Berberine decelerates glucose metabolism via suppression of mTOR‑dependent HIF‑1α protein synthesis in colon cancer cells

  • Authors:
    • Liyuan Mao
    • Qiongyun Chen
    • Ke Gong
    • Xiaolin Xu
    • Yurou Xie
    • Wenqing Zhang
    • Hanwei Cao
    • Tianhui Hu
    • Xiaoting Hong
    • Yan‑Yan Zhan
  • View Affiliations

  • Published online on: March 16, 2018     https://doi.org/10.3892/or.2018.6318
  • Pages: 2436-2442
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hyperactivated glucose uptake and glycolytic metabolism are considered as a hallmark of cancer. Berberine, a natural alkaloid with tumor‑selective anticancer effects, has been shown to promote glucose uptake in metabolic tissues and cells. However, whether and how berberine regulates the glucose metabolism of cancer cells are still poorly understood. In the present study, we revealed that berberine, which suppressed the growth of colon cancer cell lines HCT116 and KM12C, greatly inhibited the glucose uptake and the transcription of glucose metabolic genes, GLUT1, LDHA and HK2 in these two cell lines as assessed by RT‑qPCR. A mechanistic study further indicated that the protein expression but not mRNA transcription of HIF‑1α, a well‑known transcription factor critical for dysregulated cancer cell glucose metabolism, was dramatically inhibited in berberine‑treated colon cancer cell lines. Using western blot analysis, this regulation appears to occur via protein synthesis but not protein stability as blockade of HIF‑1α protein degradation by hypoxia mimic desferrioxamine (DFX) or proteasome inhibitor MG132 did not affect berberine's effect. In addition, mTOR signaling previously reported to regulate HIF‑1α protein synthesis was further found to be suppressed by berberine. Taken together, our results indicated that berberine inhibits overactive glucose metabolism of colon cancer cells via suppressing mTOR‑depended HIF‑1α protein synthesis, which provided not only a novel mechanism involved in berberine's tumor‑specific toxicity but also a theoretical basis for the development of berberine for colon cancer treatment.

Introduction

Overactive glucose metabolism termed as the ‘Warburg effect’ plays a vital role in cancer growth. Tumor cells tend to have a higher level of glucose uptake and glycolytic metabolism to satisfy themselves with a high energy need (1). Therefore, targeting glucose metabolism is considered as a therapeutic approach for cancer treatment (2). Inhibition of glucose uptake and metabolism that aim to generate an energy deprivation state can facilitate the effect of other anticancer therapies.

Berberine is a botanical alkaloid from the Ranunculaceae and Papaveraceae plant families. It is the active component of Chinese medicine Rhizoma coptidis, and has been widely used for treating metabolic diseases, including obesity and diabetes (3,4). One of the rationale for this treatment is that berberine increased cellular glucose uptake and metabolism (57). On the other hand, the effect of berberine against cancer has also been widely explored (8). Numerous studies have demonstrated that berberine inhibits the growth of broad types of tumor cells, and is thus recognized as a potential multispectrum anticancer therapeutic agent (810). Given that cancer growth needs overspeed glucose metabolism, it raised a problem that berberine's promoting effect on glucose metabolism appears to be contradictory to its anticancer effect. However, as most of the studies on berberine's glucose metabolism function were carried out in non-tumor metabolic tissues and cells, we hypothesized that berberine may have a distinct role on cellular glucose metabolism in cancer cells.

In the present study, we investigated the berberine's effects on colon cancer cell lines. We revealed that berberine inhibits glucose uptake and reduces the transcription of glucose metabolism relative genes in colon cancer cells. These effects may be mediated by the inhibition of HIF-1α protein synthesis through suppression of mTOR pathway.

Materials and methods

Materials

Berberine, anti-rabbit IgG, anti-mouse IgG and anti-β-actin IgG were purchased from Sigma-Aldrich (Shanghai, China). Antibodies for HIF-1α (cat. no. 14179), mTOR (cat. no. 2983) and phospho-mTOR (Ser2448) (cat. no. 2971) were purchased from Cell Signaling Technology Inc. (Beverly, MA, USA). 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG) was purchased from Thermo Fisher Scientific (Eugene, OR, USA). All other reagents including DFX and MG132 were obtained from Sigma-Aldrich unless stated otherwise.

Cell lines and cell culture

Human colon cancer cell lines HCT116 and KM12C were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). HCT116 cells were cultured in McCoy'5A medium (Thermo Fisher Scientific, Hudson, NH, USA). KM12C cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Thermo Fisher Scientific). All the medium was supplemented with 10% fetal bovine serum (FBS; GE Healthcare Life Sciences, HyClone Laboratories, Logan, UT, USA), 100 µg/ml of streptomycin (Life Technologies; Thermo Fisher Scientific) and 100 units of penicillin.

MTT assay

Cell viability was detected using the 3-(4,5-dimethyl thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cells were seeded at a density of 1×104 cells/well in 96-well plates and then treated with 0–100 µM berberine as indicated. At the time-points (24 h for HCT116 cells and 15 h for KM12C cells), 50 µl of 5 mg/ml MTT solution was added to each well and incubated at 37°C for 4 h. The formazan crystals formed were then dissolved in 150 µl of dimethyl sulfoxide (DMSO), and the absorbance of the solution was then obtained on a microplate reader at λ570 nm. Results are presented as percentage loss of cell viability compared with the control.

Colony-forming assay

Cells were seeded in a 6-well plate one day before the experiment. Cells were treated with berberine for 24 h (HCT116) or 15 h (KM12C), and then washed with phosphate-buffered saline (PBS), harvested by trypsinization, counted, and were seeded into 6-well dishes at 600 cells/well. The cells were incubated for another 10 days, fixed and stained with 1% crystal violet in ethanol. Then, the colonies in 6-well plates were photographed using a scanner (Epson Perfection V330; Epson Corp., Nagano, Japan).

Glucose uptake assay

Glucose uptake of cells was measured by 2-NBDG uptake as previously described (11). Briefly, the cells were seeded in a 12-well plate at a density of 70–80%, and treated with 0–100 µM of berberine for 24 h (HCT116) or 15 h (KM12C). After treatment, the cells were harvested and resuspended in Krebs-Ringer's HEPES Buffer (KRB) solution, and incubated with 100 nmol/l 2-NBDG at 37°C in 5% CO2 for 30 min. The 2-NBDG uptake reaction was terminated by removing the incubation medium and washing the cells twice with pre-cold PBS. Cells were resuspended in 1 µg/ml propidium iodide (PI) solution to exclude dead cells. Glucose uptake was determined by measuring the fluorescence intensity of 2-NBDG in PI-negative cells.

Real-time quantitative PCR

Cells were seeded in a 6-well plate at a density of 70–80% and treated with 0–100 µM berberine for 24 h (HCT116) or 15 h (KM12C). Total RNA was isolated using TRIzol reagent (Takara Biotechnology Co., Ltd., Dalian, China). RNA was reverse-transcribed into cDNA using PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd.) according to the manufacturer's protocol. Real-time quantitative polymerase chain reaction (PCR) was carried out with the SYBR-Green I fluorescent dye method (SYBR® Premix Ex Taq™ II; Takara Biotechnology Co., Ltd.) and the StepOnePlus Real-Time PCR apparatus (Applied Biosystems; Thermo Fisher Scientific). The sequences of primers used were as follows: Forward, 5′-TATTGCACTGCACAGGCCACATTC-3′ and reverse, 5′-TGATGGGTGAGGAATGGGTTCACA-3′ for HIF-1α; forward, 5′-GGCATTGATGACTCCAGTGTT-3′ and reverse, 5′-ATGGAGCCCAGCAGCAA-3′ for GLUT1; forward, 5′-TCACGGAGCTCAACCATGAC-3′ and reverse, 5′-CTGCAGTAGGGTGAGTGGTG-3′ for HK2; forward, 5′-GCCCGACGTGCATTCCCGATTCCTT-3′ and reverse, 5′-GACGGCTTTCTCCCTCTTGCTGACG-3′ for LDHA; forward, 5′-CGTGTACTACAATGAGGCTGC-3′ and reverse, 5′-CTGGTCTGAAGATCTGGCCG-3′ for β-tubulin. The amplification specificity was checked by melting curve analysis. The relative expression of miRNA was calculated by the 2−ΔΔCt method as described by Livak and Schmittgen (12).

Western blotting

Cells at 60–80% confluence were washed with PBS and lysed directly into SDS-PAGE loading buffer. A total of 20 µg of protein was analyzed by SDS-PAGE and transferred to PVDF membranes. All primary antibodies were used at 1:1,000 in 5% milk in Tris-buffered saline with 0.05% Tween. Immunopositive bands were visualized by Amersham ECL™ Plus Western Blotting Detection kit (GE Healthcare, Chicago, IL, USA).

Statistical analysis

The SigmaPlot version 11.0 software package (Systat Software, Inc., San Jose, CA, USA) was used for statistical analysis. The results are presented as mean ± standard error (SEM). Data were analyzed by one way analysis of variance (ANOVA) or the Student's t-test. P<0.05 was considered to indicate a statistically significant result.

Results

Berberine inhibits glucose uptake and cell growth in colon cancer cells

Berberine has been shown to enhance glucose uptake in cell lines including 3T3 adipose cells (5,13) and L6 myotubes (7). To test its effect on colon cancer cells, two colon cancer cell lines, HCT116 and KM12C, were treated with 0, 6.25, 12.5, 25, 50 and 100 µM of berberine for 24 or 15 h respectively, and the glucose uptake of these cell lines was assessed. In contrast to berberine's reported effect of enhancing glucose update, we unexpectedly revealed that berberine inhibited glucose uptake in the colon cancer cell lines. As shown in Fig. 1A and B, treatment with the different concentrations of berberine significantly reduced glucose uptake in these two cell lines. At concentrations between 6.25 and 100 µM, berberine decreased glucose uptake of HCT116 by 40–90%, and by 10–75% in KM12C cells, which was slightly less sensitive. We then validated the anticancer effect of berberine by detecting cell proliferation after berberine treatment. MTT assay indicated that berberine decreased the viability of the HCT116 and KM12C cells in a concentration-dependent manner (Fig. 1C and D). To further investigate the long-term effect of berberine on cell growth, colony forming assay was performed. Similarly, berberine inhibited the growth of colon cancer cells, manifested as the reduction in the number of colonies of the HCT116 and KM12C cells in a concentration-dependent manner (Fig. 1E). These results indicated that berberine inhibits glucose uptake in colon cancer cells, and these effects may contribute to its antitumor effect.

Berberine inhibits the transcription of glucose metabolism-related gene in colon cancer cells

We then further investigated the mechanism by which berberine inhibits glucose uptake. Glucose transporter 1 (GLUT1) is the dominate glucose transport gene in colon cancer cells (14,15), which facilitates the transport of glucose across the plasma membranes. We examined the transcription of GLUT1 after berberine treatment. As shown in Fig. 2A and B, treatment with berberine inhibited the mRNA level of GLUT1 in a concentration-dependent manner. At concentrations between 6.25 and 25 µM, berberine decreased the mRNA level of GLUT1 in the HCT116 cells by 50–70%, and similar to the data of glucose uptake, KM12C cells were less sensitive. Berberine (6.25–25 µM) reduced the mRNA level of GLUT1 by 10–35% in the KM12C cells. These data indicated that the inhibitory effect of berberine on glucose uptake may be mediated by the transcription inhibition of GLUT1.

The altered energy metabolism of cancer cells is not only attained by enhancing glucose uptake, but also a higher rate of glycolysis. Thus, we examined the transcription of two glycolytic enzymes following berberine treatment: Lactate dehydrogenase A (LDHA) and hexokinases 2 (HK2). LDHA catalyzes the inter-conversion of pyruvate and L-lactate with concomitant inter-conversion of NADH and NAD+. HK2 phosphorylates glucose to produce glucose-6-phosphate (G6P), the first step in most glucose metabolism pathways. As shown in Fig. 2C-F, treatment with 0–100 µM berberine inhibited the mRNA levels of LDHA and HK2 in a concentration-dependent manner as assessed by qPCR. Thus, our data indicated that berberine may downregulate glucose metabolism in colon cancer cells by inhibition of the transcription of glucose metabolism-related genes.

Berberine inhibits HIF-1α expression at the post-transcriptional level

We next explored how berberine regulates the transcription of the above glucose metabolism-related genes. It is well known that the enhanced glucose uptake and glycolysis of cancer cells is partly due to highly expressed HIF-1α. HIF-1α is upregulated in low O2 concentrations but also by oncogene activation or loss of tumor suppressors (16). Upregulated HIF-1α would further facilitate the transcription of genes involved in glucose uptake and glycolysis-related genes, including GLUT1, LDHA and HK2 (17,18). Therefore, we next examined the effect of berberine on the mRNA and protein level of HIF-1α. As determined by qPCR, we found that treatment with berberine did not alter the mRNA level of HIF1A in the HCT116 and KM12C cells (Fig. 3A and B). Western blotting demonstrated that KM12C cells expressed HIF-1α protein even in normoxic condition. Treatment with 0–100 µM berberine decreased the protein level in a concentration-dependent manner (Fig. 3C). These data indicated that berberine negatively modulates glucose metabolism through regulation of HIF-1α at the post-transcriptional level.

Berberine suppresses HIF-1α protein synthesis by inhibition of the mTOR pathway

We then aimed to resolve whether berberine affects HIF-1α degradation. It is known that HIF-1α protein degradation is regulated by prolyl hydroxylation and then the ubiquitin-dependent proteasome pathway (19). Briefly, HIF-1α is hydroxylated by prolyl hydroxylases (PHD1-3) at proline (Pro)-402 and −564. Hydroxylated HIF-1α then binds to the von Hippel-Lindau tumor-suppressor protein (VHL). VHL is the recognition component of an E3 ubiquitin-protein and ubiquitylated HIF-1α is rapidly degraded by the proteasome (20). We first treated KM12C cells with desferrioxamine (DFX), an iron chelator, which is known to inhibit hydroxylation of HIF-1α by chelating the iron required for activity of the HIF-1α-specific proline hydroxylases, and thus blocks HIF-1α degradation (21). As shown in Fig. 4A, when HIF-1α degradation was blocked by DFX, berberine was still able to decrease the expression of HIF-1α. Similarly, treatment with proteasome inhibitor, MG132, was unable to inhibit berberine's effect on the reduction of HIF-1α expression (Fig. 4B). These results demonstrated that the negative post-transcriptional regulation of berberine on HIF-1α expression was not by proteasomal degradation.

We then aimed to ascertain whether berberine interrupts HIF-1α protein translation. Mammalian target of rapamycin (mTOR) activity is a major determinant of the rate of HIF-1α protein synthesis (22,23). We then detected the activity of the mTOR pathway in KM12C cells after the treatment of berberine. As shown in Fig. 4C, berberine significantly inhibited the phosphorylation of mTOR (p-mTOR) while not altering the total expression level of mTOR protein. Taken together, our data indicated that berberine inhibits HIF-1α protein expression by inhibition of the mTOR pathway, and thereby interruption of HIF-1α protein synthesis.

Discussion

In the present study, we demonstrated that berberine inhibits glucose uptake and cell growth in colon cancer cells. This effect is mediated by the inhibition of the mTOR pathway, which leads to the suppression of HIF-1α protein synthesis. The reduction in the HIF-1α expression level may attenuate the transcription activity of HIF-1α and decrease the transcription of glucose metabolism-related genes, GLUT1, LDHA and HK2.

The main mechanism by which berberine exerts antidiabetic effects relates to its glucose-lowering activity. It has been reported that berberine improves glucose uptake in glucose-consuming tissues, such as adipose, liver or muscle cells (5,6,13,24,25). In the present study, we revealed that berberine inhibited colon cancer cell glucose uptake and metabolism, which is in contrast from what has been reported in normal cells. Berberine has been considered as an anticancer drug with low toxicity to normal cells (2628). For example, Inoue et al reported that berberine showed higher cytotoxicity against five human oral squamous cell carcinomas (HSC-2, HSC-3, HSC-4, NA and CA9-22) and one human promyelocytic leukemia (HL-60) cell line, when compared with normal human oral tissue-derived cells (gingival fibroblasts, pulp cells and periodontal ligament fibroblasts) (26). Chidambara et al reported that berberine inhibited proliferation of colon cancer cells (SW480) but showed low toxicity to normal colon cells (CCD-CoN112) (29). The mechanisms for this tumor-specific selective toxicity of berberine remains elusive. Since tumor cells exhibit higher levels of glucose uptake and glycolytic metabolism as compared to normal cells owing to upregulated HIF-1α expression, the differential effect of berberine on glucose uptake between normal and cancer cells may attribute to the selectivity of berberine for cancer-targeted treatment.

HIF-1α was initially identified due to its response to low O2 concentrations, but it can also be regulated by oncogene activation or loss of tumor suppressors. Therefore, upregulation of HIF-1α is quite common in many human cancers. HIF-1α overexpression is associated with increased patient mortality in many different types of cancer (16). Inhibition of HIF-1α activity has marked effects on tumor growth and is considered as an anticancer therapeutic strategy (30,31). In the present study, we identified berberine as a novel HIF-1α inhibitor that could be used for colon cancer treatment. Berberine reduced HIF-1α expression in normoxia and also hypoxia conditions (DFX treatment is considered as a hypoxia mimic). Given the different expression of HIF-1α in tumor and normal cells, the effect of berberine on HIF-1α may account for its selective toxicity in cancer. Consistent with our research, there are several studies that have reported the inhibitory effect of berberine on HIF-1α expression in other types of cancers, such as esophageal squamous cancer (32), gastric adenocarcinoma cell line SC-M1 (33), nasopharyngeal carcinoma (34) and prostate cancer (35).

The mTOR kinase integrates and transmits signals from a diverse array of signaling pathways to regulate cell survival and growth. Activated mTOR stimulates protein synthesis and cell growth through phosphorylation of ribosomal protein S6 kinase (p70S6K), eukaryotic initiation factor 4E binding protein 1 (4E-BP1) and eukaryotic elongation factor 2 kinase (EEF2K) (36). The mTOR pathway consists of the downstream effectors of PI3K-AKT signaling (37). It is reported that berberine inhibits PI3K-AKT signaling in colorectal cancer cells and melanoma cells (38,39). Our finding that berberine suppresses mTOR activity may be due to its effects on the PI3K/AKT pathway. Between the two colon cancer cell lines that we tested, HCT116 cells were much more sensitive to berberine than KM12C cells. One of the differences between HCT116 and KM12C cells is that KM12C cells are PTEN-loss, while HCT116 cells are PTEN-competent. PTEN is an upstream inhibitory mediator to mTOR. The loss of PTEN in KM12C cells may lead to increased mTOR expression, which may affect the sensitivity to berberine (40).

In conclusion, the present study reveals a new role of berberine in the inhibition of tumor glucose uptake. The results not only suggest a possible mechanism involved in berberine's tumor selectivity but also disclose a promising therapeutic effect of berberine in colon cancer treatment.

Acknowledgements

Not applicable.

References

1 

Hsu PP and Sabatini DM: Cancer cell metabolism: Warburg and beyond. Cell. 134:703–707. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Denko NC: Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat Rev Cancer. 8:705–713. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Cicero AF and Tartagni E: Antidiabetic properties of berberine: From cellular pharmacology to clinical effects. Hosp Pract. 40:56–63. 2012. View Article : Google Scholar

4 

Pang B, Zhao LH, Zhou Q, Zhao TY, Wang H, Gu CJ and Tong XL: Application of berberine on treating type 2 diabetes mellitus. Int J Endocrinol. 15:9057492015.

5 

Cok A, Plaisier C, Salie MJ, Oram DS, Chenge J and Louters LL: Berberine acutely activates the glucose transport activity of GLUT1. Biochimie. 93:1187–1192. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Zhou L, Yang Y, Wang X, Liu S, Shang W, Yuan G, Li F, Tang J, Chen M and Chen J: Berberine stimulates glucose transport through a mechanism distinct from insulin. Metabolism. 56:405–412. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Cheng Z, Pang T, Gu M, Gao AH, Xie CM, Li JY, Nan FJ and Li J: Berberine-stimulated glucose uptake in L6 myotubes involves both AMPK and p38 MAPK. Biochim Biophys Acta. 1760:1682–1689. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Ortiz LM, Lombardi P, Tillhon M and Scovassi AI: Berberine, an epiphany against cancer. Molecules. 19:12349–12367. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Wang N, Tan HY, Li L, Yuen MF and Feng Y: Berberine and Coptidis Rhizoma as potential anticancer agents: Recent updates and future perspectives. J Ethnopharmacol. 176:35–48. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Tan W, Li Y, Chen M and Wang Y: Berberine hydrochloride: Anticancer activity and nanoparticulate delivery system. Int J Nanomedicine. 6:1773–1777. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Zou C, Wang Y and Shen Z: 2-NBDG as a fluorescent indicator for direct glucose uptake measurement. J Biochem Biophys Methods. 64:207–215. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Kim SH, Shin EJ, Kim ED, Bayaraa T, Frost SC and Hyun CK: Berberine activates GLUT1-mediated glucose uptake in 3T3-L1 adipocytes. Biol Pharm Bull. 30:2120–2125. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Haber RS, Rathan A, Weiser KR, Pritsker A, Itzkowitz SH, Bodian C, Slater G, Weiss A and Burstein DE: GLUT1 glucose transporter expression in colorectal carcinoma: A marker for poor prognosis. Cancer. 83:34–40. 1998. View Article : Google Scholar : PubMed/NCBI

15 

Saigusa S, Toiyama Y, Tanaka K, Okugawa Y, Fujikawa H, Matsushita K, Uchida K, Inoue Y and Kusunoki M: Prognostic significance of glucose transporter-1 (GLUT1) gene expression in rectal cancer after preoperative chemoradiotherapy. Surg Today. 42:460–469. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 3:721–732. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Koppenol WH, Bounds PL and Dang CV: Otto Warburg's contributions to current concepts of cancer metabolism. Nat Rev Cancer. 11:325–337. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Semenza GL: Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin Cancer Biol. 19:12–16. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Kallio PJ, Wilson WJ, O'Brien S, Makino Y and Poellinger L: Regulation of the hypoxia-inducible transcription factor 1alpha by the ubiquitin-proteasome pathway. J Biol Chem. 274:6519–6525. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Lee JW, Bae SH, Jeong JW, Kim SH and Kim KW: Hypoxia-inducible factor (HIF-1)alpha: Its protein stability and biological functions. Exp Mol Med. 36:1–12. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Demidenko ZN, Rapisarda A, Garayoa M, Giannakakou P, Melillo G and Blagosklonny MV: Accumulation of hypoxia-inducible factor-1alpha is limited by transcription-dependent depletion. Oncogene. 24:4829–4838. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Abraham RT: mTOR as a positive regulator of tumor cell responses to hypoxia. Curr Top Microbiol Immunol. 279:299–319. 2004.PubMed/NCBI

23 

Masoud GN and Li W: HIF-1α pathway: Role, regulation and intervention for cancer therapy. Acta Pharm Sin B. 5:378–389. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Yi P, Lu FE, Xu LJ, Chen G, Dong H and Wang KF: Berberine reverses free-fatty-acid-induced insulin resistance in 3T3-L1 adipocytes through targeting IKKbeta. World J Gastroenterol. 14:876–883. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Zhang CH, Yu RY, Liu YH, Tu XY, Tu J, Wang YS and Xu GL: Interaction of baicalin with berberine for glucose uptake in 3T3-L1 adipocytes and HepG2 hepatocytes. J Ethnopharmaco. 151:864–872. 2014. View Article : Google Scholar

26 

Inoue K, Kulsum U, Chowdhury SA, Fujisawa S, Ishihara M, Yokoe I and Sakagami H: Tumor-specific cytotoxicity and apoptosis-inducing activity of berberines. Anticancer Res. 25:4053–4059. 2005.PubMed/NCBI

27 

Liu B, Wang G, Yang J, Pan X, Yang Z and Zang L: Berberine inhibits human hepatoma cell invasion without cytotoxicity in healthy hepatocytes. PLoS One. 6:e214162011. View Article : Google Scholar : PubMed/NCBI

28 

Wang L, Liu L, Shi Y, Cao H, Chaturvedi R, Calcutt MW, Hu T, Ren X, Wilson KT, Polk DB and Yan F: Berberine induces caspase-independent cell death in colon tumor cells through activation of apoptosis-inducing factor. PLoS One. 7:e364182012. View Article : Google Scholar : PubMed/NCBI

29 

Murthy Chidambara KN, Jayaprakasha GK and Patil BS: The natural alkaloid berberine targets multiple pathways to induce cell death in cultured human colon cancer cells. Eur J Pharmacol. 688:14–21. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Yu T, Tang B and Sun X: Development of Inhibitors Targeting hypoxia-inducible factor 1 and 2 for cancer therapy. Yonsei Med J. 58:489–496. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Melillo G: Targeting hypoxia cell signaling for cancer therapy. Cancer Metastasis Rev. 26:341–352. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Yang X, Yang B, Cai J, Zhang C, Zhang Q, Xu L, Qin Q, Zhu H, Ma J, Tao G, et al: Berberine enhances radiosensitivity of esophageal squamous cancer by targeting HIF-1α in vitro and in vivo. Cancer Biol Ther. 14:1068–1073. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Lin S, Tsai SC, Lee CC, Wang BW, Liou JY and Shyu KG: Berberine inhibits HIF-1 alpha expression via enhanced proteolysis. Mol Pharmacol. 66:612–619. 2004.PubMed/NCBI

34 

Zhang C, Yang X, Zhang Q, Yang B, Xu L, Qin Q, Zhu H, Liu J, Cai J, Tao G, et al: Berberine radiosensitizes human nasopharyngeal carcinoma by suppressing hypoxia-inducible factor-1α expression. Acta Otolaryngol. 134:185–192. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Zhang Q, Zhang C, Yang X, Yang B, Wang J, Kang Y, Wang Z, Li D, Huang G, Ma Z, et al: Berberine inhibits the expression of hypoxia induction factor-1alpha and increases the radiosensitivity of prostate cancer. Diagn Pathol. 9:982014. View Article : Google Scholar : PubMed/NCBI

36 

Dazert E and Hall MN: mTOR signaling in disease. Curr Opin Cell Biol. 23:744–755. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Engelman JA: Targeting PI3K signalling in cancer: Opportunities, challenges and limitations. Nat Rev Cancer. 9:550–562. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Chen ZZ: Berberine induced apoptosis of human osteosarcoma cells by inhibiting phosphoinositide 3 kinase/protein kinase B (PI3K/Akt) signal pathway activation. Iran J Public Health. 45:578–585. 2016.PubMed/NCBI

39 

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

40 

Milella M, Falcone I, Conciatori F, Matteoni S, Sacconi A, De Luca T, Bazzichetto C, Corbo V, Simbolo M, Sperduti I, et al: PTEN status is a crucial determinant of the functional outcome of combined MEK and mTOR inhibition in cancer. Sci Rep. 7:430132017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 39 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
Mao L, Chen Q, Gong K, Xu X, Xie Y, Zhang W, Cao H, Hu T, Hong X, Zhan YY, Zhan YY, et al: Berberine decelerates glucose metabolism via suppression of mTOR‑dependent HIF‑1α protein synthesis in colon cancer cells. Oncol Rep 39: 2436-2442, 2018
APA
Mao, L., Chen, Q., Gong, K., Xu, X., Xie, Y., Zhang, W. ... Zhan, Y. (2018). Berberine decelerates glucose metabolism via suppression of mTOR‑dependent HIF‑1α protein synthesis in colon cancer cells. Oncology Reports, 39, 2436-2442. https://doi.org/10.3892/or.2018.6318
MLA
Mao, L., Chen, Q., Gong, K., Xu, X., Xie, Y., Zhang, W., Cao, H., Hu, T., Hong, X., Zhan, Y."Berberine decelerates glucose metabolism via suppression of mTOR‑dependent HIF‑1α protein synthesis in colon cancer cells". Oncology Reports 39.5 (2018): 2436-2442.
Chicago
Mao, L., Chen, Q., Gong, K., Xu, X., Xie, Y., Zhang, W., Cao, H., Hu, T., Hong, X., Zhan, Y."Berberine decelerates glucose metabolism via suppression of mTOR‑dependent HIF‑1α protein synthesis in colon cancer cells". Oncology Reports 39, no. 5 (2018): 2436-2442. https://doi.org/10.3892/or.2018.6318