Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 3/2018

01-03-2018 | Original Article

Rabdocoestin B exhibits antitumor activity by inducing G2/M phase arrest and apoptosis in esophageal squamous cell carcinoma

Authors: Jingnan Wang, Zhirong Zhang, Yun Che, Zuyang Yuan, Zhiliang Lu, Yuan Li, Jun Wan, Handong Sun, Zhaoli Chen, Jianxin Pu, Jie He

Published in: Cancer Chemotherapy and Pharmacology | Issue 3/2018

Login to get access

Abstract

Purpose

Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive squamous cell carcinomas and is generally resistant to chemotherapy. In the present study, the cytotoxic activity of Rabdocoestin B (Rabd-B) against ESCC and the underlying mechanisms were investigated.

Methods

The inhibitory effect of Rabd-B on KYSE30 and KYSE450 was evaluated by Cell Counting Kit-8 (CCK8) and colony formation assays in vitro. The cell cycle distribution and apoptosis of cells treated with Rabd-B were determined by flow cytometry. The mechanisms underlying the effects of Rabd-B were systematically examined by Western blot. The in vivo anti-tumor ability of Rabd-B was measured in mouse xenograft models and cisplatin (DDP) was used as positive control.

Results

Rabd-B efficiently induced G2/M phase arrest in ESCC cells by upregulating the Chk1/Chk2-Cdc25C axis to inhibit the G2→M transition facilitated by Cdc2/Cyclin B1. Furthermore, Rabd-B suppressed ATM/ATR phosphorylation, thereby inhibiting BRCA1-mediated DNA repair, which resulted in mitotic catastrophe and induced cell apoptosis. Rabd-B also decreased the activity of the Akt and NF-κB survival signaling pathways and ultimately initiated the caspase-9-dependent intrinsic apoptotic pathway in ESCC cells. The apoptosis induced by Rabd-B could be partially reversed by a caspase-9-specific inhibitor (Z-LEHD-FMK) and a pan-caspase inhibitor (Z-VAD-FMK). Moreover, Rabd-B effectively suppressed tumor growth in mouse xenografts which was comparable to that of DDP without significant injuries to the mice.

Conclusion

Taken together, these findings indicate that Rabd-B is a promising precursor compound that may be useful as a treatment for ESCC and thus warrants further investigation.
Appendix
Available only for authorised users
Literature
12.
go back to reference Feng T, Pu J, Hu Z, Liu D, Sun H, Zhou G (2009) Rabdocoetsin B, a diterpenoid isolated from Isodon coetsa, is a potential proteasome inhibitor and induced apoptosis of t(8;21) leukemia cells. Sheng Wu Gong Cheng Xue Bao 25(8):1218–1224PubMed Feng T, Pu J, Hu Z, Liu D, Sun H, Zhou G (2009) Rabdocoetsin B, a diterpenoid isolated from Isodon coetsa, is a potential proteasome inhibitor and induced apoptosis of t(8;21) leukemia cells. Sheng Wu Gong Cheng Xue Bao 25(8):1218–1224PubMed
14.
go back to reference Wang XR, Wang ZQ, Wang HP, Hu HP, Wang DQ (1987) Chemical structures of Coetsin A and B. Zhiwu Xuebao 29(4):412–415 Wang XR, Wang ZQ, Wang HP, Hu HP, Wang DQ (1987) Chemical structures of Coetsin A and B. Zhiwu Xuebao 29(4):412–415
15.
go back to reference Shimada Y, Imamura M, Wagata T, Yamaguchi N, Tobe T (1992) Characterization of 21 newly established esophageal cancer cell lines. Cancer 69(2):277–284CrossRefPubMed Shimada Y, Imamura M, Wagata T, Yamaguchi N, Tobe T (1992) Characterization of 21 newly established esophageal cancer cell lines. Cancer 69(2):277–284CrossRefPubMed
16.
go back to reference Kanda Y, Nishiyama Y, Shimada Y, Imamura M, Nomura H, Hiai H, Fukumoto M (1994) Analysis of gene amplification and overexpression in human esophageal-carcinoma cell lines. Int J Cancer 58(2):291–297CrossRefPubMed Kanda Y, Nishiyama Y, Shimada Y, Imamura M, Nomura H, Hiai H, Fukumoto M (1994) Analysis of gene amplification and overexpression in human esophageal-carcinoma cell lines. Int J Cancer 58(2):291–297CrossRefPubMed
25.
28.
go back to reference Ding C, Zhang Y, Chen H, Yang Z, Wild C, Ye N, Ester CD, Xiong A, White MA, Shen Q, Zhou J (2013) Oridonin ring A-based diverse constructions of enone functionality: identification of novel dienone analogues effective for highly aggressive breast cancer by inducing apoptosis. J Med Chem 56(21):8814–8825. https://doi.org/10.1021/jm401248x CrossRefPubMed Ding C, Zhang Y, Chen H, Yang Z, Wild C, Ye N, Ester CD, Xiong A, White MA, Shen Q, Zhou J (2013) Oridonin ring A-based diverse constructions of enone functionality: identification of novel dienone analogues effective for highly aggressive breast cancer by inducing apoptosis. J Med Chem 56(21):8814–8825. https://​doi.​org/​10.​1021/​jm401248x CrossRefPubMed
29.
go back to reference Ma YC, Ke Y, Zi X, Zhao W, Shi XJ, Liu HM (2013) Jaridonin, a novel ent-kaurene diterpenoid from Isodon rubescens, inducing apoptosis via production of reactive oxygen species in esophageal cancer cells. Curr Cancer Drug Targets 13(6):611–624CrossRefPubMedPubMedCentral Ma YC, Ke Y, Zi X, Zhao W, Shi XJ, Liu HM (2013) Jaridonin, a novel ent-kaurene diterpenoid from Isodon rubescens, inducing apoptosis via production of reactive oxygen species in esophageal cancer cells. Curr Cancer Drug Targets 13(6):611–624CrossRefPubMedPubMedCentral
35.
37.
go back to reference Bulavin DV, Amundson SA, Fornace AJ (2002) p38 and Chk1 kinases: different conductors for the G(2)/M checkpoint symphony. Curr Opin Genet Dev 12(1):92–97CrossRefPubMed Bulavin DV, Amundson SA, Fornace AJ (2002) p38 and Chk1 kinases: different conductors for the G(2)/M checkpoint symphony. Curr Opin Genet Dev 12(1):92–97CrossRefPubMed
43.
go back to reference Li B, Xu WW, Lam AKY, Wang Y, Hu HF, Guan XY, Qin YR, Saremi N, Tsao SW, He QY, Cheung ALM (2017) Significance of PI3K/AKT signaling pathway in metastasis of esophageal squamous cell carcinoma and its potential as a target for anti-metastasis therapy. Oncotarget. https://doi.org/10.18632/oncotarget.16333 Li B, Xu WW, Lam AKY, Wang Y, Hu HF, Guan XY, Qin YR, Saremi N, Tsao SW, He QY, Cheung ALM (2017) Significance of PI3K/AKT signaling pathway in metastasis of esophageal squamous cell carcinoma and its potential as a target for anti-metastasis therapy. Oncotarget. https://​doi.​org/​10.​18632/​oncotarget.​16333
44.
go back to reference Yoshioka A, Miyata H, Doki Y, Yasuda T, Yamasaki M, Motoori M, Okada K, Matsuyama J, Makari Y, Sohma I, Takiguchi S, Fujiwara Y, Monden M (2008) The activation of Akt during preoperative chemotherapy for esophageal cancer correlates with poor prognosis. Oncol Rep 19(5):1099–1107PubMed Yoshioka A, Miyata H, Doki Y, Yasuda T, Yamasaki M, Motoori M, Okada K, Matsuyama J, Makari Y, Sohma I, Takiguchi S, Fujiwara Y, Monden M (2008) The activation of Akt during preoperative chemotherapy for esophageal cancer correlates with poor prognosis. Oncol Rep 19(5):1099–1107PubMed
53.
go back to reference Gavrieli Y, Sherman Y, Ben-Sasson SA (1992) Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol 119(3):493–501CrossRefPubMed Gavrieli Y, Sherman Y, Ben-Sasson SA (1992) Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol 119(3):493–501CrossRefPubMed
Metadata
Title
Rabdocoestin B exhibits antitumor activity by inducing G2/M phase arrest and apoptosis in esophageal squamous cell carcinoma
Authors
Jingnan Wang
Zhirong Zhang
Yun Che
Zuyang Yuan
Zhiliang Lu
Yuan Li
Jun Wan
Handong Sun
Zhaoli Chen
Jianxin Pu
Jie He
Publication date
01-03-2018
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 3/2018
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-017-3507-2

Other articles of this Issue 3/2018

Cancer Chemotherapy and Pharmacology 3/2018 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