Open Access

Circulating microRNA-194 regulates human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway

  • Authors:
    • Ming Bai
    • Mingzi Zhang
    • Fei Long
    • Nanze Yu
    • Ang Zeng
    • Ru Zhao
  • View Affiliations

  • Published online on: March 30, 2017     https://doi.org/10.3892/or.2017.5537
  • Pages: 2702-2710
  • Copyright: © Bai 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

In the present study, we analyzed the role of microRNA-194 circulating regulated human melanoma cell growth. We found that microRNA-194 expression was markedly suppressed in human melanoma patients, compared with negative control group. Next, disease-free survival (DFS) and overall survival (OS) of high expression in human melanoma patients was higher than those of low expression in human melanoma patients. MicroRNA-194 overexpression inhibited cell proliferation, induced apoptosis, increased caspase-3/-9 activities and promoted Bax/Bcl-2 of human melanoma cells. Furthermore, microRNA-194 overexpression also suppressed PI3K/AKT/FoxO3a signaling pathway and induced p53/p21 signaling pathway. PI3K inhibitor, suppressed PI3K, phosphorylation-AKT, FoxO3a protein expression and increased the effects of microRNA-194 overexpression on cell growth, apoptosis, caspase-3/-9 activities and Bax/Bcl-2 protein expression of human melanoma cells through the induction of p53/p21 signaling pathway. Taken together, these data indicate that circulating microRNA-194 regulated human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway.

Introduction

Malignant melanoma (MM) is a highly malignant tumor that mostly strikes adults aged more than 30 years, and the elderly, and it has high mortality. The morbidity of MM has increased gradually in recent years and shows a younger trend (1,2). It accounts for ~2% of malignancies in terms of morbidity and ranks the third of skin malignancies (6.8–20%); the disease can occur in any part of the skin, which is dominant by the sites that are vulnerable to friction in the four extremities, and its characteristics are early metastasis, strong invasiveness and poor prognosis (3). The treatment at present is mainly surgical treatments, and the subsequent biotherapy, radiotherapy and chemotherapy is not clinically effective, thus, it has poor prognosis and high mortality (4).

Recently, with the continuous development of molecular biology, great progresses have been achieved in research on genetics and molecular biology, such as the genesis, development and metastasis of MM, which has greatly promoted the clinical treatment and basic research on MM (5). However, there is little research on the effect of miRNA on MM as well as its mechanism of action (5).

microRNA is a type of non-coding small RNA consisting of 18–25 nucleotides that was newly discovered to be closely associated with MM (6). microRNA plays an important regulatory role in normal human physiological function, which can regulate the target gene and influence the expression of its protein; while under some conditions, the increased or decreased expression of the key protein will lead to cellular functional changes, or even the genesis of tumor (7). The expression profiles of microRNA are greatly different between tumor cells and normal tissue cells (7). A majority of microRNAs are associated with high expression, low expression or expression deletion in diverse tumor cells, the abnormal expression of which is one of the important molecular mechanisms of participating tumor cell proliferation and growth (8).

Akt is the key downstream effector molecule of PI3K, which can promote tumor growth and invasion through promoting angiogenesis and migration-associated procedures after being phosphorylated and activated. PTEN can dephosphorylate phosphatidylinositol 3,4,5-trisphosphate (PIP3), and thus, weaken the PI3K-derived activation signal and indirectly inhibit the activation of Akt (9). The low expression of PTEN results in its disability to effectively inhibit the abnormal activation of the PDK/Akt pathway, and thus leads to tumor migration, which is associated with the highest occurrence rate in prostate, endometrial cancer, glioma and MM (10). As is reported, PTEN gene changes can be seen in 30–50% of MM cell lines and 5–20% of MM cells (9,11). The activated PI3K/Akt may further activate its downstream molecule mTOR through the TSC1/2 complex, and the activated mTOR can activate its two downstream molecules subsequently, which are the translation inhibitory molecule eIF-4E binding protein 1 (4E-BP1) and ribosomal protein p70S6K (10,12). The activation of p70S6K can promote the filament reconstruction of the actin and promote cell movement (12,13).

p21, is a CDK kinase inhibitor belonging to the Cip/Kip CDK kinase inhibitor family, and has multiple activities in cancer (14). p21 has two non-overlapped domains, which are the PCNA binding domain locating in the carboxyl-terminal and the CDK-cyclin inhibiting domain in the amino-terminal. p21 binds with the cyclin through the Cy1 motif in the N-terminal, and it can also bind with CDK subunit through the CDK binding locus locating in the N-terminal which separates from the Cy1 motif. Under certain specific conditions, p21 can promote the kinase activities of CDK4 and CDK6, thus, promoting the cells to pass stage G1; p21 can inhibit the activity of CDK2 and thus, inhibit the phosphorylation of the CDK2-dependent RB protein, and the unphosphorylated RB can bind with the transcription factor E2F1 and thus, inhibit its transcriptional activity; therefore, p21 inhibits CDK2 and eventually inhibits the transcription of the gene necessary for the E2F1-dependent G1/S transformation, leading to stage G1/S arrest (2,15). Targeted knockout CDK2 demonstrates that CDK2 is not necessary for the inhibitory effect of p21 on cell cycle; and p21 can also inhibit the activity of CDK1 kinase, thus, rendering the arrest of cell cycle in the G2/M stage (2,16). The present study addressed the role of microRNA-194 circulation-regulated human melanoma cell growth.

Materials and methods

Patients and tissue samples

MM samples and matched adjacent normal tissues were obtained from patients in the department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College. The samples were stored at liquid nitrogen after the collection. The study was approved by the Ethics Committee of the department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College (Table I). Disease-free survival (DFS) and overall survival (OS) were followed up every 2 months.

Table I.

Clinicopathological factors of the malignant melanoma patients.

Table I.

Clinicopathological factors of the malignant melanoma patients.

Stage I/IIStage IIIStage IV
Gender
  Male237
  Female246
Age (years)62.12±5.1260.28±4.8961.45±5.04
Primary site
  Extremities0
  Head/neck1
  Trunk4
Distant sites
  1 0
  2 3
  3+ 10
Breslow thickness (mm)
  ≤1.00 0
  1.01–2.00 2
  2.01–4.00 4
  >4.00 7
Lymph nodes (+)
  1 2
  2–3 7
  4+ 4
RNA isolation and quantitative real-time PCR

RNA was isolated from tissue samples using TRIzol (Invitrogen, Carlsbad, CA, USA), following the manufacturer's instructions. cDNA was reverse transcripted with the Fermentas reverse transcription reagents and the Applied Biosystems® TaqMan® MicroRNA Reverse Transcription kit (Applied Biosystems, Inc., Carlsbad, CA, USA). The cycling conditions were as follows: 94°C for 30 sec, 60°C for 30 sec and 72°C for 30 sec for 40 cycles. The 2-∆∆Ct method was applied to analyze the data.

Cell culture and transfection

Melanoma cell line SK-Mel2 cells were maintained in RPMI-1640 medium (Invitrogen), supplemented with 10% heat-inactive fetal bovine serum (FBS; Invitrogen), 1% penicillin-streptomycin and 4 mmol/l L-glutamine, in a CO2 incubator at 37°C. MicroRNA-194 mimics and negative mimics were purchased from Beijing Zoman technology, co., Ltd. (Beijing, China). MicroRNA-194 mimics and negative mimics were transfected into melanoma cell lines by using Lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions.

Cell survival MTT assay

Following transfection, cells were detected using MTT (5 mg/ml, 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; Sigma-Aldrich, St. Louis, MO, USA) for 4 h at 37°C. A total of 150 µl of dimethyl sulfoxide (DMSO) assay was added into each well and optic density (OD) value was detected at 570 nm with a microplate reader (PerSeptive Biosystems, Inc., Framingham, MA, USA).

Annexin V detection of apoptosis

Following transfection, cells were washed and resuspended in 1X binding buffer (BD Biosciences, Shanghai, China). Cells were incubated with Annexin V-FITC and propidium iodide (PI) (5 µg/ml each) at darkness for 15 min. Early apoptotic cells were detected using Becton-Dickinson FACSCalibur cytometer (Becton-Dickinson, Franklin Lakes, NJ, USA).

Caspase-3/−9 activity assay

Following transfection, cells were washed and incubated on ice in RIPA buffer. Protein concentration was detected using BCA kit (Pierce, Rockford, IL, USA). Protein lysates (100–200 µg) were incubated with 100 µM of the caspase-3 substrate DEVD-pNA (Beyotime Institute of Biotechnology, Haimen, China) or 100 µM of the caspase-9 substrate Ac-LEHD-pNA (Beyotime Institute of Biotechnology) at 37°C for 2 h. Optic density (OD) value was detected at 405 nm with a microplate reader (PerSeptive Biosystems).

Western blots

Cells were washed and incubated on ice in RIPA buffer. Protein concentration was detected using BCA kit (Pierce). Protein (30–50 µg) was separated by 6–10% sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) and transferred onto nitrocellulose (NC) membranes (Bio-Rad Laboratories) followed by incubation with primary antibodies: Bax (1:500; Santa Cruz Biotechnology, Santa Cruz, CA, USA), Bcl-2 (1:500; Santa Cruz Biotechnology), PI3K (1:500; Santa Cruz Biotechnology), phosphorylation-AKT (p-AKT, 1:500; Santa Cruz Biotechnology), FoxO3a (1:500; Santa Cruz Biotechnology), p53 (1:500; Santa Cruz Biotechnology), p21 (1:500; Santa Cruz Biotechnology) and GAPDH (1:500; Santa Cruz Biotechnology) overnight at 4°C. Protein bands were visualized using anti-rabbit horseradish peroxidase (HRP)-conjugated secondary antibodies (1:5,000; Santa Cruz Biotechnology) and by the enhanced chemiluminescence (ECL) reagents (Bio-Rad Laboratories, Hercules, ca, usa). The bands were visualized with the Fusion FX7 system (Vilber Lourmat, Marne la Vallée, France).

Statistical analysis

All data were presented as mean ± standard deviation (SD). The statistical significance between the groups was analyzed using Student's t-test or one-way ANOVA. Values of p<0.05 were considered to indicate a statistically different result.

Results

MicroRNA-194 expression of human melanoma patients

In the present study, we measured the microRNA-194 expression in MM tissues or para-carcinoma tissue using quantitative real-time PCR. As shown in Fig. 1, microRNA-194 expression of MM tissues samples was suppressed compared to that of the para-carcinoma tissue.

Disease-free survival (DFS) and overall survival (OS) of human melanoma patients

At 10 years of follow-up, we analyzed the DFS and OS of human MM patients and microRNA-194 expression. Basic information of these MM patients is shown in Table I. Fig. 2 shows that DFS and OS of high expression in human MM patients were higher than those of low expression in human MM patients.

MicroRNA-194 overexpression inhibits cell proliferation and induces apoptosis of human melanoma cells

We used microRNA-194 mimics to promote the expression of microRNA-194 in SK-Mel2 cells and analyzed cell proliferation and apoptosis of human melanoma cells. As shown in Fig. 3, microRNA-194 overexpression inhibited cell proliferation and induced apoptosis of human melanoma cells, compared with the negative control group.

MicroRNA-194 overexpression increases caspase-3/−9 activities of human melanoma cells

We explored the function of microRNA-194 in apoptosis of human melanoma cells, and caspase-3/−9 activities were measured using commercial kits. There was a significant increase of caspase-3/−9 activities in human melanoma cells by microRNA-194 overexpression, compared with negative control group (Fig. 4).

MicroRNA-194 overexpression promotes Bax/Bcl-2 of human melanoma cells

To explore the apoptosis mechanism of microRNA-194 on human melanoma cells, Bax and Bcl-2 protein was measured using western blots. As shown in Fig. 5, microRNA-194 overexpression promoted Bax/Bcl-2 of human melanoma cells, compared with the negative control group.

MicroRNA-194 overexpression suppresses PI3K/AKT/FoxO3a signaling pathway of human melanoma cells

To determine whether PI3K/AKT/FoxO3a signaling pathway is a functional target of miR-194, we analyzed PI3K, p-AKT and FoxO3a protein expression using western blots. The results of western blots showed that PI3K, p-AKT and FoxO3a protein expression was significantly suppressed by microRNA-194 overexpression in human melanoma cells, compared with the negative control group (Fig. 6).

MicroRNA-194 overexpression induces p53/p21 signaling pathway of human melanoma cells

We determined p53/p21 signaling pathway function effect of microRNA-194 on human melanoma cells. Fig. 7 indicated that microRNA-194 overexpression significantly induced p53/p21 signaling pathway in human melanoma cells, compared with the negative control group.

Suppression of PI3K affects microRNA-194 overexpression suppressed PI3K/AKT/FoxO3a signaling pathway of human melanoma cells

Then, we employed whether the suppression of PI3K affects microRNA-194 overexpression on apoptosis of human melanoma cells. We used PI3K inhibitor, 3-Methyladenine (3-MA, 1 Μm, 48 h), significantly suppressed PI3K/AKT/FoxO3a signaling pathway in SK-Mel2 cells following microRNA-194 overexpression, compared with the microRNA-194 overexpression group (Fig. 8).

Suppression of PI3K affects microRNA-194 overexpression inhibited cell proliferation and induced apoptosis of human melanoma cells

As expected, the suppression of PI3K accelerated the anticancer effect of microRNA-194 on the cell proliferation inhibition and apoptosis induction of SK-Mel2 cells, compared with the microRNA-194 overexpression group (Fig. 9).

Suppression of PI3K affects microRNA-194 overexpression increased caspase-3/−9 activities of human melanoma cells

PI3K inhibitor significantly increased caspase-3/−9 activities in human melanoma cells by microRNA-194 overexpression, compared with microRNA-194 overexpression group (Fig. 10).

Suppression of PI3K affects microRNA-194 overexpression promoted Bax/Bcl-2 of human melanoma cells

We determined whether PI3K inhibition affects microRNA-194 overexpression promoted Bax/Bcl-2 of human melanoma cells. As shown in Fig. 11, PI3K inhibition significantly increased Bax/Bcl-2 of human melanoma cells following microRNA-194 overexpression, compared with the microRNA-194 overexpression group (Fig. 11).

Suppression of PI3K affects microRNA-194 overexpression induced p53/p21 signaling pathway of human melanoma cells

We assess whether PI3K inhibition affects p53/p21 signaling pathway of human melanoma cells following microRNA-194 overexpression. Compared with microRNA-194 overexpression, PI3K inhibition significantly induced p53/p21 signaling pathway of human melanoma cells following microRNA-194 overexpression (Fig. 12).

Discussion

Malignant melanoma (MM), is derived from the melanocytes in the neural crest, and it is the most fatal skin tumor (17). It is associated with extremely high grade of malignancy and hidden onset, and the majority already has metastases when discovered in clinic, thus, it has poor chemoradiotherapy effects and great treatment difficulties (18). MM patients account for 80% among the patients who die of skin cancer (19). In recent years, the morbidity and mortality of MM show an increasing trend year by year, which greatly threaten human health (19). Our results found that microRNA-194 expression of MM tissues samples was suppressed compared to that of para-carcinoma tissue, and DFS and OS of high expression in MM patients were higher than those of low expression in MM patients.

Akt is also called protein kinase B, which is a kind of serine/threonine protein kinase. As previously reported, the PI3K pathway in a variety of malignancies is active, and the downstream signaling molecules of PI3K (Akt, mTOR, PDK and ILK) are the potential targets for treating malignancies (20,21). Human Foxo transcription factor family (forkhead box protein family) has four members, the Foxo1, Foxo2, Foxo3a and Foxo4, which participate in regulating multiple cellular processes, including cell proliferation, apoptosis, differentiation and metabolism (22). Foxo family is an important downstream signaling molecule of the PI3K/Akt signal pathway (22). Foxo family transfers from the nucleus to the cytoplasm after being phosphorylated by Akt and its transcriptional activity is inhibited (23). We demonstrated that microRNA-194 overexpression inhibited cell proliferation, induced apoptosis, increased caspase-3/−9 activities and promoted Bax/Bcl-2 of human melanoma cells through suppression of PI3K/AKT/FoxO3a signaling pathway.

Foxo has the function of inhibiting the intracellular reactive oxygen, which is mainly through promoting the expression of reactive oxygen scavenger (23). The reactive oxygen scavengers include superoxide dismutase (SOD2, MnSOD) and catalase. Akt has long been considered as the signal pathway for promoting tumor survival and proliferation, besides, in multiple tumor cells, Akt is in the excessively activated state (24). However, it has also been considered that the excessively activated Akt could inhibit cell apoptosis due to numerous reasons, except for the reactive oxygen-induced apoptosis; furthermore, the activation of Akt made the cells more sensitive to the reactive oxygen-induced apoptosis (25). Altogether, these data indicated that PI3K inhibition significantly suppressed PI3K/AKT/FoxO3a signaling pathway in SK-Mel2 cells following microRNA-194 overexpression. Chi (26) provided compelling evidence that microRNA-194 inhibited cell proliferation of oral squamous cell carcinoma by reducing PI3K-Akt-FoxO3a signaling. Our results were consistent with previous studies demonstrating that microRNA-194 inhibits biological features of human MM through PI3K/AKT/FoxO3a signaling pathway. Krützfeldt et al (27) provided compelling evidence that microRNA-194 regulates cell proliferation of oral squamous cell carcinoma via PI3K/AKT/FoxO3a signaling pathway.

The transcription of p53, as well as mRNA splicing and translation is regulated, the mRNA splicing prompts the p53 gene to express several isomeric molecules which have various activities (28). p53 has extremely short half-life in normal cells, which is only 5–30 min. The intracellular p53 level is mainly regulated by the rigid negative feedback of its target gene murine double minute 2 (MDM2), and its transcript MDM2 is a kind of E3 ubiquitin ligase, which can promote its degradation through promoting the ubiquitination of p53 (29). However, MDM2 has the function of regulating the ubiquitin-independent p53 activity, and it can prompt p53 to adopt the mutant conformation through binding its acidic domain with p53, and thus inhibit the DNA binding activity of p53 (30). Furthermore, MDM2 is a target gene of p53 activation; therefore, p53-MDM2 forms a negative feedback circuit, which means that p53 regulates the expression of MDM2 at the transcription level, while MDM2 regulates the transcriptional activity of p53 as well as the degradation of the proteasome pathway that it mediates (29). In the present study, we demonstrated that microRNA-194 overexpression induced p53/p21 signaling pathway of human melanoma cells.

p21 is an apoptosis regulatory factor, the activated AKT1 can phosphorylate the Thr145 residue of p21, and thus block the nuclear translocation of p21, render the arrest of p21 in the cytoplasm; and the phosphorylated p21 which locates in cytoplasm has anti-apoptotic activity, which is due to inhibition of the activity of proteins that are involved in apoptosis, such as procaspase-3, caspase-8 and caspase-10; in addition, it can also upregulate the expression of the cytokines that have anti-apoptotic activity, and inhibit the expression of the pro-apoptotic genes transcribed by MYC and E2F1 (31,32). The transcription of p21 is regulated by multiple signals and factors. Under the induction of various stimulations, such as DNA damage and nucleotide loss, the transcription of p21 depends on p53 as well as its family member p73 (33). Furthermore, many other factors can directly or indirectly induce p53-independent p21 transcription (34).

It is currently considered that the p53-p21 pathway can induce aging, while the p16-pRB pathway maintains aging. In-depth research reveals that p53 induces aging through its downstream p21 and ROS; to be more specific, certain level and duration of ROS is the determinant of p53 initiating the p21 transcription, while the increased p21 expression can exert its transcription regulatory function through the interaction with the transcriptional coactivator p300, which does not rely on PCNA binding or CDK kinase inhibitory activity; induce PIG3 expression and thus induce elevated ROS in cells; and the elevated ROS can maintain long-term cell cycle arrest (14,35). If ROS is eliminated from the p21-induced aging cells, the cells can recover from aging (14). Though p16 is also CDK inhibitory factor, it does not have transcription regulatory function and cannot induce elevated ROS level in the cell; therefore, it can induce irreversible cell cycle arrest (36). In the present study, our in vitro studies showed that PI3K inhibition significantly induced p53/p2 signaling pathway in SK-Mel2 cells following microRNA-194 overexpression. Sundaram et al (37) reported that miR-194 inhibits thrombospondin-1 and promotes angiogenesis of colon cancers through p53 signaling pathway. Taken together, these data provide solid evidence to support that microRNA-194 exerts it inhibitory effect on human melanoma cells, at least in part, through p53/p21 signaling pathway. Krützfeldt et al (27) found that miR-194 is a target of transcription factor 1 and regulates Dgcr8 and p53 in liver tumorigenesis.

In conclusion, our results indicate that microRNA-194 overexpression inhibited cell proliferation, induced apoptosis, increased caspase-3/−9 activities and promoted Bax/Bcl-2 of human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway. Therefore, the microRNA-194/PI3K/AKT/FoxO3a and p53/p21 should be further studied for possible treatment of melanoma.

References

1 

Greene JM, Schneble EJ, Jackson DO, Hale DF, Vreeland TJ, Flores M, Martin J, Herbert GS, Hardin MO, Yu X, et al: A phase I/IIa clinical trial in stage IV melanoma of an autologous tumor-dendritic cell fusion (dendritoma) vaccine with low dose interleukin-2. Cancer Immunol Immunother. 65:383–392. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Ong CC, Jubb AM, Jakubiak D, Zhou W, Rudolph J, Haverty PM, Kowanetz M, Yan Y, Tremayne J, Lisle R, et al: P21-activated kinase 1 (PAK1) as a therapeutic target in BRAF wild-type melanoma. J Natl Cancer Inst. 105:606–607. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Benyi E, Kieler H, Linder M, Ritzén M, Carlstedt-Duke J, Tuvemo T, Westphal O and Sävendahl L: Risks of malignant and non-malignant tumours in tall women treated with high-dose oestrogen during adolescence. Horm Res Paediatr. 82:89–96. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Eggermont AM, Suciu S, Rutkowski P, Kruit WH, Punt CJ, Dummer R, Salès F, Keilholz U, de Schaetzen G and Testori A: EORTC Melanoma Group: Long term follow up of the EORTC 18952 trial of adjuvant therapy in resected stage IIB-III cutaneous melanoma patients comparing intermediate doses of interferon-alpha-2b (IFN) with observation: Ulceration of primary is key determinant for IFN-sensitivity. Eur J Cancer. 55:111–121. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Varamo C, Occelli M, Vivenza D, Merlano M and Lo Nigro C: MicroRNAs role as potential biomarkers and key regulators in melanoma. Genes Chromosomes Cancer. 56:3–10. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Babapoor S, Horwich M, Wu R, Levinson S, Gandhi M, Makkar H, Kristjansson A, Chang M and Dadras SS: microRNA in situ hybridization for miR-211 detection as an ancillary test in melanoma diagnosis. Mod Pathol. 29:461–475. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Venkatesan N, Kanwar J, Deepa PR, Khetan V, Crowley TM, Raguraman R, Sugneswari G, Rishi P, Natarajan V, Biswas J, et al: Clinico-pathological association of delineated miRNAs in uveal melanoma with monosomy 3/disomy 3 chromosomal aberrations. PLoS One. 11:e01461282016. View Article : Google Scholar : PubMed/NCBI

8 

Dror S, Sander L, Schwartz H, Sheinboim D, Barzilai A, Dishon Y, Apcher S, Golan T, Greenberger S, Barshack I, et al: Melanoma miRNA trafficking controls tumour primary niche formation. Nat Cell Biol. 18:1006–1017. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Kou Y, Li L, Li H, Tan Y, Li B, Wang K and Du B: Berberine suppressed epithelial mesenchymal transition through cross-talk regulation of PI3K/AKT and RARα/RARβ in melanoma cells. Biochem Biophys Res Commun. 479:290–296. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Xu G, Zhang W, Bertram P, Zheng XF and McLeod H: Pharmacogenomic profiling of the PI3K/PTEN-AKT-mTOR pathway in common human tumors. Int J Oncol. 24:893–900. 2004.PubMed/NCBI

11 

Dong MH, Zhang Q, Wang YY, Zhou BS, Sun YF and Fu Q: Euphorbia fischeriana Steud inhibits malignant melanoma via modulation of the phosphoinositide-3-kinase/Akt signaling pathway. Exp Ther Med. 11:1475–1480. 2016.PubMed/NCBI

12 

Doscas ME, Williamson AJ, Usha L, Bogachkov Y, Rao GS, Xiao F, Wang Y, Ruby C, Kaufman H, Zhou J, et al: Inhibition of p70 S6 kinase (S6K1) activity by A77 1726 and its effect on cell proliferation and cell cycle progress. Neoplasia. 16:824–834. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Guo Y, Chang H, Li J, Xu XY, Shen L, Yu ZB and Liu WC: Thymosin alpha 1 suppresses proliferation and induces apoptosis in breast cancer cells through PTEN-mediated inhibition of PI3K/Akt/mTOR signaling pathway. Apoptosis. 20:1109–1121. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Guterres FA, Martinez GR, Rocha ME and Winnischofer SM: Simvastatin rises reactive oxygen species levels and induces senescence in human melanoma cells by activation of p53/p21 pathway. Exp Cell Res. 319:2977–2988. 2013. View Article : Google Scholar : PubMed/NCBI

15 

de Andrade BA, León JE, Carlos R, Delgado-Azañero W, Mosqueda-Taylor A and de Almeida OP: Immunohistochemical expression of p16, p21, p27 and cyclin D1 in oral nevi and melanoma. Head Neck Pathol. 6:297–304. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Jang GH and Lee M: BH3-mimetic gossypol-induced autophagic cell death in mutant BRAF melanoma cells with high expression of p21Cip¹). Life Sci. 102:41–48. 2014. View Article : Google Scholar : PubMed/NCBI

17 

van der Ploeg AP, Haydu LE, Spillane AJ, Scolyer RA, Quinn MJ, Saw RP, Shannon KF, Stretch JR and Thompson JF: Melanoma patients with an unknown primary tumor site have a better outcome than those with a known primary following therapeutic lymph node dissection for macroscopic (clinically palpable) nodal disease. Ann Surg Oncol. 21:3108–3116. 2014. View Article : Google Scholar : PubMed/NCBI

18 

18. Plimack ER, Desai JR, Issa JP, Jelinek J, Sharma P, Vence LM, Bassett RL, Ilagan JL, Papadopoulos NE and Hwu WJ: A phase I study of decitabine with pegylated interferon α-2b in advanced melanoma: Impact on DNA methylation and lymphocyte populations. Invest New Drugs. 32:969–975. 2014. View Article : Google Scholar : PubMed/NCBI

19 

19. Robert C, Dummer R, Gutzmer R, Lorigan P, Kim KB, Nyakas M, Arance A, Liszkay G, Schadendorf D, Cantarini M, et al: Selumetinib plus dacarbazine versus placebo plus dacarbazine as first-line treatment for BRAF-mutant metastatic melanoma: A phase 2 double-blind randomised study. Lancet Oncol. 14:733–740. 2013. View Article : Google Scholar : PubMed/NCBI

20 

20. Wang K, Gao W, Dou Q, Li Q, Nice EC and Huang C: Ivermectin induces PAK1-mediated cytostatic autophagy in breast cancer. Autophagy. 12:2498–2499. 2016. View Article : Google Scholar : PubMed/NCBI

21 

21. Vucicevic L, Misirkic M, Janjetovic K, Vilimanovich U, Sudar E, Isenovic E, Prica M, Harhaji-Trajkovic L, Kravic-Stevovic T, Bumbasirevic V, et al: Compound C induces protective autophagy in cancer cells through AMPK inhibition-independent blockade of Akt/mTOR pathway. Autophagy. 7:40–50. 2011. View Article : Google Scholar : PubMed/NCBI

22 

22. Syed DN, Chamcheu JC, Khan MI, Sechi M, Lall RK, Adhami VM and Mukhtar H: Fisetin inhibits human melanoma cell growth through direct binding to p70S6K and mTOR: Findings from 3-D melanoma skin equivalents and computational modeling. Biochem Pharmacol. 89:349–360. 2014. View Article : Google Scholar : PubMed/NCBI

23 

23. Babchia N, Calipel A, Mouriaux F, Faussat AM and Mascarelli F: The PI3K/Akt and mTOR/P70S6K signaling pathways in human uveal melanoma cells: Interaction with B-Raf/ERK. Invest Ophthalmol Vis Sci. 51:421–429. 2010. View Article : Google Scholar : PubMed/NCBI

24 

24. Caporali S, Alvino E, Lacal PM, Levati L, Giurato G, Memoli D, Caprini E, Cappellini Antonini GC and D'Atri S: Targeting the PI3K/AKT/mTOR pathway overcomes the stimulating effect of dabrafenib on the invasive behavior of melanoma cells with acquired resistance to the BRAF inhibitor. Int J Oncol. 49:1164–1174. 2016.PubMed/NCBI

25 

25. Pappalardo F, Russo G, Candido S, Pennisi M, Cavalieri S, Motta S, McCubrey JA, Nicoletti F and Libra M: Computational modeling of PI3K/AKT and MAPK signaling pathways in melanoma cancer. PLoS One. 11:e01521042016. View Article : Google Scholar : PubMed/NCBI

26 

26. Chi H: miR-194 regulated AGK and inhibited cell proliferation of oral squamous cell carcinoma by reducing PI3K-Akt-FoxO3a signaling. Biomed Pharmacother. 71:53–57. 2015. View Article : Google Scholar : PubMed/NCBI

27 

27. Krützfeldt J, Rösch N, Hausser J, Manoharan M, Zavolan M and Stoffel M: MicroRNA-194 is a target of transcription factor 1 (Tcf1, HNF1α) in adult liver and controls expression of frizzled-6. Hepatology. 55:98–107. 2012. View Article : Google Scholar : PubMed/NCBI

28 

28. Lawrence NF, Hammond MR, Frederick DT, Su Y, Dias-Santagata D, Deng A, Selim MA, Mahalingam M, Flaherty KT and Hoang MP: Ki-67, p53, and p16 expression, and G691S RET polymorphism in desmoplastic melanoma (DM): A clinicopathologic analysis of predictors of outcome. J Am Acad Dermatol. 75:595–602. 2016. View Article : Google Scholar : PubMed/NCBI

29 

29. Perrotta C, Buonanno F, Zecchini S, Giavazzi A, Serafini Proietti F, Catalani E, Guerra L, Belardinelli MC, Picchietti S, Fausto AM, et al: Climacostol reduces tumour progression in a mouse model of melanoma via the p53-dependent intrinsic apoptotic programme. Sci Rep. 6:272812016. View Article : Google Scholar : PubMed/NCBI

30 

30. Zhao S, Niu F, Xu CY, Ye L, Bi GB, Chen L, Gong P, Tian G and Nie TH: Microarray and ChIP-seq data analysis revealed changes in p53-mediated transcriptional regulation in Nutlin-3-treated U2OS cells. Mol Med Rep. 12:4284–4290. 2015.PubMed/NCBI

31 

31. Kaluzki I, Hrgovic I, Hailemariam-Jahn T, Doll M, Kleemann J, Valesky EM, Kippenberger S, Kaufmann R, Zoeller N and Meissner M: Dimethylfumarate inhibits melanoma cell proliferation via p21 and p53 induction and bcl-2 and cyclin B1 downregulation. Tumour Biol. 37:13627–13635. 2016. View Article : Google Scholar : PubMed/NCBI

32 

32. Fan T, Jiang S, Chung N, Alikhan A, Ni C, Lee CC and Hornyak TJ: EZH2-dependent suppression of a cellular senescence phenotype in melanoma cells by inhibition of p21/CDKN1A expression. Mol Cancer Res. 9:418–429. 2011. View Article : Google Scholar : PubMed/NCBI

33 

33. Genov M, Kreiseder B, Nagl M, Drucker E, Wiederstein M, Muellauer B, Krebs J, Grohmann T, Pretsch D, Baumann K, et al: Tetrahydroanthraquinone derivative (±)-4-deoxyaustrocortilutein induces cell cycle arrest and apoptosis in melanoma cells via upregulation of p21 and p53 and downregulation of NF-kappaB. J Cancer. 7:555–568. 2016. View Article : Google Scholar : PubMed/NCBI

34 

34. Yun CY, You ST, Kim JH, Chung JH, Han SB, Shin EY and Kim EG: p21-activated kinase 4 critically regulates melanogenesis via activation of the CREB/MITF and β-catenin/MITF pathways. J Invest Dermatol. 135:1385–1394. 2015. View Article : Google Scholar : PubMed/NCBI

35 

35. Pelletier J, Dayan F, Durivault J, Ilc K, Pécou E, Pouysségur J and Mazure NM: The asparaginyl hydroxylase factor-inhibiting HIF is essential for tumor growth through suppression of the p53-p21 axis. Oncogene. 31:2989–3001. 2012. View Article : Google Scholar : PubMed/NCBI

36 

36. Nicolau-Galmés F, Asumendi A, Alonso-Tejerina E, Pérez-Yarza G, Jangi SM, Gardeazabal J, Arroyo-Berdugo Y, Careaga JM, Díaz-Ramón JL, Apraiz A, et al: Terfenadine induces apoptosis and autophagy in melanoma cells through ROS-dependent and -independent mechanisms. Apoptosis. 16:1253–1267. 2011. View Article : Google Scholar : PubMed/NCBI

37 

37. Sundaram P, Hultine S, Smith LM, Dews M, Fox JL, Biyashev D, Schelter JM, Huang Q, Cleary MA, Volpert OV, et al: p53-responsive miR-194 inhibits thrombospondin-1 and promotes angiogenesis in colon cancers. Cancer Res. 71:7490–7501. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 37 Issue 5

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
Bai M, Zhang M, Long F, Yu N, Zeng A and Zhao R: Circulating microRNA-194 regulates human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway. Oncol Rep 37: 2702-2710, 2017
APA
Bai, M., Zhang, M., Long, F., Yu, N., Zeng, A., & Zhao, R. (2017). Circulating microRNA-194 regulates human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway. Oncology Reports, 37, 2702-2710. https://doi.org/10.3892/or.2017.5537
MLA
Bai, M., Zhang, M., Long, F., Yu, N., Zeng, A., Zhao, R."Circulating microRNA-194 regulates human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway". Oncology Reports 37.5 (2017): 2702-2710.
Chicago
Bai, M., Zhang, M., Long, F., Yu, N., Zeng, A., Zhao, R."Circulating microRNA-194 regulates human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway". Oncology Reports 37, no. 5 (2017): 2702-2710. https://doi.org/10.3892/or.2017.5537