Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 2/2017

01-08-2017 | Original Article

JS-K, a GST-activated nitric oxide donor prodrug, enhances chemo-sensitivity in renal carcinoma cells and prevents cardiac myocytes toxicity induced by Doxorubicin

Authors: Mingning Qiu, Longzhi Ke, Sai Zhang, Xin Zeng, Zesong Fang, Jianjun Liu

Published in: Cancer Chemotherapy and Pharmacology | Issue 2/2017

Login to get access

Abstract

Purpose

Doxorubicin, a highly effective and widely used anthracycline antibiotic in multiple chemotherapy regimens, has been limited by its cardiotoxicity. The aim of this study is to investigate the effect of nitric oxide donor prodrug JS-K on proliferation and apoptosis in renal carcinoma cells and cardiac myocytes toxicity induced by Doxorubicin and to explore possible p53-related mechanism in renal carcinoma cells.

Methods

The effect of JS-K on anti-cancer activity of Doxorubicin was investigated in renal carcinoma cells via detecting cell proliferation, cytotoxicity, cell death and apoptosis and expressions of apoptotic-related proteins. Effect of p53 on the combination of JS-K and Doxorubicin was determined using p53 inhibitor Pifithrin-α and p53 activator III. Furthermore, the effect of JS-K on cardiac myocytes toxicity of Doxorubicin was investigated in H9c2 (2-1) cardiac myocytes via measuring cell growth, cell death and apoptosis, expressions of proteins involved in apoptosis and intracellular reactive oxygen species.

Results

We demonstrated that JS-K could increase Doxorubicin-induced renal carcinoma cell growth suppression and apoptosis and could increase expressions of proteins that are involved in apoptosis. Additionally, Pifithrin-α reversed the promoting effect of JS-K on Doxorubicin-induced renal carcinoma cell apoptosis; conversely, the p53 activator III exacerbated the promoting effect of JS-K on Doxorubicin-induced renal carcinoma cell apoptosis. Furthermore, JS-K protected H9c2 (2-1) cardiac myocytes against Doxorubicin-induced toxicity and decreased Doxorubicin-induced reactive oxygen species production.

Conclusions

JS-K enhances the anti-cancer activity of Doxorubicin in renal carcinoma cells by upregulating p53 expression and prevents cardiac myocytes toxicity of Doxorubicin by decreasing oxidative stress.
Literature
2.
go back to reference Zhu H, Luo P, Fu Y, Wang J, Dai J, Shao J, Yang X, Chang L, Weng Q, Yang B, He Q (2015) Dihydromyricetin prevents cardiotoxicity and enhances anticancer activity induced by adriamycin. Oncotarget 6(5):3254–3267. doi:10.18632/oncotarget.2410 CrossRefPubMed Zhu H, Luo P, Fu Y, Wang J, Dai J, Shao J, Yang X, Chang L, Weng Q, Yang B, He Q (2015) Dihydromyricetin prevents cardiotoxicity and enhances anticancer activity induced by adriamycin. Oncotarget 6(5):3254–3267. doi:10.​18632/​oncotarget.​2410 CrossRefPubMed
3.
go back to reference Becker PS, Gooley TA, Green DJ, Burwick N, Kim TY, Kojouri K, Inoue Y, Moore DJ, Nelli E, Dennie T, Bensinger WI (2016) A phase 2 study of bortezomib, cyclophosphamide, pegylated liposomal Doxorubicin and dexamethasone for newly diagnosed multiple myeloma. Blood Cancer J 6:e422. doi:10.1038/bcj.2016.31 CrossRefPubMedPubMedCentral Becker PS, Gooley TA, Green DJ, Burwick N, Kim TY, Kojouri K, Inoue Y, Moore DJ, Nelli E, Dennie T, Bensinger WI (2016) A phase 2 study of bortezomib, cyclophosphamide, pegylated liposomal Doxorubicin and dexamethasone for newly diagnosed multiple myeloma. Blood Cancer J 6:e422. doi:10.​1038/​bcj.​2016.​31 CrossRefPubMedPubMedCentral
4.
go back to reference Ahmadi F, Mojarrab M, Ghazi-Khansari M, Hosseinzadeh L (2015) A semipolar fraction of petroleum ether extract of Artemisia aucheri induces apoptosis and enhances the apoptotic response to Doxorubicin in human neuroblastoma SKNMC cell line. Res Pharm Sci 10(4):335–344PubMedPubMedCentral Ahmadi F, Mojarrab M, Ghazi-Khansari M, Hosseinzadeh L (2015) A semipolar fraction of petroleum ether extract of Artemisia aucheri induces apoptosis and enhances the apoptotic response to Doxorubicin in human neuroblastoma SKNMC cell line. Res Pharm Sci 10(4):335–344PubMedPubMedCentral
5.
go back to reference Muggia FM, Speyer JL (1999) Doxorubicin-induced cardiomyopathy. N Engl J Med 340(8):654–655PubMed Muggia FM, Speyer JL (1999) Doxorubicin-induced cardiomyopathy. N Engl J Med 340(8):654–655PubMed
8.
go back to reference Wang S, Konorev EA, Kotamraju S, Joseph J, Kalivendi S, Kalyanaraman B (2004) Doxorubicin induces apoptosis in normal and tumor cells via distinctly different mechanisms. intermediacy of H(2)O(2)- and p53-dependent pathways. J Biol Chem 279(24):25535–25543. doi:10.1074/jbc.M400944200 CrossRefPubMed Wang S, Konorev EA, Kotamraju S, Joseph J, Kalivendi S, Kalyanaraman B (2004) Doxorubicin induces apoptosis in normal and tumor cells via distinctly different mechanisms. intermediacy of H(2)O(2)- and p53-dependent pathways. J Biol Chem 279(24):25535–25543. doi:10.​1074/​jbc.​M400944200 CrossRefPubMed
9.
go back to reference Hovorka O, Subr V, Vetvicka D, Kovar L, Strohalm J, Strohalm M, Benda A, Hof M, Ulbrich K, Rihova B (2010) Spectral analysis of Doxorubicin accumulation and the indirect quantification of its DNA intercalation. Eur J Pharm Biopharm 76(3):514–524. doi:10.1016/j.ejpb.2010.07.008 CrossRefPubMed Hovorka O, Subr V, Vetvicka D, Kovar L, Strohalm J, Strohalm M, Benda A, Hof M, Ulbrich K, Rihova B (2010) Spectral analysis of Doxorubicin accumulation and the indirect quantification of its DNA intercalation. Eur J Pharm Biopharm 76(3):514–524. doi:10.​1016/​j.​ejpb.​2010.​07.​008 CrossRefPubMed
10.
go back to reference Kim KH, Oudit GY, Backx PH (2008) Erythropoietin protects against Doxorubicin-induced cardiomyopathy via a phosphatidylinositol 3-kinase-dependent pathway. J Pharm Exp Ther 324(1):160–169. doi:10.1124/jpet.107.125773 CrossRef Kim KH, Oudit GY, Backx PH (2008) Erythropoietin protects against Doxorubicin-induced cardiomyopathy via a phosphatidylinositol 3-kinase-dependent pathway. J Pharm Exp Ther 324(1):160–169. doi:10.​1124/​jpet.​107.​125773 CrossRef
12.
go back to reference Kiziltepe T, Hideshima T, Ishitsuka K, Ocio EM, Raje N, Catley L, Li CQ, Trudel LJ, Yasui H, Vallet S, Kutok JL, Chauhan D, Mitsiades CS, Saavedra JE, Wogan GN, Keefer LK, Shami PJ, Anderson KC (2007) JS-K, a GST-activated nitric oxide generator, induces DNA double-strand breaks, activates DNA damage response pathways, and induces apoptosis in vitro and in vivo in human multiple myeloma cells. Blood 110(2):709–718. doi:10.1182/blood-2006-10-052845 CrossRefPubMedPubMedCentral Kiziltepe T, Hideshima T, Ishitsuka K, Ocio EM, Raje N, Catley L, Li CQ, Trudel LJ, Yasui H, Vallet S, Kutok JL, Chauhan D, Mitsiades CS, Saavedra JE, Wogan GN, Keefer LK, Shami PJ, Anderson KC (2007) JS-K, a GST-activated nitric oxide generator, induces DNA double-strand breaks, activates DNA damage response pathways, and induces apoptosis in vitro and in vivo in human multiple myeloma cells. Blood 110(2):709–718. doi:10.​1182/​blood-2006-10-052845 CrossRefPubMedPubMedCentral
13.
go back to reference McMurtry V, Saavedra JE, Nieves-Alicea R, Simeone AM, Keefer LK, Tari AM (2011) JS-K, a nitric oxide-releasing prodrug, induces breast cancer cell death while sparing normal mammary epithelial cells. Int J Oncol 38(4):963–971. doi:10.3892/ijo.2011.925 PubMed McMurtry V, Saavedra JE, Nieves-Alicea R, Simeone AM, Keefer LK, Tari AM (2011) JS-K, a nitric oxide-releasing prodrug, induces breast cancer cell death while sparing normal mammary epithelial cells. Int J Oncol 38(4):963–971. doi:10.​3892/​ijo.​2011.​925 PubMed
14.
go back to reference Laschak M, Spindler KD, Schrader AJ, Hessenauer A, Streicher W, Schrader M, Cronauer MV (2012) JS-K, a glutathione/glutathione S-transferase-activated nitric oxide releasing prodrug inhibits androgen receptor and WNT-signaling in prostate cancer cells. BMC Cancer 12:130. doi:10.1186/1471-2407-12-130 CrossRefPubMedPubMedCentral Laschak M, Spindler KD, Schrader AJ, Hessenauer A, Streicher W, Schrader M, Cronauer MV (2012) JS-K, a glutathione/glutathione S-transferase-activated nitric oxide releasing prodrug inhibits androgen receptor and WNT-signaling in prostate cancer cells. BMC Cancer 12:130. doi:10.​1186/​1471-2407-12-130 CrossRefPubMedPubMedCentral
16.
go back to reference Liu L, Wang D, Wang J, Wang S (2016) The Nitric oxide prodrug JS-K induces Ca(2+)-mediated apoptosis in human hepatocellular carcinoma HepG2 Cells. J Biochem Mol Toxicol 30(4):192–199. doi:10.1002/jbt.21778 CrossRefPubMed Liu L, Wang D, Wang J, Wang S (2016) The Nitric oxide prodrug JS-K induces Ca(2+)-mediated apoptosis in human hepatocellular carcinoma HepG2 Cells. J Biochem Mol Toxicol 30(4):192–199. doi:10.​1002/​jbt.​21778 CrossRefPubMed
18.
go back to reference Issaeva N, Bozko P, Enge M, Protopopova M, Verhoef LG, Masucci M, Pramanik A, Selivanova G (2004) Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors. Nat Med 10(12):1321–1328. doi:10.1038/nm1146 CrossRefPubMed Issaeva N, Bozko P, Enge M, Protopopova M, Verhoef LG, Masucci M, Pramanik A, Selivanova G (2004) Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors. Nat Med 10(12):1321–1328. doi:10.​1038/​nm1146 CrossRefPubMed
21.
22.
go back to reference Johnstone RW, Ruefli AA, Lowe SW (2002) Apoptosis: a link between cancer genetics and chemotherapy. Cell 108(2):153–164CrossRefPubMed Johnstone RW, Ruefli AA, Lowe SW (2002) Apoptosis: a link between cancer genetics and chemotherapy. Cell 108(2):153–164CrossRefPubMed
23.
go back to reference Roberts AM, Watson IR, Evans AJ, Foster DA, Irwin MS, Ohh M (2009) Suppression of hypoxia-inducible factor 2alpha restores p53 activity via Hdm2 and reverses chemoresistance of renal carcinoma cells. Can Res 69(23):9056–9064. doi:10.1158/0008-5472.CAN-09-1770 CrossRef Roberts AM, Watson IR, Evans AJ, Foster DA, Irwin MS, Ohh M (2009) Suppression of hypoxia-inducible factor 2alpha restores p53 activity via Hdm2 and reverses chemoresistance of renal carcinoma cells. Can Res 69(23):9056–9064. doi:10.​1158/​0008-5472.​CAN-09-1770 CrossRef
24.
go back to reference Liu T, Xiong J, Yi S, Zhang H, Zhou S, Gu L, Zhou M (2016) FKBP12 enhances sensitivity to chemotherapy-induced cancer cell apoptosis by inhibiting MDM2. Oncogene. doi:10.1038/onc.2016.331 Liu T, Xiong J, Yi S, Zhang H, Zhou S, Gu L, Zhou M (2016) FKBP12 enhances sensitivity to chemotherapy-induced cancer cell apoptosis by inhibiting MDM2. Oncogene. doi:10.​1038/​onc.​2016.​331
25.
go back to reference Walia V, Kakar S, Elble R (2011) Micromanagement of the mitochondrial apoptotic pathway by p53. Front Biosci (Landmark Ed) 16:749–758CrossRef Walia V, Kakar S, Elble R (2011) Micromanagement of the mitochondrial apoptotic pathway by p53. Front Biosci (Landmark Ed) 16:749–758CrossRef
27.
go back to reference Bulucu F, Ocal R, Karadurmus N, Sahin M, Kenar L, Aydin A, Oktenli C, Koc B, Inal V, Yamanel L, Yaman H (2009) Effects of N-acetylcysteine, deferoxamine and selenium on Doxorubicin-induced hepatotoxicity. Biol Trace Elem Res 132(1–3):184–196. doi:10.1007/s12011-009-8377-y CrossRefPubMed Bulucu F, Ocal R, Karadurmus N, Sahin M, Kenar L, Aydin A, Oktenli C, Koc B, Inal V, Yamanel L, Yaman H (2009) Effects of N-acetylcysteine, deferoxamine and selenium on Doxorubicin-induced hepatotoxicity. Biol Trace Elem Res 132(1–3):184–196. doi:10.​1007/​s12011-009-8377-y CrossRefPubMed
29.
go back to reference Maciag AE, Chakrapani H, Saavedra JE, Morris NL, Holland RJ, Kosak KM, Shami PJ, Anderson LM, Keefer LK (2011) The nitric oxide prodrug JS-K is effective against non-small-cell lung cancer cells in vitro and in vivo: involvement of reactive oxygen species. J Pharm Exp Ther 336(2):313–320. doi:10.1124/jpet.110.174904 CrossRef Maciag AE, Chakrapani H, Saavedra JE, Morris NL, Holland RJ, Kosak KM, Shami PJ, Anderson LM, Keefer LK (2011) The nitric oxide prodrug JS-K is effective against non-small-cell lung cancer cells in vitro and in vivo: involvement of reactive oxygen species. J Pharm Exp Ther 336(2):313–320. doi:10.​1124/​jpet.​110.​174904 CrossRef
30.
go back to reference Shami PJ, Saavedra JE, Wang LY, Bonifant CL, Diwan BA, Singh SV, Gu Y, Fox SD, Buzard GS, Citro ML, Waterhouse DJ, Davies KM, Ji X, Keefer LK (2003) JS-K, a glutathione/glutathione S-transferase-activated nitric oxide donor of the diazeniumdiolate class with potent antineoplastic activity. Mol Cancer Ther 2(4):409–417PubMed Shami PJ, Saavedra JE, Wang LY, Bonifant CL, Diwan BA, Singh SV, Gu Y, Fox SD, Buzard GS, Citro ML, Waterhouse DJ, Davies KM, Ji X, Keefer LK (2003) JS-K, a glutathione/glutathione S-transferase-activated nitric oxide donor of the diazeniumdiolate class with potent antineoplastic activity. Mol Cancer Ther 2(4):409–417PubMed
33.
go back to reference Esparza-Lopez J, Ramos-Elias PA, Castro-Sanchez A, Rocha-Zavaleta L, Escobar-Arriaga E, Zentella-Dehesa A, Leon-Rodriguez E, Medina-Franco H, Ibarra-Sanchez Mde J (2016) Primary breast cancer cell culture yields intra-tumor heterogeneous subpopulations expressing exclusive patterns of receptor tyrosine kinases. BMC Cancer 16(1):740. doi:10.1186/s12885-016-2769-0 CrossRefPubMedPubMedCentral Esparza-Lopez J, Ramos-Elias PA, Castro-Sanchez A, Rocha-Zavaleta L, Escobar-Arriaga E, Zentella-Dehesa A, Leon-Rodriguez E, Medina-Franco H, Ibarra-Sanchez Mde J (2016) Primary breast cancer cell culture yields intra-tumor heterogeneous subpopulations expressing exclusive patterns of receptor tyrosine kinases. BMC Cancer 16(1):740. doi:10.​1186/​s12885-016-2769-0 CrossRefPubMedPubMedCentral
Metadata
Title
JS-K, a GST-activated nitric oxide donor prodrug, enhances chemo-sensitivity in renal carcinoma cells and prevents cardiac myocytes toxicity induced by Doxorubicin
Authors
Mingning Qiu
Longzhi Ke
Sai Zhang
Xin Zeng
Zesong Fang
Jianjun Liu
Publication date
01-08-2017
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 2/2017
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-017-3359-9

Other articles of this Issue 2/2017

Cancer Chemotherapy and Pharmacology 2/2017 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