Skip to main content
Top
Published in: Tumor Biology 2/2016

01-02-2016 | Original Article

Fangchinoline suppresses the growth and invasion of human glioblastoma cells by inhibiting the kinase activity of Akt and Akt-mediated signaling cascades

Authors: Bingyu Guo, Peng Xie, Jingyuan Su, Tingting Zhang, Xiaoming Li, Guobiao Liang

Published in: Tumor Biology | Issue 2/2016

Login to get access

Abstract

Glioblastoma multiforme (GBM) is one of the most palindromic and malignant central nervous system neoplasms, and the current treatment is not effectual for GBM. Research of specific medicine for GBM is significant. Fangchinoline possesses a wide range of pharmacological activities and attracts more attentions due to its anti-tumor effects. In this study, two WHO grade IV human GBM cell lines (U87 MG and U118 MG) were exposed to fangchinoline, and we found that fangchinoline specifically inhibits the kinase activity of Akt and markedly suppresses the phosphorylation of Thr308 and Ser473 of Akt in human GBM cells. We also observed that fangchinoline inhibits tumor cell proliferation and invasiveness and induces apoptosis through suppressing the Akt-mediated signaling cascades, including Akt/p21, Akt/Bad, and Akt/matrix metalloproteinases (MMPs). These data demonstrated that fangchinoline exerts its anti-tumor effects in human glioblastoma cells, at least partly by inhibiting the kinase activity of Akt and suppressing Akt-mediated signaling cascades.
Literature
1.
go back to reference Ahmed KA, Chinnaiyan P. Applying metabolomics to understand the aggressive phenotype and identify novel therapeutic targets in glioblastoma. Metabolites. 2014;4:740–50.CrossRefPubMedPubMedCentral Ahmed KA, Chinnaiyan P. Applying metabolomics to understand the aggressive phenotype and identify novel therapeutic targets in glioblastoma. Metabolites. 2014;4:740–50.CrossRefPubMedPubMedCentral
2.
go back to reference Agrawal NS, Miller R, Jr., Lal R, Mahanti H, Dixon-Mah YN, DeCandio ML, et al. Current studies of immunotherapy on glioblastoma. J Neurol Neurosurg. 2014;5:1(1). Agrawal NS, Miller R, Jr., Lal R, Mahanti H, Dixon-Mah YN, DeCandio ML, et al. Current studies of immunotherapy on glioblastoma. J Neurol Neurosurg. 2014;5:1(1).
3.
go back to reference Kazlauskas A, Cooper JA. Phosphorylation of the PDGF receptor beta subunit creates a tight binding site for phosphatidylinositol 3 kinase. EMBO J. 1990;9:3279–86.PubMedPubMedCentral Kazlauskas A, Cooper JA. Phosphorylation of the PDGF receptor beta subunit creates a tight binding site for phosphatidylinositol 3 kinase. EMBO J. 1990;9:3279–86.PubMedPubMedCentral
4.
go back to reference Chiang EP, Tsai SY, Kuo YH, Pai MH, Chiu HL, Rodriguez RL, et al. Caffeic acid derivatives inhibit the growth of colon cancer: involvement of the PI3-K/Akt and AMPK signaling pathways. PloS one. 2014;9:e99631.CrossRefPubMedPubMedCentral Chiang EP, Tsai SY, Kuo YH, Pai MH, Chiu HL, Rodriguez RL, et al. Caffeic acid derivatives inhibit the growth of colon cancer: involvement of the PI3-K/Akt and AMPK signaling pathways. PloS one. 2014;9:e99631.CrossRefPubMedPubMedCentral
5.
go back to reference Gustin JP, Karakas B, Weiss MB, Abukhdeir AM, Lauring J, Garay JP, et al. Knockin of mutant pik3ca activates multiple oncogenic pathways. Proc Natl Acad Sci U S A. 2009;106:2835–40.CrossRefPubMedPubMedCentral Gustin JP, Karakas B, Weiss MB, Abukhdeir AM, Lauring J, Garay JP, et al. Knockin of mutant pik3ca activates multiple oncogenic pathways. Proc Natl Acad Sci U S A. 2009;106:2835–40.CrossRefPubMedPubMedCentral
6.
go back to reference Kang S, Bader AG, Vogt PK. Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc Natl Acad Sci U S A. 2005;102:802–7.CrossRefPubMedPubMedCentral Kang S, Bader AG, Vogt PK. Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc Natl Acad Sci U S A. 2005;102:802–7.CrossRefPubMedPubMedCentral
7.
go back to reference Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, et al. High frequency of mutations of the pik3ca gene in human cancers. Science. 2004;304:554.CrossRefPubMed Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, et al. High frequency of mutations of the pik3ca gene in human cancers. Science. 2004;304:554.CrossRefPubMed
8.
go back to reference Cully M, You H, Levine AJ, Mak TW. Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat Rev Cancer. 2006;6:184–92.CrossRefPubMed Cully M, You H, Levine AJ, Mak TW. Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat Rev Cancer. 2006;6:184–92.CrossRefPubMed
10.
go back to reference McDowell KA, Riggins GJ, Gallia GL. Targeting the Akt pathway in glioblastoma. Curr Pharm Des. 2011;17:2411–20.CrossRefPubMed McDowell KA, Riggins GJ, Gallia GL. Targeting the Akt pathway in glioblastoma. Curr Pharm Des. 2011;17:2411–20.CrossRefPubMed
11.
go back to reference Suzuki Y, Shirai K, Oka K, Mobaraki A, Yoshida Y, Noda SE, et al. Higher pAkt expression predicts a significant worse prognosis in glioblastomas. J Radiat Res. 2010;51:343–8.CrossRefPubMed Suzuki Y, Shirai K, Oka K, Mobaraki A, Yoshida Y, Noda SE, et al. Higher pAkt expression predicts a significant worse prognosis in glioblastomas. J Radiat Res. 2010;51:343–8.CrossRefPubMed
12.
13.
go back to reference Wang Y, Chen J, Wang L, Huang Y, Leng Y, Wang G. Fangchinoline induces G0/G1 arrest by modulating the expression of CDKN1A and CCND2 in K562 human chronic myelogenous leukemia cells. Exp Ther Med. 2013;5:1105–12.PubMedPubMedCentral Wang Y, Chen J, Wang L, Huang Y, Leng Y, Wang G. Fangchinoline induces G0/G1 arrest by modulating the expression of CDKN1A and CCND2 in K562 human chronic myelogenous leukemia cells. Exp Ther Med. 2013;5:1105–12.PubMedPubMedCentral
14.
go back to reference Wang CD, Yuan CF, Bu YQ, Wu XM, Wan JY, Zhang L, et al. Fangchinoline inhibits cell proliferation via Akt/gsk-3beta/cyclin d1 signaling and induces apoptosis in MDA-MB-231 breast cancer cells. Asian Pac J Cancer Prev. 2014;15:769–73.CrossRefPubMed Wang CD, Yuan CF, Bu YQ, Wu XM, Wan JY, Zhang L, et al. Fangchinoline inhibits cell proliferation via Akt/gsk-3beta/cyclin d1 signaling and induces apoptosis in MDA-MB-231 breast cancer cells. Asian Pac J Cancer Prev. 2014;15:769–73.CrossRefPubMed
15.
go back to reference Wang N, Pan W, Zhu M, Zhang M, Hao X, Liang G, et al. Fangchinoline induces autophagic cell death via p53/sestrin2/AMPK signalling in human hepatocellular carcinoma cells. Br J Pharmacol. 2011;164:731–42.CrossRefPubMedPubMedCentral Wang N, Pan W, Zhu M, Zhang M, Hao X, Liang G, et al. Fangchinoline induces autophagic cell death via p53/sestrin2/AMPK signalling in human hepatocellular carcinoma cells. Br J Pharmacol. 2011;164:731–42.CrossRefPubMedPubMedCentral
16.
go back to reference Wang CD, Huang JG, Gao X, Li Y, Zhou SY, Yan X, et al. Fangchinoline induced G1/S arrest by modulating expression of p27, PCNA, and cyclin D in human prostate carcinoma cancer PC3 cells and tumor xenograft. Biosci Biotechnol Biochem. 2010;74:488–93.CrossRefPubMed Wang CD, Huang JG, Gao X, Li Y, Zhou SY, Yan X, et al. Fangchinoline induced G1/S arrest by modulating expression of p27, PCNA, and cyclin D in human prostate carcinoma cancer PC3 cells and tumor xenograft. Biosci Biotechnol Biochem. 2010;74:488–93.CrossRefPubMed
17.
go back to reference Guo B, Su J, Zhang T, Wang K, Li X. Fangchinoline as a kinase inhibitor targets FAK and suppresses FAK-mediated signaling pathway in A549. J Drug Target. 2015;23:266–74.CrossRefPubMed Guo B, Su J, Zhang T, Wang K, Li X. Fangchinoline as a kinase inhibitor targets FAK and suppresses FAK-mediated signaling pathway in A549. J Drug Target. 2015;23:266–74.CrossRefPubMed
18.
go back to reference Zhang YH, Fang LH, Ku BS. Fangchinoline inhibits rat aortic vascular smooth muscle cell proliferation and cell cycle progression through inhibition of ERK1/2 activation and c-fos expression. Biochem Pharmacol. 2003;66:1853–60.CrossRefPubMed Zhang YH, Fang LH, Ku BS. Fangchinoline inhibits rat aortic vascular smooth muscle cell proliferation and cell cycle progression through inhibition of ERK1/2 activation and c-fos expression. Biochem Pharmacol. 2003;66:1853–60.CrossRefPubMed
19.
go back to reference Meng LH, Zhang H, Hayward L, Takemura H, Shao RG, Pommier Y. Tetrandrine induces early G1 arrest in human colon carcinoma cells by down-regulating the activity and inducing the degradation of G1-S-specific cyclin-dependent kinases and by inducing p53 and p21Cip1. Cancer Res. 2004;64:9086–92.CrossRefPubMed Meng LH, Zhang H, Hayward L, Takemura H, Shao RG, Pommier Y. Tetrandrine induces early G1 arrest in human colon carcinoma cells by down-regulating the activity and inducing the degradation of G1-S-specific cyclin-dependent kinases and by inducing p53 and p21Cip1. Cancer Res. 2004;64:9086–92.CrossRefPubMed
20.
go back to reference Sun X, Xu R, Deng Y, Cheng H, Ma J, Ji J, et al. Effects of tetrandrine on apoptosis and radiosensitivity of nasopharyngeal carcinoma cell line CNE. Acta Biochim Biophys Sin. 2007;39:869–78.CrossRefPubMed Sun X, Xu R, Deng Y, Cheng H, Ma J, Ji J, et al. Effects of tetrandrine on apoptosis and radiosensitivity of nasopharyngeal carcinoma cell line CNE. Acta Biochim Biophys Sin. 2007;39:869–78.CrossRefPubMed
21.
go back to reference Xing Z, Zhang Y, Zhang X, Yang Y, Ma Y, Pang D. Fangchinoline induces G1 arrest in breast cancer cells through cell-cycle regulation. Phytother Res. 2013;27:1790–4.CrossRefPubMed Xing Z, Zhang Y, Zhang X, Yang Y, Ma Y, Pang D. Fangchinoline induces G1 arrest in breast cancer cells through cell-cycle regulation. Phytother Res. 2013;27:1790–4.CrossRefPubMed
22.
go back to reference Xing ZB, Yao L, Zhang GQ, Zhang XY, Zhang YX, Pang D. Fangchinoline inhibits breast adenocarcinoma proliferation by inducing apoptosis. Chem Pharm Bull. 2011;59:1476–80.CrossRefPubMed Xing ZB, Yao L, Zhang GQ, Zhang XY, Zhang YX, Pang D. Fangchinoline inhibits breast adenocarcinoma proliferation by inducing apoptosis. Chem Pharm Bull. 2011;59:1476–80.CrossRefPubMed
23.
go back to reference Baki A, Bielik A, Molnar L, Szendrei G, Keseru GM. A high throughput luminescent assay for glycogen synthase kinase-3beta inhibitors. Assay Drug Dev Technol. 2007;5:75–83.CrossRefPubMed Baki A, Bielik A, Molnar L, Szendrei G, Keseru GM. A high throughput luminescent assay for glycogen synthase kinase-3beta inhibitors. Assay Drug Dev Technol. 2007;5:75–83.CrossRefPubMed
24.
25.
go back to reference Tian F, Ding D, Li D. Fangchinoline targets PI3K and suppresses PI3K/Akt signaling pathway in SGC7901 cells. Int J Oncol. 2015;46:2355–63.PubMedPubMedCentral Tian F, Ding D, Li D. Fangchinoline targets PI3K and suppresses PI3K/Akt signaling pathway in SGC7901 cells. Int J Oncol. 2015;46:2355–63.PubMedPubMedCentral
26.
go back to reference Chautard E, Ouedraogo ZG, Biau J, Verrelle P. Role of Akt in human malignant glioma: from oncogenesis to tumor aggressiveness. J Neurooncol. 2014;117:205–15.CrossRefPubMed Chautard E, Ouedraogo ZG, Biau J, Verrelle P. Role of Akt in human malignant glioma: from oncogenesis to tumor aggressiveness. J Neurooncol. 2014;117:205–15.CrossRefPubMed
27.
go back to reference Sonoda Y, Ozawa T, Aldape KD, Deen DF, Berger MS, Pieper RO. Akt pathway activation converts anaplastic astrocytoma to glioblastoma multiforme in a human astrocyte model of glioma. Cancer Res. 2001;61:6674–8.PubMed Sonoda Y, Ozawa T, Aldape KD, Deen DF, Berger MS, Pieper RO. Akt pathway activation converts anaplastic astrocytoma to glioblastoma multiforme in a human astrocyte model of glioma. Cancer Res. 2001;61:6674–8.PubMed
28.
go back to reference Nicholson KM, Anderson NG. The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal. 2002;14:381–95.CrossRefPubMed Nicholson KM, Anderson NG. The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal. 2002;14:381–95.CrossRefPubMed
Metadata
Title
Fangchinoline suppresses the growth and invasion of human glioblastoma cells by inhibiting the kinase activity of Akt and Akt-mediated signaling cascades
Authors
Bingyu Guo
Peng Xie
Jingyuan Su
Tingting Zhang
Xiaoming Li
Guobiao Liang
Publication date
01-02-2016
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 2/2016
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-015-3990-1

Other articles of this Issue 2/2016

Tumor Biology 2/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine