Skip to main content
Top
Published in: Breast Cancer Research and Treatment 1/2018

01-11-2018 | Preclinical study

Oestrogen receptor-regulated glutathione S-transferase mu 3 expression attenuates hydrogen peroxide-induced cytotoxicity, which confers tamoxifen resistance on breast cancer cells

Authors: Juo-Han Lin, Shih-Hsin Tu, Li-Ching Chen, Chi-Cheng Huang, Hang-Lung Chang, Tzu-Chun Cheng, Hui-Wen Chang, Chih-Hsiung Wu, Han-Chung Wu, Yuan-Soon Ho

Published in: Breast Cancer Research and Treatment | Issue 1/2018

Login to get access

Abstract

Purpose

Glutathione S-transferase mu 3 (GSTM3) is an enzyme involving in the detoxification of electrophilic compounds by conjugation with glutathione. Higher GSTM3 mRNA levels were reported in patients with ERα-positive breast cancer who received only tamoxifen therapy after surgery. Thus, this study aimed to clarify the oncogenic characteristics of GSTM3 in breast cancer and the mechanism of tamoxifen resistance.

Methods

GSTM3 expression in human breast tumour tissues (n = 227) was analysed by RT-PCR and quantitative PCR. Western blot, promoter activity assays, and chromatin immunoprecipitation (ChIP) assays were used to investigate the mechanism of GSTM3 gene regulation. Hydrogen peroxide (H2O2)-induced cytotoxicity in breast cancer cells was detected by MTT assays and flow cytometry. The oncogenic characteristics of GSTM3 in MCF-7 cells were examined by siRNA knockdown in soft agar assays and a xenograft animal model.

Results

GSTM3 mRNA was highly expressed in ER- and HER2-positive breast cancers. Moreover, patients who received adjuvant Herceptin had increased GSTM3 mRNA levels in tumour tissue. Oestrogen-activated GSTM3 gene expression through ERα-mediated recruitment of SP1, EP300, and AP-1 complexes. GSTM3-silenced MCF-7 cells were more sensitive to H2O2, with significantly inhibited proliferation and colony formation abilities. Tamoxifen-resistant (Tam-R) cells lacking GSTM3 showed enhanced sensitivity to H2O2, but this result was contrary to that obtained after short-term tamoxifen exposure. The animal model suggested that GSTM3 silencing might suppress the tumourigenic ability of MCF-7 cells and increase tumour cell apoptosis.

Conclusions

ROS production is one mechanism by which cancer drugs kill tumour cells, and according to our evidence, GSTM3 may play an important role in preventing breast cancer treatment-induced cellular cytotoxicity.
Appendix
Available only for authorised users
Literature
1.
go back to reference Pink JJ, Jordan VC (1996) Models of estrogen receptor regulation by estrogens and antiestrogens in breast cancer cell lines. Cancer Res 56:2321–2330PubMed Pink JJ, Jordan VC (1996) Models of estrogen receptor regulation by estrogens and antiestrogens in breast cancer cell lines. Cancer Res 56:2321–2330PubMed
3.
go back to reference Marino M, Galluzzo P, Ascenzi P (2006) Estrogen signaling multiple pathways to impact gene transcription. Curr Genomics 7:497–508CrossRef Marino M, Galluzzo P, Ascenzi P (2006) Estrogen signaling multiple pathways to impact gene transcription. Curr Genomics 7:497–508CrossRef
4.
go back to reference Zhu BT, Conney AH (1998) Functional role of estrogen metabolism in target cells: review and perspectives. Carcinogenesis 19:1–27CrossRef Zhu BT, Conney AH (1998) Functional role of estrogen metabolism in target cells: review and perspectives. Carcinogenesis 19:1–27CrossRef
5.
go back to reference Feigelson HS, Henderson BE (1996) Estrogens and breast cancer. Carcinogenesis 17:2279–2284CrossRef Feigelson HS, Henderson BE (1996) Estrogens and breast cancer. Carcinogenesis 17:2279–2284CrossRef
6.
go back to reference Strange RC, Spiteri MA, Ramachandran S, Fryer AA (2001) Glutathione-S-transferase family of enzymes. Mutat Res 482:21–26CrossRef Strange RC, Spiteri MA, Ramachandran S, Fryer AA (2001) Glutathione-S-transferase family of enzymes. Mutat Res 482:21–26CrossRef
7.
go back to reference Chasseaud LF (1979) The role of glutathione and glutathione S-transferases in the metabolism of chemical carcinogens and other electrophilic agents. Adv Cancer Res 29:175–274CrossRef Chasseaud LF (1979) The role of glutathione and glutathione S-transferases in the metabolism of chemical carcinogens and other electrophilic agents. Adv Cancer Res 29:175–274CrossRef
8.
go back to reference Shea TC, Claflin G, Comstock KE, Sanderson BJ, Burstein NA, Keenan EJ et al (1990) Glutathione transferase activity and isoenzyme composition in primary human breast cancers. Cancer Res 50:6848–6853PubMed Shea TC, Claflin G, Comstock KE, Sanderson BJ, Burstein NA, Keenan EJ et al (1990) Glutathione transferase activity and isoenzyme composition in primary human breast cancers. Cancer Res 50:6848–6853PubMed
9.
go back to reference Xu S, Wang Y, Roe B, Pearson WR (1998) Characterization of the human class Mu glutathione S-transferase gene cluster and the GSTM1 deletion. J Biol Chem 273:3517–3527CrossRef Xu S, Wang Y, Roe B, Pearson WR (1998) Characterization of the human class Mu glutathione S-transferase gene cluster and the GSTM1 deletion. J Biol Chem 273:3517–3527CrossRef
10.
go back to reference Inskip A, Elexperu-Camiruaga J, Buxton N, Dias PS, MacIntosh J, Campbell D et al (1995) Identification of polymorphism at the glutathione S-transferase, GSTM3 locus: evidence for linkage with GSTM1*A. Biochem J 312(Pt 3):713–716CrossRef Inskip A, Elexperu-Camiruaga J, Buxton N, Dias PS, MacIntosh J, Campbell D et al (1995) Identification of polymorphism at the glutathione S-transferase, GSTM3 locus: evidence for linkage with GSTM1*A. Biochem J 312(Pt 3):713–716CrossRef
11.
go back to reference Jourenkova-Mironova N, Voho A, Bouchardy C, Wikman H, Dayer P, Benhamou S et al (1999) Glutathione S-transferase GSTM1, GSTM3, GSTP1 and GSTT1 genotypes and the risk of smoking-related oral and pharyngeal cancers. Int J Cancer 81:44–48CrossRef Jourenkova-Mironova N, Voho A, Bouchardy C, Wikman H, Dayer P, Benhamou S et al (1999) Glutathione S-transferase GSTM1, GSTM3, GSTP1 and GSTT1 genotypes and the risk of smoking-related oral and pharyngeal cancers. Int J Cancer 81:44–48CrossRef
12.
go back to reference Yengi L, Inskip A, Gilford J, Alldersea J, Bailey L, Smith A et al (1996) Polymorphism at the glutathione S-transferase locus GSTM3: interactions with cytochrome P450 and glutathione S-transferase genotypes as risk factors for multiple cutaneous basal cell carcinoma. Cancer Res 56:1974–1977PubMed Yengi L, Inskip A, Gilford J, Alldersea J, Bailey L, Smith A et al (1996) Polymorphism at the glutathione S-transferase locus GSTM3: interactions with cytochrome P450 and glutathione S-transferase genotypes as risk factors for multiple cutaneous basal cell carcinoma. Cancer Res 56:1974–1977PubMed
18.
go back to reference Huerta S, Harris DM, Jazirehi A, Bonavida B, Elashoff D, Livingston EH et al (2003) Gene expression profile of metastatic colon cancer cells resistant to cisplatin-induced apoptosis. Int J Oncol 22:663–670PubMed Huerta S, Harris DM, Jazirehi A, Bonavida B, Elashoff D, Livingston EH et al (2003) Gene expression profile of metastatic colon cancer cells resistant to cisplatin-induced apoptosis. Int J Oncol 22:663–670PubMed
20.
25.
go back to reference Gordon MN, Osterburg HH, May PC, Finch CE (1986) Effective oral administration of 17 beta-estradiol to female C57BL/6J mice through the drinking water. Biol Reprod 35:1088–1095CrossRef Gordon MN, Osterburg HH, May PC, Finch CE (1986) Effective oral administration of 17 beta-estradiol to female C57BL/6J mice through the drinking water. Biol Reprod 35:1088–1095CrossRef
26.
go back to reference Kang JS, Kang MR, Han SB, Yoon WK, Kim JH, Lee TC et al (2009) Low dose estrogen supplementation reduces mortality of mice in estrogen-dependent human tumor xenograft model. Biol Pharm Bull 32:150–152CrossRef Kang JS, Kang MR, Han SB, Yoon WK, Kim JH, Lee TC et al (2009) Low dose estrogen supplementation reduces mortality of mice in estrogen-dependent human tumor xenograft model. Biol Pharm Bull 32:150–152CrossRef
28.
go back to reference Kushner PJ, Agard DA, Greene GL, Scanlan TS, Shiau AK, Uht RM et al (2000) Estrogen receptor pathways to AP-1. J Steroid Biochem Mol Biol 74:311–317CrossRef Kushner PJ, Agard DA, Greene GL, Scanlan TS, Shiau AK, Uht RM et al (2000) Estrogen receptor pathways to AP-1. J Steroid Biochem Mol Biol 74:311–317CrossRef
35.
go back to reference Yang GP, Ross DT, Kuang WW, Brown PO, Weigel RJ (1999) Combining SSH and cDNA microarrays for rapid identification of differentially expressed genes. Nucleic Acids Res 27:1517–1523CrossRef Yang GP, Ross DT, Kuang WW, Brown PO, Weigel RJ (1999) Combining SSH and cDNA microarrays for rapid identification of differentially expressed genes. Nucleic Acids Res 27:1517–1523CrossRef
37.
go back to reference Anttila S, Luostarinen L, Hirvonen A, Elovaara E, Karjalainen A, Nurminen T et al (1995) Pulmonary expression of glutathione S-transferase M3 in lung cancer patients: association with GSTM1 polymorphism, smoking, and asbestos exposure. Cancer Res 55:3305–3309PubMed Anttila S, Luostarinen L, Hirvonen A, Elovaara E, Karjalainen A, Nurminen T et al (1995) Pulmonary expression of glutathione S-transferase M3 in lung cancer patients: association with GSTM1 polymorphism, smoking, and asbestos exposure. Cancer Res 55:3305–3309PubMed
44.
go back to reference Mitrunen K, Hirvonen A (2003) Molecular epidemiology of sporadic breast cancer. The role of polymorphic genes involved in oestrogen biosynthesis and metabolism. Mutat Res 544:9–41CrossRef Mitrunen K, Hirvonen A (2003) Molecular epidemiology of sporadic breast cancer. The role of polymorphic genes involved in oestrogen biosynthesis and metabolism. Mutat Res 544:9–41CrossRef
Metadata
Title
Oestrogen receptor-regulated glutathione S-transferase mu 3 expression attenuates hydrogen peroxide-induced cytotoxicity, which confers tamoxifen resistance on breast cancer cells
Authors
Juo-Han Lin
Shih-Hsin Tu
Li-Ching Chen
Chi-Cheng Huang
Hang-Lung Chang
Tzu-Chun Cheng
Hui-Wen Chang
Chih-Hsiung Wu
Han-Chung Wu
Yuan-Soon Ho
Publication date
01-11-2018
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 1/2018
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-018-4897-5

Other articles of this Issue 1/2018

Breast Cancer Research and Treatment 1/2018 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