Skip to main content
Top
Published in: Breast Cancer Research and Treatment 3/2012

01-02-2012 | Preclinical Study

Inhibitors of histone demethylation and histone deacetylation cooperate in regulating gene expression and inhibiting growth in human breast cancer cells

Authors: Yi Huang, Shauna N. Vasilatos, Lamia Boric, Patrick G. Shaw, Nancy E. Davidson

Published in: Breast Cancer Research and Treatment | Issue 3/2012

Login to get access

Abstract

Abnormal activities of histone lysine demethylases (KDMs) and lysine deacetylases (HDACs) are associated with aberrant gene expression in breast cancer development. However, the precise molecular mechanisms underlying the crosstalk between KDMs and HDACs in chromatin remodeling and regulation of gene transcription are still elusive. In this study, we showed that treatment of human breast cancer cells with inhibitors targeting the zinc cofactor dependent class I/II HDAC, but not NAD+ dependent class III HDAC, led to significant increase of H3K4me2 which is a specific substrate of histone lysine-specific demethylase 1 (LSD1) and a key chromatin mark promoting transcriptional activation. We also demonstrated that inhibition of LSD1 activity by a pharmacological inhibitor, pargyline, or siRNA resulted in increased acetylation of H3K9 (AcH3K9). However, siRNA knockdown of LSD2, a homolog of LSD1, failed to alter the level of AcH3K9, suggesting that LSD2 activity may not be functionally connected with HDAC activity. Combined treatment with LSD1 and HDAC inhibitors resulted in enhanced levels of H3K4me2 and AcH3K9, and exhibited synergistic growth inhibition of breast cancer cells. Finally, microarray screening identified a unique subset of genes whose expression was significantly changed by combination treatment with inhibitors of LSD1 and HDAC. Our study suggests that LSD1 intimately interacts with histone deacetylases in human breast cancer cells. Inhibition of histone demethylation and deacetylation exhibits cooperation and synergy in regulating gene expression and growth inhibition, and may represent a promising and novel approach for epigenetic therapy of breast cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Stearns V, Zhou Q, Davidson NE (2007) Epigenetic regulation as a new target for breast cancer therapy. Cancer Invest 25(8):659–665PubMedCrossRef Stearns V, Zhou Q, Davidson NE (2007) Epigenetic regulation as a new target for breast cancer therapy. Cancer Invest 25(8):659–665PubMedCrossRef
2.
go back to reference Marks PA, Richon VM, Miller T, Kelly WK (2004) Histone deacetylase inhibitors. Adv Cancer Res 91:137–168PubMedCrossRef Marks PA, Richon VM, Miller T, Kelly WK (2004) Histone deacetylase inhibitors. Adv Cancer Res 91:137–168PubMedCrossRef
3.
go back to reference Ficner R (2009) Novel structural insights into class I and II histone deacetylases. Curr Top Med Chem 9(3):235–240PubMedCrossRef Ficner R (2009) Novel structural insights into class I and II histone deacetylases. Curr Top Med Chem 9(3):235–240PubMedCrossRef
4.
go back to reference Keen JC, Yan L, Mack KM, Pettit C, Smith D, Sharma D, Davidson NE (2003) A novel histone deacetylase inhibitor, scriptaid, enhances expression of functional estrogen receptor alpha (ER) in ER negative human breast cancer cells in combination with 5-aza 2′-deoxycytidine. Breast Cancer Res Treat 81(3):177–186PubMedCrossRef Keen JC, Yan L, Mack KM, Pettit C, Smith D, Sharma D, Davidson NE (2003) A novel histone deacetylase inhibitor, scriptaid, enhances expression of functional estrogen receptor alpha (ER) in ER negative human breast cancer cells in combination with 5-aza 2′-deoxycytidine. Breast Cancer Res Treat 81(3):177–186PubMedCrossRef
5.
go back to reference Zhou Q, Atadja P, Davidson NE (2007) Histone deacetylase inhibitor LBH589 reactivates silenced estrogen receptor alpha (ER) gene expression without loss of DNA hypermethylation. Cancer Biol Ther 6(1):64–69PubMedCrossRef Zhou Q, Atadja P, Davidson NE (2007) Histone deacetylase inhibitor LBH589 reactivates silenced estrogen receptor alpha (ER) gene expression without loss of DNA hypermethylation. Cancer Biol Ther 6(1):64–69PubMedCrossRef
6.
go back to reference Yang X, Ferguson AT, Nass SJ, Phillips DL, Butash KA, Wang SM, Herman JG, Davidson NE (2000) Transcriptional activation of estrogen receptor alpha in human breast cancer cells by histone deacetylase inhibition. Cancer Res 60(24):6890–6894PubMed Yang X, Ferguson AT, Nass SJ, Phillips DL, Butash KA, Wang SM, Herman JG, Davidson NE (2000) Transcriptional activation of estrogen receptor alpha in human breast cancer cells by histone deacetylase inhibition. Cancer Res 60(24):6890–6894PubMed
7.
go back to reference Sharma D, Saxena NK, Davidson NE, Vertino PM (2006) Restoration of tamoxifen sensitivity in estrogen receptor-negative breast cancer cells: tamoxifen-bound reactivated ER recruits distinctive corepressor complexes. Cancer Res 66(12):6370–6378PubMedCrossRef Sharma D, Saxena NK, Davidson NE, Vertino PM (2006) Restoration of tamoxifen sensitivity in estrogen receptor-negative breast cancer cells: tamoxifen-bound reactivated ER recruits distinctive corepressor complexes. Cancer Res 66(12):6370–6378PubMedCrossRef
8.
go back to reference Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA, Shi Y (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, Shi Y (2004) Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119(7):941–953PubMedCrossRef
9.
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
10.
go back to reference Kahl P, Gullotti L, Heukamp LC, Wolf S, Friedrichs N, Vorreuther R, Solleder G, Bastian PJ, Ellinger J, Metzger E et al (2006) Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res 66(23):11341–11347PubMedCrossRef Kahl P, Gullotti L, Heukamp LC, Wolf S, Friedrichs N, Vorreuther R, Solleder G, Bastian PJ, Ellinger J, Metzger E et al (2006) Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res 66(23):11341–11347PubMedCrossRef
11.
go back to reference Scoumanne A, Chen X (2007) The lysine-specific demethylase 1 is required for cell proliferation in both p53-dependent and -independent manners. J Biol Chem 282(21):15471–15475PubMedCrossRef Scoumanne A, Chen X (2007) The lysine-specific demethylase 1 is required for cell proliferation in both p53-dependent and -independent manners. J Biol Chem 282(21):15471–15475PubMedCrossRef
12.
go back to reference Bradley C, van der Meer R, Roodi N, Yan H, Chandrasekharan MB, Sun ZW, Mernaugh RL, Parl FF (2007) Carcinogen-induced histone alteration in normal human mammary epithelial cells. Carcinogenesis 28(10):2184–2192PubMedCrossRef Bradley C, van der Meer R, Roodi N, Yan H, Chandrasekharan MB, Sun ZW, Mernaugh RL, Parl FF (2007) Carcinogen-induced histone alteration in normal human mammary epithelial cells. Carcinogenesis 28(10):2184–2192PubMedCrossRef
13.
go back to reference Huang Y, Greene E, Murray Stewart T, Goodwin AC, Baylin SB, Woster PM, Casero RA Jr (2007) Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc Natl Acad Sci USA 104(19):8023–8028PubMedCrossRef Huang Y, Greene E, Murray Stewart T, Goodwin AC, Baylin SB, Woster PM, Casero RA Jr (2007) Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc Natl Acad Sci USA 104(19):8023–8028PubMedCrossRef
14.
go back to reference Huang Y, Stewart TM, Wu Y, Baylin SB, Marton LJ, Perkins B, Jones RJ, Woster PM, Casero RA Jr (2009) Novel oligoamine analogues inhibit lysine-specific demethylase 1 and induce reexpression of epigenetically silenced genes. Clin Cancer Res 15(23):7217–7228PubMedCrossRef Huang Y, Stewart TM, Wu Y, Baylin SB, Marton LJ, Perkins B, Jones RJ, Woster PM, Casero RA Jr (2009) Novel oligoamine analogues inhibit lysine-specific demethylase 1 and induce reexpression of epigenetically silenced genes. Clin Cancer Res 15(23):7217–7228PubMedCrossRef
15.
go back to reference Huang Y, Marton LJ, Woster PM, Casero RA (2009) Polyamine analogues targeting epigenetic gene regulation. Essays Biochem 46:95–110PubMedCrossRef Huang Y, Marton LJ, Woster PM, Casero RA (2009) Polyamine analogues targeting epigenetic gene regulation. Essays Biochem 46:95–110PubMedCrossRef
16.
go back to reference Karytinos A, Forneris F, Profumo A, Ciossani G, Battaglioli E, Binda C, Mattevi A (2009) A novel mammalian flavin-dependent histone demethylase. J Biol Chem 284(26):17775–17782PubMedCrossRef Karytinos A, Forneris F, Profumo A, Ciossani G, Battaglioli E, Binda C, Mattevi A (2009) A novel mammalian flavin-dependent histone demethylase. J Biol Chem 284(26):17775–17782PubMedCrossRef
17.
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):415–418PubMedCrossRef 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):415–418PubMedCrossRef
18.
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 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
19.
go back to reference Huang Y, Hager ER, Phillips DL, Dunn VR, Hacker A, Frydman B, Kink JA, Valasinas AL, Reddy VK, Marton LJ et al (2003) A novel polyamine analog inhibits growth and induces apoptosis in human breast cancer cells. Clin Cancer Res 9(7):2769–2777PubMed Huang Y, Hager ER, Phillips DL, Dunn VR, Hacker A, Frydman B, Kink JA, Valasinas AL, Reddy VK, Marton LJ et al (2003) A novel polyamine analog inhibits growth and induces apoptosis in human breast cancer cells. Clin Cancer Res 9(7):2769–2777PubMed
20.
go back to reference Huang Y, Keen JC, Hager E, Smith R, Hacker A, Frydman B, Valasinas AL, Reddy VK, Marton LJ, Casero RA Jr et al (2004) Regulation of polyamine analogue cytotoxicity by c-Jun in human MDA-MB-435 cancer cells. Mol Cancer Res 2(2):81–88PubMed Huang Y, Keen JC, Hager E, Smith R, Hacker A, Frydman B, Valasinas AL, Reddy VK, Marton LJ, Casero RA Jr et al (2004) Regulation of polyamine analogue cytotoxicity by c-Jun in human MDA-MB-435 cancer cells. Mol Cancer Res 2(2):81–88PubMed
21.
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
22.
go back to reference Hahm HA, Dunn VR, Butash KA, Deveraux WL, Woster PM, Casero RA Jr, Davidson NE (2001) Combination of standard cytotoxic agents with polyamine analogues in the treatment of breast cancer cell lines. Clin Cancer Res 7(2):391–399PubMed Hahm HA, Dunn VR, Butash KA, Deveraux WL, Woster PM, Casero RA Jr, Davidson NE (2001) Combination of standard cytotoxic agents with polyamine analogues in the treatment of breast cancer cell lines. Clin Cancer Res 7(2):391–399PubMed
23.
go back to reference Tusher VG, Tibshirani R, Chu G (2001) Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA 98(9):5116–5121PubMedCrossRef Tusher VG, Tibshirani R, Chu G (2001) Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA 98(9):5116–5121PubMedCrossRef
24.
go back to reference Blander G, Guarente L (2004) The Sir2 family of protein deacetylases. Annu Rev Biochem 73:417–435PubMedCrossRef Blander G, Guarente L (2004) The Sir2 family of protein deacetylases. Annu Rev Biochem 73:417–435PubMedCrossRef
25.
go back to reference Shi YJ, Matson C, Lan F, Iwase S, Baba T, Shi Y (2005) Regulation of LSD1 histone demethylase activity by its associated factors. Mol Cell 19(6):857–864PubMedCrossRef Shi YJ, Matson C, Lan F, Iwase S, Baba T, Shi Y (2005) Regulation of LSD1 histone demethylase activity by its associated factors. Mol Cell 19(6):857–864PubMedCrossRef
26.
go back to reference Lan F, Collins RE, De Cegli R, Alpatov R, Horton JR, Shi X, Gozani O, Cheng X, Shi Y (2007) Recognition of unmethylated histone H3 lysine 4 links BHC80 to LSD1-mediated gene repression. Nature 448(7154):718–722PubMedCrossRef Lan F, Collins RE, De Cegli R, Alpatov R, Horton JR, Shi X, Gozani O, Cheng X, Shi Y (2007) Recognition of unmethylated histone H3 lysine 4 links BHC80 to LSD1-mediated gene repression. Nature 448(7154):718–722PubMedCrossRef
27.
go back to reference Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB (1999) Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet 21(1):103–107PubMedCrossRef Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB (1999) Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet 21(1):103–107PubMedCrossRef
28.
go back to reference Gore SD, Baylin S, Sugar E, Carraway H, Miller CB, Carducci M, Grever M, Galm O, Dauses T, Karp JE et al (2006) Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res 66(12):6361–6369PubMedCrossRef Gore SD, Baylin S, Sugar E, Carraway H, Miller CB, Carducci M, Grever M, Galm O, Dauses T, Karp JE et al (2006) Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res 66(12):6361–6369PubMedCrossRef
29.
go back to reference Alexopoulou AN, Leao M, Caballero OL, Da Silva L, Reid L, Lakhani SR, Simpson AJ, Marshall JF, Neville AM, Jat PS (2010) Dissecting the transcriptional networks underlying breast cancer: NR4A1 reduces the migration of normal and breast cancer cell lines. Breast Cancer Res 12(4):R51 Alexopoulou AN, Leao M, Caballero OL, Da Silva L, Reid L, Lakhani SR, Simpson AJ, Marshall JF, Neville AM, Jat PS (2010) Dissecting the transcriptional networks underlying breast cancer: NR4A1 reduces the migration of normal and breast cancer cell lines. Breast Cancer Res 12(4):R51
30.
go back to reference Wu Q, Dawson MI, Zheng Y, Hobbs PD, Agadir A, Jong L, Li Y, Liu R, Lin B, Zhang XK (1997) Inhibition of trans-retinoic acid-resistant human breast cancer cell growth by retinoid X receptor-selective retinoids. Mol Cell Biol 17(11):6598–6608PubMed Wu Q, Dawson MI, Zheng Y, Hobbs PD, Agadir A, Jong L, Li Y, Liu R, Lin B, Zhang XK (1997) Inhibition of trans-retinoic acid-resistant human breast cancer cell growth by retinoid X receptor-selective retinoids. Mol Cell Biol 17(11):6598–6608PubMed
31.
go back to reference Novak P, Jensen T, Oshiro MM, Watts GS, Kim CJ, Futscher BW (2008) Agglomerative epigenetic aberrations are a common event in human breast cancer. Cancer Res 68(20):8616–8625PubMedCrossRef Novak P, Jensen T, Oshiro MM, Watts GS, Kim CJ, Futscher BW (2008) Agglomerative epigenetic aberrations are a common event in human breast cancer. Cancer Res 68(20):8616–8625PubMedCrossRef
32.
go back to reference Liang G, Bansal G, Xie Z, Druey KM (2009) RGS16 inhibits breast cancer cell growth by mitigating phosphatidylinositol 3-kinase signaling. J Biol Chem 284(32):21719–21727PubMedCrossRef Liang G, Bansal G, Xie Z, Druey KM (2009) RGS16 inhibits breast cancer cell growth by mitigating phosphatidylinositol 3-kinase signaling. J Biol Chem 284(32):21719–21727PubMedCrossRef
33.
go back to reference Rao R, Nalluri S, Kolhe R, Yang Y, Fiskus W, Chen J, Ha K, Buckley KM, Balusu R, Coothankandaswamy V et al (2010) Treatment with panobinostat induces glucose-regulated protein 78 acetylation and endoplasmic reticulum stress in breast cancer cells. Mol Cancer Ther 9(4):942–952 Rao R, Nalluri S, Kolhe R, Yang Y, Fiskus W, Chen J, Ha K, Buckley KM, Balusu R, Coothankandaswamy V et al (2010) Treatment with panobinostat induces glucose-regulated protein 78 acetylation and endoplasmic reticulum stress in breast cancer cells. Mol Cancer Ther 9(4):942–952
34.
go back to reference Ho TF, Ma CJ, Lu CH, Tsai YT, Wei YH, Chang JS, Lai JK, Cheuh PJ, Yeh CT, Tang PC et al (2007) Undecylprodigiosin selectively induces apoptosis in human breast carcinoma cells independent of p53. Toxicol Appl Pharmacol 225(3):318–328PubMedCrossRef Ho TF, Ma CJ, Lu CH, Tsai YT, Wei YH, Chang JS, Lai JK, Cheuh PJ, Yeh CT, Tang PC et al (2007) Undecylprodigiosin selectively induces apoptosis in human breast carcinoma cells independent of p53. Toxicol Appl Pharmacol 225(3):318–328PubMedCrossRef
Metadata
Title
Inhibitors of histone demethylation and histone deacetylation cooperate in regulating gene expression and inhibiting growth in human breast cancer cells
Authors
Yi Huang
Shauna N. Vasilatos
Lamia Boric
Patrick G. Shaw
Nancy E. Davidson
Publication date
01-02-2012
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 3/2012
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-011-1480-8

Other articles of this Issue 3/2012

Breast Cancer Research and Treatment 3/2012 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