Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2020

Open Access 01-12-2020 | Gastric Cancer | Research

Lycorine hydrochloride inhibits cell proliferation and induces apoptosis through promoting FBXW7-MCL1 axis in gastric cancer

Authors: Chongyang Li, Chaowei Deng, Guangzhao Pan, Xue Wang, Kui Zhang, Zhen Dong, Gaichao Zhao, Mengqin Tan, Xiaosong Hu, Shaomin Shi, Juan Du, Haoyan Ji, Xiaowen Wang, Liqun Yang, Hongjuan Cui

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2020

Login to get access

Abstract

Background

Lycorine hydrochloride (LH), an alkaloid extracted from the bulb of the Lycoris radiata, is considered to have anti-viral, anti-malarial, and anti-tumorous effects. At present, the underlying mechanisms of LH in gastric cancer remain unclear. MCL1, an anti-apoptotic protein of BCL2 family, is closely related to drug resistance of tumor. Therefore, MCL1 is considered as a potential target for cancer treatment.

Methods

The effect of LH on gastric cancer was assessed in vitro (by MTT, BrdU, western blotting…) and in vivo (by immunohistochemistry).

Results

In this study, we showed that LH has an anti-tumorous effect by down-regulating MCL1 in gastric cancer. Besides, we unveiled that LH reduced the protein stability of MCL1 by up-regulating ubiquitin E3 ligase FBXW7, arrested cell cycle at S phase and triggered apoptosis of gastric cancer cells. Meanwhile, we also demonstrated that LH could induce apoptosis of the BCL2-drug-resistant-cell-lines. Moreover, PDX (Patient-Derived tumor xenograft) model experiment proved that LH combined with HA14–1 (inhibitor of BCL2), had a more significant therapeutic effect on gastric cancer.

Conclusions

The efficacy showed in our data suggests that lycorine hydrochloride is a promising anti-tumor compound for gastric cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference EC S, M N, HI G, van Grieken NC, F L. Gastric cancer. Lancet (London, England). 2020;396:635–48. EC S, M N, HI G, van Grieken NC, F L. Gastric cancer. Lancet (London, England). 2020;396:635–48.
3.
go back to reference Li W, Zhang X, Wu F, et al. Gastric cancer-derived mesenchymal stromal cells trigger M2 macrophage polarization that promotes metastasis and EMT in gastric cancer. Cell Death Dis. 2019;10:918.PubMedPubMedCentral Li W, Zhang X, Wu F, et al. Gastric cancer-derived mesenchymal stromal cells trigger M2 macrophage polarization that promotes metastasis and EMT in gastric cancer. Cell Death Dis. 2019;10:918.PubMedPubMedCentral
4.
go back to reference Montagnani F, Crivelli F, Aprile G, et al. Long-term survival after liver metastasectomy in gastric cancer: systematic review and meta-analysis of prognostic factors. Cancer Treat Rev. 2018;69:11–20.PubMed Montagnani F, Crivelli F, Aprile G, et al. Long-term survival after liver metastasectomy in gastric cancer: systematic review and meta-analysis of prognostic factors. Cancer Treat Rev. 2018;69:11–20.PubMed
5.
go back to reference Kitayama J, Ishigami H, Yamaguchi H, et al. Treatment of patients with peritoneal metastases from gastric cancer. Ann Gastroenterol Surg. 2018;2:116–23.PubMedPubMedCentral Kitayama J, Ishigami H, Yamaguchi H, et al. Treatment of patients with peritoneal metastases from gastric cancer. Ann Gastroenterol Surg. 2018;2:116–23.PubMedPubMedCentral
6.
go back to reference Song Y, Wang Y, Tong C, et al. A unified model of the hierarchical and stochastic theories of gastric cancer. Br J Cancer. 2017;116:973–89.PubMedPubMedCentral Song Y, Wang Y, Tong C, et al. A unified model of the hierarchical and stochastic theories of gastric cancer. Br J Cancer. 2017;116:973–89.PubMedPubMedCentral
7.
go back to reference Jakubek M, Kejik Z, Kaplanek R, et al. Strategy for improved therapeutic efficiency of curcumin in the treatment of gastric cancer. Biomed Pharmacother. 2019;118:109278.PubMed Jakubek M, Kejik Z, Kaplanek R, et al. Strategy for improved therapeutic efficiency of curcumin in the treatment of gastric cancer. Biomed Pharmacother. 2019;118:109278.PubMed
8.
go back to reference Wu Q, Yang Z, Nie Y, Shi Y, Fan D. Multi-drug resistance in cancer chemotherapeutics: mechanisms and lab approaches. Cancer Lett. 2014;347:159–66.PubMed Wu Q, Yang Z, Nie Y, Shi Y, Fan D. Multi-drug resistance in cancer chemotherapeutics: mechanisms and lab approaches. Cancer Lett. 2014;347:159–66.PubMed
9.
go back to reference Wang JH, Du JP, Zhang YH, et al. Dynamic changes and surveillance function of prion protein expression in gastric cancer drug resistance. World J Gastroenterol. 2011;17:3986–93.PubMedPubMedCentral Wang JH, Du JP, Zhang YH, et al. Dynamic changes and surveillance function of prion protein expression in gastric cancer drug resistance. World J Gastroenterol. 2011;17:3986–93.PubMedPubMedCentral
10.
go back to reference Leist M, Jaattela M. Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol. 2001;2:589–98.PubMed Leist M, Jaattela M. Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol. 2001;2:589–98.PubMed
11.
go back to reference Kaufmann SH, Gores GJ. Apoptosis in cancer: cause and cure. BIOESSAYS. 2000;22:1007–17.PubMed Kaufmann SH, Gores GJ. Apoptosis in cancer: cause and cure. BIOESSAYS. 2000;22:1007–17.PubMed
12.
go back to reference Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between cancer genetics and chemotherapy. Cell. 2002;108:153–64.PubMed Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between cancer genetics and chemotherapy. Cell. 2002;108:153–64.PubMed
13.
go back to reference Park H, Cho SY, Kim H, et al. Genomic alterations in BCL2L1 and DLC1 contribute to drug sensitivity in gastric cancer. Proc Natl Acad Sci U S A. 2015;112:12492–7.PubMedPubMedCentral Park H, Cho SY, Kim H, et al. Genomic alterations in BCL2L1 and DLC1 contribute to drug sensitivity in gastric cancer. Proc Natl Acad Sci U S A. 2015;112:12492–7.PubMedPubMedCentral
14.
go back to reference Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008;9:47–59.PubMed Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008;9:47–59.PubMed
15.
16.
go back to reference Belmar J, Fesik SW. Small molecule mcl-1 inhibitors for the treatment of cancer. Pharmacol Ther. 2015;145:76–84.PubMed Belmar J, Fesik SW. Small molecule mcl-1 inhibitors for the treatment of cancer. Pharmacol Ther. 2015;145:76–84.PubMed
17.
go back to reference Chen L, Willis SN, Wei A, et al. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 2005;17:393–403.PubMed Chen L, Willis SN, Wei A, et al. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 2005;17:393–403.PubMed
18.
go back to reference Ashkenazi A, Fairbrother WJ, Leverson JD, Souers AJ. From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors. Nat Rev Drug Discov. 2017;16:273–84.PubMed Ashkenazi A, Fairbrother WJ, Leverson JD, Souers AJ. From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors. Nat Rev Drug Discov. 2017;16:273–84.PubMed
19.
go back to reference Tse C, Shoemaker AR, Adickes J, et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68:3421–8.PubMed Tse C, Shoemaker AR, Adickes J, et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68:3421–8.PubMed
20.
go back to reference Zhao Y, He J, Li J, et al. Demethylzeylasteral inhibits cell proliferation and induces apoptosis through suppressing MCL1 in melanoma cells. Cell Death Dis. 2017;8:e3133.PubMedPubMedCentral Zhao Y, He J, Li J, et al. Demethylzeylasteral inhibits cell proliferation and induces apoptosis through suppressing MCL1 in melanoma cells. Cell Death Dis. 2017;8:e3133.PubMedPubMedCentral
21.
go back to reference Shoemaker AR, Mitten MJ, Adickes J, et al. Activity of the Bcl-2 family inhibitor ABT-263 in a panel of small cell lung cancer xenograft models. Clin Cancer Res. 2008;14:3268–77.PubMed Shoemaker AR, Mitten MJ, Adickes J, et al. Activity of the Bcl-2 family inhibitor ABT-263 in a panel of small cell lung cancer xenograft models. Clin Cancer Res. 2008;14:3268–77.PubMed
22.
go back to reference van Delft MF, Wei AH, Mason KD, et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if mcl-1 is neutralized. Cancer Cell. 2006;10:389–99.PubMedPubMedCentral van Delft MF, Wei AH, Mason KD, et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if mcl-1 is neutralized. Cancer Cell. 2006;10:389–99.PubMedPubMedCentral
23.
go back to reference Akgul C. Mcl-1 is a potential therapeutic target in multiple types of cancer. Cell Mol Life Sci. 2009;66:1326–36.PubMed Akgul C. Mcl-1 is a potential therapeutic target in multiple types of cancer. Cell Mol Life Sci. 2009;66:1326–36.PubMed
24.
go back to reference Quinn BA, Dash R, Azab B, et al. Targeting mcl-1 for the therapy of cancer. Expert Opin Investig Drugs. 2011;20:1397–411.PubMedPubMedCentral Quinn BA, Dash R, Azab B, et al. Targeting mcl-1 for the therapy of cancer. Expert Opin Investig Drugs. 2011;20:1397–411.PubMedPubMedCentral
25.
go back to reference Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013;19:202–8.PubMed Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013;19:202–8.PubMed
26.
go back to reference Lamoral-Theys D, Decaestecker C, Mathieu V, et al. Lycorine and its derivatives for anticancer drug design. Mini-Rev Med Chem. 2010;10:41–50.PubMed Lamoral-Theys D, Decaestecker C, Mathieu V, et al. Lycorine and its derivatives for anticancer drug design. Mini-Rev Med Chem. 2010;10:41–50.PubMed
27.
go back to reference Cedron JC, Gutierrez D, Flores N, Ravelo AG, Estevez-Braun A. Synthesis and antiplasmodial activity of lycorine derivatives. Bioorg Med Chem. 2010;18:4694–701.PubMed Cedron JC, Gutierrez D, Flores N, Ravelo AG, Estevez-Braun A. Synthesis and antiplasmodial activity of lycorine derivatives. Bioorg Med Chem. 2010;18:4694–701.PubMed
28.
go back to reference Nair JJ, van Staden J. Acetylcholinesterase inhibition within the lycorine series of Amaryllidaceae alkaloids. Nat Prod Commun. 2012;7:959–62.PubMed Nair JJ, van Staden J. Acetylcholinesterase inhibition within the lycorine series of Amaryllidaceae alkaloids. Nat Prod Commun. 2012;7:959–62.PubMed
29.
go back to reference Cao Z, Yu D, Fu S, et al. Lycorine hydrochloride selectively inhibits human ovarian cancer cell proliferation and tumor neovascularization with very low toxicity. Toxicol Lett. 2013;218:174–85.PubMed Cao Z, Yu D, Fu S, et al. Lycorine hydrochloride selectively inhibits human ovarian cancer cell proliferation and tumor neovascularization with very low toxicity. Toxicol Lett. 2013;218:174–85.PubMed
30.
go back to reference Zhang K, Fu G, Pan G, et al. Demethylzeylasteral inhibits glioma growth by regulating the miR-30e-5p/MYBL2 axis. Cell Death Dis. 2018;9:1035.PubMedPubMedCentral Zhang K, Fu G, Pan G, et al. Demethylzeylasteral inhibits glioma growth by regulating the miR-30e-5p/MYBL2 axis. Cell Death Dis. 2018;9:1035.PubMedPubMedCentral
31.
go back to reference Zhao Y, He J, Li Y, Lv S, Cui H. NUSAP1 potentiates chemoresistance in glioblastoma through its SAP domain to stabilize ATR. Signal Transduct Target Ther. 2020;5:44.PubMedPubMedCentral Zhao Y, He J, Li Y, Lv S, Cui H. NUSAP1 potentiates chemoresistance in glioblastoma through its SAP domain to stabilize ATR. Signal Transduct Target Ther. 2020;5:44.PubMedPubMedCentral
32.
go back to reference Cao J, Zhao E, Zhu Q, et al. Tubeimoside-1 inhibits Glioblastoma growth, migration, and invasion via inducing Ubiquitylation of MET. Cells-Basel. 2019;8:774. Cao J, Zhao E, Zhu Q, et al. Tubeimoside-1 inhibits Glioblastoma growth, migration, and invasion via inducing Ubiquitylation of MET. Cells-Basel. 2019;8:774.
33.
go back to reference Zhang G, Zhu Q, Fu G, et al. TRIP13 promotes the cell proliferation, migration and invasion of glioblastoma through the FBXW7/c-MYC axis. Br J Cancer. 2019;121:1069–78.PubMedPubMedCentral Zhang G, Zhu Q, Fu G, et al. TRIP13 promotes the cell proliferation, migration and invasion of glioblastoma through the FBXW7/c-MYC axis. Br J Cancer. 2019;121:1069–78.PubMedPubMedCentral
34.
go back to reference Jin ZJ. Addition in drug combination. Acta Pharmacol Sin. 1980;1:70–6. Jin ZJ. Addition in drug combination. Acta Pharmacol Sin. 1980;1:70–6.
35.
go back to reference Likui W, Qun L, Wanqing Z, et al. Prognostic role of myeloid cell leukemia-1 protein (mcl-1) expression in human gastric cancer. J Surg Oncol. 2009;100:396–400.PubMed Likui W, Qun L, Wanqing Z, et al. Prognostic role of myeloid cell leukemia-1 protein (mcl-1) expression in human gastric cancer. J Surg Oncol. 2009;100:396–400.PubMed
36.
go back to reference Ramsey HE, Fischer MA, Lee T, et al. A novel MCL1 inhibitor combined with Venetoclax rescues Venetoclax-resistant acute Myelogenous leukemia. Cancer Discov. 2018;8:1566–81.PubMedPubMedCentral Ramsey HE, Fischer MA, Lee T, et al. A novel MCL1 inhibitor combined with Venetoclax rescues Venetoclax-resistant acute Myelogenous leukemia. Cancer Discov. 2018;8:1566–81.PubMedPubMedCentral
37.
go back to reference Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.PubMed Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.PubMed
38.
go back to reference Sieghart W, Losert D, Strommer S, et al. Mcl-1 overexpression in hepatocellular carcinoma: a potential target for antisense therapy. J Hepatol. 2006;44:151–7.PubMed Sieghart W, Losert D, Strommer S, et al. Mcl-1 overexpression in hepatocellular carcinoma: a potential target for antisense therapy. J Hepatol. 2006;44:151–7.PubMed
39.
go back to reference Bierbrauer A, Jacob M, Vogler M, Fulda S. A direct comparison of selective BH3-mimetics reveals BCL-XL, BCL-2 and MCL-1 as promising therapeutic targets in neuroblastoma. Br J Cancer. 2020;122:1544–51.PubMedPubMedCentral Bierbrauer A, Jacob M, Vogler M, Fulda S. A direct comparison of selective BH3-mimetics reveals BCL-XL, BCL-2 and MCL-1 as promising therapeutic targets in neuroblastoma. Br J Cancer. 2020;122:1544–51.PubMedPubMedCentral
40.
go back to reference Maurer U, Charvet C, Wagman AS, Dejardin E, Green DR. Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1. Mol Cell. 2006;21:749–60.PubMed Maurer U, Charvet C, Wagman AS, Dejardin E, Green DR. Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1. Mol Cell. 2006;21:749–60.PubMed
41.
go back to reference Tong J, Wang P, Tan S, et al. Mcl-1 degradation is required for targeted therapeutics to eradicate Colon Cancer cells. Cancer Res. 2017;77:2512–21.PubMedPubMedCentral Tong J, Wang P, Tan S, et al. Mcl-1 degradation is required for targeted therapeutics to eradicate Colon Cancer cells. Cancer Res. 2017;77:2512–21.PubMedPubMedCentral
42.
go back to reference Zhong Q, Gao W, Du F, Wang X. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of mcl-1 and regulates apoptosis. CELL. 2005;121:1085–95.PubMed Zhong Q, Gao W, Du F, Wang X. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of mcl-1 and regulates apoptosis. CELL. 2005;121:1085–95.PubMed
43.
go back to reference Ding Q, He X, Hsu JM, et al. Degradation of mcl-1 by beta-TrCP mediates glycogen synthase kinase 3-induced tumor suppression and chemosensitization. Mol Cell Biol. 2007;27:4006–17.PubMed Ding Q, He X, Hsu JM, et al. Degradation of mcl-1 by beta-TrCP mediates glycogen synthase kinase 3-induced tumor suppression and chemosensitization. Mol Cell Biol. 2007;27:4006–17.PubMed
44.
go back to reference Inuzuka H, Shaik S, Onoyama I, et al. SCF(FBW7) regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruction. NATURE. 2011;471:104–9.PubMedPubMedCentral Inuzuka H, Shaik S, Onoyama I, et al. SCF(FBW7) regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruction. NATURE. 2011;471:104–9.PubMedPubMedCentral
45.
go back to reference Wertz IE, Kusam S, Lam C, et al. Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature. 2011;471:110–4.PubMed Wertz IE, Kusam S, Lam C, et al. Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature. 2011;471:110–4.PubMed
46.
go back to reference Wu X, Luo Q, Zhao P, et al. JOSD1 inhibits mitochondrial apoptotic signalling to drive acquired chemoresistance in gynaecological cancer by stabilizing MCL1. Cell Death Differ. 2020;27:55–70.PubMed Wu X, Luo Q, Zhao P, et al. JOSD1 inhibits mitochondrial apoptotic signalling to drive acquired chemoresistance in gynaecological cancer by stabilizing MCL1. Cell Death Differ. 2020;27:55–70.PubMed
47.
go back to reference Wu X, Luo Q, Zhao P, et al. MGMT-activated DUB3 stabilizes MCL1 and drives chemoresistance in ovarian cancer. Proc Natl Acad Sci U S A. 2019;116:2961–6.PubMedPubMedCentral Wu X, Luo Q, Zhao P, et al. MGMT-activated DUB3 stabilizes MCL1 and drives chemoresistance in ovarian cancer. Proc Natl Acad Sci U S A. 2019;116:2961–6.PubMedPubMedCentral
48.
go back to reference Zhang S, Zhang M, Jing Y, et al. Deubiquitinase USP13 dictates MCL1 stability and sensitivity to BH3 mimetic inhibitors. Nat Commun. 2018;9:215.PubMedPubMedCentral Zhang S, Zhang M, Jing Y, et al. Deubiquitinase USP13 dictates MCL1 stability and sensitivity to BH3 mimetic inhibitors. Nat Commun. 2018;9:215.PubMedPubMedCentral
49.
go back to reference Schwickart M, Huang X, Lill JR, et al. Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival. Nature. 2010;463:103–7.PubMed Schwickart M, Huang X, Lill JR, et al. Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival. Nature. 2010;463:103–7.PubMed
50.
go back to reference Wei SH, Dong K, Lin F, et al. Inducing apoptosis and enhancing chemosensitivity to gemcitabine via RNA interference targeting mcl-1 gene in pancreatic carcinoma cell. Cancer Chemother Pharmacol. 2008;62:1055–64.PubMed Wei SH, Dong K, Lin F, et al. Inducing apoptosis and enhancing chemosensitivity to gemcitabine via RNA interference targeting mcl-1 gene in pancreatic carcinoma cell. Cancer Chemother Pharmacol. 2008;62:1055–64.PubMed
51.
go back to reference Xia X, Huang C, Liao Y, et al. Inhibition of USP14 enhances the sensitivity of breast cancer to enzalutamide. J Exp Clin Cancer Res. 2019;38:220.PubMedPubMedCentral Xia X, Huang C, Liao Y, et al. Inhibition of USP14 enhances the sensitivity of breast cancer to enzalutamide. J Exp Clin Cancer Res. 2019;38:220.PubMedPubMedCentral
Metadata
Title
Lycorine hydrochloride inhibits cell proliferation and induces apoptosis through promoting FBXW7-MCL1 axis in gastric cancer
Authors
Chongyang Li
Chaowei Deng
Guangzhao Pan
Xue Wang
Kui Zhang
Zhen Dong
Gaichao Zhao
Mengqin Tan
Xiaosong Hu
Shaomin Shi
Juan Du
Haoyan Ji
Xiaowen Wang
Liqun Yang
Hongjuan Cui
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2020
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01743-3

Other articles of this Issue 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 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