Downregulation of histone H2A and H2B pathways is associated with anthracycline sensitivity in breast cancer

Marsela Braunstein, Linda Liao, Nicola Lyttle, Nazleen Lobo, Karen J Taylor, Paul M Krzyzanowski, Irina Kalatskaya, Cindy Q Yao, Lincoln D Stein, Paul C Boutros, Christopher J Twelves, Richard Marcellus, John M S Bartlett, Melanie Spears, Marsela Braunstein, Linda Liao, Nicola Lyttle, Nazleen Lobo, Karen J Taylor, Paul M Krzyzanowski, Irina Kalatskaya, Cindy Q Yao, Lincoln D Stein, Paul C Boutros, Christopher J Twelves, Richard Marcellus, John M S Bartlett, Melanie Spears

Abstract

Background: Drug resistance in breast cancer is the major obstacle to effective treatment with chemotherapy. While upregulation of multidrug resistance genes is an important component of drug resistance mechanisms in vitro, their clinical relevance remains to be determined. Therefore, identifying pathways that could be targeted in the clinic to eliminate anthracycline-resistant breast cancer remains a major challenge.

Methods: We generated paired native and epirubicin-resistant MDA-MB-231, MCF7, SKBR3 and ZR-75-1 epirubicin-resistant breast cancer cell lines to identify pathways contributing to anthracycline resistance. Native cell lines were exposed to increasing concentrations of epirubicin until resistant cells were generated. To identify mechanisms driving epirubicin resistance, we used a complementary approach including gene expression analyses to identify molecular pathways involved in resistance, and small-molecule inhibitors to reverse resistance. In addition, we tested its clinical relevance in a BR9601 adjuvant clinical trial.

Results: Characterisation of epirubicin-resistant cells revealed that they were cross-resistant to doxorubicin and SN-38 and had alterations in apoptosis and cell-cycle profiles. Gene expression analysis identified deregulation of histone H2A and H2B genes in all four cell lines. Histone deacetylase small-molecule inhibitors reversed resistance and were cytotoxic for epirubicin-resistant cell lines, confirming that histone pathways are associated with epirubicin resistance. Gene expression of a novel 18-gene histone pathway module analysis of the BR9601 adjuvant clinical trial revealed that patients with low expression of the 18-gene histone module benefited from anthracycline treatment more than those with high expression (hazard ratio 0.35, 95 % confidence interval 0.13-0.96, p = 0.042).

Conclusions: This study revealed a key pathway that contributes to anthracycline resistance and established model systems for investigating drug resistance in all four major breast cancer subtypes. As the histone modification can be targeted with small-molecule inhibitors, it represents a possible means of reversing clinical anthracycline resistance.

Trial registration: ClinicalTrials.gov identifier NCT00003012 . Registered on 1 November 1999.

Figures

Fig. 1
Fig. 1
Characterisation of epirubicin-resistant cell lines. Native and resistant cells were exposed to drug concentrations ranging from 0.3 to 3000 nM. Cell viability was determined 72 h later by Cell Counting Kit-8 assay. a Percentage of live cells relative to dimethyl sulphoxide control was plotted against epirubicin concentration. Black = native cells, magenta = resistant cells. b Half-maximal inhibitory concentration (IC50) values in nanomolar concentrations ± standard deviation. Resistance factor is shown in parentheses and represents resistant IC50/native IC50
Fig. 2
Fig. 2
Expression of conventional breast cancer biomarkers and selected multidrug resistance genes. Cell lysates were prepared in radioimmunoprecipitation assay buffer supplemented with cOmplete Mini protease inhibitor and PhosSTOP phosphatase inhibitor. Quantities (10–50 μg) of total protein were run on a 10 % gel (MDR1), 4–20 % precast gels (epidermal growth factor receptor [EGFR], oestrogen receptor [ER], progesterone receptor [PR], type II topoisomerase [TOPO IIα]) and Any kD Mini-PROTEAN TGX Precast Protein Gels (human epidermal growth factor receptor 2 [HER2], HER3), transferred onto polyvinylidene fluoride membrane and developed using chemiluminescence substrate. Nat native, Epi-R epirubicin-resistant, GAPDH glyceraldehyde-3-phosphate dehydrogenase
Fig. 3
Fig. 3
Resistant cell lines overcome epirubicin-induced G2/M arrest. ad Cells were synchronised by a double-thymidine block and treated with dimethyl sulphoxide or epirubicin at selection doses established for each resistant cell line: 25 nM epirubicin to MDA-MB-231, 30 nM epirubicin to MCF7, 15 nM epirubicin to SKBR3 and 15 nM epirubicin to ZR-75-1. Epirubicin concentration was increased to 100 nM for MCF7 and SKBR3 cells because G2/M block was not observed at the lower doses of epirubicin. Cells were collected at 48 h, stained with propidium iodide and analysed by flow cytometry. Debris was gated out. GAPDH glyceraldehyde-3-phosphate dehydrogenase
Fig. 4
Fig. 4
Network-based analysis of epirubicin-resistant (EpiR) cell lines. a Venn diagram of genes with significant changes in expression in breast cancer cell lines. b Histone module identified in Functional Interaction network analysis. Coloured rings denote genes demonstrating consistent changes across all four cell lines. Red rings = upregulated genes, green rings = downregulated genes, diamonds = linker genes. c Quantitative real-time polymerase chain reactions performed on RNA isolated from native and epirubicin-resistant cell lines. Bar graphs indicate average quantitative means, while error bars represent standard error of the mean. p Values were calculated using unpaired t test. ns non-significant. d Immunoblotting of total H2A and H2B histone proteins in native and epirubicin-resistant cell lines. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a housekeeping control. e Reactome pathways significantly enriched within the module shown in (b)
Fig. 5
Fig. 5
Histone gene knockdown is not sufficient to resensitise breast cancer cells to epirubicin. a A total of 7 × 104 ZR75-1 epirubicin-resistant (EpiR) cells and MDA-MB-231 EpiR cells were transfected with 30 nM of each siRNA (Dharmacon; GE Healthcare, Lafayette, CO, USA) targeting HIST1H2AC and HIST1H2BK (individual knockdowns not shown for simplicity). Negative controls included media only, Lipofectamine only or mock transfection with non-targeting siRNA. Percentage gene expression knock-down is shown in Additional file 1: Table S3. b IC50 values were generated using non-linear regression analysis, and average values of two independent experiments are graphed. Error bars represent standard deviation. c Fold changes in gene expression of each histone variant relative to the housekeeping gene, RPL37A. d-e Histone knockdown effects on cell cycle and apoptosis for MDA-MB-231 and ZR-75-1. Tables show average percentages of cells in G1, S and G2 stages of cell cycle, and precent live versus dead cells in each experimental and control condition. Numbers in parenthesis indicate standard deviation between two experiments
Fig. 6
Fig. 6
Histone module is a biological marker for anthracycline therapy. High expression and low expression of histone module were tested for association with distant recurrence-free survival (DRFS) and overall survival (OS) in the BR9601 trial, in which patients were treated with standard chemotherapy (CMF) or anthracycline-containing chemotherapy (E-CMF). a DRFS and OS for patients treated with E-CMF vs. CMF split into high or low histone gene expression groups. b Multivariate, treatment by marker analysis after adjustment for human epidermal growth factor receptor 2 (HER2) status, oestrogen receptor (ER) status, nodal status, grade and age. HR hazard ratio, CI confidence interval
Fig. 7
Fig. 7
Histone deacetylase inhibitors (HDACi) induce cytotoxicity in epirubicin-resistant (EpiR) cell lines. a An example of the effect of panobinostat on all cell lines. Half-maximal inhibitory concentration values for panobinostat and the remaining HDACi are shown in Additional file 1: Table S4. b A schematic indicating that HDACi could offer a viable treatment option for patients who do not respond to anthracyclines (high histone score). In addition, patients with low histone score who would initially benefit from epirubicin treatment may in due course develop de novo resistance, and, if diagnosed with “high histone score” at recurrence, may be offered HDACi as a subsequent treatment option. c Working models of molecular mechanisms involved in epirubicin resistance. There are three proposed mechanisms by which HDACi sensitise cells to epirubicin: (1) by transcriptional activation of repressors and pro-apoptotic genes, (2) by repression of resistance genes and (3) due to increased accessibility to DNA

References

    1. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365:1687–1717. doi: 10.1016/S0140-6736(05)66544-0.
    1. Guo B, Villeneuve DJ, Hembruff SL, Kirwan AF, Blais DE, Bonin M, et al. Cross-resistance studies of isogenic drug-resistant breast tumor cell lines support recent clinical evidence suggesting that sensitivity to paclitaxel may be strongly compromised by prior doxorubicin exposure. Breast Cancer Res Treat. 2004;85:31–51. doi: 10.1023/B:BREA.0000021046.29834.12.
    1. Ringel I, Horwitz SB. Studies with RP 56976 (Taxotere): a semisynthetic analogue of Taxol. J Natl Cancer Inst. 1991;83:288–291. doi: 10.1093/jnci/83.4.288.
    1. Chazard M, Pellae-Cosset B, Garet F, Soares JA, Lucidi B, Lavail Y, et al. Taxol (paclitaxel), first molecule of a new class of cytotoxic agents: taxanes. Bull Cancer. 1994;81:173–181.
    1. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004;56:185–229. doi: 10.1124/pr.56.2.6.
    1. Schinkel AH, Roelofs EM, Borst P. Characterization of the human MDR3 P-glycoprotein and its recognition by P-glycoprotein-specific monoclonal antibodies. Cancer Res. 1991;51:2628–2635.
    1. Van der Bliek AM, Baas F, Van der Velde-Koerts T, Biedler JL, Meyers MB, Ozols RF, et al. Genes amplified and overexpressed in human multidrug-resistant cell lines. Cancer Res. 1988;48:5927–5932.
    1. Giaccone G, Gazdar AF, Beck H, Zunino F, Capranico G. Multidrug sensitivity phenotype of human lung cancer cells associated with topoisomerase II expression. Cancer Res. 1992;52:1666–1674.
    1. de Jong S, Zijlstra JG, de Vries EG, Mulder NH. Reduced DNA topoisomerase II activity and drug-induced DNA cleavage activity in an Adriamycin-resistant human small cell lung carcinoma cell line. Cancer Res. 1990;50:304–309.
    1. Friesen C, Fulda S, Debatin KM. Deficient activation of the CD95 (APO-1/Fas) system in drug-resistant cells. Leukemia. 1997;11:1833–1841. doi: 10.1038/sj.leu.2400827.
    1. Lowe SW, Ruley HE, Jacks T, Housman DE. p53-Dependent apoptosis modulates the cytotoxicity of anticancer agents. Cell. 1993;74:957–967. doi: 10.1016/0092-8674(93)90719-7.
    1. Munro AF, Twelves C, Thomas JS, Cameron DA, Bartlett JM. Chromosome instability and benefit from adjuvant anthracyclines in breast cancer. Br J Cancer. 2012;107:71–74. doi: 10.1038/bjc.2012.232.
    1. Pritchard KI, Munro A, O’Malley FP, Tu D, Li X, Levine MN, et al. Chromosome 17 centromere (CEP17) duplication as a predictor of anthracycline response: evidence from the NCIC Clinical Trials Group (NCIC CTG) MA.5 Trial. Breast Cancer Res Treat. 2012;131:541–551. doi: 10.1007/s10549-011-1840-4.
    1. Bartlett JM, McConkey CC, Munro AF, Desmedt C, Dunn JA, Larsimont DP, et al. Predicting anthracycline benefit: TOP2A and CEP17—not only but also. J Clin Oncol. 2015;33:1680–1687. doi: 10.1200/JCO.2013.54.7869.
    1. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–752. doi: 10.1038/35021093.
    1. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98:10869–10874. doi: 10.1073/pnas.191367098.
    1. Poole CJ, Earl HM, Hiller L, Dunn JA, Bathers S, Grieve RJ, et al. Epirubicin and cyclophosphamide, methotrexate, and fluorouracil as adjuvant therapy for early breast cancer. N Engl J Med. 2006;355:1851–1862. doi: 10.1056/NEJMoa052084.
    1. Whitfield ML, Zheng LX, Baldwin A, Ohta T, Hurt MM, Marzluff WF. Stem-loop binding protein, the protein that binds the 3′ end of histone mRNA, is cell cycle regulated by both translational and posttranslational mechanisms. Mol Cell Biol. 2000;20:4188–4198. doi: 10.1128/MCB.20.12.4188-4198.2000.
    1. Kanehisa M, Goto S, Sato Y, Kawashima M, Furumichi M, Tanabe M. Data, information, knowledge and principle: back to metabolism in KEGG. Nucleic Acids Res. 2014;42(Database issue):D199–D205. doi: 10.1093/nar/gkt1076.
    1. Subik K, Lee JF, Baxter L, Strzepek T, Costello D, Crowley P, et al. The expression patterns of ER, PR, HER2, CK5/6, EGFR, Ki-67 and AR by immunohistochemical analysis in breast cancer cell lines. Breast Cancer (Auckl) 2010;4:35–41.
    1. Győrffy B, Lanczky A, Eklund AC, Denkert C, Budczies J, Li Q, et al. An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res Treat. 2010;123:725–731. doi: 10.1007/s10549-009-0674-9.
    1. Nayak SR, Harrington E, Boone D, Hartmaier R, Chen J, Pathiraja TN, et al. A role for histone H2B variants in endocrine-resistant breast cancer. Horm Cancer. 2015;6:214–224. doi: 10.1007/s12672-015-0230-5.
    1. Vardabasso C, Gaspar-Maia A, Hasson D, Pünzeler S, Valle-Garcia D, Straub T, et al. Histone variant H2A.Z.2 mediates proliferation and drug sensitivity of malignant melanoma. Mol Cell. 2015;59:75–88. doi: 10.1016/j.molcel.2015.05.009.
    1. Bonenfant D, Coulot M, Towbin H, Schindler P, van Oostrum J. Characterization of histone H2A and H2B variants and their post-translational modifications by mass spectrometry. Mol Cell Proteomics. 2006;5:541–552. doi: 10.1074/mcp.M500288-MCP200.
    1. Hembruff SL, Laberge ML, Villeneuve DJ, Guo B, Veitch Z, Cecchetto M, et al. Role of drug transporters and drug accumulation in the temporal acquisition of drug resistance. BMC Cancer. 2008;8:318. doi: 10.1186/1471-2407-8-318.
    1. Wyrick JJ, Parra MA. The role of histone H2A and H2B post-translational modifications in transcription: a genomic perspective. Biochim Biophys Acta. 2009;1789:37–44. doi: 10.1016/j.bbagrm.2008.07.001.
    1. Harris ME, Bohni R, Schneiderman MH, Ramamurthy L, Schumperli D, Marzluff WF. Regulation of histone mRNA in the unperturbed cell cycle: evidence suggesting control at two posttranscriptional steps. Mol Cell Biol. 1991;11:2416–2424. doi: 10.1128/MCB.11.5.2416.
    1. Parra MA, Wyrick JJ. Regulation of gene transcription by the histone H2A N-terminal domain. Mol Cell Biol. 2007;27:7641–7648. doi: 10.1128/MCB.00742-07.
    1. Parra MA, Kerr D, Fahy D, Pouchnik DJ, Wyrick JJ. Deciphering the roles of the histone H2B N-terminal domain in genome-wide transcription. Mol Cell Biol. 2006;26:3842–3852. doi: 10.1128/MCB.26.10.3842-3852.2006.
    1. Beck HC, Nielsen EC, Matthiesen R, Jensen LH, Sehested M, Finn P, et al. Quantitative proteomic analysis of post-translational modifications of human histones. Mol Cell Proteomics. 2006;5:1314–1325. doi: 10.1074/mcp.M600007-MCP200.
    1. Swierniak A, Kimmel M, Smieja J. Mathematical modeling as a tool for planning anticancer therapy. Eur J Pharmacol. 2009;625:108–121. doi: 10.1016/j.ejphar.2009.08.041.
    1. Norton L, Simon R. Tumor size, sensitivity to therapy, and design of treatment schedules. Cancer Treat Rep. 1977;61:1307–1317.
    1. Norton L, Simon R, Brereton HD, Bogden AE. Predicting the course of Gompertzian growth. Nature. 1976;264:542–545. doi: 10.1038/264542a0.
    1. Regel I, Merkl L, Friedrich T, Burgermeister E, Zimmermann W, Einwachter H, et al. Pan-histone deacetylase inhibitor panobinostat sensitizes gastric cancer cells to anthracyclines via induction of CITED2. Gastroenterology. 2012;143:99–109. doi: 10.1053/j.gastro.2012.03.035.
    1. Groselj B, Sharma NL, Hamdy FC, Kerr M, Kiltie AE. Histone deacetylase inhibitors as radiosensitisers: effects on DNA damage signalling and repair. Br J Cancer. 2013;108:748–754. doi: 10.1038/bjc.2013.21.
    1. Wagner JM, Hackanson B, Lubbert M, Jung M. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin Epigenetics. 2010;1:117–136. doi: 10.1007/s13148-010-0012-4.
    1. Lee JH, Choy ML, Marks PA. Mechanisms of resistance to histone deacetylase inhibitors. Adv Cancer Res. 2012;116:39–86. doi: 10.1016/B978-0-12-394387-3.00002-1.
    1. Gosland MP, Gillespie MN, Tsuboi CP, Tofiq S, Olson JW, Crooks PA, et al. Reversal of doxorubicin, etoposide, vinblastine, and taxol resistance in multidrug resistant human sarcoma cells by a polymer of spermine. Cancer Chemother Pharmacol. 1996;37:593–600. doi: 10.1007/s002800050434.
    1. Paridaens R, Biganzoli L, Bruning P, Klijn JG, Gamucci T, Houston S, et al. Paclitaxel versus doxorubicin as first-line single-agent chemotherapy for metastatic breast cancer: a European Organization for Research and Treatment of Cancer Randomized Study with cross-over. J Clin Oncol. 2000;18:724–733.
    1. Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst. 2009;101:1446–1452. doi: 10.1093/jnci/djp335.

Source: PubMed

3
Iratkozz fel