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Ma H, Hu Z, Wang H, Jin G, Wang Y, Sun W, Chen D, Tian T, Jin L, Wei Q, et al: ERCC6/CSB gene polymorphisms and lung cancer risk. Cancer Lett. 273:172–176. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Chiu CF, Tsai MH, Tseng HC, Wang CL, Tsai FJ, Lin CC and Bau DT: A novel single nucleotide polymorphism in ERCC6 gene is associated with oral cancer susceptibility in Taiwanese patients. Oral Oncol. 44:582–586. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Liu JW, He CY, Sun LP, Xu Q, Xing CZ and Yuan Y: The DNA repair gene ERCC6 rs1917799 polymorphism is associated with gastric cancer risk in Chinese. Asian Pac J Cancer Prev. 14:6103–6108. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Leung HW, Wu CH, Lin CH and Lee HZ: Luteolin induced DNA damage leading to human lung squamous carcinoma CH27 cell apoptosis. Eur J Pharmacol. 508:77–83. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Kaina B: DNA damage-triggered apoptosis: Critical role of DNA repair, double-strand breaks, cell proliferation and signaling. Biochem Pharmacol. 66:1547–1554. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Minocherhomji S, Ying S, Bjerregaard VA, Bursomanno S, Aleliunaite A, Wu W, Mankouri HW, Shen H, Liu Y and Hickson ID: Replication stress activates DNA repair synthesis in mitosis. Nature. 528:286–290. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Burrell RA, McClelland SE, Endesfelder D, Groth P, Weller MC, Shaikh N, Domingo E, Kanu N, Dewhurst SM, Gronroos E, et al: Replication stress links structural and numerical cancer chromosomal instability. Nature. 494:492–496. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Kasyanenko N: DNA as a target for anticancer drugs based on the coordination compounds of metals. FEBS J. 280:89. 2013.

34 

O'Grady S, Finn SP, Cuffe S, Richard DJ, O'Byrne KJ and Barr MP: The role of DNA repair pathways in cisplatin resistant lung cancer. Cancer Treat Rev. 40:1161–1170. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Zhu H, Luo H, Zhang W, Shen Z, Hu X and Zhu X: Molecular mechanisms of cisplatin resistance in cervical cancer. Drug Des Devel Ther. 10:1885–1895. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 38 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fu X, Mao X, Wang Y, Ding X and Li Y: Let-7c-5p inhibits cell proliferation and induces cell apoptosis by targeting ERCC6 in breast cancer. Oncol Rep 38: 1851-1856, 2017
APA
Fu, X., Mao, X., Wang, Y., Ding, X., & Li, Y. (2017). Let-7c-5p inhibits cell proliferation and induces cell apoptosis by targeting ERCC6 in breast cancer. Oncology Reports, 38, 1851-1856. https://doi.org/10.3892/or.2017.5839
MLA
Fu, X., Mao, X., Wang, Y., Ding, X., Li, Y."Let-7c-5p inhibits cell proliferation and induces cell apoptosis by targeting ERCC6 in breast cancer". Oncology Reports 38.3 (2017): 1851-1856.
Chicago
Fu, X., Mao, X., Wang, Y., Ding, X., Li, Y."Let-7c-5p inhibits cell proliferation and induces cell apoptosis by targeting ERCC6 in breast cancer". Oncology Reports 38, no. 3 (2017): 1851-1856. https://doi.org/10.3892/or.2017.5839