Skip to main content
Top
Published in: Breast Cancer Research 1/2017

Open Access 01-12-2017 | Research article

Glucocorticoids induce production of reactive oxygen species/reactive nitrogen species and DNA damage through an iNOS mediated pathway in breast cancer

Authors: Renée L. Flaherty, Matthew Owen, Aidan Fagan-Murphy, Haya Intabli, David Healy, Anika Patel, Marcus C. Allen, Bhavik A. Patel, Melanie S. Flint

Published in: Breast Cancer Research | Issue 1/2017

Login to get access

Abstract

Background

Psychological stress increases the circulating levels of the stress hormones cortisol and norepinephrine (NE). Chronic exposure to elevated stress hormones has been linked to a reduced response to chemotherapy through induction of DNA damage. We hypothesize that stress hormone signalling may induce DNA damage through the production of reactive oxygen species (ROS)/reactive nitrogen species (RNS) and interference in DNA repair processes, promoting tumourigenesis.

Methods

Breast cancer cell lines were incubated with physiological levels of cortisol and NE in the presence and absence of receptor antagonists and inducible nitric oxide synthase (iNOS) inhibitors and DNA damage measured using phosphorylated γ-H2AX. The rate of DNA repair was measured using comet assays and electrochemical sensors were used to detect ROS/RNS in the cell lysates from cells exposed to stress hormones. A syngeneic mouse model was used to assess the presence of iNOS in mammary tumours in stressed versus control animals and expression of iNOS was examined using western blotting and qRT-PCR.

Results

Acute exposure to cortisol and NE significantly increased levels of ROS/RNS and DNA damage and this effect was diminished in the presence of receptor antagonists. Cortisol induced DNA damage and the production of RNS was further attenuated in the presence of an iNOS inhibitor. An increase in the expression of iNOS in response to psychological stress was observed in vivo and in cortisol-treated cells. Inhibition of glucocorticoid receptor-associated Src kinase also produced a decrease in cortisol-induced RNS.

Conclusion

These results demonstrate that glucocorticoids may interact with iNOS in a non-genomic manner to produce damaging levels of RNS, thus allowing an insight into the potential mechanisms by which psychological stress may impact breast cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Reiche EMV, Nunes SOV, Morimoto HK. Stress, depression, the immune system, and cancer. Lancet Oncol. 2004;5(10):617–25.CrossRefPubMed Reiche EMV, Nunes SOV, Morimoto HK. Stress, depression, the immune system, and cancer. Lancet Oncol. 2004;5(10):617–25.CrossRefPubMed
2.
go back to reference Costanzo ES, Sood AK, Lutgendorf SK. Biobehavioral influences on cancer progression. Immunol Allergy Clin N Am. 2011;31(1):109.CrossRef Costanzo ES, Sood AK, Lutgendorf SK. Biobehavioral influences on cancer progression. Immunol Allergy Clin N Am. 2011;31(1):109.CrossRef
3.
go back to reference Vanltallie TB. Stress: a risk factor for serious illness. Metab Clin Exp. 2002;51(6):40–5.CrossRef Vanltallie TB. Stress: a risk factor for serious illness. Metab Clin Exp. 2002;51(6):40–5.CrossRef
4.
5.
go back to reference Armaiz-Pena GN, Lutgendorf SK, Cole SW, Sood AK. Neuroendocrine modulation of cancer progression. Brain Behav Immun. 2009;23(1):10–5.CrossRefPubMed Armaiz-Pena GN, Lutgendorf SK, Cole SW, Sood AK. Neuroendocrine modulation of cancer progression. Brain Behav Immun. 2009;23(1):10–5.CrossRefPubMed
6.
go back to reference Wu W, Chaudhuri S, Brickley DR, Pang D, Karrison T, Conzen SD. Microarray analysis reveals glucocorticoid-regulated survival genes that are associated with inhibition of apoptosis in breast epithelial cells. Cancer Res. 2004;64(5):1757–64.CrossRefPubMed Wu W, Chaudhuri S, Brickley DR, Pang D, Karrison T, Conzen SD. Microarray analysis reveals glucocorticoid-regulated survival genes that are associated with inhibition of apoptosis in breast epithelial cells. Cancer Res. 2004;64(5):1757–64.CrossRefPubMed
8.
go back to reference Flint MS, Baum A, Chambers WH, Jenkins FJ. Induction of DNA damage, alteration of DNA repair and transcriptional activation by stress hormones. Psychoneuroendocrinology. 2007;32(5):470–9.CrossRefPubMed Flint MS, Baum A, Chambers WH, Jenkins FJ. Induction of DNA damage, alteration of DNA repair and transcriptional activation by stress hormones. Psychoneuroendocrinology. 2007;32(5):470–9.CrossRefPubMed
9.
go back to reference Hara MR, Kovacs JJ, Whalen EJ, Rajagopal S, Strachan RT, Grant W, et al. A stress response pathway regulates DNA damage through beta(2)-adrenoreceptors and beta-arrestin-1. Nature. 2011;477(7364):349–U129.CrossRefPubMedPubMedCentral Hara MR, Kovacs JJ, Whalen EJ, Rajagopal S, Strachan RT, Grant W, et al. A stress response pathway regulates DNA damage through beta(2)-adrenoreceptors and beta-arrestin-1. Nature. 2011;477(7364):349–U129.CrossRefPubMedPubMedCentral
10.
go back to reference Gidron Y, Russ K, Tissarchondou H, Warner J. The relation between psychological factors and DNA-damage: a critical review. Biol Psychol. 2006;72(3):291–304.CrossRefPubMed Gidron Y, Russ K, Tissarchondou H, Warner J. The relation between psychological factors and DNA-damage: a critical review. Biol Psychol. 2006;72(3):291–304.CrossRefPubMed
11.
go back to reference Bagchi D, Carryl OR, Tran MX, Bagchi M, Garg A, Milnes MM, et al. Acute and chronic stress-induced oxidative gastrointestinal mucosal injury in rats and protection by bismuth subsalicylate. Mol Cell Biochem. 1999;196(1-2):109–16.CrossRefPubMed Bagchi D, Carryl OR, Tran MX, Bagchi M, Garg A, Milnes MM, et al. Acute and chronic stress-induced oxidative gastrointestinal mucosal injury in rats and protection by bismuth subsalicylate. Mol Cell Biochem. 1999;196(1-2):109–16.CrossRefPubMed
12.
go back to reference Boonstra J, Post JA. Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells. Gene. 2004;337:1–13.CrossRefPubMed Boonstra J, Post JA. Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells. Gene. 2004;337:1–13.CrossRefPubMed
13.
go back to reference Nogueira V, Hay N. Molecular Pathways: Reactive oxygen species homeostasis in cancer cells and implications for cancer therapy. Clin Cancer Res. 2013;19(16):4309–14.CrossRefPubMedPubMedCentral Nogueira V, Hay N. Molecular Pathways: Reactive oxygen species homeostasis in cancer cells and implications for cancer therapy. Clin Cancer Res. 2013;19(16):4309–14.CrossRefPubMedPubMedCentral
14.
go back to reference Trachootham D, Zhou Y, Zhang H, Demizu Y, Chen Z, Pelicano H, et al. Selective killing of oncogenically transformed cells through a ROS-mediated mechanism by beta-phenylethyl isothiocyanate. Cancer Cell. 2006;10(3):241–52.CrossRefPubMed Trachootham D, Zhou Y, Zhang H, Demizu Y, Chen Z, Pelicano H, et al. Selective killing of oncogenically transformed cells through a ROS-mediated mechanism by beta-phenylethyl isothiocyanate. Cancer Cell. 2006;10(3):241–52.CrossRefPubMed
15.
16.
go back to reference Thomsen LL, Miles DW, Happerfield L, Bobrow LG, Knowles RG, Moncada S. Nitric-oxide synthase activity in human breast-cancer. Br J Cancer. 1995;72(1):41–4.CrossRefPubMedPubMedCentral Thomsen LL, Miles DW, Happerfield L, Bobrow LG, Knowles RG, Moncada S. Nitric-oxide synthase activity in human breast-cancer. Br J Cancer. 1995;72(1):41–4.CrossRefPubMedPubMedCentral
17.
go back to reference Fukumura D, Kashiwagi S, Jain RK. The role of nitric oxide in tumour progression. Nat Rev Cancer. 2006;6(7):521–34.CrossRefPubMed Fukumura D, Kashiwagi S, Jain RK. The role of nitric oxide in tumour progression. Nat Rev Cancer. 2006;6(7):521–34.CrossRefPubMed
18.
go back to reference Granados-Principal S, Liu Y, Guevara ML, Blanco E, Choi DS, Qian W, et al. Inhibition of iNOS as a novel effective targeted therapy against triple negative breast cancer. Breast Cancer Res. 2015;17(1):527.CrossRef Granados-Principal S, Liu Y, Guevara ML, Blanco E, Choi DS, Qian W, et al. Inhibition of iNOS as a novel effective targeted therapy against triple negative breast cancer. Breast Cancer Res. 2015;17(1):527.CrossRef
19.
go back to reference Kiecoltglaser JK, Stephens RE, Lipetz PD, Speicher CE, Glaser R. Distress and DNA-repair in human-lymphocytes. J Behav Med. 1985;8(4):311–20.CrossRef Kiecoltglaser JK, Stephens RE, Lipetz PD, Speicher CE, Glaser R. Distress and DNA-repair in human-lymphocytes. J Behav Med. 1985;8(4):311–20.CrossRef
20.
go back to reference Sikora AG, Gelbard A, Davies MA, Sano D, Ekmekcioglu S, Kwon J, et al. Targeted inhibition of inducible nitric oxide synthase inhibits growth of human melanoma in vivo and synergizes with chemotherapy. Clin Cancer Res. 2010;16(6):1834–44.CrossRefPubMedPubMedCentral Sikora AG, Gelbard A, Davies MA, Sano D, Ekmekcioglu S, Kwon J, et al. Targeted inhibition of inducible nitric oxide synthase inhibits growth of human melanoma in vivo and synergizes with chemotherapy. Clin Cancer Res. 2010;16(6):1834–44.CrossRefPubMedPubMedCentral
21.
go back to reference Reeder A, Attar M, Nazario L, Bathula C, Zhang A, Hochbaum D, et al. Stress hormones reduce the efficacy of paclitaxel in triple negative breast cancer through induction of DNA damage. Br J Cancer. 2015;112(9):1461–70.CrossRefPubMedPubMedCentral Reeder A, Attar M, Nazario L, Bathula C, Zhang A, Hochbaum D, et al. Stress hormones reduce the efficacy of paclitaxel in triple negative breast cancer through induction of DNA damage. Br J Cancer. 2015;112(9):1461–70.CrossRefPubMedPubMedCentral
22.
go back to reference Li Y, Sella C, Lemaitre F, Guille-Collignon M, Thouin L, Amatore C. Electrochemical detection of nitric oxide and peroxynitrite anion in microchannels at highly sensitive platinum-black coated electrodes. Application to ROS and RNS mixtures prior to biological investigations. Electrochim Acta. 2014;144:111–8.CrossRef Li Y, Sella C, Lemaitre F, Guille-Collignon M, Thouin L, Amatore C. Electrochemical detection of nitric oxide and peroxynitrite anion in microchannels at highly sensitive platinum-black coated electrodes. Application to ROS and RNS mixtures prior to biological investigations. Electrochim Acta. 2014;144:111–8.CrossRef
23.
go back to reference Budiu RA, Vlad AM, Nazario L, Bathula C, Cooper KL, Edmed J, et al. Restraint and social isolation stressors differentially regulate adaptive immunity and tumor angiogenesis in a breast cancer mouse model. Cancer Clin Oncol. 2016;6(1):12.CrossRef Budiu RA, Vlad AM, Nazario L, Bathula C, Cooper KL, Edmed J, et al. Restraint and social isolation stressors differentially regulate adaptive immunity and tumor angiogenesis in a breast cancer mouse model. Cancer Clin Oncol. 2016;6(1):12.CrossRef
26.
go back to reference Marchetti MC, Di Marco B, Cifone G, Migliorati G, Riccardi C. Dexamethasone-induced apoptosis of thymocytes: role of glucocorticoid receptor-associated Src kinase and caspase-8 activation. Blood. 2003;101(2):585–93.CrossRefPubMed Marchetti MC, Di Marco B, Cifone G, Migliorati G, Riccardi C. Dexamethasone-induced apoptosis of thymocytes: role of glucocorticoid receptor-associated Src kinase and caspase-8 activation. Blood. 2003;101(2):585–93.CrossRefPubMed
27.
go back to reference Croxtall JD, Choudhary Q, Flower RJ. Glucocorticoids act within minutes to inhibit recruitment of signalling factors to activated EGF receptors through a receptor-dependent, transcription-independent mechanism. Br J Pharmacol. 2000;130(2):289–98.CrossRefPubMedPubMedCentral Croxtall JD, Choudhary Q, Flower RJ. Glucocorticoids act within minutes to inhibit recruitment of signalling factors to activated EGF receptors through a receptor-dependent, transcription-independent mechanism. Br J Pharmacol. 2000;130(2):289–98.CrossRefPubMedPubMedCentral
28.
go back to reference Lu C, Heldt J-M, Guille-Collignon M, Lemaitre F, Jaouen G, Vessieres A, et al. Quantitative analyses of ROS and RNS production in breast cancer cell lines incubated with ferrocifens. ChemMedChem. 2014;9(6):1286–93.CrossRefPubMed Lu C, Heldt J-M, Guille-Collignon M, Lemaitre F, Jaouen G, Vessieres A, et al. Quantitative analyses of ROS and RNS production in breast cancer cell lines incubated with ferrocifens. ChemMedChem. 2014;9(6):1286–93.CrossRefPubMed
29.
go back to reference Ranganathan S, Krishnan A, Sivasithambaram ND. Significance of twist and iNOS expression in human breast carcinoma. Mol Cell Biochem. 2016;412(1-2):41–7.CrossRefPubMed Ranganathan S, Krishnan A, Sivasithambaram ND. Significance of twist and iNOS expression in human breast carcinoma. Mol Cell Biochem. 2016;412(1-2):41–7.CrossRefPubMed
30.
go back to reference Loibl S, Buck A, Strank C, von Minckwitz G, Roller M, Sinn HP, et al. The role of early expression of inducible nitric oxide synthase in human breast cancer. Eur J Cancer. 2005;41(2):265–71.CrossRefPubMed Loibl S, Buck A, Strank C, von Minckwitz G, Roller M, Sinn HP, et al. The role of early expression of inducible nitric oxide synthase in human breast cancer. Eur J Cancer. 2005;41(2):265–71.CrossRefPubMed
31.
go back to reference Bulut A, Erden E, Sak SD, Doruk H, Kursun N, Dincol D. Significance of inducible nitric oxide synthase expression in benign and malignant breast epithelium: an immunohistochemical study of 151 cases. Virchows Arch. 2005;447(1):24–30.CrossRefPubMed Bulut A, Erden E, Sak SD, Doruk H, Kursun N, Dincol D. Significance of inducible nitric oxide synthase expression in benign and malignant breast epithelium: an immunohistochemical study of 151 cases. Virchows Arch. 2005;447(1):24–30.CrossRefPubMed
32.
go back to reference Mocellin S, Bronte V, Nitti D. Nitric oxide, a double edged sword in cancer biology: searching for therapeutic opportunities. Med Res Rev. 2007;27(3):317–52.CrossRefPubMed Mocellin S, Bronte V, Nitti D. Nitric oxide, a double edged sword in cancer biology: searching for therapeutic opportunities. Med Res Rev. 2007;27(3):317–52.CrossRefPubMed
33.
go back to reference Xu WM, Liu LZ, Loizidou M, Ahmed M, Charles IG. The role of nitric oxide in cancer. Cell Res. 2002;12(5-6):311–20.CrossRefPubMed Xu WM, Liu LZ, Loizidou M, Ahmed M, Charles IG. The role of nitric oxide in cancer. Cell Res. 2002;12(5-6):311–20.CrossRefPubMed
34.
35.
go back to reference Song IH, Buttgereit F. Non-genomic glucocorticoid effects to provide the basis for new drug developments. Mol Cell Endocrinol. 2006;246(1-2):142–6.CrossRefPubMed Song IH, Buttgereit F. Non-genomic glucocorticoid effects to provide the basis for new drug developments. Mol Cell Endocrinol. 2006;246(1-2):142–6.CrossRefPubMed
36.
go back to reference Stahn C, Buttgereit F. Genomic and nongenomic effects of glucocorticoids. Nat Clin Pract Rheumatol. 2008;4(10):525–33.CrossRefPubMed Stahn C, Buttgereit F. Genomic and nongenomic effects of glucocorticoids. Nat Clin Pract Rheumatol. 2008;4(10):525–33.CrossRefPubMed
37.
go back to reference Ishizawar R, Parsons SJ. c-Src and cooperating partners in human cancer. Cancer Cell. 2004;6(3):209–14.CrossRefPubMed Ishizawar R, Parsons SJ. c-Src and cooperating partners in human cancer. Cancer Cell. 2004;6(3):209–14.CrossRefPubMed
38.
go back to reference Gonzalez L, Agullo-Ortuno MT, Garcia-Martinez JM, Calcabrini A, Gamallo C, Palacios J, et al. Role of c-Src in human MCF7 breast cancer cell tumorigenesis. J Biol Chem. 2006;281(30):20851–64.CrossRefPubMed Gonzalez L, Agullo-Ortuno MT, Garcia-Martinez JM, Calcabrini A, Gamallo C, Palacios J, et al. Role of c-Src in human MCF7 breast cancer cell tumorigenesis. J Biol Chem. 2006;281(30):20851–64.CrossRefPubMed
39.
go back to reference Larsen SL, Laenkholm A-V, Duun-Henriksen AK, Bak M, Lykkesfeldt AE, Kirkegaard T. Src drives growth of antiestrogen resistant breast cancer cell lines and is a marker for reduced benefit of tamoxifen treatment. PLoS One. 2015;10(2):e0118346. doi:10.1371/journal.pone.0118346. Larsen SL, Laenkholm A-V, Duun-Henriksen AK, Bak M, Lykkesfeldt AE, Kirkegaard T. Src drives growth of antiestrogen resistant breast cancer cell lines and is a marker for reduced benefit of tamoxifen treatment. PLoS One. 2015;10(2):e0118346. doi:10.​1371/​journal.​pone.​0118346.
40.
go back to reference Armaiz-Pena GN, Allen JK, Cruz A, Stone RL, Nick AM, Lin YG, et al. Src activation by beta-adrenoreceptors is a key switch for tumour metastasis. Nat Commun. 2013;4:11. Armaiz-Pena GN, Allen JK, Cruz A, Stone RL, Nick AM, Lin YG, et al. Src activation by beta-adrenoreceptors is a key switch for tumour metastasis. Nat Commun. 2013;4:11.
41.
go back to reference Tyryshkin A, Gorgun FM, Fattah EA, Mazumdar T, Pandit L, Zeng SY, et al. Src Kinase-mediated phosphorylation stabilizes inducible nitric-oxide synthase in normal cells and cancer cells. J Biol Chem. 2010;285(1):784–92.CrossRefPubMed Tyryshkin A, Gorgun FM, Fattah EA, Mazumdar T, Pandit L, Zeng SY, et al. Src Kinase-mediated phosphorylation stabilizes inducible nitric-oxide synthase in normal cells and cancer cells. J Biol Chem. 2010;285(1):784–92.CrossRefPubMed
42.
go back to reference Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, et al. The selectivity of protein kinase inhibitors: a further update. Biochem J. 2007;408:297–315.CrossRefPubMedPubMedCentral Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, et al. The selectivity of protein kinase inhibitors: a further update. Biochem J. 2007;408:297–315.CrossRefPubMedPubMedCentral
Metadata
Title
Glucocorticoids induce production of reactive oxygen species/reactive nitrogen species and DNA damage through an iNOS mediated pathway in breast cancer
Authors
Renée L. Flaherty
Matthew Owen
Aidan Fagan-Murphy
Haya Intabli
David Healy
Anika Patel
Marcus C. Allen
Bhavik A. Patel
Melanie S. Flint
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2017
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-017-0823-8

Other articles of this Issue 1/2017

Breast Cancer Research 1/2017 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