Int J Stem Cells.  2018 Jun;11(1):131-140. 10.15283/ijsc18021.

The Bromodomain Inhibitor JQ1 Enhances the Responses to All-trans Retinoic Acid in HL-60 and MV4-11 Leukemia Cells

Affiliations
  • 1Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea. stemchung@gmail.com
  • 2Department of Immunology and Physiology, School of Medicine, Konkuk University, Seoul, Korea.
  • 3Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Korea.

Abstract

All-trans retinoic acid (ATRA) is a highly effective treatment for acute promyelocytic leukemia (APL), a cytogenetically distinct subtype of acute myeloid leukemia (AML). However, ATRA-based treatment is not effective in other subtypes of AML. In non-APL AML, ATRA signaling pathway is impaired or downmodulated, and consequently fails to respond to pharmacological doses of ATRA. Therefore, complementary treatment strategies are needed to improve ATRA responsiveness in non-APL AML. In this study, we investigated the combined effect of ATRA and bromodomain inhibitor JQ1, proven to have potent anti-cancer activity mainly through inhibition of c-Myc. We showed that the combination of ATRA with JQ1 synergistically inhibited proliferation of AML cells. The synergistic growth inhibition was resulted from differentiation or apoptosis depending on the kind of AML cells. Concomitantly, the combined treatment of ATRA and JQ1 caused greater depletion of c-Myc and hTERT expression than each agent alone in AML cells. Taken together, these findings support the rationale for the use of the combination of ATRA and JQ1 as a therapeutic strategy for the treatment of AML.

Keyword

Acute myeloid leukemia; All-trans retinoic acid; JQ1; c-Myc; Differentiation; Apoptosis

MeSH Terms

Apoptosis
Leukemia*
Leukemia, Myeloid, Acute
Leukemia, Promyelocytic, Acute
Tretinoin*
Tretinoin

Figure

  • Fig. 1 JQ1 synergizes with ATRA to reduce proliferation of AML cells. (A) HL-60 cells and MV4-11 cells were treated with ATRA (20~100 nM) for the indicated days. Cell proliferation was measured using a CCK-8 assay. The data represent the means±SD of 3 independent experiments. *ATRA versus control (***p<0.001). (B) HL-60 cells and MV4-11 cells were treated with the indicated concentrations of JQ1 and/or 100 nM ATRA for 3 days. Cell proliferation was measured using a CCK-8 assay. The data represent the means±SD of 3 independent experiments. *combination versus ATRA (***p<0.001); †combination versus JQ1 (†††p<0.001). (C) CDI values at different concentrations of JQ1 and 100 nM ATRA were calculated as described in materials and methods.

  • Fig. 2 Effect of JQ1 on ATRA-induced HL-60 cell differentiation. HL-60 cells were treated with the indicated concentrations of JQ1 and/or 100 nM ATRA for 3 days. (A) Histogram (left) and bar graph (right) represented CD11b expression. Values were expressed as Mean fluorescence of intensity (MFI). Experiments were independently repeated three times and results were shown as mean±SD. (B) The mRNA levels of C/EBPɛ were determined by real-time PCR. The relative expression of C/EBPɛ mRNA was normalized to GAPDH and was reported as a percentage of untreated cells. (C) The protein levels of C/EBPɛ were measured by Western blot analysis using 20 μg protein. Similar results were obtained in three independent experiments. *combination versus ATRA (***p<0.001; **p<0.01).

  • Fig. 3 Effect of combined treatment with JQ1 and ATRA on apoptosis of AML cells. (A) HL-60 cells and (B) MV4-11 cells were treated with the indicated concentrations of JQ1 and/or 100 nM ATRA for 48 h. At the end of treatment, cells were washed with staining buffer and stained with Annexin V-FITC. Annexin-V positive, apoptotic cells was determined by flow cytometry. Numbers in each plot represented the percentage of Annexin-V positive cells. Bar graph showed means±SD of 3 independent experiments. *combination versus ATRA (**p<0.01; *p<0.05; ns, not significant).

  • Fig. 4 Effect of JQ1 and ATRA on the expression of c-Myc and hTERT mRNA. HL-60 cells (left) and MV4-11 cells (right) were treated with 100 nM ATRA and/or the indicated concentrations of JQ1 for 24 h. At the end of treatment, cells were harvest and analyzed in the following experiments. (A) The mRNA levels of c-Myc were determined by real-time PCR. The relative expression of c-Myc mRNA was normalized to GAPDH and was reported as a percentage of untreated cells. (B) The protein levels of c-Myc were measured by Western blot analysis using 20 μg protein. Similar results were obtained in three independent experiments. (C) The mRNA levels of hTERT were determined by real-time PCR. The relative expression of hTERT mRNA was normalized to GAPDH and was reported as a percentage of untreated cells. *combination versus ATRA (**p<0.01; *p<0.05); †combination versus JQ1 (††p<0.01; †p<0.05).


Reference

References

1. Balmer JE, Blomhoff R. Gene expression regulation by retinoic acid. J Lipid Res. 2002; 43:1773–1808. DOI: 10.1194/jlr.R100015-JLR200. PMID: 12401878.
Article
2. Chambon P. A decade of molecular biology of retinoic acid receptors. FASEB J. 1996; 10:940–954. DOI: 10.1096/fasebj.10.9.8801176. PMID: 8801176.
Article
3. de Thé H, Lavau C, Marchio A, Chomienne C, Degos L, Dejean A. The PML-RAR alpha fusion mRNA generated by the t(15;17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR. Cell. 1991; 66:675–684. DOI: 10.1016/0092-8674(91)90113-D. PMID: 1652369.
Article
4. Lin RJ, Nagy L, Inoue S, Shao W, Miller WH Jr, Evans RM. Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature. 1998; 391:811–814. DOI: 10.1038/35895.
Article
5. Grignani F, De Matteis S, Nervi C, Tomassoni L, Gelmetti V, Cioce M, Fanelli M, Ruthardt M, Ferrara FF, Zamir I, Seiser C, Grignani F, Lazar MA, Minucci S, Pelicci PG. Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature. 1998; 391:815–818. DOI: 10.1038/35901. PMID: 9486655.
Article
6. Schenk T, Stengel S, Zelent A. Unlocking the potential of retinoic acid in anticancer therapy. Br J Cancer. 2014; 111:2039–2045. DOI: 10.1038/bjc.2014.412. PMID: 25412233. PMCID: 4260020.
Article
7. Scholl S, Müller R, Clement JH, Loncarevic IF, Böhmer FD, Höffken K. ATRA can enhance apoptosis that is induced by Flt3 tyrosine kinase inhibition in Flt3-ITD positive cells. Leuk Res. 2006; 30:633–642. DOI: 10.1016/j.leukres.2005.10.005. PMID: 16473406.
Article
8. Schenk T, Chen WC, Göllner S, Howell L, Jin L, Hebestreit K, Klein HU, Popescu AC, Burnett A, Mills K, Casero RA Jr, Marton L, Woster P, Minden MD, Dugas M, Wang JC, Dick JE, Müller-Tidow C, Petrie K, Zelent A. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-transretinoic acid differentiation pathway in acute myeloid leukemia. Nat Med. 2012; 18:605–611. DOI: 10.1038/nm.2661. PMID: 22406747. PMCID: 3539284.
Article
9. Ma HS, Greenblatt SM, Shirley CM, Duffield AS, Bruner JK, Li L, Nguyen B, Jung E, Aplan PD, Ghiaur G, Jones RJ, Small D. All-trans retinoic acid synergizes with FLT3 inhibition to eliminate FLT3/ITD+ leukemia stem cells in vitro and in vivo. Blood. 2016; 127:2867–2878. DOI: 10.1182/blood-2015-05-646786. PMID: 27103744. PMCID: 4900954.
Article
10. Adhikary S, Eilers M. Transcriptional regulation and transformation by Myc proteins. Nat Rev Mol Cell Biol. 2005; 6:635–645. DOI: 10.1038/nrm1703. PMID: 16064138.
Article
11. Delgado MD, León J. Myc roles in hematopoiesis and leukemia. Genes Cancer. 2010; 1:605–616. DOI: 10.1177/1947601910377495.
Article
12. Uribesalgo I, Buschbeck M, Gutiérrez A, Teichmann S, Demajo S, Kuebler B, Nomdedéu JF, Martín-Caballero J, Roma G, Benitah SA, Di Croce L. E-box-independent regulation of transcription and differentiation by MYC. Nat Cell Biol. 2011; 13:1443–1449. DOI: 10.1038/ncb2355. PMID: 22020439.
Article
13. Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J, Chesi M, Schinzel AC, McKeown MR, Heffernan TP, Vakoc CR, Bergsagel PL, Ghobrial IM, Richardson PG, Young RA, Hahn WC, Anderson KC, Kung AL, Bradner JE, Mitsiades CS. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011; 146:904–917. DOI: 10.1016/j.cell.2011.08.017. PMCID: 3187920.
Article
14. Zuber J, Shi J, Wang E, Rappaport AR, Herrmann H, Sison EA, Magoon D, Qi J, Blatt K, Wunderlich M, Taylor MJ, Johns C, Chicas A, Mulloy JC, Kogan SC, Brown P, Valent P, Bradner JE, Lowe SW, Vakoc CR. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature. 2011; 478:524–528. DOI: 10.1038/nature10334. PMID: 21814200. PMCID: 3328300.
Article
15. Jang MK, Mochizuki K, Zhou M, Jeong HS, Brady JN, Ozato K. The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Mol Cell. 2005; 19:523–534. DOI: 10.1016/j.molcel.2005.06.027. PMID: 16109376.
Article
16. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I, Philpott M, Munro S, McKeown MR, Wang Y, Christie AL, West N, Cameron MJ, Schwartz B, Heightman TD, La Thangue N, French CA, Wiest O, Kung AL, Knapp S, Bradner JE. Selective inhibition of BET bromodomains. Nature. 2010; 468:1067–1073. DOI: 10.1038/nature09504. PMID: 20871596. PMCID: 3010259.
Article
17. Berthon C, Raffoux E, Thomas X, Vey N, Gomez-Roca C, Yee K, Taussig DC, Rezai K, Roumier C, Herait P, Kahatt C, Quesnel B, Michallet M, Recher C, Lokiec F, Preudhomme C, Dombret H. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 2016; 3:e186–e195. DOI: 10.1016/S2352-3026(15)00247-1. PMID: 27063977.
Article
18. Amorim S, Stathis A, Gleeson M, Iyengar S, Magarotto V, Leleu X, Morschhauser F, Karlin L, Broussais F, Rezai K, Herait P, Kahatt C, Lokiec F, Salles G, Facon T, Palumbo A, Cunningham D, Zucca E, Thieblemont C. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol. 2016; 3:e196–e204. DOI: 10.1016/S2352-3026(16)00021-1. PMID: 27063978.
Article
19. Stathis A, Zucca E, Bekradda M, Gomez-Roca C, Delord JP, de La Motte Rouge T, Uro-Coste E, de Braud F, Pelosi G, French CA. Clinical response of carcinomas harboring the BRD4-NUT oncoprotein to the targeted bromodomain inhibitor OTX015/MK-8628. Cancer Discov. 2016; 6:492–500. DOI: 10.1158/2159-8290.CD-15-1335. PMID: 26976114. PMCID: 4854801.
Article
20. Fong CY, Gilan O, Lam EY, Rubin AF, Ftouni S, Tyler D, Stanley K, Sinha D, Yeh P, Morison J, Giotopoulos G, Lugo D, Jeffrey P, Lee SC, Carpenter C, Gregory R, Ramsay RG, Lane SW, Abdel-Wahab O, Kouzarides T, Johnstone RW, Dawson SJ, Huntly BJ, Prinjha RK, Papenfuss AT, Dawson MA. BET inhibitor resistance emerges from leukaemia stem cells. Nature. 2015; 525:538–542. DOI: 10.1038/nature14888. PMID: 26367796.
Article
21. Rathert P, Roth M, Neumann T, Muerdter F, Roe JS, Muhar M, Deswal S, Cerny-Reiterer S, Peter B, Jude J, Hoffmann T, Boryń ŁM, Axelsson E, Schweifer N, Tontsch-Grunt U, Dow LE, Gianni D, Pearson M, Valent P, Stark A, Kraut N, Vakoc CR, Zuber J. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature. 2015; 525:543–547. DOI: 10.1038/nature14898. PMID: 26367798. PMCID: 4921058.
Article
22. Fiskus W, Sharma S, Qi J, Valenta JA, Schaub LJ, Shah B, Peth K, Portier BP, Rodriguez M, Devaraj SG, Zhan M, Sheng J, Iyer SP, Bradner JE, Bhalla KN. Highly active combination of BRD4 antagonist and histone deacetylase inhibitor against human acute myelogenous leukemia cells. Mol Cancer Ther. 2014; 13:1142–1154. DOI: 10.1158/1535-7163.MCT-13-0770. PMID: 24435446.
Article
23. Moros A, Rodríguez V, Saborit-Villarroya I, Montraveta A, Balsas P, Sandy P, Martínez A, Wiestner A, Normant E, Campo E, Pérez-Galán P, Colomer D, Roué G. Synergistic antitumor activity of lenalidomide with the BET bromodomain inhibitor CPI203 in bortezomib-resistant mantle cell lymphoma. Leukemia. 2014; 28:2049–2059. DOI: 10.1038/leu.2014.106. PMID: 24721791.
Article
24. Siegel MB, Liu SQ, Davare MA, Spurgeon SE, Loriaux MM, Druker BJ, Scott EC, Tyner JW. Small molecule inhibitor screen identifies synergistic activity of the bromodomain inhibitor CPI203 and bortezomib in drug resistant myeloma. Oncotarget. 2015; 6:18921–18932. DOI: 10.18632/oncotarget.4214. PMID: 26254279. PMCID: 4662464.
Article
25. Park DJ, Chumakov AM, Vuong PT, Chih DY, Gombart AF, Miller WH Jr, Koeffler HP. CCAAT/enhancer binding protein epsilon is a potential retinoid target gene in acute promyelocytic leukemia treatment. J Clin Invest. 1999; 103:1399–1408. DOI: 10.1172/JCI2887. PMID: 10330422. PMCID: 408448.
Article
26. Brondfield S, Umesh S, Corella A, Zuber J, Rappaport AR, Gaillard C, Lowe SW, Goga A, Kogan SC. Direct and indirect targeting of MYC to treat acute myeloid leukemia. Cancer Chemother Pharmacol. 2015; 76:35–46. DOI: 10.1007/s00280-015-2766-z. PMID: 25956709. PMCID: 4485702.
Article
27. Greenberg RA, O’Hagan RC, Deng H, Xiao Q, Hann SR, Adams RR, Lichtsteiner S, Chin L, Morin GB, DePinho RA. Telomerase reverse transcriptase gene is a direct target of c-Myc but is not functionally equivalent in cellular transformation. Oncogene. 1999; 18:1219–1226. DOI: 10.1038/sj.onc.1202669. PMID: 10022128.
Article
28. Tallman MS, Andersen JW, Schiffer CA, Appelbaum FR, Feusner JH, Ogden A, Shepherd L, Willman C, Bloomfield CD, Rowe JM, Wiernik PH. All-trans-retinoic acid in acute promyelocytic leukemia. N Engl J Med. 1997; 337:1021–1028. DOI: 10.1056/NEJM199710093371501. PMID: 9321529.
Article
29. Warrell RP Jr, Maslak P, Eardley A, Heller G, Miller WH Jr, Frankel SR. Treatment of acute promyelocytic leukemia with all-trans retinoic acid: an update of the New York experience. Leukemia. 1994; 8:929–933. PMID: 8207986.
30. Huang MJ, Cheng YC, Liu CR, Lin S, Liu HE. A small-molecule c-Myc inhibitor, 10058-F4, induces cell-cycle arrest, apoptosis, and myeloid differentiation of human acute myeloid leukemia. Exp Hematol. 2006; 34:1480–1489. DOI: 10.1016/j.exphem.2006.06.019. PMID: 17046567.
Article
31. Pan XN, Chen JJ, Wang LX, Xiao RZ, Liu LL, Fang ZG, Liu Q, Long ZJ, Lin DJ. Inhibition of c-Myc overcomes cytotoxic drug resistance in acute myeloid leukemia cells by promoting differentiation. PLos One. 2014; 9:e105381. DOI: 10.1371/journal.pone.0105381. PMID: 25127121. PMCID: 4134294.
Article
32. Bentley DL, Groudine M. A block to elongation is largely responsible for decreased transcription of c-myc in differentiated HL60 cells. Nature. 1986; 321:702–706. DOI: 10.1038/321702a0. PMID: 3520340.
Article
33. Counter CM, Meyerson M, Eaton EN, Ellisen LW, Caddle SD, Haber DA, Weinberg RA. Telomerase activity is restored in human cells by ectopic expression of hTERT (hEST2), the catalytic subunit of telomerase. Oncogene. 1998; 16:1217–1222. DOI: 10.1038/sj.onc.1201882. PMID: 9528864.
Article
34. Röth A, Vercauteren S, Sutherland HJ, Lansdorp PM. Telomerase is limiting the growth of acute myeloid leukemia cells. Leukemia. 2003; 17:2410–2417. DOI: 10.1038/sj.leu.2403177. PMID: 14562114.
Article
35. Bruedigam C, Bagger FO, Heidel FH, Paine Kuhn C, Guignes S, Song A, Austin R, Vu T, Lee E, Riyat S, Moore AS, Lock RB, Bullinger L, Hill GR, Armstrong SA, Williams DA, Lane SW. Telomerase inhibition effectively targets mouse and human AML stem cells and delays relapse following chemotherapy. Cell Stem Cell. 2014; 15:775–790. DOI: 10.1016/j.stem.2014.11.010. PMID: 25479751. PMCID: 4317339.
Article
36. Blagitko-Dorfs N, Jiang Y, Duque-Afonso J, Hiller J, Yalcin A, Greve G, Abdelkarim M, Hackanson B, Lübbert M. Epigenetic priming of AML blasts for all-trans retinoic acid-induced differentiation by the HDAC class-I selective inhibitor entinostat. PLoS One. 2013; 8:e75258. DOI: 10.1371/journal.pone.0075258. PMID: 24116031. PMCID: 3792939.
Article
37. Shao X, Liu Y, Li Y, Xian M, Zhou Q, Yang B, Ying M, He Q. The HER2 inhibitor TAK165 sensitizes human acute myeloid leukemia cells to retinoic acid-induced myeloid differentiation by activating MEK/ERK mediated RARα/STAT1 axis. Sci Rep. 2016; 6:24589. DOI: 10.1038/srep24589.
Article
38. Boutzen H, Saland E, Larrue C, de Toni F, Gales L, Castelli FA, Cathebas M, Zaghdoudi S, Stuani L, Kaoma T, Riscal R, Yang G, Hirsch P, David M, De Mas-Mansat V, Delabesse E, Vallar L, Delhommeau F, Jouanin I, Ouerfelli O, Le Cam L, Linares LK, Junot C, Portais JC, Vergez F, Récher C, Sarry JE. Isocitrate dehydrogenase 1 mutations prime the all-trans retinoic acid myeloid differentiation pathway in acute myeloid leukemia. J Exp Med. 2016; 213:483–497. DOI: 10.1084/jem.20150736. PMID: 26951332. PMCID: 4821643.
Article
Full Text Links
  • IJSC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr