c-Myc modulates glucose metabolism via regulation of miR-184/PKM2 pathway in clear-cell renal cell carcinoma

  • Authors:
    • Jiwei Huang
    • Wen Kong
    • Jin Zhang
    • Yonghui Chen
    • Wei Xue
    • Dongming Liu
    • Yiran Huang
  • View Affiliations

  • Published online on: July 14, 2016     https://doi.org/10.3892/ijo.2016.3622
  • Pages: 1569-1575
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Renal cell carcinoma (RCC) is one of the most malignant tumors worldwide. Among all subtypes of RCC, clear-cell RCC (ccRCC) is the most common and aggressive one. The difficulty in overcoming resistance of traditional treatment is a threat for ccRCC therapies. Therefore, to understand the mechanism that underlies ccRCC progression is critical for new drug development. In the present study, we identified that miR-184 could be downregulated by c-Myc, which is different from the standard opinion that c-Myc is a target of miR-184. Overexpression of pre-miR-184 changed the metabolic and proliferation features of ccRCC cells by reducing cell glucose consumption, lactate production and cell proliferation. Further analysis by computer bioinformatics revealed that PKM2 is a target of miR-184. Both PKM2 mRNA and protein were significantly affected by addition of miR-184. Importantly, the PKM2 expression level was indeed increased in ccRCC samples, which is totally reverse compared to the decreased miR-184 expression level. Interestingly, we found that when PKM2 was knocked down in ccRCC cells, the rapid proliferation, high glucose consumption and high lactate production were all clearly inhibited, which indicates metabolic reprogramming and cancer progression blocking the in ccRCC cells. Our findings shed new light on ccRCC molecular study and provide a new and solid basis for developing ccRCC therapy.

Introduction

Renal cell carcinoma (RCC) is identified as the seventh most common cancer for adult humans and account for 2% of all cancers worldwide (1,2). The lack of efficient clinical diagnostic and curative plan is one of the major reasons for RCC lethality (3,4). Among all the subtypes of RCC, clear-cell renal cell carcinoma (ccRCC) is the most frequently discovered subtype. Patients with ccRCC usually have the lowest probability of recovery compared with other RCC subtypes (5).

ccRCC is a metabolic disease characterized by the dysregulation of many metabolic pathways, including abnormal oxygen and energy consumption, as well as aberrant nutrient sensing (6). During ccRCC progression, inactivation of certain genes (such as von Hippel-Lindau, VHL) and signaling pathway activation (such as MYC pathway) were reported to take part in the abnormal metabolic profiles (710). Recently, a large number of abnormally expressed miRNAs were reported to be involved in the progression of ccRCC (11). Among these miRNAs, miR-184 is widely dysregulated in various human cancers (1215). Previous studies have demonstrated that miR-184 was downregulated and functions as a tumor suppressor in RCC (16,17). However, the molecular mechanism of miR-184 in regulating RCC progression is still unknown. It is worth mentioning that c-Myc, a well-known oncogene that promotes cell cycle, cell differentiation and apoptosis (18), is reported to be a target of miR-184 in many human malignancies, including non-small cell lung cancer (19,20), nasopharyngeal carcinoma (14), tongue squamous cell carcinoma (12) and hepatocellular carcinoma (15). Notably, the expression of c-Myc was indeed upregulated in ccRCC (10,21). However, whether downregulated miR-184 is related to upregulated c-Myc in ccRCC is still unknown.

In this study, we indentified PKM2 as a new target of miR-184. Furthermore, the expression of miR-184 can be negatively regulated by a well-known oncogene c-Myc. PKM2 is highly expressed in human ccRCC samples. Remarkably, PKM2 knockdown leads to metabolism reprogramming and growth inhibition in ccRCC cell lines.

Materials and methods

Patient samples

Fifty pairs of human ccRCC and normal kidney samples were collected from our institution, where the samples were evaluated by a comprehensive clinical assessment. All the paraffin-embedded sections and frozen samples used in the present study were approved by ethics license of RJ2014N017.

Cell culture and transient transfection

The 786-O and RCC4 cell lines were from the American Type Culture Collection (ATCC; Manassas, VA, USA). The basic medium for 786-O and RCC4 cells were RPMI and Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 2 mM glutamine, respectively. The culture medium consisted of basic medium supplemented with 10% fetal bovine serum (FBS), 1X penicillin/streptomycin (both from Invitrogen, Grand Island, NY, USA). Cells were incubated at 37°C in a humidified atmosphere of 5% CO2 and 95% air. For transfection, cells were plated subconfluently onto each well of 24-well tissue culture plates 24 h before transfection. Transient transfection of pre-miR-184 or control plasmid at a final concentration of 20 nM per well was accomplished with Lipofectamine 2000 (Invitrogen) according to the manufacturer’s protocol. The human PKM2 siRNA and control siRNA were purchased from OriGene. The human c-Myc plasmid was generated according to standard molecular biology techniques and verified by sequencing. c-Myc plasmids were transfected with Lipofectamine 2000 according to the manufacturer’s instructions.

RNA isolation and miRNA detection

Total RNA of cells and tissues were extracted using TRIzol reagent (Invitrogen) according to the manufacturer’s instructions. For isolation of small RNAs the mirVana miRNA isolation kit (Ambion) was used. Detecting the miRNA mature form was performed using the mirVana qRT-PCR miRNA detection kit (Ambion). qRT-PCR primer sets were according to the manufacturer’s instructions. We chose U6 small nuclear RNA as an internal control. The real-time RT-PCR assays were performed using the 7500 Fast Real-Time PCR System for quantitative mRNA detection and with iTaq Fast SYBR-Green Supermix (Bio-Rad Laboratories). The primers for real-time PCR were human actin: 5′-AAGGAGCCCCACGAGAAAAAT-3′ (forward) and 5′-ACCGAACTTGCATTGATTCCAG-3′ (reverse); human c-Myc: 5′-TCAAGAGGTGCCACGTCTCC-3′ (forward) and 5′-TCTTGGCAGCAGGATAGTCCTT-3′ (reverse); human PKM2: 5′-ATAACGCCTACATGGAAAAGTGT-3′ (forward) and 5′-TAAGCCCATCATCCACGTAGA-3′ (reverse).

MTT experiments

Cells were seeded in 96-well microtitre plates with 1×103 cells/well and incubated for 24 h in 100 μl culture medium. MTT [100 μl (5 g/l)] was added to the cells, which were then cultured for a further 4 h. Following the removal of the supernatant fluid, 100 μl/well DMSO was added to the cells which were agitated for 15 min. The absorbance was measured at 570 nm by a microplate reader. Each assay was repeated three times.

Metabolism assays

786-O and RCC4 cells were seeded into 24-well plate with 200 ml of medium in each well. To determine the levels of glucose and lactate, the supernatants of cell culture media were collected and assayed for glucose and lactate levels by using glucose assay kit and lactate assay kit (BioVision Inc., Milpitas, CA, USA) according to the manufacturer’s instructions. Glucose consumption and lactate production were calculated based on the standard curve, and normalized to the cell number.

Colony formation assay

Anchorage-independent growth was determined by the ability of cells to form colonies in soft agar. 786-O and RCC4 cells were diluted in fresh medium, reseeded in 0.3% agar in 6-well plates. Cell growth was allowed to proceed for 25 days. Formed colonies were fixed with 10% formaldehyde and counted using the Col Count instrument (Oxford Optronix Ltd., Abingdon, UK).

Luciferase reporter assay

Luciferase assay was performed as previously described (22). Briefly, 786-O cells of 50% confluence in 24-well plates were transfected with FuGENE HD reagent (Roche). In each well, 200 ng pRL-TK-PKM2-3′-UTR reporter firefly luciferase construct was co-transfected with 1 ng Renilla plasmid and pre-miR-184, or Ctrl microRNA (Ambion) into cells. Cell extracts were prepared 48 h after transfection and luciferase activity was measured with the Dual-Luciferase reporter assay system (Promega). PKM2 3′-UTR-luciferase or PKM2 3′-UTR-luciferase mutant activities were normalized by first dividing the luciferase activity in each well by Renilla activity.

Western blot analysis

Lysates were prepared from 1×107 cells by dissolving cell pellets in 100 μl of lysis buffer containing protease inhibitors. Lysates were centrifuged at 18,000 × g for 15 min and the supernatant was collected. The protein concentration was estimated with the Bio-Rad protein assay kit (Bio-Rad Laboratories, Hercules, CA, USA) using bovine serum albumin as a standard. Sample proteins were resolved by 10% sodium dodecylsulfate polyacrylamide gel (Bio-Rad Laboratories) electrophoresis and then electrophoretically transferred to PVDF membrane (Millipore) and blocked with 5% BSA (Sigma-Aldrich). Subsequently the primary antibodies PKM2 (1:1,000; Cell Signalling Technology), c-Myc (1:2,000; Santa Cruz Biotechnology) and actin (1:10,000; Sigma-Aldrich) were added. After overnight incubation at 4°C the blots were washed, exposed to HRP-conjugated corresponding secondary antibodies for one hour and finally were visualized by ECL Advanced Solution (GE Healthcare Life Sciences). Digital images were captured by Gel Doc™ gel documentation system (Bio-Rad Laboratories).

Immunocytochemistry and in situ hybridization

The procedures of immunohistochemical studies for paraffin-embedded sections were performed as previously described (23). Anti-human PKM2 antibody (1:100; GeneTex Inc.) and anti-human c-Myc antibody (1:100; Santa Cruz Biotechnology) were used as primary antibody. miR-184 in situ hybridization assay was conducted by using locked nucleic acid (LNA)-digoxigenin (DIG)-labeled probes (Exiqon, Vedbæk, Denmark) as previously described (24). Briefly, deparaffinized sections were treated with Proteinase K for 40 min. The hybridization was performed at 45°C for 75 min in a hybridization buffer containing Dual-DIG-labelled LNA probes miR-184 detection probe (probe sequence: ACCCTTATCAGTTCTCCGTCCA) or scramble-miR (probe sequence: GTGTAACACGTCTAT ACGCCCA) (Exiqon). Sections were blocked in a blocking buffer for 30 min and washed in PBS three times.

miR-184 target gene prediction algorithms

The presence of miR-184 target gene binding sites was analyzed using miRWalk, which collates data from multiple prediction programs as previously described (25).

Statistical analysis

Statistical analyses were performed using the SPSS software package. Results are expressed as mean ± SD of at least three independent experiments. Statistical significance was assessed by two-tailed Student’s t-test. Differences were considered statistically significant at P<0.05. P<0.05, P<0.01 and P<0.001.

Results

c-Myc negatively regulates miR-184 expression in ccRCC

In order to investigate the relationship between c-Myc and miR-184, we first examined their expression in 50 matched pairs of human ccRCC samples and tumor adjacent normal kidney tissue. As shown in Fig. 1A, consistent with previous reports, the expression of c-Myc was greatly increased in ccRCC samples, whereas miR-184 was decreased comparing with the normal tissues. To confirm these results, we detected c-Myc and miR-184 expression by immunohistochemistry and in situ hybridization, respectively. Consistently, increased c-Myc and decreased miR-34a were observed in ccRCC samples (Fig. 1C and D). As c-Myc was reported to be a target of miR-184 (12,14,15,19,20), we assume that the opposite expression profile between c-Myc and miR-184 may be a result of miR-184 targeting. However, we also noted that inhibition of MYCN, another member of MYC gene family, causes significantly upregulation of miR-184 (26). It is known that c-Myc functions as a transcription factor to influence the expression of a broad range of human genes involved in the progression of cell carcinogenesis (18). We wondered whether the downregulated expression of miR-184 could also be regulated by c-Myc. To test this possibility, we transfected c-Myc into the ccRCC cell line 786-O. Surprisingly, overexpression of c-Myc significantly downregulated the expression of miR-184 (Fig. 1B), which indicated miR-184 is a probable target of c-Myc in ccRCC cells.

miR-184 inhibits glucose metabolism in ccRCC cells

As ccRCC is featured by abnormal cell proliferation and metabolism, including increased glucose consumption, lactate production (6), we next wondered whether the abnormal proliferative and metabolic features in ccRCC cells could be improved by miR-184 overexpression. We transfected either the pre-miR negative control or the pre-miR-184 into the ccRCC cell lines 786-O and RCC4. As shown in Fig. 2A, transfection of pre-miR-184 led to a ~3-fold increase of the miR-184 expression level both in 786-O and RCC4 cells. Next, we measured glucose consumption and lactate production in these cells. As shown Fig. 2B, we observed significant glucose uptake reduction in pre-miR-184 transfected 786-O and RCC4 cells; in addition, lactate production were also decreased ~30% in 786-O cells and ~20% in RCC4 cells with miR-184 over-expression (Fig. 2C), indicating the metabolic features were improved by miR-184 overexpression. The effect of miR-184 on RCC cell proliferation was then analyzed. As shown in the MTT assay of Fig. 2D, the cell proliferation was markedly reduced by miR-184 overexpression in 786-O and RCC4 cells on the fourth day after transfection. Similarly, miR-184 overexpression impaired colony formation of both 786-O and RCC4 cells (Fig. 2E). Thus, miR-184 inhibited ccRCC cell proliferation, glucose uptake and lactate production.

miR-184 specifically represses PKM2 expression

To uncover the mechanism of miR-184 contribution to ccRCC progression, we investigated putative miR-184 targets that directly regulate ccRCC metabolism and proliferation using bioinformatics methods. The bioinformatics analysis revealed that PKM2 (M2 isoform of pyruvate kinase) contains a potential miRNA response element (MRE) for miR-184 in its 3′-untranslated region (3′-UTR) (Fig. 3A). PKM2 is a ‘glycolytic valve’ in deciding the direction of glucose flux in glycolysis (27). It catalyzes the conversion of phosphoenolpyruvate (PEP) to pyruvate with concomitant production of ATP (28), which is independent of oxygen consumption. Thus, the high expression of PKM2 in tumor tissues helps tumor cells to grow rapidly even in hypoxic conditions (29). To determine whether PKM2 is indeed a target that is directly affected by miR-184, we examined the luciferase activity driven by PKM2-3′UTR containing the MRE sequence. As shown in Fig. 3B, in cells transfected with promoter containing PKM2 WT MRE sequences, pre-miR-184 could significantly reduce the luminescence comparing with the control group. However, when the PKM2 MRE was mutated, miR-184 could not change the luciferase activity (Fig. 3A and B). To directly monitor the target effect of miR-184 on PKM2, we tested PKM2 expression both at mRNA level and protein expression level. In accordance, pre-miR-184 decreased PKM2 expression by 50% at mRNA level (Fig. 3C), and repressed PKM2 protein expression obviously (Fig. 3D). We also examined the effects of c-Myc overexpression on PKM2 expression. As shown in Fig. 3E, PKM2 expression was enhanced by c-Myc over-expression. However, when c-Myc and pre-miR-184 were co-expressed, the upregulated PKM2 by c-Myc overexpression was abolished in response to miR-184 overexpression, suggesting that c-Myc indeed increased PKM2 expression by inhibiting miR-184 expression. In brief, our data indicated PKM2 is a new target of miR-184 in ccRCC cells.

Knockdown of PKM2 suppressed the glycolysis and cell proliferation

After discovering PKM2 as a direct target of miR-184, we next asked whether the altered metabolic and proliferative features in ccRCC were the results of increased PKM2 level, as the miR-184 was downregulated. To test this hypothesis, we transfected PKM2 siRNA into 786-O cells to lower PKM2 expression level (Fig. 4A). Firstly we analyzed the proliferation of PKM2 knockdown cells, and found a profound decrease in proliferative cell numbers by MTT on the fourth day post-PKM2 siRNA transfection (Fig. 4B). Furthermore, consistent with our expectations, knockdown of PKM2 remarkably decreased glucose uptake and lactate production in 786-O cells (Fig. 4C and D). Furthermore, we found pre-miR-184 overexpression in 786-O cells resulted in impaired glucose uptake, which could be rescued by PKM2 overexpression. This result, therefore, suggested that miR-184 inhibited glycolysis in a PKM2-dependent manner (Fig. 4E and F). These data supported the conclusion that knockdown of PKM2 inhibits abnormal cell proliferation and metabolism in ccRCC cell line.

Expression of PKM2 is increased in ccRCC

Finally, we examined the expression of PKM2 in human ccRCC samples. As shown in Fig. 5A, the expression of PKM2 mRNA doubled in the ccRCC samples compared with the normal tissue. Consistently, high expression of PKM2 protein level was also observed in ccRCC samples examined by immunohistochemistry (Fig. 5B). Then we analyzed the protein expression level of PKM2 and c-Myc in paired ccRCC samples. Strikingly, increased expression of both PKM2 and c-Myc were observed in one same ccRCC sample comparing with paired normal tissue (Fig. 5C). Thus, our results indicated that the high expression of PKM2 in ccRCC tissues might be a reason leading to hyperproliferation and metabolic disorder of ccRCC.

Discussion

ccRCC is one of the most malignant tumors worldwide. It consists of several histologic subtypes, including clear cell, papillary, chromophobe and collecting duct (3). Each subtype is different in molecular profiling, and ccRCC is the most common and aggressive one. One of the most important difficulties for ccRCC treatment is its resistance to traditional therapies (4,30). Therefore, the mechanism study in ccRCC progression is critical for uncovering the putative molecular targets for ccRCC therapy.

To date, many proteins and RNAs have been reported to take part in the progression of ccRCC, such as c-Myc, VHL, miR-34 and miR-184 (7,10,21,31). However, the exact mechanism was not clarified. In the present study, we identified that miR-184 could be regulated by c-Myc, which is contrary to general opinion that c-Myc is a target of miR-184.

miR-184 has been reported to be downregulated in many cancer types, including ccRCC. In this study, we found over-expression of pre-miR-184 could reduce ccRCC cell glucose consumption, lactate production and cell proliferation. Further analysis by computer bioinformatics revealed that PKM2 is a target of miR-184. Both the mRNA and protein level of PKM2 was markedly reduced in response to miR-184 overexpression. Most importantly, the PKM2 expression level was indeed increased in ccRCC samples, which is totally reverse to the decreased miR-184 expression level. This result indirectly verified that PKM2 is a target of miR-184 in human ccRCC samples.

PKM2 is a key regulator of the metabolic fate of the glycolytic intermediates, its high expression in many cancers favored the cancer cells to proliferate rapidly even in anaerobic conditions. We found that knockdown of PKM2 in ccRCC cells, inhibited the rapid proliferation, high glucose consumption and high lactate production to a certain extent, which indicated metabolic reprogramming in ccRCC cells. In summary, in the present study, we identified PKM2 as a new target of miR-184, and that its expression can be inhibited by c-Myc. As a result, PKM2 expression can be upregulated by c-Myc overexpression. Knockdown the high PKM2 expression profile in ccRCC leads to metabolism reprogramming in these cancer cells, and then inhibits cancer progression of ccRCC (Fig. 6). Our findings shed new light on ccRCC molecular study and provide a new and solid basis for developing ccRCC therapy.

Acknowledgements

The design and conduct of the project was supported by the Award Number 81402084, 81272841, 81472378 from the National Natural Science Foundation of China and the Award Number PYXJS16-008 from the Incubating Program for Clinical Research and Innovation of Renji Hospital.

References

1 

Chow WH, Dong LM and Devesa SS: Epidemiology and risk factors for kidney cancer. Nat Rev Urol. 7:245–257. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Majer W, Kluzek K, Bluyssen H and Wesoły J: Potential approaches and recent advances in biomarker discovery in clear-cell renal cell carcinoma. J Cancer. 6:1105–1113. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Cohen HT and McGovern FJ: Renal-cell carcinoma. N Engl J Med. 353:2477–2490. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Amato RJ: Chemotherapy for renal cell carcinoma. Semin Oncol. 27:177–186. 2000.PubMed/NCBI

5 

Beck SD, Patel MI, Snyder ME, Kattan MW, Motzer RJ, Reuter VE and Russo P: Effect of papillary and chromophobe cell type on disease-free survival after nephrectomy for renal cell carcinoma. Ann Surg Oncol. 11:71–77. 2004. View Article : Google Scholar

6 

Massari F, Ciccarese C, Santoni M, Brunelli M, Piva F, Modena A, Bimbatti D, Fantinel E, Santini D, Cheng L, et al: Metabolic alterations in renal cell carcinoma. Cancer Treat Rev. 41:767–776. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Linehan WM, Srinivasan R and Schmidt LS: The genetic basis of kidney cancer: A metabolic disease. Nat Rev Urol. 7:277–285. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Gossage L, Eisen T and Maher ER: VHL, the story of a tumour suppressor gene. Nat Rev Cancer. 15:55–64. 2015. View Article : Google Scholar

9 

Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 499:43–49. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Tang SW, Chang WH, Su YC, Chen YC, Lai YH, Wu PT, Hsu CI, Lin WC, Lai MK and Lin JY: MYC pathway is activated in clear cell renal cell carcinoma and essential for proliferation of clear cell renal cell carcinoma cells. Cancer Lett. 273:35–43. 2009. View Article : Google Scholar

11 

Ma L and Qu L: The function of microRNAs in renal development and pathophysiology. J Genet Genomics. 40:143–152. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Wong TS, Liu XB, Wong BY, Ng RW, Yuen AP and Wei WI: Mature miR-184 as potential oncogenic microRNA of squamous cell carcinoma of tongue. Clin Cancer Res. 14:2588–2592. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Foley NH, Bray IM, Tivnan A, Bryan K, Murphy DM, Buckley PG, Ryan J, O’Meara A, O’Sullivan M and Stallings RL: MicroRNA-184 inhibits neuroblastoma cell survival through targeting the serine/threonine kinase AKT2. Mol Cancer. 9:832010. View Article : Google Scholar : PubMed/NCBI

14 

Zhen Y, Liu Z, Yang H, Yu X, Wu Q, Hua S, Long X, Jiang Q, Song Y, Cheng C, et al: Tumor suppressor PDCD4 modulates miR-184-mediated direct suppression of C-MYC and BCL2 blocking cell growth and survival in nasopharyngeal carcinoma. Cell Death Dis. 4:e8722013. View Article : Google Scholar : PubMed/NCBI

15 

Wu GG, Li WH, He WG, Jiang N, Zhang GX, Chen W, Yang HF, Liu QL, Huang YN, Zhang L, et al: Mir-184 post-transcriptionally regulates SOX7 expression and promotes cell proliferation in human hepatocellular carcinoma. PLoS One. 9:e887962014. View Article : Google Scholar : PubMed/NCBI

16 

Leng HM, Qian WP, Zhou L, Zhai QN, Li XX, Guan ZC, Gui YT and Cai ZM: Abnormal expression and significance of MIR-184 in human renal carcinoma. Beijing Da Xue Xue Bao. 43:509–513. 2011.(In Chinese). PubMed/NCBI

17 

Su Z, Chen D, Li Y, Zhang E, Yu Z, Chen T, Jiang Z, Ni L, Yang S, Gui Y, et al: microRNA-184 functions as tumor suppressor in renal cell carcinoma. Exp Ther Med. 9:961–966. 2015.PubMed/NCBI

18 

Lebofsky R and Walter JC: New Myc-anisms for DNA replication and tumorigenesis? Cancer Cell. 12:102–103. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Lin TC, Lin PL, Cheng YW, Wu TC, Chou MC, Chen CY and Lee H: MicroRNA-184 deregulated by the microRNA-21 promotes tumor malignancy and poor outcomes in non-small cell lung cancer via targeting CDC25A and c-Myc. Ann Surg Oncol. 22(Suppl 3): 1532–1539. 2015. View Article : Google Scholar

20 

Liu Z, Mai C, Yang H, Zhen Y, Yu X, Hua S, Wu Q, Jiang Q, Zhang Y, Song X, et al: Candidate tumour suppressor CCDC19 regulates miR-184 direct targeting of C-Myc thereby suppressing cell growth in non-small cell lung cancers. J Cell Mol Med. 18:1667–1679. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Shroff EH, Eberlin LS, Dang VM, Gouw AM, Gabay M, Adam SJ, Bellovin DI, Tran PT, Philbrick WM, Garcia-Ocana A, et al: MYC oncogene overexpression drives renal cell carcinoma in a mouse model through glutamine metabolism. Proc Natl Acad Sci USA. 112:6539–6544. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Liu W, Cao H, Ye C, Chang C, Lu M, Jing Y, Zhang D, Yao X, Duan Z, Xia H, et al: Hepatic miR-378 targets p110α and controls glucose and lipid homeostasis by modulating hepatic insulin signalling. Nat Commun. 5:56842014. View Article : Google Scholar

23 

Jo HS, Kang KH, Joe CO and Kim JW: Pten coordinates retinal neurogenesis by regulating Notch signalling. EMBO J. 31:817–828. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Jia LF, Wei SB, Gong K, Gan YH and Yu GY: Prognostic implications of micoRNA miR-195 expression in human tongue squamous cell carcinoma. PLoS One. 8:e566342013. View Article : Google Scholar : PubMed/NCBI

25 

Dweep H, Sticht C, Pandey P and Gretz N: miRWalk - database: Prediction of possible miRNA binding sites by ‘walking’ the genes of three genomes. J Biomed Inform. 44:839–847. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Chen Y and Stallings RL: Differential patterns of microRNA expression in neuroblastoma are correlated with prognosis, differentiation, and apoptosis. Cancer Res. 67:976–983. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Iqbal MA, Gupta V, Gopinath P, Mazurek S and Bamezai RN: Pyruvate kinase M2 and cancer: An updated assessment. FEBS Lett. 588:2685–2692. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Gupta V and Bamezai RN: Human pyruvate kinase M2: a multi-functional protein. Protein Sci. 19:2031–2044. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Wong N, Ojo D, Yan J and Tang D: PKM2 contributes to cancer metabolism. Cancer Lett. 356:184–191. 2015. View Article : Google Scholar

30 

Lane BR, Rini BI, Novick AC and Campbell SC: Targeted molecular therapy for renal cell carcinoma. Urology. 69:3–10. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Redova M, Svoboda M and Slaby O: MicroRNAs and their target gene networks in renal cell carcinoma. Biochem Biophys Res Commun. 405:153–156. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 49 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang J, Kong W, Zhang J, Chen Y, Xue W, Liu D and Huang Y: c-Myc modulates glucose metabolism via regulation of miR-184/PKM2 pathway in clear-cell renal cell carcinoma. Int J Oncol 49: 1569-1575, 2016
APA
Huang, J., Kong, W., Zhang, J., Chen, Y., Xue, W., Liu, D., & Huang, Y. (2016). c-Myc modulates glucose metabolism via regulation of miR-184/PKM2 pathway in clear-cell renal cell carcinoma. International Journal of Oncology, 49, 1569-1575. https://doi.org/10.3892/ijo.2016.3622
MLA
Huang, J., Kong, W., Zhang, J., Chen, Y., Xue, W., Liu, D., Huang, Y."c-Myc modulates glucose metabolism via regulation of miR-184/PKM2 pathway in clear-cell renal cell carcinoma". International Journal of Oncology 49.4 (2016): 1569-1575.
Chicago
Huang, J., Kong, W., Zhang, J., Chen, Y., Xue, W., Liu, D., Huang, Y."c-Myc modulates glucose metabolism via regulation of miR-184/PKM2 pathway in clear-cell renal cell carcinoma". International Journal of Oncology 49, no. 4 (2016): 1569-1575. https://doi.org/10.3892/ijo.2016.3622