Skip to main content
Top
Published in: BMC Cancer 1/2016

Open Access 01-12-2016 | Research article

Molecular features of the cytotoxicity of an NHE inhibitor: Evidence of mitochondrial alterations, ROS overproduction and DNA damage

Authors: Francesca Aredia, Sebastian Czaplinski, Simone Fulda, A. Ivana Scovassi

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

NH exchangers (NHEs) play a crucial role in regulating intra/extracellular pH, which is altered in cancer cells, and are therefore suitable targets to alter cancer cell metabolism in order to inhibit cell survival and proliferation. Among NHE inhibitors, amiloride family members are commonly used in clinical practice as diuretics; we focused on the amiloride HMA, reporting a net cytotoxic effect on a panel of human cancer cell lines; now we aim to provide new insights into the molecular events leading to cell death by HMA.

Methods

Colon cancer cell lines were treated with HMA and analysed with: morphological and cellular assays for cell viability and death, and autophagy; biochemical approaches to evaluate mitochondrial function and ROS production; in situ detection of DNA damage; molecular tools to silence crucial autophagy/necroptosis factors.

Results

HMA affects cellular morphology, alters mitochondrial structure and function, causes an increase in ROS, which is detrimental to DNA integrity, stimulates poly(ADP-ribose) synthesis, activates RIPK3-dependent death and triggers autophagy, which is unable to rescue cell survival. These features are hot points of an intricate network of processes, including necroptosis and autophagy, regulating the homeostasis between survival and death.

Conclusion

Our results allow the identification of multiple events leading to cell death in cancer cells treated with HMA. The here-defined intricate network activated by HMA could be instrumental to selectively target the key players of each pathway in the attempt to improve the global response to HMA. Our data could be the starting point for developing a newly designed targeted therapy.
Literature
1.
go back to reference Spill F, Reynolds DS, Kamm RD, Zaman MH. Impact of the physical microenvironment on tumor progression and metastasis. Curr Opin Biotechnol. 2016;40:41–8.CrossRefPubMed Spill F, Reynolds DS, Kamm RD, Zaman MH. Impact of the physical microenvironment on tumor progression and metastasis. Curr Opin Biotechnol. 2016;40:41–8.CrossRefPubMed
3.
go back to reference Aredia F, Scovassi AI. Multiple effects of intracellular pH modulation in cancer cells. Cancer Cell Microenv. 2014;1:72–9. Aredia F, Scovassi AI. Multiple effects of intracellular pH modulation in cancer cells. Cancer Cell Microenv. 2014;1:72–9.
4.
go back to reference Spugnini EP, Sonveaux P, Stock C, Perez-Sayans M, De Milito A, Avnet S, et al. Proton channels and exchangers in cancer. Biochim Biophys Acta. 1848;2015:2715–26. Spugnini EP, Sonveaux P, Stock C, Perez-Sayans M, De Milito A, Avnet S, et al. Proton channels and exchangers in cancer. Biochim Biophys Acta. 1848;2015:2715–26.
5.
go back to reference Harguindey S, Arranz JL, Polo Orozco JD, Rauch C, Fais S, Cardone RA, et al. Cariporide and other new and powerful NHE1 inhibitors as potentially selective anticancer drugs--an integral molecular/biochemical/metabolic/clinical approach after one hundred years of cancer research. J Transl Med. 2013;11:282.CrossRefPubMedPubMedCentral Harguindey S, Arranz JL, Polo Orozco JD, Rauch C, Fais S, Cardone RA, et al. Cariporide and other new and powerful NHE1 inhibitors as potentially selective anticancer drugs--an integral molecular/biochemical/metabolic/clinical approach after one hundred years of cancer research. J Transl Med. 2013;11:282.CrossRefPubMedPubMedCentral
6.
go back to reference Kimura K, Nakao K, Shibata Y, Sone T, Takayama T, Fukuzawa S, et al. Randomized controlled trial of TY-51924, a novel hydrophilic NHE inhibitor, in acute myocardial infarction. J Cardiol. 2016;67:307–13.CrossRefPubMed Kimura K, Nakao K, Shibata Y, Sone T, Takayama T, Fukuzawa S, et al. Randomized controlled trial of TY-51924, a novel hydrophilic NHE inhibitor, in acute myocardial infarction. J Cardiol. 2016;67:307–13.CrossRefPubMed
7.
go back to reference Giansanti V, Santamaria G, Torriglia A, Aredia F, Scovassi AI, Bottiroli G, et al. Fluorescence properties of the Na+/H + exchanger inhibitor HMA (5-(N, N-hexamethylene)amiloride) are modulated by intracellular pH. Eur J Histochem. 2012;56:e3.CrossRefPubMedPubMedCentral Giansanti V, Santamaria G, Torriglia A, Aredia F, Scovassi AI, Bottiroli G, et al. Fluorescence properties of the Na+/H + exchanger inhibitor HMA (5-(N, N-hexamethylene)amiloride) are modulated by intracellular pH. Eur J Histochem. 2012;56:e3.CrossRefPubMedPubMedCentral
8.
go back to reference Giansanti V, Rodriguez GEV, Savoldelli M, Gioia R, Forlino A, Mazzini G, et al. Characterization of stress response in human retinal epithelial cells. J Cell Mol Med. 2013;17:103–15.CrossRefPubMed Giansanti V, Rodriguez GEV, Savoldelli M, Gioia R, Forlino A, Mazzini G, et al. Characterization of stress response in human retinal epithelial cells. J Cell Mol Med. 2013;17:103–15.CrossRefPubMed
9.
go back to reference Rowson-Hodel AR, Berg AL, Wald JH, Hatakeyama J, VanderVorst K, Curiel DA, et al. Hexamethylene amiloride engages a novel reactive oxygen species- and lysosome-dependent programmed necrotic mechanism to selectively target breast cancer cells. Cancer Lett. 2016;375:62–72.CrossRefPubMed Rowson-Hodel AR, Berg AL, Wald JH, Hatakeyama J, VanderVorst K, Curiel DA, et al. Hexamethylene amiloride engages a novel reactive oxygen species- and lysosome-dependent programmed necrotic mechanism to selectively target breast cancer cells. Cancer Lett. 2016;375:62–72.CrossRefPubMed
10.
go back to reference Aredia F, Giansanti V, Mazzini G, Savio M, Ortiz LMG, Jaadane I, et al. Multiple effects of the Na(+)/H (+) antiporter inhibitor HMA on cancer cells. Apoptosis. 2013;18:1586–98.CrossRefPubMed Aredia F, Giansanti V, Mazzini G, Savio M, Ortiz LMG, Jaadane I, et al. Multiple effects of the Na(+)/H (+) antiporter inhibitor HMA on cancer cells. Apoptosis. 2013;18:1586–98.CrossRefPubMed
11.
go back to reference Leon LJ, Pasupuleti N, Gorin F, Carraway KL. A cell-permeant amiloride derivative induces caspase-independent, AIF-mediated programmed necrotic death of breast cancer cells. PLoS One. 2013;8:e63038.CrossRefPubMedPubMedCentral Leon LJ, Pasupuleti N, Gorin F, Carraway KL. A cell-permeant amiloride derivative induces caspase-independent, AIF-mediated programmed necrotic death of breast cancer cells. PLoS One. 2013;8:e63038.CrossRefPubMedPubMedCentral
12.
go back to reference Pasupuleti N, Leon L, Carraway KL, Gorin F. 5-Benzylglycinyl-amiloride kills proliferating and nonproliferating malignant glioma cells through caspase-independent necroptosis mediated by apoptosis-inducing factor. J Pharmacol Exp Ther. 2013;344:600–15.CrossRefPubMedPubMedCentral Pasupuleti N, Leon L, Carraway KL, Gorin F. 5-Benzylglycinyl-amiloride kills proliferating and nonproliferating malignant glioma cells through caspase-independent necroptosis mediated by apoptosis-inducing factor. J Pharmacol Exp Ther. 2013;344:600–15.CrossRefPubMedPubMedCentral
13.
go back to reference Hawn MT, Umar A, Carethers JM, Marra G, Kunkel TA, Boland CR, et al. Evidence for a connection between the mismatch repair system and the G2 cell cycle checkpoint. Cancer Res. 1995;55:3721–5.PubMed Hawn MT, Umar A, Carethers JM, Marra G, Kunkel TA, Boland CR, et al. Evidence for a connection between the mismatch repair system and the G2 cell cycle checkpoint. Cancer Res. 1995;55:3721–5.PubMed
14.
go back to reference Croce AC, Bottiroli G, Supino R, Favini E, Zuco V, Zunino F. Subcellular localization of the camptothecin analogues, topotecan and gimatecan. Biochem Pharmacol. 2004;67:1035–45.CrossRefPubMed Croce AC, Bottiroli G, Supino R, Favini E, Zuco V, Zunino F. Subcellular localization of the camptothecin analogues, topotecan and gimatecan. Biochem Pharmacol. 2004;67:1035–45.CrossRefPubMed
15.
go back to reference Malatesta M, Giagnacovo M, Costanzo M, Conti B, Genta I, Dorati R, et al. Diaminobenzidine photoconversion is a suitable tool for tracking the intracellular location of fluorescently labelled nanoparticles at transmission electron microscopy. Eur J Histochem. 2012;56:e20.CrossRefPubMedPubMedCentral Malatesta M, Giagnacovo M, Costanzo M, Conti B, Genta I, Dorati R, et al. Diaminobenzidine photoconversion is a suitable tool for tracking the intracellular location of fluorescently labelled nanoparticles at transmission electron microscopy. Eur J Histochem. 2012;56:e20.CrossRefPubMedPubMedCentral
16.
go back to reference Guamán Ortiz LM, Tillhon M, Parks M, Dutto I, Prosperi E, Savio M, et al. Multiple effects of berberine derivatives on colon cancer cells. Biomed Res Int. 2014;2014:924585.CrossRefPubMedPubMedCentral Guamán Ortiz LM, Tillhon M, Parks M, Dutto I, Prosperi E, Savio M, et al. Multiple effects of berberine derivatives on colon cancer cells. Biomed Res Int. 2014;2014:924585.CrossRefPubMedPubMedCentral
17.
go back to reference Grecchi S, Mazzini G, Lisa A, Armentero M-T, Bergamaschi R, Romani A, et al. Search for cellular stress biomarkers in lymphocytes from patients with multiple sclerosis: a pilot study. PLoS One. 2012;7:e44935.CrossRefPubMedPubMedCentral Grecchi S, Mazzini G, Lisa A, Armentero M-T, Bergamaschi R, Romani A, et al. Search for cellular stress biomarkers in lymphocytes from patients with multiple sclerosis: a pilot study. PLoS One. 2012;7:e44935.CrossRefPubMedPubMedCentral
18.
go back to reference Briand JP, Van Dorsselaer A, Raboy B, Muller S. Total chemical synthesis of ubiquitin using BOP reagent: biochemical and immunochemical properties of the purified synthetic product. Pept Res. 1989;2:381–8.PubMed Briand JP, Van Dorsselaer A, Raboy B, Muller S. Total chemical synthesis of ubiquitin using BOP reagent: biochemical and immunochemical properties of the purified synthetic product. Pept Res. 1989;2:381–8.PubMed
19.
go back to reference Sandell JH, Masland RH. Photoconversion of some fluorescent markers to a diaminobenzidine product. J Histochem Cytochem. 1988;36:555–9.CrossRefPubMed Sandell JH, Masland RH. Photoconversion of some fluorescent markers to a diaminobenzidine product. J Histochem Cytochem. 1988;36:555–9.CrossRefPubMed
20.
21.
go back to reference Wang H, Joseph JA. Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic Biol Med. 1999;27:612–6.CrossRefPubMed Wang H, Joseph JA. Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic Biol Med. 1999;27:612–6.CrossRefPubMed
22.
go back to reference Aguilera-Aguirre L, Bacsi A, Radak Z, Hazra TK, Mitra S, Sur S, et al. Innate inflammation induced by the 8-oxoguanine DNA glycosylase-1-KRAS-NF-κB pathway. J Immunol. 2014;193:4643–53.CrossRefPubMedPubMedCentral Aguilera-Aguirre L, Bacsi A, Radak Z, Hazra TK, Mitra S, Sur S, et al. Innate inflammation induced by the 8-oxoguanine DNA glycosylase-1-KRAS-NF-κB pathway. J Immunol. 2014;193:4643–53.CrossRefPubMedPubMedCentral
23.
go back to reference Campalans A, Kortulewski T, Amouroux R, Menoni H, Vermeulen W, Radicella JP. Distinct spatiotemporal patterns and PARP dependence of XRCC1 recruitment to single-strand break and base excision repair. Nucleic Acids Res. 2013;41:3115–29.CrossRefPubMedPubMedCentral Campalans A, Kortulewski T, Amouroux R, Menoni H, Vermeulen W, Radicella JP. Distinct spatiotemporal patterns and PARP dependence of XRCC1 recruitment to single-strand break and base excision repair. Nucleic Acids Res. 2013;41:3115–29.CrossRefPubMedPubMedCentral
24.
go back to reference Fernandez-Capetillo O, Chen H-T, Celeste A, Ward I, Romanienko PJ, Morales JC, et al. DNA damage-induced G2-M checkpoint activation by histone H2AX and 53BP1. Nat Cell Biol. 2002;4:993–7.CrossRefPubMed Fernandez-Capetillo O, Chen H-T, Celeste A, Ward I, Romanienko PJ, Morales JC, et al. DNA damage-induced G2-M checkpoint activation by histone H2AX and 53BP1. Nat Cell Biol. 2002;4:993–7.CrossRefPubMed
25.
go back to reference Aredia F, Scovassi AI. Poly(ADP-ribose): a signaling molecule in different paradigms of cell death. Biochem Pharmacol. 2014;92:157–63.CrossRefPubMed Aredia F, Scovassi AI. Poly(ADP-ribose): a signaling molecule in different paradigms of cell death. Biochem Pharmacol. 2014;92:157–63.CrossRefPubMed
27.
go back to reference Moriwaki K, Bertin J, Gough PJ, Orlowski GM, Chan FK. Differential roles of RIPK1 and RIPK3 in TNF-induced necroptosis and chemotherapeutic agent-induced cell death. Cell Death Dis. 2015;6:e1636.CrossRefPubMedPubMedCentral Moriwaki K, Bertin J, Gough PJ, Orlowski GM, Chan FK. Differential roles of RIPK1 and RIPK3 in TNF-induced necroptosis and chemotherapeutic agent-induced cell death. Cell Death Dis. 2015;6:e1636.CrossRefPubMedPubMedCentral
28.
go back to reference Lalaoui N, Lindqvist LM, Sandow JJ, Ekert PG. The molecular relationships between apoptosis, autophagy and necroptosis. Semin Cell Dev Biol. 2015;39:63–9.CrossRefPubMed Lalaoui N, Lindqvist LM, Sandow JJ, Ekert PG. The molecular relationships between apoptosis, autophagy and necroptosis. Semin Cell Dev Biol. 2015;39:63–9.CrossRefPubMed
29.
go back to reference Nagelkerke A, Bussink J, Sweep FCGJ, Span PN. The unfolded protein response as a target for cancer therapy. Biochim Biophys Acta. 1846;2014:277–84. Nagelkerke A, Bussink J, Sweep FCGJ, Span PN. The unfolded protein response as a target for cancer therapy. Biochim Biophys Acta. 1846;2014:277–84.
30.
go back to reference Virág L, Robaszkiewicz A, Rodriguez-Vargas JM, Oliver FJ. Poly(ADP-ribose) signaling in cell death. Mol Aspects Med. 2013;34:1153–67.CrossRefPubMed Virág L, Robaszkiewicz A, Rodriguez-Vargas JM, Oliver FJ. Poly(ADP-ribose) signaling in cell death. Mol Aspects Med. 2013;34:1153–67.CrossRefPubMed
31.
go back to reference Liberti MV, Locasale JW. The Warburg effect: How does it benefit cancer cells? Trends Biochem Sci. 2016;41:211–8.CrossRefPubMed Liberti MV, Locasale JW. The Warburg effect: How does it benefit cancer cells? Trends Biochem Sci. 2016;41:211–8.CrossRefPubMed
32.
go back to reference Matthews H, Ranson M, Kelso MJ. Anti-tumour/metastasis effects of the potassium-sparing diuretic amiloride: an orally active anti-cancer drug waiting for its call-of-duty? Int J Cancer. 2011;129:2051–61.CrossRefPubMed Matthews H, Ranson M, Kelso MJ. Anti-tumour/metastasis effects of the potassium-sparing diuretic amiloride: an orally active anti-cancer drug waiting for its call-of-duty? Int J Cancer. 2011;129:2051–61.CrossRefPubMed
33.
go back to reference Altairac S, Zeggai S, Perani P, Courtois Y, Torriglia A. Apoptosis induced by Na+/H+ antiport inhibition activates the LEI/L-DNase II pathway. Cell Death Differ. 2003;10:548–57.CrossRefPubMed Altairac S, Zeggai S, Perani P, Courtois Y, Torriglia A. Apoptosis induced by Na+/H+ antiport inhibition activates the LEI/L-DNase II pathway. Cell Death Differ. 2003;10:548–57.CrossRefPubMed
34.
go back to reference Basello DA, Scovassi AI. Poly(ADP-ribosylation) and neurodegenerative disorders. Mitochondrion. 2015;24:56–63.CrossRefPubMed Basello DA, Scovassi AI. Poly(ADP-ribosylation) and neurodegenerative disorders. Mitochondrion. 2015;24:56–63.CrossRefPubMed
35.
go back to reference Acevedo-Olvera LF, Diaz-Garcia H, Parra-Barrera A, Caceres-Perez AA, Gutierrez-Iglesias G, Rangel-Corona R, et al. Inhibition of the Na+/H+ antiporter induces cell death in TF-1 erythroleukemia cells stimulated by the stem cell factor. Cytokine. 2015;75:142–50.CrossRefPubMed Acevedo-Olvera LF, Diaz-Garcia H, Parra-Barrera A, Caceres-Perez AA, Gutierrez-Iglesias G, Rangel-Corona R, et al. Inhibition of the Na+/H+ antiporter induces cell death in TF-1 erythroleukemia cells stimulated by the stem cell factor. Cytokine. 2015;75:142–50.CrossRefPubMed
36.
go back to reference Pasupuleti N, Grodzki AC, Gorin F. Mis-trafficking of endosomal urokinase proteins triggers drug-induced glioma nonapoptotic cell death. Mol Pharmacol. 2015;87:683–96.CrossRefPubMedPubMedCentral Pasupuleti N, Grodzki AC, Gorin F. Mis-trafficking of endosomal urokinase proteins triggers drug-induced glioma nonapoptotic cell death. Mol Pharmacol. 2015;87:683–96.CrossRefPubMedPubMedCentral
37.
go back to reference Murai M, Murakami S, Ito T, Miyoshi H. Amilorides bind to the quinone binding pocket of bovine mitochondrial complex I. Biochemistry. 2015;54:2739–46.CrossRefPubMed Murai M, Murakami S, Ito T, Miyoshi H. Amilorides bind to the quinone binding pocket of bovine mitochondrial complex I. Biochemistry. 2015;54:2739–46.CrossRefPubMed
38.
go back to reference Ahmad T, Aggarwal K, Pattnaik B, Mukherjee S, Sethi T, Tiwari BK, et al. Computational classification of mitochondrial shapes reflects stress and redox state. Cell Death Dis. 2013;4:e461.CrossRefPubMedPubMedCentral Ahmad T, Aggarwal K, Pattnaik B, Mukherjee S, Sethi T, Tiwari BK, et al. Computational classification of mitochondrial shapes reflects stress and redox state. Cell Death Dis. 2013;4:e461.CrossRefPubMedPubMedCentral
39.
go back to reference Holmström KM, Finkel T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat Rev Mol Cell Biol. 2014;15:411–21.CrossRefPubMed Holmström KM, Finkel T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat Rev Mol Cell Biol. 2014;15:411–21.CrossRefPubMed
40.
go back to reference Wang Y, Nartiss Y, Steipe B, McQuibban GA, Kim PK. ROS-induced mitochondrial depolarization initiates PARK2/PARKIN-dependent mitochondrial degradation by autophagy. Autophagy. 2012;8:1462–76.CrossRefPubMed Wang Y, Nartiss Y, Steipe B, McQuibban GA, Kim PK. ROS-induced mitochondrial depolarization initiates PARK2/PARKIN-dependent mitochondrial degradation by autophagy. Autophagy. 2012;8:1462–76.CrossRefPubMed
41.
42.
go back to reference Wang L, Yeung JH, Hu T, Lee WY, Lu L, Zhang L, et al. Dihydrotanshinone induces p53-independent but ROS-dependent apoptosis in colon cancer cells. Life Sci. 2013;93:344–51.CrossRefPubMed Wang L, Yeung JH, Hu T, Lee WY, Lu L, Zhang L, et al. Dihydrotanshinone induces p53-independent but ROS-dependent apoptosis in colon cancer cells. Life Sci. 2013;93:344–51.CrossRefPubMed
43.
go back to reference Khamphio M, Barusrux S, Weerapreeyakul N. Sesamol induces mitochondrial apoptosis pathway in HCT-116 human colon cancer cells via pro-oxidant effect. Life Sci. 2016;158:46–56.CrossRefPubMed Khamphio M, Barusrux S, Weerapreeyakul N. Sesamol induces mitochondrial apoptosis pathway in HCT-116 human colon cancer cells via pro-oxidant effect. Life Sci. 2016;158:46–56.CrossRefPubMed
44.
go back to reference Yang Y, Luo H, Hui K, Ci Y, Shi K, Chen G, et al. Selenite-induced autophagy antagonizes apoptosis in colorectal cancer cells in vitro and in vivo. Oncol Rep. 2016;35:1255–64.PubMed Yang Y, Luo H, Hui K, Ci Y, Shi K, Chen G, et al. Selenite-induced autophagy antagonizes apoptosis in colorectal cancer cells in vitro and in vivo. Oncol Rep. 2016;35:1255–64.PubMed
45.
go back to reference Galluzzi L, Kroemer G. Necroptosis: a specialized pathway of programmed necrosis. Cell. 2008;135:1161–3.CrossRefPubMed Galluzzi L, Kroemer G. Necroptosis: a specialized pathway of programmed necrosis. Cell. 2008;135:1161–3.CrossRefPubMed
47.
48.
go back to reference Zhao Y, Qu T, Wang P, Li X, Qiang J, Xia Z, et al. Unravelling the relationship between macroautophagy and mitochondrial ROS in cancer therapy. Apoptosis. 2016;21:517–31.CrossRefPubMed Zhao Y, Qu T, Wang P, Li X, Qiang J, Xia Z, et al. Unravelling the relationship between macroautophagy and mitochondrial ROS in cancer therapy. Apoptosis. 2016;21:517–31.CrossRefPubMed
Metadata
Title
Molecular features of the cytotoxicity of an NHE inhibitor: Evidence of mitochondrial alterations, ROS overproduction and DNA damage
Authors
Francesca Aredia
Sebastian Czaplinski
Simone Fulda
A. Ivana Scovassi
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2016
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2878-9

Other articles of this Issue 1/2016

BMC Cancer 1/2016 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