Skip to main content
Top
Published in: Breast Cancer Research and Treatment 1/2014

01-07-2014 | Preclinical study

Inhibition of histone demethylase, LSD2 (KDM1B), attenuates DNA methylation and increases sensitivity to DNMT inhibitor-induced apoptosis in breast cancer cells

Authors: Tiffany A. Katz, Shauna N. Vasilatos, Emily Harrington, Steffi Oesterreich, Nancy E. Davidson, Yi Huang

Published in: Breast Cancer Research and Treatment | Issue 1/2014

Login to get access

Abstract

Increasing evidence suggests that dysfunction of histone lysine demethylase is associated with abnormal chromatin remodeling and gene silencing, contributing to breast tumorigenesis. In silico analysis shows that the newly identified histone demethylase lysine-specific demethylase 2 is highly expressed in breast cancer, especially in invasive tumors. However, it is currently unknown how LSD2 regulates chromatin remodeling and gene expression regulation in breast cancer. Using short hairpin RNA, we stably knocked down LSD2 (LSD2-KD) in MDA-MB-231 breast cancer cells. LSD2-KD led to accumulation of H3K4me1/2 without changing methylation levels of other key histone lysine residues, suggesting that LSD2 acts as a bona fide H3K4 demethylase in breast cancer cells. LSD2-KD resulted in decreased colony formation and attenuated global DNA methylation in MDA-MB-231 cells. Additionally, treatment with the DNMT inhibitor, 5-aza-deoxycytidine (DAC), synergistically increased mRNA expression of aberrantly silenced genes important in breast cancer development, including PR, RARβ, ERα, SFRP1, SFRP2, and E-cadherin in LSD2-KD cells. Furthermore, LSD2-KD cells are more susceptible to cell death than scramble controls, and combined treatment with tranylcypromine, an LSD2 inhibitor, and DAC resulted in synergistic growth inhibition of breast cancer cells. DNMT inhibition by DAC in LSD2-KD cells led to internucleosomal DNA fragmentation, enhanced PARP cleavage and increased sub-G1 apoptotic cell population. These results demonstrate an important role for LSD2 in regulation of DNA methylation and gene silencing in breast cancer, and suggest that inhibition of LSD2 in combination with DNA methyltransferase inhibition represents a novel approach for epigenetic therapy of breast cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA (2004) Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119(7):941–953PubMedCrossRef Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA (2004) Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119(7):941–953PubMedCrossRef
2.
go back to reference Lee MG, Wynder C, Cooch N, Shiekhattar R (2005) An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature 437(7057):432–435PubMed Lee MG, Wynder C, Cooch N, Shiekhattar R (2005) An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature 437(7057):432–435PubMed
6.
go back to reference Yang Z, Jiang J, Stewart DM, Qi S, Yamane K, Li J, Zhang Y, Wong J (2010) AOF1 is a histone H3K4 demethylase possessing demethylase activity-independent repression function. Cell Res 20(3):276–287. doi:10.1038/cr.2010.12 PubMedCrossRef Yang Z, Jiang J, Stewart DM, Qi S, Yamane K, Li J, Zhang Y, Wong J (2010) AOF1 is a histone H3K4 demethylase possessing demethylase activity-independent repression function. Cell Res 20(3):276–287. doi:10.​1038/​cr.​2010.​12 PubMedCrossRef
7.
go back to reference Ciccone DN, Su H, Hevi S, Gay F, Lei H, Bajko J, Xu G, Li E, Chen T (2009) KDM1B is a histone H3K4 demethylase required to establish maternal genomic imprints. Nature 461(7262):415–418. doi:10.1038/nature08315 PubMedCrossRef Ciccone DN, Su H, Hevi S, Gay F, Lei H, Bajko J, Xu G, Li E, Chen T (2009) KDM1B is a histone H3K4 demethylase required to establish maternal genomic imprints. Nature 461(7262):415–418. doi:10.​1038/​nature08315 PubMedCrossRef
9.
go back to reference Munster PN, Thurn KT, Thomas S, Raha P, Lacevic M, Miller A, Melisko M, Ismail-Khan R, Rugo H, Moasser M, Minton SE (2011) A phase II study of the histone deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of patients with hormone therapy-resistant breast cancer. Br J Cancer 104(12):1828–1835. doi:10.1038/bjc.2011.156 PubMedCentralPubMedCrossRef Munster PN, Thurn KT, Thomas S, Raha P, Lacevic M, Miller A, Melisko M, Ismail-Khan R, Rugo H, Moasser M, Minton SE (2011) A phase II study of the histone deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of patients with hormone therapy-resistant breast cancer. Br J Cancer 104(12):1828–1835. doi:10.​1038/​bjc.​2011.​156 PubMedCentralPubMedCrossRef
12.
13.
go back to reference Lim S, Janzer A, Becker A, Zimmer A, Schule R, Buettner R, Kirfel J (2009) Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive tumor biology. Carcinogenesis 31(3):512–520. doi:10.1093/carcin/bgp324 PubMedCrossRef Lim S, Janzer A, Becker A, Zimmer A, Schule R, Buettner R, Kirfel J (2009) Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive tumor biology. Carcinogenesis 31(3):512–520. doi:10.​1093/​carcin/​bgp324 PubMedCrossRef
14.
go back to reference Zhu Q, Huang Y, Marton LJ, Woster PM, Davidson NE, Casero RA Jr (2012) Polyamine analogs modulate gene expression by inhibiting lysine-specific demethylase 1 (LSD1) and altering chromatin structure in human breast cancer cells. Amino Acids 42(2–3):887–898. doi:10.1007/s00726-011-1004-1 PubMedCentralPubMedCrossRef Zhu Q, Huang Y, Marton LJ, Woster PM, Davidson NE, Casero RA Jr (2012) Polyamine analogs modulate gene expression by inhibiting lysine-specific demethylase 1 (LSD1) and altering chromatin structure in human breast cancer cells. Amino Acids 42(2–3):887–898. doi:10.​1007/​s00726-011-1004-1 PubMedCentralPubMedCrossRef
15.
go back to reference Vasilatos SN, Katz TA, Oesterreich S, Wan Y, Davidson NE, Huang Y (2013) Crosstalk between lysine specific demethylase 1 (LSD1) and histone demethylases mediates antineoplastic efficacy of HDAC inhibitors in breast cancer cells. Carcinogenesis 34 (6):1196–1207. doi:10.1093/carcin/bgt033 Vasilatos SN, Katz TA, Oesterreich S, Wan Y, Davidson NE, Huang Y (2013) Crosstalk between lysine specific demethylase 1 (LSD1) and histone demethylases mediates antineoplastic efficacy of HDAC inhibitors in breast cancer cells. Carcinogenesis 34 (6):1196–1207. doi:10.​1093/​carcin/​bgt033
16.
go back to reference Huang Y, Hager ER, Phillips DL, Dunn VR, Hacker A, Frydman B, Kink JA, Valasinas AL, Reddy VK, Marton LJ, Casero RA Jr, Davidson NE (2003) A novel polyamine analog inhibits growth and induces apoptosis in human breast cancer cells. Clin Cancer Res 9(7):2769–2777PubMedCentralPubMed Huang Y, Hager ER, Phillips DL, Dunn VR, Hacker A, Frydman B, Kink JA, Valasinas AL, Reddy VK, Marton LJ, Casero RA Jr, Davidson NE (2003) A novel polyamine analog inhibits growth and induces apoptosis in human breast cancer cells. Clin Cancer Res 9(7):2769–2777PubMedCentralPubMed
17.
go back to reference Chou TC, Talalay P (1984) Quantitative analysis of dose–effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 22:27–55PubMedCrossRef Chou TC, Talalay P (1984) Quantitative analysis of dose–effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 22:27–55PubMedCrossRef
19.
go back to reference Seward DJ, Cubberley G, Kim S, Schonewald M, Zhang L, Tripet B, Bentley DL (2007) Demethylation of trimethylated histone H3 Lys4 in vivo by JARID1 JmjC proteins. Nat Struct Mol Biol 14(3):240–242. doi:10.1038/nsmb1200 PubMedCrossRef Seward DJ, Cubberley G, Kim S, Schonewald M, Zhang L, Tripet B, Bentley DL (2007) Demethylation of trimethylated histone H3 Lys4 in vivo by JARID1 JmjC proteins. Nat Struct Mol Biol 14(3):240–242. doi:10.​1038/​nsmb1200 PubMedCrossRef
20.
go back to reference Katoh Y, Katoh M (2007) Comparative integromics on JMJD2A, JMJD2B and JMJD2C: preferential expression of JMJD2C in undifferentiated ES cells. Int J Mol Med 20(2):269–273PubMed Katoh Y, Katoh M (2007) Comparative integromics on JMJD2A, JMJD2B and JMJD2C: preferential expression of JMJD2C in undifferentiated ES cells. Int J Mol Med 20(2):269–273PubMed
21.
go back to reference Fodor BD, Kubicek S, Yonezawa M, O’Sullivan RJ, Sengupta R, Perez-Burgos L, Opravil S, Mechtler K, Schotta G, Jenuwein T (2006) Jmjd2b antagonizes H3K9 trimethylation at pericentric heterochromatin in mammalian cells. Genes Dev 20(12):1557–1562. doi:10.1101/gad.388206 PubMedCentralPubMedCrossRef Fodor BD, Kubicek S, Yonezawa M, O’Sullivan RJ, Sengupta R, Perez-Burgos L, Opravil S, Mechtler K, Schotta G, Jenuwein T (2006) Jmjd2b antagonizes H3K9 trimethylation at pericentric heterochromatin in mammalian cells. Genes Dev 20(12):1557–1562. doi:10.​1101/​gad.​388206 PubMedCentralPubMedCrossRef
24.
go back to reference Lapidus RG, Ferguson AT, Ottaviano YL, Parl FF, Smith HS, Weitzman SA, Baylin SB, Issa JP, Davidson NE (1996) Methylation of estrogen and progesterone receptor gene 5′ CpG islands correlates with lack of estrogen and progesterone receptor gene expression in breast tumors. Clin Cancer Res 2(5):805–810PubMed Lapidus RG, Ferguson AT, Ottaviano YL, Parl FF, Smith HS, Weitzman SA, Baylin SB, Issa JP, Davidson NE (1996) Methylation of estrogen and progesterone receptor gene 5′ CpG islands correlates with lack of estrogen and progesterone receptor gene expression in breast tumors. Clin Cancer Res 2(5):805–810PubMed
25.
go back to reference Pathiraja TN, Shetty PB, Jelinek J, He R, Hartmaier R, Margossian AL, Hilsenbeck SG, Issa JP, Oesterreich S (2011) Progesterone receptor isoform-specific promoter methylation: association of PRA promoter methylation with worse outcome in breast cancer patients. Clin Cancer Res 17(12):4177–4186. doi:10.1158/1078-0432.CCR-10-2950 PubMedCentralPubMedCrossRef Pathiraja TN, Shetty PB, Jelinek J, He R, Hartmaier R, Margossian AL, Hilsenbeck SG, Issa JP, Oesterreich S (2011) Progesterone receptor isoform-specific promoter methylation: association of PRA promoter methylation with worse outcome in breast cancer patients. Clin Cancer Res 17(12):4177–4186. doi:10.​1158/​1078-0432.​CCR-10-2950 PubMedCentralPubMedCrossRef
26.
go back to reference Ooi SK, Qiu C, Bernstein E, Li K, Jia D, Yang Z, Erdjument-Bromage H, Tempst P, Lin SP, Allis CD, Cheng X, Bestor TH (2007) DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448(7154):714–717. doi:10.1038/nature05987 PubMedCentralPubMedCrossRef Ooi SK, Qiu C, Bernstein E, Li K, Jia D, Yang Z, Erdjument-Bromage H, Tempst P, Lin SP, Allis CD, Cheng X, Bestor TH (2007) DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448(7154):714–717. doi:10.​1038/​nature05987 PubMedCentralPubMedCrossRef
28.
go back to reference Wang J, Hevi S, Kurash JK, Lei H, Gay F, Bajko J, Su H, Sun W, Chang H, Xu G, Gaudet F, Li E, Chen T (2009) The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat Genet 41(1):125–129. doi:10.1038/ng.268 PubMedCrossRef Wang J, Hevi S, Kurash JK, Lei H, Gay F, Bajko J, Su H, Sun W, Chang H, Xu G, Gaudet F, Li E, Chen T (2009) The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat Genet 41(1):125–129. doi:10.​1038/​ng.​268 PubMedCrossRef
29.
Metadata
Title
Inhibition of histone demethylase, LSD2 (KDM1B), attenuates DNA methylation and increases sensitivity to DNMT inhibitor-induced apoptosis in breast cancer cells
Authors
Tiffany A. Katz
Shauna N. Vasilatos
Emily Harrington
Steffi Oesterreich
Nancy E. Davidson
Yi Huang
Publication date
01-07-2014
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 1/2014
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-014-3012-9

Other articles of this Issue 1/2014

Breast Cancer Research and Treatment 1/2014 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