miR-24-3p regulates bladder cancer cell proliferation, migration, invasion and autophagy by targeting DEDD

  • Authors:
    • Guoqiang Yu
    • Zhaohui Jia
    • Zhongling Dou
  • View Affiliations

  • Published online on: December 16, 2016     https://doi.org/10.3892/or.2016.5326
  • Pages: 1123-1131
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

microRNAs (miRNAs), a class of small non-coding RNA molecules, can regulate gene expression by interacting with the 3'-untranslated regions (3'UTR) of target genes and influence various biological processes. We investigated the potential role of miR-24-3p in the development of bladder cancer by regulating DEDD, a member of the death effector domain-containing protein family. First, we found that miR-24-3p was highly expressed and that DEDD was expressed at a low level in bladder cancer tissues compared with that in adjacent bladder tissues by qRT-PCR (P<0.0001). Second, we found that miR-24-3p promoted the proliferation ability of bladder cancer cells using the MTT assay and colony forming assay; and showed that miR-24-3p accelerated the migration and invasion of bladder cancer cells using migration and invasion assays (P<0.05). Moreover, miR-24-3p inhibited apoptosis of bladder cancer cells, as shown by flow cytometry (P<0.05). Western blot results demonstrated that miR-24-3p participated in autophagy of bladder cancer cells by DEDD. In addition, the tumor formation assay showed that miR-24-3p promoted the growth of bladder tumor in vivo. Furthermore, the luciferase reporter gene assay indicated that miR-24-3p suppressed DEDD gene transcription. Therefore, our study indicated that miR-24-3p promoted bladder cancer progression by inhibiting DEDD.

Introduction

Human bladder cancer is prevalent throughout the world. According to statistics, human bladder cancer is the fourth leading cause of malignancy in men and currently the tenth most malignant tumor in women (1). According to its pathological features, bladder cancer can be divided into two major groups: urothelial carcinoma (UC) and invasive bladder cancer. UC is the most widespread bladder cancer, and it frequently recurs, but rarely progresses (2,3); invasive bladder cancer easily progresses to distant metastases (4). However, because the pathophysiological mechanisms underlying bladder cancer are not clear, the appropriate treatment to increase bladder cancer survival rates is limited. Consequently, molecular mechanism studies concerning the development mechanisms of bladder cancer are urgent.

microRNAs (miRNAs) are a subgroup of endogenous non-coding small RNAs that are 19–23 nucleotides in length and are involved in post-transcriptional regulation by targeting the 3′UTR of mRNA to affect biological processes (58). An increasing number of studies have shown that miRNAs act as a group of regulatory genes and participate in the development and progression of various diseases, such as the progression of tumorigenesis (6,911). Many studies have revealed that various miRNAs, such as miR-126, miR-143, miR-145, miR-23b, and miR-144-5p, are related to the development of bladder cancer (1216). The mechanism and function of miR-24-3p have also been studied in glioma (17), lung cancer (18), hepatocellular carcinoma (19), colorectal cancer (20), and breast cancer (21). However, the functional relevance of miR-24-3p in bladder cancer is not fully understood.

DEDD, a member of the death effector domain-containing protein family, participates in biological processes, such as cell apoptosis, cell cycle, and cell mitosis (22,23). Increasing evidence has shown that the epithelial-mesenchymal transition (EMT), a crucial development process, promotes cancer invasion and metastasis (24,25). Studies have found that DEDD serves as a novel tumor repressor and can suppress EMT and metastasis in breast and colon cancers (26,27). Therefore, DEDD may act as a prognostic marker and potential therapeutic target for the treatment of cancer metastasis. However, the mechanism and function of DEDD have not been reported in bladder cancer. To further study the anti-metastatic roles of DEDD in bladder cancer, our study investigate the expression level of DEDD in bladder cancer tissues and the interaction relationship between DEDD and miR-24-3p in bladder cancer.

Here, we confirmed that miR-24-3p was highly expressed and that DEDD was expressed at a low level in bladder cancer tissues. miR-24-3p promoted the abilities of proliferation, migration and invasion; inhibited apoptosis; participated in autophagy of bladder cancer cells by LC3, DEDD, and p62 in vitro; and promoted the growth of bladder tumor in vivo. Furthermore, miR-24-3p suppressed DEDD gene transcription and accelerated proliferation and invasion through DEDD in bladder cancer cells. Therefore, miR-24-3p could be a pivotal potential therapeutic target for the treatment of bladder cancer.

Materials and methods

Cell lines and transfection

Human ureter epithelium (HCV29) cells, the human bladder cancer cell line T24, human bladder transitional cell carcinoma (UM-UC-3) cells, human bladder cancer (HBC) cells, and malignant bladder carcinoma (BLU87) cells were purchased from College of Life Science, Hunan Normal University, China. All of the cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco-Invitrogen, Carlsbad, CA, USA) containing 10% fetal bovine serum (FBS; Invitrogen, Carlsbad, CA, USA), penicillin (100 U/ml; Invitrogen), and streptomycin (100 µg/ml; Invitrogen). All of the cultures were maintained at 37°C with 5% CO2. For treatment, according to the manufacturer's protocol, T24 and HBC cells (2×105 cells/well) were seeded in 6-well plates and were transfected with 200 µl of mature miR-24-3p mock, mimic, scramble, or inhibitor (GenePharma Co., Ltd., Shanghai, China) using Lipofectamine™ 3000 (Invitrogen, Carlsbad, CA, USA) for 72 h. Similarly, T24 and HBC cells were transfected with scramble and vector, miR-24-3p and pcMV6, and miR-24-3p and DEDD with Lipofectamine 3000, respectively.

Clinical specimens

Our study obtained ethics committee approval from the First Affiliated Hospital of Henan University of Science and Technology. We collected the bladder cancer tissues and adjacent non-cancerous tissues samples from the First Affiliated Hospital of Henan University of Science and Technology between 2014 and 2016. All of the tissue samples were snap-frozen at −80°C. Each patient provided written informed consent.

Quantitative real-time reverse transcription PCR (qRT-PCR)

According to the manufacturer's instructions, total RNA was extracted from bladder cancer tissues, matched adjacent noncancerous tissues and treated T24 and HBC cells using the RNeasy Plus Mini kit. cDNAs were synthesized using the RevertAid First Strand cDNA Synthesis kit (Thermo Fisher). The samples were normalized to U6 according to their miRNA and GAPDH for mRNA. As previously described (28), the mRNA expression levels were analyzed using the SYBR-Green PCR Master Mix kit (Takara). For the detection of mRNA, the primer sequences for GAPDH were: 5′-TGT TCG TCA TGG GTG TGA AC-3′ (the forward primer) and 5′-ATG GCA TGG ACT GTG GTC AT-3′ (the reverse primer); the primer sequences for DEDD were: 5′-TCC CCA GCC CTC TAA AAC AG-3′ (the forward primer) and 5′-CCG CAG TCT GAT GTC ACA TG-3′ (the reverse primer); the primer sequences for PAK1 were: 5′- TTG GGA ATG GAT GGC TCT GT-3′ (the forward primer) and 5′-ATG TCA ACC TTG GGC CCA TA-3′ (the reverse primer). For the detection of miRNA, the primer sequence for miR-24-3p was 5′-CCC ATT CAG CAG GAA CAG AAA-3′ and that for U6 was 5′-ACG CAA ATT CGT GAA GCG TT-3′.

Western blot analysis

Treated T24 and HBC cells were lysed using RIPA buffer containing a protease inhibitor cocktail (P8340; Sigma). Equivalent protein samples were separated on 8% SDS/PAGE gels and were then transferred to PVDF membranes (Millipore, Billerica, MA, USA). The PVDF membranes were incubated in 5% skim milk (BD Biosciences) for 2 h with a primary antibody at 4°C overnight, followed by incubation with horseradish peroxidase-conjugated secondary antibodies for 1 h at room temperature. The results were obtained using an enhanced chemiluminescence detection system (Amersham Biosciences, Piscataway, NJ, USA). In this study, the used primary antibodies were anti-LC3 (1:200; MBL International Co., Woburn, MA, USA), anti-DEDD (1:200; ab56480, Abcam), and anti-P62 (1:5000, Progen, GP62-C); the anti-GAPDH antibody (1:4000, Beverly, MA, USA) was used as an internal control.

Luciferase reporter assays

Treated T24 and HBC cells (5×104 cells/well) were cultured in 24-well plates and were co-transfected with miR-24-3p and the wild-type pGL3-promoter-DEDD reporter vector or mutant pGL3-promoter-DEDD reporter vector as well as a renilla plasmid (RL-SV40) using Lipofectamine 3000 (Invitrogen). The renilla plasmid was used as an internal control. According to the manufacturer's instructions, the luciferase activities were measured by the Dual-Luciferase reporter assay system (Promega, Madison, WI, USA).

Colony forming unit (CFU) assay

For the colony forming unit assay, T24 and HBC cells (5×103 cells/well) transfected with miR-24-3p mock, mimics, scramble, or inhibitors were cultured into a 10-cm culture dish with complete DMEM medium for 12 days. Next, the colonies were fixed using methanol and dyed using Giemsa dye solution. The colony forming units were observed and photographed.

MTT assay

T24 and HBC cells (3×103 cells/well) transfected with miR-24-3p mock, mimics, scramble, or inhibitors were seeded in 96-well plates for 0, 12, 24, 48, and 72 h. Next, 20 µl of a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT, 5 mg/ml) solution was added into each well. After 4 h, formazan was dissolved in a dimethyl sulfoxide (DMSO) solution. A microplate reader was used to measure the absorbance at 570 nm.

Flow cytometric analysis of the cell apoptosis

The treated T24 and HBC cells were resuspended in PBS and were stained with FITC-Annexin V and propidium iodide (PI). The flow cytometry results were analyzed using FlowJo software. The cells were segmented into four types, viable cells, dead cells, early-stage apoptotic cells, and late-stage apoptotic cells.

Migration and invasion assays

According to the manufacturer's instructions, the migration abilities of the treated T24 and HBC cells were carried out in 24-well Transwell chambers with 8-µm pore cell culture inserts (Costar). The treated cells (5×105 cells/well) in serum-free media were seeded into the upper chamber, and DMEM medium with 10% FBS was seeded into the lower chambers for 24 h in an incubator at 37°C with 5% CO2. Thereafter, the migratory cells were fixed, stained and counted under a microscope. For the invasion assay, 10 µl of 1:5 diluted matrigel (BD Biosciences) was pre-paved into Transwell inserts before the experiment at 37°C for 2 h.

Tumor formation in nude mice

The Institutional Committee for Animal Research approved the animal experiments of this study, and we performed tumor formation in nude mice according to the Institutional Animal Care and Use Committee. The flanks of 5-week-old BALB/c athymic nude mice were subcutaneously implanted with T24 cells (1×107 cells in 100 µl) transfected with miR-24-3p mimics, mock, inhibitor or scramble for 0, 8, 16, 24, 32, 40, and 48 days. The tumor volume and tumor weight were measured.

Statistical analysis

The data were analyzed by Student's t-test and analysis of variance (ANOVA) using Prism6 (GraphPad Software, Inc., San Diego, CA, USA) and SPSS 15.0 software (SPSS, Chicago, IL, USA). All of the results are presented as the means ± SD. A P-value <0.05 was considered to indicate a statistically significant difference.

Results

miR-24-3p is overexpressed, and DEDD expression is low in bladder cancer

We collected bladder cancer tissues and paired non-carcinoma tissues from 26 patients. The mRNA expression levels of miR-24-3p and DEDD were measured by qRT-PCR. The results indicated that the expression level of miR-24-3p was increased in bladder cancer tissues (n=26) compared with that in para-carcinoma tissues (n=26) (P<0.0001) (Fig. 1A); the mRNA expression level of DEDD was decreased in bladder cancer tissues (n=26) compared with that in para-carcinoma tissues (n=26) (P<0.0001) (Fig. 1B). We also found that the mRNA expression level of miR-24-3p was increased in human ureter epithelium (HCV29) cells compared with that in bladder cancer cells (T24, UM-UC-3, HBC, and BLU87) (P<0.0001). The highest level of miR-24-3p was found in T24 cells (P<0.0001) (Fig. 1C); the mRNA expression level of DEDD was decreased in human ureter epithelium (HCV29) cells compared with that in bladder cancer cells (T24, UM-UC-3, HBC, and BLU87) (P<0.0001). The lowest level of DEDD was found in T24 cells (P<0.0001) (Fig. 1D).

miR-24-3p promotes cell proliferation and invasion in bladder cancer cells

The impact of miR-24-3p on the proliferation and invasion abilities of bladder cancer cells was detected in T24 and HBC cells transfected with miR-24-3p mimics, mock (control), inhibitor or scramble (control). First, the mRNA expression level of miR-24-3p was measured by qRT-PCR. Our results indicated that the transfection effects of miR-24-3p mimics and inhibitor were high (P<0.05) (Fig. 2A). The bladder cell proliferation ability was detected by the MTT assay, and the results indicated that miR-24-3p promoted the proliferation of T24 cells (P<0.05) (Fig. 2B) and HBC cells (P<0.05) (Fig. 2C). Similarly, the colony forming unit assay was performed to determine the bladder cell proliferation. Our results found that the proliferation ability was significantly increased in T24 and HBC cells transfected with miR-24-3p mimics relative to mock and that the proliferation ability was significantly decreased in T24 and HBC cells transfected with miR-24-3p inhibitor relative to scramble (P<0.05) (Fig. 2D and E). Furthermore, we found that miR-24-3p promoted the invasion ability of T24 and HBC cells (Fig. 2F and G).

miR-24-3p promotes cell migration, inhibits apoptosis, and participates in autophagy

We further studied the cell migration and apoptosis that were affected by miR-24-3p in bladder cancer cells. First, the migration ability of T24 and HBC cells transfected with miR-24-3p mimics, mock, inhibitor or scramble were measured by the migration assay. The results indicated that the migration of T24 and HBC cells transfected with miR-24-3p mimics were significantly increased compared with mock (P<0.05); the cell migration capacity of T24 and HBC cells transfected with the miR-24-3p inhibitor was significantly decreased compared with scramble (P<0.05) (Fig. 3A and B). Second, cell apoptosis was measured by Annexin V-FITC/PI staining. The results showed that apoptosis T24 and HBC cells transfected with miR-24-3p mimics was significantly decreased compared with that of the mock (P<0.05); apoptosis of T24 and HBC transfected with miR-24-3p inhibitor was significantly increased compared with that of scramble (P<0.05) (Fig. 3C and D). Furthermore, the western blot results showed that miR-24-3p upregulated the protein expression levels of LC3 and p62 and downregulated the protein expression level of DEDD in T24 and HBC cells (Fig. 3E). These results indicated that miR-24-3p participated in autophagy by repressing DEDD and then increasing LC3 and p62 in bladder cancer cells.

miR-24-3p suppresses DEDD gene transcription in bladder cancer

To further explore the mechanism of miR-24-3p in bladder cancer, we studied the relationship between miR-24-3p and DEDD. According to TargetScan, the miR-24-3p target sites in the sequence of DEDD were found (Fig. 4A). As shown in Fig. 4B, the target sites between miR-24-3p and the DEDD 3′UTR among human and other species were also analyzed. Therefore, we hypothesized that miR-24-3p may directly bind to DEDD. The luciferase reporter gene assay was used to measure the luciferase activity of the DEDD 3′UTR. The results showed a significant decrease in fluorescence activity after cotransfection of miR-24-3p and the wild-type DEDD 3′UTR vector (P<0.01), but a not significant change in the mutant DEDD 3′UTR vector (Fig. 4C). The qRT-PCR results revealed that miR-24-3p inhibited the mRNA expression level of DEDD T24 and HBC cells (P<0.05) (Fig. 4D).

miR-24-3p promotes the growth of bladder tumor in vivo

To assess the effect of miR-24-3p on tumorigenesis in vivo, T24 cells transfected with miR-24-3p mimics, mock, inhibitor or scramble were implanted subcutaneously into nude mice. The tumor volume was calculated, and the tumor weight was measured at 0, 8, 16, 24, 32, 40, and 48 days. The results showed that mice injected with cells transfected with miR-24-3p mimics were larger than control mice (P<0.05) (Fig. 5A). Mice injected with cells transfected with miR-24-3p mimics were heavier than control mice (P<0.05) (Fig. 5B). The tumors are shown in Fig. 5C, and RNA was extracted from the tumors. The qRT-PCR results indicated that the mRNA expression level of miR-24-3p was decreased significantly in mice injected with cells transfected with miR-24-3p mimics compared with mock (P<0.05); the mRNA expression level of miR-24-3p was increased significantly in mice injected with cells transfected with the miR-24-3p inhibitor compared with scramble (P<0.05) (Fig. 5D).

miR-24-3p accelerates the proliferation and invasion through DEDD in bladder cancer cells

We further investigated whether miR-24-3p expression influenced the bladder cancer cell proliferation and invasion through DEDD. T24 and HBC cells were transfected with scramble and vector, miR-24-3p and pcMV6, and miR-24-3p and DEDD, respectively. The qRT-PCR results indicated that the mRNA expression level of DEDD was decreased significantly in T24 and HBC cells transfected with miR-24-3p and pcMV6 compared with scramble and vector (P<0.05); the mRNA expression level of DEDD was increased significantly in T24 and HBC cells transfected with miR-24-3p and DEDD compared with those transfected with miR-24-3p and pcMV6 (P<0.05) (Fig. 6A). The western blot results also indicated that miR-24-3p inhibited the protein expression level of DEDD (Fig. 6B).

The qRT-PCR results showed that the mRNA expression level of miR-24-3p was increased significantly in T24 and HBC cells transfected with miR-24-3p and pcMV6 compared with that in scramble and vector (P<0.05); the mRNA expression level of miR-24-3p was decreased significantly in T24 and HBC cells transfected with miR-24-3p and DEDD compared with miR-24-3p and pcMV6 (P<0.05) (Fig. 6C). The bladder cell proliferation ability was further detected by the colony forming unit assay. We found that miR-24-3p inhibited the proliferation ability of T24 and HBC cells and that DEDD promoted the proliferation ability of T24 and HBC cells mediated by miR-24-3p (P<0.05) (Fig. 6D). We also found that miR-24-3p promoted the invasion ability of T24 and HBC cells and that DEDD inhibited the invasion ability of T24 and HBC cells mediated by miR-24-3p (P<0.05) (Fig. 6E). Therefore, as shown in Fig. 6F, miR-24-3p accelerated cell proliferation, migration, invasion and autophagy and inhibited apoptosis by inhibiting DEDD and activating p62 in bladder cancer.

Discussion

miRNAs serve as a class of small non-coding RNAs that regulate mRNAs according to the 3′UTR. Several studies have demonstrated that miRNAs are involved in multiple biological functions, including cell proliferation, migration, metastasis, and inflammation, as well as tumor angiogenesis, by targeting mRNAs in cancer (29,30). Various studies have indicated that miRNAs participate in the occurrence and development of diversified diseases (6,9,11). A handful of studies has found that various miRNAs are involved in the development of bladder cancer (31). For example, miR-126 and miR-182 are related to urinary bladder cancer (12); miR-125b inhibits the development of bladder cancer through E2F3 (32); miR-143 serves as a tumor inhibitor of bladder cancer (13); miR-200 regulates EMT in bladder cancer cells and reverses the resistance to epidermal growth factor (EGF) receptor (33).

miR-24-3p has been studied in many cancers. For example, miR-24-3p promotes cell proliferation in glioma cells via MXI1 (17); the diagnosis and prognosis of miR-24-3p have been investigated in HBV-related hepatocellular carcinoma (19); miR-24-3p inhibits VP16-DDP resistance in small cell lung cancer via ATG4A(18). However, the functional relevance of miR-24-3p in bladder cancer is still not known. In our study, we indicated that the expression level of miR-24-3p was increased in bladder cancer tissues compared with para-carcinoma tissues. We also found that miR-24-3p promotes cell proliferation, migration and invasion and inhibited apoptosis in bladder cancer cells. In addition, we found that miR-24-3p promotes the growth of bladder tumors in vivo.

DEDD, an important effector molecule for cell death signaling receptors, plays critical roles in cell apoptosis, the cell cycle, and cell mitosis (22,23). A recent study also reported that DEDD can weaken the EMT and inhibits tumor growth and metastasis (26). Studies have found that DEDD inhibits transforming growth factor-β1 signaling (34). DEDD has also been correlated with apoptosis (35,36) and reverses EMT by activating selective autophagy (26,27). In our study, we found that miR-24-3p suppressed DEDD gene transcription. miR-24-3p accelerated proliferation and invasion through DEDD in bladder cancer cells.

Autophagy, also called type 2 non-apoptotic cell death, is a prominent mechanism of self-destruction, distinguished by a catabolic process beginning with the formation of autophagosomes and degradation of cytoplasmic material in the autophagy-lysosome by lysosomal enzymes to affect health and disease (3740). Various studies have suggested that LC3 can serve as an autophagosome marker, and p62 has been shown to be related to autophagy (4143). Our study showed that miR-24-3p participated in autophagy through DEDD, LC3 and p62 in bladder cancer cells.

In summary, we found that miR-24-3p was highly expressed and that DEDD was expressed at a low level in bladder cancer tissues. miR-24-3p promoted proliferation, migration and invasion and inhibited apoptosis in bladder cancer cells. miR-24-3p also participated in the autophagy of bladder cancer cells through LC3, DEDD, and p62. We found that miR-24-3p promoted the growth of bladder tumors in vivo. Furthermore, miR-24-3p suppressed DEDD gene transcription. miR-24-3p accelerated proliferation and invasion through DEDD in bladder cancer cells. Therefore, our study indicated that miR-24-3p promoted bladder cancer progression by inhibiting DEDD. Thus, miR-24-3p could be a pivotal potential therapeutic target for the treatment of bladder cancer.

Acknowledgements

The study was supported by the Key Project of Education Department of Henan Science and Technology (13A320243).

References

1 

Ying L, Chen Q, Wang Y, Zhou Z, Huang Y and Qiu F: Upregulated MALAT-1 contributes to bladder cancer cell migration by inducing epithelial-to-mesenchymal transition. Mol Biosyst. 8:2289–2294. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Bo J, Yang G, Huo K, Jiang H, Zhang L, Liu D and Huang Y: microRNA-203 suppresses bladder cancer development by repressing bcl-w expression. FEBS J. 278:786–792. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Egerod FL, Bartels A, Fristrup N, Borre M, Ørntoft TF, Oleksiewicz MB, Brünner N and Dyrskjøt L: High frequency of tumor cells with nuclear Egr-1 protein expression in human bladder cancer is associated with disease progression. BMC Cancer. 9:3852009. View Article : Google Scholar : PubMed/NCBI

4 

Said N, Sanchez-Carbayo M, Smith SC and Theodorescu D: RhoGDI2 suppresses lung metastasis in mice by reducing tumor versican expression and macrophage infiltration. J Clin Invest. 122:1503–1518. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Iorio MV and Croce CM: MicroRNAs in cancer: Small molecules with a huge impact. J Clin Oncol. 27:5848–5856. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Farazi TA, Hoell JI, Morozov P and Tuschl T: MicroRNAs in human cancer. Adv Exp Med Biol. 774:1–20. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Djuranovic S, Nahvi A and Green R: A parsimonious model for gene regulation by miRNAs. Science. 331:550–553. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Kasinski AL and Slack FJ: Epigenetics and genetics. MicroRNAs en route to the clinic: Progress in validating and targeting microRNAs for cancer therapy. Nat Rev Cancer. 11:849–864. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Baer C, Claus R and Plass C: Genome-wide epigenetic regulation of miRNAs in cancer. Cancer Res. 73:473–477. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Zaman MS, Maher DM, Khan S, Jaggi M and Chauhan SC: Current status and implications of microRNAs in ovarian cancer diagnosis and therapy. J Ovarian Res. 5:442012. View Article : Google Scholar : PubMed/NCBI

11 

Di Leva G and Croce CM: The role of microRNAs in the tumorigenesis of ovarian cancer. Front Oncol. 3:1532013. View Article : Google Scholar : PubMed/NCBI

12 

Hanke M, Hoefig K, Merz H, Feller AC, Kausch I, Jocham D, Warnecke JM and Sczakiel G: A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol Oncol. 28:655–661. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Lin T, Dong W, Huang J, Pan Q, Fan X, Zhang C and Huang L: MicroRNA-143 as a tumor suppressor for bladder cancer. J Urol. 181:1372–1380. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Noguchi S, Yasui Y, Iwasaki J, Kumazaki M, Yamada N, Naito S and Akao Y: Replacement treatment with microRNA-143 and −145 induces synergistic inhibition of the growth of human bladder cancer cells by regulating PI3K/Akt and MAPK signaling pathways. Cancer Lett. 328:353–361. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Majid S, Dar AA, Saini S, Deng G, Chang I, Greene K, Tanaka Y, Dahiya R and Yamamura S: MicroRNA-23b functions as a tumor suppressor by regulating Zeb1 in bladder cancer. PLoS One. 8:e676862013. View Article : Google Scholar : PubMed/NCBI

16 

Matsushita R, Seki N, Chiyomaru T, Inoguchi S, Ishihara T, Goto Y, Nishikawa R, Mataki H, Tatarano S, Itesako T, et al: Tumour-suppressive microRNA-144-5p directly targets CCNE1/2 as potential prognostic markers in bladder cancer. Br J Cancer. 113:282–289. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Xu W, Liu M, Peng X, Zhou P, Zhou J, Xu K, Xu H and Jiang S: miR-24-3p and miR-27a-3p promote cell proliferation in glioma cells via cooperative regulation of MXI1. Int J Oncol. 42:757–766. 2013.PubMed/NCBI

18 

Pan B, Chen Y, Song H, Xu Y, Wang R and Chen L: Mir-24-3p downregulation contributes to VP16-DDP resistance in small-cell lung cancer by targeting ATG4A. Oncotarget. 6:317–331. 2015.PubMed/NCBI

19 

Meng F-L, Wang W and Jia W-D: Diagnostic and prognostic significance of serum miR-24-3p in HBV-related hepatocellular carcinoma. Med Oncol. 31:1772014. View Article : Google Scholar : PubMed/NCBI

20 

Gao Y, Liu Y, Du L, Li J, Qu A, Zhang X, Wang L and Wang C: Down-regulation of miR-24-3p in colorectal cancer is associated with malignant behavior. Med Oncol. 32:3622015. View Article : Google Scholar : PubMed/NCBI

21 

Lu K, Wang J, Song Y, Zhao S, Liu H, Tang D, Pan B, Zhao H and Zhang Q: miRNA-24-3p promotes cell proliferation and inhibits apoptosis in human breast cancer by targeting p27Kip1. Oncol Rep. 34:995–1002. 2015.PubMed/NCBI

22 

Hua F, Xue J, Lü X and Hu Z: DEDD decreases Smad3 activity, promotes tumor cell apoptosis and inhibits proliferation. Yao Xue Xue Bao. 48:680–685. 2013.(In Chinese). PubMed/NCBI

23 

He L, Zhong G and Zhu B: microRNA-15b expressed in lung cancer-infiltrated CD8-positive memory T cell inhibit cell apoptosis by repressing DEDD. Eur J Cancer (Suppl). S147. 2013.

24 

Carstens JL, Lovisa S and Kalluri R: Microenvironment-dependent cues trigger miRNA-regulated feedback loop to facilitate the EMT/MET switch. J Clin Invest. 124:1458–1460. 2014. View Article : Google Scholar : PubMed/NCBI

25 

De Craene B and Berx G: Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 13:97–110. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Lv Q, Hua F and Hu Z-W: DEDD, a novel tumor repressor, reverses epithelial-mesenchymal transition by activating selective autophagy. Autophagy. 8:1675–1676. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Lv Q, Wang W, Xue J, Hua F, Mu R, Lin H, Yan J, Lv X, Chen X and Hu ZW: DEDD interacts with PI3KC3 to activate autophagy and attenuate epithelial-mesenchymal transition in human breast cancer. Cancer Res. 72:3238–3250. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Jiang L, Lai YK, Zhang J, Wang H, Lin MC, He ML and Kung HF: Targeting S100P inhibits colon cancer growth and metastasis by Lentivirus-mediated RNA interference and proteomic analysis. Mol Med. 17:709–716. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Wang J, Paris PL, Chen J, Ngo V, Yao H, Frazier ML, Killary AM, Liu CG, Liang H, Mathy C, et al: Next generation sequencing of pancreatic cyst fluid microRNAs from low grade-benign and high grade-invasive lesions. Cancer Lett. 356:404–409. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Stahlhut C and Slack FJ: MicroRNAs and the cancer phenotype: Profiling, signatures and clinical implications. Genome Med. 5:1112013. View Article : Google Scholar : PubMed/NCBI

31 

Ichimi T, Enokida H, Okuno Y, Kunimoto R, Chiyomaru T, Kawamoto K, Kawahara K, Toki K, Kawakami K, Nishiyama K, et al: Identification of novel microRNA targets based on microRNA signatures in bladder cancer. Int J Cancer. 125:345–352. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Huang L, Luo J, Cai Q, Pan Q, Zeng H, Guo Z, Dong W, Huang J and Lin T: MicroRNA-125b suppresses the development of bladder cancer by targeting E2F3. Int J Cancer. 128:1758–1769. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Adam L, Zhong M, Choi W, Qi W, Nicoloso M, Arora A, Calin G, Wang H, Siefker-Radtke A, McConkey D, et al: miR-200 expression regulates epithelial-to-mesenchymal transition in bladder cancer cells and reverses resistance to epidermal growth factor receptor therapy. Clin Cancer Res. 15:5060–5072. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Xue J-F, Hua F, Lv Q, Lin H, Wang ZY, Yan J, Liu JW, Lv XX, Yang HZ and Hu ZW: DEDD negatively regulates transforming growth factor-β1 signaling by interacting with Smad3. FEBS Lett. 584:3028–3034. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Lee JC, Schickling O, Stegh AH, Oshima RG, Dinsdale D, Cohen GM and Peter ME: DEDD regulates degradation of intermediate filaments during apoptosis. J Cell Biol. 158:1051–1066. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Schutte B, Henfling M and Ramaekers FC: DEDD association with cytokeratin filaments correlates with sensitivity to apoptosis. Apoptosis. 11:1561–1572. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Klionsky DJ: Autophagy: From phenomenology to molecular understanding in less than a decade. Nat Rev Mol Cell Biol. 8:931–937. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Huett A, Goel G and Xavier RJ: A systems biology viewpoint on autophagy in health and disease. Curr Opin Gastroenterol. 26:302–309. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Li ZY, Yang Y, Ming M and Liu B: Mitochondrial ROS generation for regulation of autophagic pathways in cancer. Biochem Biophys Res Commun. 414:5–8. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Liu JJ, Lin M, Yu JY, Liu B and Bao JK: Targeting apoptotic and autophagic pathways for cancer therapeutics. Cancer Lett. 300:105–114. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Kuma A, Matsui M and Mizushima N: LC3, an autophagosome marker, can be incorporated into protein aggregates independent of autophagy: Caution in the interpretation of LC3 localization. Autophagy. 3:323–328. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Øvervatn A, Bjørkøy G and Johansen T: p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 282:24131–24145. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Ichimura Y, Kumanomidou T, Sou YS, Mizushima T, Ezaki J, Ueno T, Kominami E, Yamane T, Tanaka K and Komatsu M: Structural basis for sorting mechanism of p62 in selective autophagy. J Biol Chem. 283:22847–22857. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2017
Volume 37 Issue 2

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
Yu G, Jia Z and Dou Z: miR-24-3p regulates bladder cancer cell proliferation, migration, invasion and autophagy by targeting DEDD. Oncol Rep 37: 1123-1131, 2017
APA
Yu, G., Jia, Z., & Dou, Z. (2017). miR-24-3p regulates bladder cancer cell proliferation, migration, invasion and autophagy by targeting DEDD. Oncology Reports, 37, 1123-1131. https://doi.org/10.3892/or.2016.5326
MLA
Yu, G., Jia, Z., Dou, Z."miR-24-3p regulates bladder cancer cell proliferation, migration, invasion and autophagy by targeting DEDD". Oncology Reports 37.2 (2017): 1123-1131.
Chicago
Yu, G., Jia, Z., Dou, Z."miR-24-3p regulates bladder cancer cell proliferation, migration, invasion and autophagy by targeting DEDD". Oncology Reports 37, no. 2 (2017): 1123-1131. https://doi.org/10.3892/or.2016.5326