Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Colorectal Cancer | Research

Targeting AKT with costunolide suppresses the growth of colorectal cancer cells and induces apoptosis in vitro and in vivo

Authors: Hai Huang, Song Park, Haibo Zhang, Sijun Park, Wookbong Kwon, Enugyung Kim, Xiujuan Zhang, Soyoung Jang, Duhak Yoon, Seong-Kyoon Choi, Jun-koo Yi, Sung-hyun Kim, Zigang Dong, Mee-hyun Lee, Zaeyoung Ryoo, Myoung Ok Kim

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Colorectal cancer (CRC) is a clinically challenging malignant tumor worldwide. As a natural product and sesquiterpene lactone, Costunolide (CTD) has been reported to possess anticancer activities. However, the regulation mechanism and precise target of this substance remain undiscovered in CRC. In this study, we found that CTD inhibited CRC cell proliferation in vitro and in vivo by targeting AKT.

Methods

Effects of CTD on colon cancer cell growth in vitro were evaluated in cell proliferation assays, migration and invasion, propidium iodide, and annexin V-staining analyses. Targets of CTD were identified utilizing phosphoprotein-specific antibody array; Costunolide-sepharose conjugated bead pull-down analysis and knockdown techniques. We investigated the underlying mechanisms of CTD by ubiquitination, immunofluorescence staining, and western blot assays. Cell-derived tumour xenografts (CDX) in nude mice and immunohistochemistry were used to assess anti-tumour effects of CTD in vivo.

Results

CTD suppressed the proliferation, anchorage-independent colony growth and epithelial-mesenchymal transformation (EMT) of CRC cells including HCT-15, HCT-116 and DLD1. Besides, the CTD also triggered cell apoptosis and cell cycle arrest at the G2/M phase. The CTD activates and induces p53 stability by inhibiting MDM2 ubiquitination via the suppression of AKT’s phosphorylation in vitro. The CTD suppresses cell growth in a p53-independent fashion manner; p53 activation may contribute to the anticancer activity of CTD via target AKT. Finally, the CTD decreased the volume of CDX tumors without of the body weight loss and reduced the expression of AKT-MDM2-p53 signaling pathway in xenograft tumors.

Conclusions

Our project has uncovered the mechanism underlying the biological activity of CTD in colon cancer and confirmed the AKT is a directly target of CTD. All of which These results revealed that CTD might be a new AKT inhibitor in colon cancer treatment, and CTD is worthy of further exploration in preclinical and clinical trials.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Schmoll HJ, Tabernero J, Maroun J, de Braud F, Price T, Van Cutsem E, Hill M, Hoersch S, Rittweger K, Haller DG. Capecitabine plus oxaliplatin compared with fluorouracil/folinic acid as adjuvant therapy for stage III colon cancer: final results of the NO16968 randomized controlled phase III trial. J Clin Oncol. 2015;33(32):3733–+.PubMedCrossRef Schmoll HJ, Tabernero J, Maroun J, de Braud F, Price T, Van Cutsem E, Hill M, Hoersch S, Rittweger K, Haller DG. Capecitabine plus oxaliplatin compared with fluorouracil/folinic acid as adjuvant therapy for stage III colon cancer: final results of the NO16968 randomized controlled phase III trial. J Clin Oncol. 2015;33(32):3733–+.PubMedCrossRef
3.
go back to reference Sasaki T, Kuniyasu H, Luo Y, Kitayoshi M, Tanabe E, Kato D, Shinya S, Fujii K, Ohmori H, Yamashita Y. Increased phosphorylation of AKT in high-risk gastric mucosa. Anticancer Res. 2013;33(8):3295–300.PubMed Sasaki T, Kuniyasu H, Luo Y, Kitayoshi M, Tanabe E, Kato D, Shinya S, Fujii K, Ohmori H, Yamashita Y. Increased phosphorylation of AKT in high-risk gastric mucosa. Anticancer Res. 2013;33(8):3295–300.PubMed
4.
go back to reference Liu X, Song M, Wang P, Zhao R, Chen H, Zhang M, Shi Y, Liu K, Liu F, Yang R, et al. Targeted therapy of the AKT kinase inhibits esophageal squamous cell carcinoma growth in vitro and in vivo. Int J Cancer. 2019;145(4):1007–19.PubMedPubMedCentralCrossRef Liu X, Song M, Wang P, Zhao R, Chen H, Zhang M, Shi Y, Liu K, Liu F, Yang R, et al. Targeted therapy of the AKT kinase inhibits esophageal squamous cell carcinoma growth in vitro and in vivo. Int J Cancer. 2019;145(4):1007–19.PubMedPubMedCentralCrossRef
5.
go back to reference Zhu YP, Dai B, Zhang HL, Shi GH, Shen YJ, Ye DW. Long non-coding RNA LOC572558 inhibits bladder cancer cell proliferation and tumor growth by regulating the AKT-MDM2-p53 signaling axis. Cancer Lett. 2016;380(2):369–74.PubMedCrossRef Zhu YP, Dai B, Zhang HL, Shi GH, Shen YJ, Ye DW. Long non-coding RNA LOC572558 inhibits bladder cancer cell proliferation and tumor growth by regulating the AKT-MDM2-p53 signaling axis. Cancer Lett. 2016;380(2):369–74.PubMedCrossRef
6.
go back to reference Tu YY, Kim E, Gao Y, Rankin GO, Li B, Chen YC. Theaflavin-3, 3′-digallate induces apoptosis and G2 cell cycle arrest through the Akt/MDM2/p53 pathway in cisplatin-resistant ovarian cancer A2780/CP70 cells. Int J Oncol. 2016;48(6):2657–65.PubMedPubMedCentralCrossRef Tu YY, Kim E, Gao Y, Rankin GO, Li B, Chen YC. Theaflavin-3, 3′-digallate induces apoptosis and G2 cell cycle arrest through the Akt/MDM2/p53 pathway in cisplatin-resistant ovarian cancer A2780/CP70 cells. Int J Oncol. 2016;48(6):2657–65.PubMedPubMedCentralCrossRef
7.
go back to reference Choi HJ, Chung TW, Kang SK, Lee YC, Ko JH, Kim JG, Kim CH. Ganglioside GM3 modulates tumor suppressor PTEN-mediated cell cycle progression--transcriptional induction of p21(WAF1) and p27(kip1) by inhibition of PI-3K/AKT pathway. Glycobiology. 2006;16(7):573–83.PubMedCrossRef Choi HJ, Chung TW, Kang SK, Lee YC, Ko JH, Kim JG, Kim CH. Ganglioside GM3 modulates tumor suppressor PTEN-mediated cell cycle progression--transcriptional induction of p21(WAF1) and p27(kip1) by inhibition of PI-3K/AKT pathway. Glycobiology. 2006;16(7):573–83.PubMedCrossRef
8.
go back to reference Jabbarzadeh Kaboli P, Salimian F, Aghapour S, Xiang S, Zhao Q, Li M, Wu X, Du F, Zhao Y, Shen J, et al. Akt-targeted therapy as a promising strategy to overcome drug resistance in breast cancer - a comprehensive review from chemotherapy to immunotherapy. Pharmacol Res. 2020;156:104806.PubMedCrossRef Jabbarzadeh Kaboli P, Salimian F, Aghapour S, Xiang S, Zhao Q, Li M, Wu X, Du F, Zhao Y, Shen J, et al. Akt-targeted therapy as a promising strategy to overcome drug resistance in breast cancer - a comprehensive review from chemotherapy to immunotherapy. Pharmacol Res. 2020;156:104806.PubMedCrossRef
9.
go back to reference Ray RM, Bhattacharya S, Johnson LR. Mdm2 inhibition induces apoptosis in p53 deficient human colon cancer cells by activating p73-and E2F1-mediated expression of PUMA and Siva-1. Apoptosis. 2011;16(1):35–44.PubMedCrossRef Ray RM, Bhattacharya S, Johnson LR. Mdm2 inhibition induces apoptosis in p53 deficient human colon cancer cells by activating p73-and E2F1-mediated expression of PUMA and Siva-1. Apoptosis. 2011;16(1):35–44.PubMedCrossRef
10.
go back to reference Abraham AG, O'Neill E. PI3K/Akt-mediated regulation of p53 in cancer. Biochem Soc Trans. 2014;42:798–803.PubMedCrossRef Abraham AG, O'Neill E. PI3K/Akt-mediated regulation of p53 in cancer. Biochem Soc Trans. 2014;42:798–803.PubMedCrossRef
11.
go back to reference Ogawara Y, Kishishita S, Obata T, Isazawa Y, Suzuki T, Tanaka K, Masuyama N, Gotoh Y. Akt enhances Mdm2-mediated ubiquitination and degradation of p53. J Biol Chem. 2002;277(24):21843–50.PubMedCrossRef Ogawara Y, Kishishita S, Obata T, Isazawa Y, Suzuki T, Tanaka K, Masuyama N, Gotoh Y. Akt enhances Mdm2-mediated ubiquitination and degradation of p53. J Biol Chem. 2002;277(24):21843–50.PubMedCrossRef
12.
go back to reference Matsuda S, Nakagawa Y, Kitagishi Y, Nakanishi A, Murai T. Reactive oxygen species, superoxide dimutases, and PTEN-p53-AKT-MDM2 signaling loop network in mesenchymal stem/stromal cells regulation. Cells. 2018;7(5):36.PubMedCentralCrossRef Matsuda S, Nakagawa Y, Kitagishi Y, Nakanishi A, Murai T. Reactive oxygen species, superoxide dimutases, and PTEN-p53-AKT-MDM2 signaling loop network in mesenchymal stem/stromal cells regulation. Cells. 2018;7(5):36.PubMedCentralCrossRef
13.
go back to reference Drakos E, Atsaves V, Li J, Leventaki V, Andreeff M, Medeiros LJ, Rassidakis GZ. Stabilization and activation of p53 downregulates mTOR signaling through AMPK in mantle cell lymphoma. Leukemia. 2009;23(4):784–90.PubMedCrossRef Drakos E, Atsaves V, Li J, Leventaki V, Andreeff M, Medeiros LJ, Rassidakis GZ. Stabilization and activation of p53 downregulates mTOR signaling through AMPK in mantle cell lymphoma. Leukemia. 2009;23(4):784–90.PubMedCrossRef
14.
go back to reference Mayo LD, Donner DB. The PTEN, Mdm2, p53 tumor suppressor-oncoprotein network. Trends Biochem Sci. 2002;27(9):462–7.PubMedCrossRef Mayo LD, Donner DB. The PTEN, Mdm2, p53 tumor suppressor-oncoprotein network. Trends Biochem Sci. 2002;27(9):462–7.PubMedCrossRef
15.
go back to reference Sun WC, Tang LL. MDM2 increases drug resistance in cancer cells by inducing EMT independent of p53. Curr Med Chem. 2016;23(40):4529–39.PubMedCrossRef Sun WC, Tang LL. MDM2 increases drug resistance in cancer cells by inducing EMT independent of p53. Curr Med Chem. 2016;23(40):4529–39.PubMedCrossRef
16.
go back to reference Harris SL, Levine AJ. The p53 pathway: positive and negative feedback loops. Oncogene. 2005;24(17):2899–908.PubMedCrossRef Harris SL, Levine AJ. The p53 pathway: positive and negative feedback loops. Oncogene. 2005;24(17):2899–908.PubMedCrossRef
17.
go back to reference Astle MV, Hannan KM, Ng PY, Lee RS, George AJ, Hsu AK, Haupt Y, Hannan RD, Pearson RB. AKT induces senescence in human cells via mTORC1 and p53 in the absence of DNA damage: implications for targeting mTOR during malignancy. Oncogene. 2012;31(15):1949–62.PubMedCrossRef Astle MV, Hannan KM, Ng PY, Lee RS, George AJ, Hsu AK, Haupt Y, Hannan RD, Pearson RB. AKT induces senescence in human cells via mTORC1 and p53 in the absence of DNA damage: implications for targeting mTOR during malignancy. Oncogene. 2012;31(15):1949–62.PubMedCrossRef
18.
go back to reference Cao Z, Xue J, Cheng Y, Wang J, Liu Y, Li H, Jiang W, Li G, Gui Y, Zhang X. MDM2 promotes genome instability by ubiquitinating the transcription factor HBP1. Oncogene. 2019;38(24):4835–55.PubMedPubMedCentralCrossRef Cao Z, Xue J, Cheng Y, Wang J, Liu Y, Li H, Jiang W, Li G, Gui Y, Zhang X. MDM2 promotes genome instability by ubiquitinating the transcription factor HBP1. Oncogene. 2019;38(24):4835–55.PubMedPubMedCentralCrossRef
19.
go back to reference Wen W, Peng C, Kim MO, Jeong CH, Zhu F, Yao K, Zykova T, Ma W, Carper A, Langfald A, et al. Knockdown of RNF2 induces apoptosis by regulating MDM2 and p53 stability. Oncogene. 2014;33(4):421–8.PubMedCrossRef Wen W, Peng C, Kim MO, Jeong CH, Zhu F, Yao K, Zykova T, Ma W, Carper A, Langfald A, et al. Knockdown of RNF2 induces apoptosis by regulating MDM2 and p53 stability. Oncogene. 2014;33(4):421–8.PubMedCrossRef
20.
go back to reference Wang SM, Zhao YJ, Aguilar A, Bernard D, Yang CY. Targeting the MDM2-p53 protein-protein interaction for new cancer therapy: progress and challenges. Cold Spring Harb Perspect Med. 2017;7(5):a026245.PubMedPubMedCentralCrossRef Wang SM, Zhao YJ, Aguilar A, Bernard D, Yang CY. Targeting the MDM2-p53 protein-protein interaction for new cancer therapy: progress and challenges. Cold Spring Harb Perspect Med. 2017;7(5):a026245.PubMedPubMedCentralCrossRef
21.
go back to reference He SL, Wang WP, Yang YS, Li EM, Xu LY, Chen LQ. FAM3B promotes progression of oesophageal carcinoma via regulating the AKT-MDM2-p53 signalling axis and the epithelial-mesenchymal transition. J Cell Mol Med. 2019;23(2):1375–85.PubMedCrossRef He SL, Wang WP, Yang YS, Li EM, Xu LY, Chen LQ. FAM3B promotes progression of oesophageal carcinoma via regulating the AKT-MDM2-p53 signalling axis and the epithelial-mesenchymal transition. J Cell Mol Med. 2019;23(2):1375–85.PubMedCrossRef
22.
go back to reference Fenouille N, Puissant A, Tichet M, Zimniak G, Abbe P, Mallavialle A, Rocchi S, Ortonne JP, Deckert M, Ballotti R, et al. SPARC functions as an anti-stress factor by inactivating p53 through Akt-mediated MDM2 phosphorylation to promote melanoma cell survival. Oncogene. 2011;30(49):4887–900.PubMedCrossRef Fenouille N, Puissant A, Tichet M, Zimniak G, Abbe P, Mallavialle A, Rocchi S, Ortonne JP, Deckert M, Ballotti R, et al. SPARC functions as an anti-stress factor by inactivating p53 through Akt-mediated MDM2 phosphorylation to promote melanoma cell survival. Oncogene. 2011;30(49):4887–900.PubMedCrossRef
23.
go back to reference Jin M, Park SJ, Kim SW, Kim HR, Hyun JW, Lee JH. PIG3 regulates p53 stability by suppressing its MDM2-mediated ubiquitination. Biomol Ther. 2017;25(4):396–403.CrossRef Jin M, Park SJ, Kim SW, Kim HR, Hyun JW, Lee JH. PIG3 regulates p53 stability by suppressing its MDM2-mediated ubiquitination. Biomol Ther. 2017;25(4):396–403.CrossRef
24.
go back to reference Zeng KX, Chen XX, Hu XX, Liu XX, Xu T, Sun HL, Pan YQ, He BS, Wang SK. LACTB, a novel epigenetic silenced tumor suppressor, inhibits colorectal cancer progression by attenuating MDM2-mediated p53 ubiquitination and degradation. Oncogene. 2018;37(41):5534–51.PubMedCrossRef Zeng KX, Chen XX, Hu XX, Liu XX, Xu T, Sun HL, Pan YQ, He BS, Wang SK. LACTB, a novel epigenetic silenced tumor suppressor, inhibits colorectal cancer progression by attenuating MDM2-mediated p53 ubiquitination and degradation. Oncogene. 2018;37(41):5534–51.PubMedCrossRef
25.
go back to reference Holzer P. Discovery of potent and selective p53-MDM2 protein-protein interaction inhibitors as anticancer drugs. Chimia. 2017;71(10):716–21.PubMedCrossRef Holzer P. Discovery of potent and selective p53-MDM2 protein-protein interaction inhibitors as anticancer drugs. Chimia. 2017;71(10):716–21.PubMedCrossRef
26.
go back to reference Zhuge W, Chen R, Vladimir K, Dong X, Zia K, Sun X, Dai X, Bao M, Shen X, Liang G. Costunolide specifically binds and inhibits thioredoxin reductase 1 to induce apoptosis in colon cancer. Cancer Lett. 2018;412:46–58.PubMedCrossRef Zhuge W, Chen R, Vladimir K, Dong X, Zia K, Sun X, Dai X, Bao M, Shen X, Liang G. Costunolide specifically binds and inhibits thioredoxin reductase 1 to induce apoptosis in colon cancer. Cancer Lett. 2018;412:46–58.PubMedCrossRef
27.
go back to reference Peng ZX, Wang Y, Fan JH, Lin XJ, Liu CY, Xu Y, Ji WD, Yan C, Su CQ. Costunolide and dehydrocostuslactone combination treatment inhibit breast cancer by inducing cell cycle arrest and apoptosis through c-Myc/p53 and AKT/14-3-3 pathway. Sci Rep. 2017;7:41254.PubMedPubMedCentralCrossRef Peng ZX, Wang Y, Fan JH, Lin XJ, Liu CY, Xu Y, Ji WD, Yan C, Su CQ. Costunolide and dehydrocostuslactone combination treatment inhibit breast cancer by inducing cell cycle arrest and apoptosis through c-Myc/p53 and AKT/14-3-3 pathway. Sci Rep. 2017;7:41254.PubMedPubMedCentralCrossRef
28.
go back to reference Hua PY, Sun M, Zhang GX, Zhang YF, Song G, Liu ZY, Li X, Zhang XY, Li BJ. Costunolide induces apoptosis through generation of ROS and activation of P53 in human esophageal cancer Eca-109 cells. J Biochem Mol Toxicol. 2016;30(9):462–9.PubMedCrossRef Hua PY, Sun M, Zhang GX, Zhang YF, Song G, Liu ZY, Li X, Zhang XY, Li BJ. Costunolide induces apoptosis through generation of ROS and activation of P53 in human esophageal cancer Eca-109 cells. J Biochem Mol Toxicol. 2016;30(9):462–9.PubMedCrossRef
29.
go back to reference Chen JS, Chen BS, Zou ZH, Li W, Zhang YM, Xie JL, Liu CX. Costunolide enhances doxorubicin-induced apoptosis in prostate cancer cells via activated mitogen-activated protein kinases and generation of reactive oxygen species. Oncotarget. 2017;8(64):107701–15.PubMedPubMedCentralCrossRef Chen JS, Chen BS, Zou ZH, Li W, Zhang YM, Xie JL, Liu CX. Costunolide enhances doxorubicin-induced apoptosis in prostate cancer cells via activated mitogen-activated protein kinases and generation of reactive oxygen species. Oncotarget. 2017;8(64):107701–15.PubMedPubMedCentralCrossRef
30.
go back to reference Kim DY, Choi BY. Costunolide-a bioactive sesquiterpene lactone with diverse therapeutic potential. Int J Mol Sci. 2019;20(12):2926.PubMedCentralCrossRef Kim DY, Choi BY. Costunolide-a bioactive sesquiterpene lactone with diverse therapeutic potential. Int J Mol Sci. 2019;20(12):2926.PubMedCentralCrossRef
31.
go back to reference Chen L, Bi SN, Hou JZ, Zhao ZJ, Wang CJ, Xie SQ. Targeting p21-activated kinase 1 inhibits growth and metastasis via Raf1/MEK1/ERK signaling in esophageal squamous cell carcinoma cells. Cell Commun Signal. 2019;17(1):1–7.PubMedPubMedCentralCrossRef Chen L, Bi SN, Hou JZ, Zhao ZJ, Wang CJ, Xie SQ. Targeting p21-activated kinase 1 inhibits growth and metastasis via Raf1/MEK1/ERK signaling in esophageal squamous cell carcinoma cells. Cell Commun Signal. 2019;17(1):1–7.PubMedPubMedCentralCrossRef
32.
go back to reference Whipple RA, Vitolo MI, Boggs AE, Charpentier MS, Thompson K, Martin SS. Parthenolide and costunolide reduce microtentacles and tumor cell attachment by selectively targeting detyrosinated tubulin independent from NF-kappaB inhibition. Breast Cancer Res. 2013;15(5):R83.PubMedPubMedCentralCrossRef Whipple RA, Vitolo MI, Boggs AE, Charpentier MS, Thompson K, Martin SS. Parthenolide and costunolide reduce microtentacles and tumor cell attachment by selectively targeting detyrosinated tubulin independent from NF-kappaB inhibition. Breast Cancer Res. 2013;15(5):R83.PubMedPubMedCentralCrossRef
33.
go back to reference Liu YC, Feng N, Li WW, Tu PF, Chen JP, Han JY, Zeng KW. Costunolide plays an anti-neuroinflammation role in lipopolysaccharide-induced BV2 microglial activation by targeting cyclin-dependent kinase 2. Molecules. 2020;25(12):2840.PubMedCentralCrossRef Liu YC, Feng N, Li WW, Tu PF, Chen JP, Han JY, Zeng KW. Costunolide plays an anti-neuroinflammation role in lipopolysaccharide-induced BV2 microglial activation by targeting cyclin-dependent kinase 2. Molecules. 2020;25(12):2840.PubMedCentralCrossRef
34.
go back to reference Park E, Song JH, Kim MS, Park SH, Kim TS. Costunolide, a sesquiterpene lactone, inhibits the differentiation of pro-inflammatory CD4(+) T cells through the modulation of mitogen-activated protein kinases. Int Immunopharmacol. 2016;40:508–16.PubMedCrossRef Park E, Song JH, Kim MS, Park SH, Kim TS. Costunolide, a sesquiterpene lactone, inhibits the differentiation of pro-inflammatory CD4(+) T cells through the modulation of mitogen-activated protein kinases. Int Immunopharmacol. 2016;40:508–16.PubMedCrossRef
35.
go back to reference Dong GZ, Shim AR, Hyeon JS, Lee HJ, Ryu JH. Inhibition of Wnt/beta-catenin pathway by dehydrocostus lactone and costunolide in colon cancer cells. Phytother Res. 2015;29(5):680–6.PubMedCrossRef Dong GZ, Shim AR, Hyeon JS, Lee HJ, Ryu JH. Inhibition of Wnt/beta-catenin pathway by dehydrocostus lactone and costunolide in colon cancer cells. Phytother Res. 2015;29(5):680–6.PubMedCrossRef
36.
go back to reference Ge MX, Liu HT, Zhang N, Niu WX, Lu ZN, Bao YY, Huang R, Yu DK, Shao RG, He HW. Costunolide represses hepatic fibrosis through WW domain-containing protein 2-mediated Notch3 degradation. Br J Pharmacol. 2020;177(2):372–87.PubMedCrossRef Ge MX, Liu HT, Zhang N, Niu WX, Lu ZN, Bao YY, Huang R, Yu DK, Shao RG, He HW. Costunolide represses hepatic fibrosis through WW domain-containing protein 2-mediated Notch3 degradation. Br J Pharmacol. 2020;177(2):372–87.PubMedCrossRef
37.
go back to reference Song MQ, Bode AM, Dong ZG, Lee MH. AKT as a therapeutic target for cancer. Cancer Res. 2019;79(6):1019–31.PubMedCrossRef Song MQ, Bode AM, Dong ZG, Lee MH. AKT as a therapeutic target for cancer. Cancer Res. 2019;79(6):1019–31.PubMedCrossRef
38.
go back to reference Itoh N, Semba S, Ito M, Takeda H, Kawata S, Yamakawa M. Phosphorylation of Akt/PKB is required for suppression of cancer cell apoptosis and tumor progression in human colorectal carcinoma. Cancer. 2002;94(12):3127–34.PubMedCrossRef Itoh N, Semba S, Ito M, Takeda H, Kawata S, Yamakawa M. Phosphorylation of Akt/PKB is required for suppression of cancer cell apoptosis and tumor progression in human colorectal carcinoma. Cancer. 2002;94(12):3127–34.PubMedCrossRef
39.
go back to reference Jin X, Wang C, Wang L. Costunolide inhibits osteosarcoma growth and metastasis via suppressing STAT3 signal pathway. Biomed Pharmacother. 2019;121:109659.PubMedCrossRef Jin X, Wang C, Wang L. Costunolide inhibits osteosarcoma growth and metastasis via suppressing STAT3 signal pathway. Biomed Pharmacother. 2019;121:109659.PubMedCrossRef
40.
go back to reference Hu M, Liu L, Yao W. Activation of p53 by costunolide blocks glutaminolysis and inhibits proliferation in human colorectal cancer cells. Gene. 2018;678:261–9.PubMedCrossRef Hu M, Liu L, Yao W. Activation of p53 by costunolide blocks glutaminolysis and inhibits proliferation in human colorectal cancer cells. Gene. 2018;678:261–9.PubMedCrossRef
41.
go back to reference Hua PY, Zhang GX, Zhang YF, Sun M, Cui RJ, Li X, Li BJ, Zhang XY. Costunolide induces G1/S phase arrest and activates mitochondrial-mediated apoptotic pathways in SK-MES 1 human lung squamous carcinoma cells. Oncol Lett. 2016;11(4):2780–6.PubMedPubMedCentralCrossRef Hua PY, Zhang GX, Zhang YF, Sun M, Cui RJ, Li X, Li BJ, Zhang XY. Costunolide induces G1/S phase arrest and activates mitochondrial-mediated apoptotic pathways in SK-MES 1 human lung squamous carcinoma cells. Oncol Lett. 2016;11(4):2780–6.PubMedPubMedCentralCrossRef
42.
go back to reference Bond GL, Hu WW, Bond EE, Robins H, Lutzker SG, Arva NC, Bargonetti J, Bartel F, Taubert H, Wuerl P, et al. A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell. 2004;119(5):591–602.PubMedCrossRef Bond GL, Hu WW, Bond EE, Robins H, Lutzker SG, Arva NC, Bargonetti J, Bartel F, Taubert H, Wuerl P, et al. A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell. 2004;119(5):591–602.PubMedCrossRef
43.
go back to reference Mendrysa SM, O'Leary KA, McElwee MK, Michalowski J, Eisenman RN, Powell DA, Perry ME. Tumor suppression and normal aging in mice with constitutively high p53 activity. Genes Dev. 2006;20(1):16–21.PubMedPubMedCentralCrossRef Mendrysa SM, O'Leary KA, McElwee MK, Michalowski J, Eisenman RN, Powell DA, Perry ME. Tumor suppression and normal aging in mice with constitutively high p53 activity. Genes Dev. 2006;20(1):16–21.PubMedPubMedCentralCrossRef
44.
go back to reference Wang P, Lushnikova T, Odvody J, Greiner TC, Jones SN, Eischen CM. Elevated Mdm2 expression induces chromosomal instability and confers a survival and growth advantage to B cells. Oncogene. 2008;27(11):1590–8.PubMedCrossRef Wang P, Lushnikova T, Odvody J, Greiner TC, Jones SN, Eischen CM. Elevated Mdm2 expression induces chromosomal instability and confers a survival and growth advantage to B cells. Oncogene. 2008;27(11):1590–8.PubMedCrossRef
46.
go back to reference Maya R, Balass M, Kim ST, Shkedy D, Leal JF, Shifman O, Moas M, Buschmann T, Ronai Z, Shiloh Y, et al. ATM-dependent phosphorylation of Mdm2 on serine 395: role in p53 activation by DNA damage. Genes Dev. 2001;15(9):1067–77.PubMedPubMedCentralCrossRef Maya R, Balass M, Kim ST, Shkedy D, Leal JF, Shifman O, Moas M, Buschmann T, Ronai Z, Shiloh Y, et al. ATM-dependent phosphorylation of Mdm2 on serine 395: role in p53 activation by DNA damage. Genes Dev. 2001;15(9):1067–77.PubMedPubMedCentralCrossRef
47.
go back to reference Zhang HB, Somasundaram K, Peng Y, Tian H, Zhang HX, Bi DK, Weber BL, El-Deiry WS. BRCA1 physically associates with p53 and stimulates its transcriptional activity. Oncogene. 1998;16(13):1713–21.PubMedCrossRef Zhang HB, Somasundaram K, Peng Y, Tian H, Zhang HX, Bi DK, Weber BL, El-Deiry WS. BRCA1 physically associates with p53 and stimulates its transcriptional activity. Oncogene. 1998;16(13):1713–21.PubMedCrossRef
48.
go back to reference Liao WL, Lin JY, Shieh JC, Yeh HF, Hsieh YH, Cheng YC, Lee HJ, Shen CY, Cheng CW. Induction of G2/M phase arrest by diosgenin via activation of Chk1 kinase and Cdc25C regulatory pathways to promote apoptosis in human breast cancer cells. Int J Mol Sci. 2020;21(1):172.CrossRef Liao WL, Lin JY, Shieh JC, Yeh HF, Hsieh YH, Cheng YC, Lee HJ, Shen CY, Cheng CW. Induction of G2/M phase arrest by diosgenin via activation of Chk1 kinase and Cdc25C regulatory pathways to promote apoptosis in human breast cancer cells. Int J Mol Sci. 2020;21(1):172.CrossRef
49.
go back to reference Giono LE, Resnick-Silverman L, Carvajal LA, St Clair S, Manfredi JJ. Mdm2 promotes Cdc25C protein degradation and delays cell cycle progression through the G2/M phase. Oncogene. 2017;36(49):6762–73.PubMedPubMedCentralCrossRef Giono LE, Resnick-Silverman L, Carvajal LA, St Clair S, Manfredi JJ. Mdm2 promotes Cdc25C protein degradation and delays cell cycle progression through the G2/M phase. Oncogene. 2017;36(49):6762–73.PubMedPubMedCentralCrossRef
50.
go back to reference Phesse TJ, Myant KB, Cole AM, Ridgway RA, Pearson H, Muncan V, van den Brink GR, Vousden KH, Sears R, Vassilev LT, et al. Endogenous c-Myc is essential for p53-induced apoptosis in response to DNA damage in vivo. Cell Death Differ. 2014;21(6):956–66.PubMedPubMedCentralCrossRef Phesse TJ, Myant KB, Cole AM, Ridgway RA, Pearson H, Muncan V, van den Brink GR, Vousden KH, Sears R, Vassilev LT, et al. Endogenous c-Myc is essential for p53-induced apoptosis in response to DNA damage in vivo. Cell Death Differ. 2014;21(6):956–66.PubMedPubMedCentralCrossRef
51.
go back to reference Rogulski K, Li YJ, Rothermund K, Pu LX, Watkins S, Yi FH, Prochownik EV. Onzin, a c-Myc-repressed target, promotes survival and transformation by modulating the Akt-Mdm2-p53 pathway. Oncogene. 2005;24(51):7524–41.PubMedCrossRef Rogulski K, Li YJ, Rothermund K, Pu LX, Watkins S, Yi FH, Prochownik EV. Onzin, a c-Myc-repressed target, promotes survival and transformation by modulating the Akt-Mdm2-p53 pathway. Oncogene. 2005;24(51):7524–41.PubMedCrossRef
52.
go back to reference Han CT, Schoene NW, Lei KY. Influence of zinc deficiency on Akt-Mdm2-p53 and Akt-p21 signaling axes in normal and malignant human prostate cells. Am J Phys Cell Phys. 2009;297(5):C1188–99. Han CT, Schoene NW, Lei KY. Influence of zinc deficiency on Akt-Mdm2-p53 and Akt-p21 signaling axes in normal and malignant human prostate cells. Am J Phys Cell Phys. 2009;297(5):C1188–99.
53.
go back to reference Chou TC, Zhang X, Zhong ZY, Li Y, Feng L, Eng S, Myles DR, Johnson R, Wu N, Yin YI, et al. Therapeutic effect against human xenograft tumors in nude mice by the third generation microtubule stabilizing epothilones. Proc Natl Acad Sci U S A. 2008;105(35):13157–62.PubMedPubMedCentralCrossRef Chou TC, Zhang X, Zhong ZY, Li Y, Feng L, Eng S, Myles DR, Johnson R, Wu N, Yin YI, et al. Therapeutic effect against human xenograft tumors in nude mice by the third generation microtubule stabilizing epothilones. Proc Natl Acad Sci U S A. 2008;105(35):13157–62.PubMedPubMedCentralCrossRef
54.
55.
go back to reference Wang SM, Sun W, Zhao YJ, McEachern D, Meaux I, Barriere C, Stuckey JA, Meagher JL, Bai LC, Liu L, et al. SAR405838: an optimized inhibitor of MDM2-p53 interaction that induces complete and durable tumor regression. Cancer Res. 2014;74(20):5855–65.PubMedPubMedCentralCrossRef Wang SM, Sun W, Zhao YJ, McEachern D, Meaux I, Barriere C, Stuckey JA, Meagher JL, Bai LC, Liu L, et al. SAR405838: an optimized inhibitor of MDM2-p53 interaction that induces complete and durable tumor regression. Cancer Res. 2014;74(20):5855–65.PubMedPubMedCentralCrossRef
56.
go back to reference Lu JF, McEachern D, Li SQ, Ellis MJ, Wang SM. Reactivation of p53 by MDM2 inhibitor MI-77301 for the treatment of endocrine-resistant breast cancer. Mol Cancer Ther. 2016;15(12):2887–93.PubMedPubMedCentralCrossRef Lu JF, McEachern D, Li SQ, Ellis MJ, Wang SM. Reactivation of p53 by MDM2 inhibitor MI-77301 for the treatment of endocrine-resistant breast cancer. Mol Cancer Ther. 2016;15(12):2887–93.PubMedPubMedCentralCrossRef
57.
go back to reference Liu S, Tackmann NR, Yang J, Zhang Y. Disruption of the RP-MDM2-p53 pathway accelerates APC loss-induced colorectal tumorigenesis. Oncogene. 2017;36(10):1374–83.PubMedCrossRef Liu S, Tackmann NR, Yang J, Zhang Y. Disruption of the RP-MDM2-p53 pathway accelerates APC loss-induced colorectal tumorigenesis. Oncogene. 2017;36(10):1374–83.PubMedCrossRef
58.
Metadata
Title
Targeting AKT with costunolide suppresses the growth of colorectal cancer cells and induces apoptosis in vitro and in vivo
Authors
Hai Huang
Song Park
Haibo Zhang
Sijun Park
Wookbong Kwon
Enugyung Kim
Xiujuan Zhang
Soyoung Jang
Duhak Yoon
Seong-Kyoon Choi
Jun-koo Yi
Sung-hyun Kim
Zigang Dong
Mee-hyun Lee
Zaeyoung Ryoo
Myoung Ok Kim
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01895-w

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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