Azacytidine sensitizes acute myeloid leukemia cells to arsenic trioxide by up-regulating the arsenic transporter aquaglyceroporin 9

David Chau, Karen Ng, Thomas Sau-Yan Chan, Yuen-Yee Cheng, Bonnie Fong, Sidney Tam, Yok-Lam Kwong, Eric Tse, David Chau, Karen Ng, Thomas Sau-Yan Chan, Yuen-Yee Cheng, Bonnie Fong, Sidney Tam, Yok-Lam Kwong, Eric Tse

Abstract

Background: The therapeutic efficacy of arsenic trioxide (As2O3) in acute myeloid leukemia (AML) is modest, which is partly related to its limited intracellular uptake into the leukemic cells. As2O3 enters cells via the transmembrane protein aquaglyceroporin 9 (AQP9). Azacytidine, a demethylating agent that is approved for the treatment of AML, has been shown to have synergistic effect with As2O3. We tested the hypothesis that azacytidine might up-regulate AQP9 and enhances As2O3-mediated cytotoxicity in AML.

Methods: Arsenic-induced cytotoxicity, the expression of AQP9, and the intracellular uptake of As2O3 were determined in AML cell lines and primary AML cells with or without azacytidine pre-treatment. The mechanism of AQP9 up-regulation was then investigated by examining the expression of transcription factors for AQP9 gene and the methylation status of their gene promoters.

Results: As2O3-induced cytotoxicity in AML cell lines was significantly enhanced after azacytidine pre-treatment as a result of AQP9 up-regulation, leading to increased arsenic uptake and hence intracellular concentration. Blocking AQP9-mediated As2O3 uptake with mercury chloride abrogated the sensitization effect of azacytidine. AQP9 promoter does not contain CpG islands. Instead, azacytidine pre-treatment led to increased expression of HNF1A, a transcription activator of AQP9, through demethylation of HNF1A promoter. HNF1 knockdown abrogated azacytidine-induced AQP9 up-regulation and almost completely blocked intracellular As2O3 entry, confirming that azacytidine enhanced As2O3-mediated cell death via up-regulation of HNF1A and hence increased AQP9 and As2O3 intracellular concentration. Azacytidine sensitization to As2O3 treatment was re-capitulated also in primary AML samples. Finally, azacytidine did not enhance arsenic toxicity in a liver cell line, where HNF1A was largely unmethylated.

Conclusions: Azacytidine sensitizes AML cells to As2O3 treatment, and our results provide proof-of-principle evidence that pharmacological up-regulation of AQP9 potentially expands the therapeutic spectrum of As2O3. Further clinical trial should evaluate the efficacy of azacytidine in combination with As2O3 in the treatment of AML.

Figures

Figure 1
Figure 1
Sensitization of leukemia cells to As2O3-induced cytotoxicity with azacytidine (5′Aza) pre-treatment. (A) Dose-dependent cytotoxicity of As2O3 with or without 5′Aza treatment in the human AML cell lines, HL-60, K562, NB4, and OCI-AML3. As determined by MTT assay, all four cell lines pre-treated with 5′Aza exhibited significant decrease in cell viability as compared with their untreated controls. Data were acquired from three independent experiments, and relative survival of each individual cell line was normalized to its respective control. (B) Representative plot of flow cytometric analysis, showing a significant increase in As2O3-mediated cytotoxicity (annexin V-positive cells) in K562 cells pre-treatment with 5′Aza, as compared with control cells not treated with 5′Aza. (C) Bar charts showing the fold changes of As2O3-induced cell death (annexin V-positive cells) in each AML lines with or without 5′Aza pre-treatment. Quantitative analysis by flow cytometry further confirmed a significant increase in As2O3-induced apoptotic cell death after 5′Aza pre-treatment.
Figure 2
Figure 2
Azacytidine-mediated up-regulation of AQP9 expression and intracellular arsenic uptake. (A) AQP9 mRNA expression was significantly up-regulated with 5′Aza treatment in HL-60, K562, NB4, and OCI-AML3 cells, as determined by RT-PCR (upper panel) and quantitative RT-PCR (lower panel). Sample data were normalized to the control of each cell line individually by the ΔΔCT method. (B) Up-regulation of membrane AQP9 protein expression was also confirmed by flow cytometric analysis. (C) Intracellular elemental arsenic concentration is significantly increased in K562 cells after pre-treatment with 5′Aza. (D) Treatment of K562 cells with HgCl2 completely abrogated the cytotoxicity induced by As2O3, either alone (left panel) or in cells pre-treatment with 5′Aza (right panel).
Figure 3
Figure 3
Quantitative RT-PCR analysis of the expression of potential transcription factors for the AQP9 gene after 5′Aza treatment in HL-60 and K562 cells. The expressions of five transcription factors, including HNF1A, CEBPα, CEBPγ, NF-1, and JUN, with or without 5′Aza treatment in HL-60 and K562 cells, were determined by quantitative RT-PCR. Sample data were normalized to the control gene GAPDH for each cell line individually.
Figure 4
Figure 4
Transcription factor HNF1A was involved in azacytidine-induced up-regulation of AQP9. (A). HNF1A mRNA and protein expressions were markedly up-regulated after treatment of 5′Aza as examined by semi-quantitative and quantitative RT-PCR (upper panel) and western immunoblotting (lower panel). (B) Diagram depicting the regions of the HNF1A promoter analyzed by Methylation-specific PCR (MSP) and Combined Bisulfite Restriction Analysis (COBRA). (C) The methylation status of HNF1A gene promoter was extensively studied and determined using combined bisulfite restriction analysis. 5′Aza treatment resulted in demethylation of the HNF1A gene promoter. (C: control; IVD: universal methylated DNA). (D) Methylation-specific PCR (MSP) was performed using PCR primers specific for methylated or unmethylated HNF1A gene promoter. The results showed that the HNF1A gene promoter was highly methylated, and 5′Aza treatment led to demethylation of the HNF1A gene promoter. (E) Specific HNF1A siRNA abrogated 5′Aza-induced AQP9 up-regulation as determined by RT-PCR (upper panel) and quantitative RT-PCR (lower panel). (F) In K562 cells treated with 5′Aza, specific HNF1A siRNA almost completely blocked the intracellular entry of As2O3 as compared with the control siRNA-treated cells. The results showed that HNF1A was the mediator for up-regulation of AQP9 after 5′Aza treatment.
Figure 5
Figure 5
Methylation status of HNF1A promoter and correlation between the expressions of AQP9 and HNF1A in primary AML samples. (A) MSP showing highly methylated HNF1A promoter in 16 human AML samples. (B) Quantification of AQP9 and HNF1A expressions in 16 human AML samples. Result was analyzed using Pearson test. AQP9 level was found to correlate positively with HNF1A level (p = 0.0045).
Figure 6
Figure 6
Pre-treatment with azacytidine sensitized primary AML cells to subsequent treatment with As2O3. (A) Treatment with 5′Aza resulted in significant up-regulation of HNF1A (left panel) and AQP9 (right panel) as determined by quantitative RT-PCR. (B) As2O3-mediated cytotoxicity in primary AML cells with or without 5′Aza pre-treatment as determined by MTT analysis. Sample data were normalized to the controls of each patient individually. Results represented triplicates of seven samples of primary AML cells.
Figure 7
Figure 7
Azacytidine did not enhance hepatotoxicity of As2O3in vitro. (A) Basal expression of AQP9 was much higher in MIHA compared with those of AML cells. (B) 5′Aza treatment did not increase AQP9 expression significantly in MIHA. (C) HNF1A promoter was hypomethylated in MIHA (upper panel), and 5′Aza treatment did not further increase HNF1A expression (lower panel). Sample data were normalized to the respective control. (D) Pre-treatment with 5′Aza did not significantly enhance the cytotoxicity of As2O3 in MIHA cells. Sample data were normalized to the respective control individually.

References

    1. Tallman MS, Altman JK. How I treat acute promyelocytic leukemia. Blood. 2009;114:5126–35. doi: 10.1182/blood-2009-07-216457.
    1. Sanz MA, Lo-Coco F. Modern approaches to treating acute promyelocytic leukemia. J Clin Oncol. 2011;29:495–503. doi: 10.1200/JCO.2010.32.1067.
    1. Soignet SL, Frankel SR, Douer D, Tallman MS, Kantarjian H, Calleja E, et al. United States multicenter study of arsenic trioxide in relapsed acute promyelocytic leukemia. J Clin Oncol. 2001;19:3852–60.
    1. Shen ZX, Shi ZZ, Fang J, Gu BW, Li JM, Zhu YM, et al. All-trans retinoic acid/As2O3 combination yields a high quality remission and survival in newly diagnosed acute promyelocytic leukemia. Proc Natl Acad Sci U S A. 2004;101:5328–35. doi: 10.1073/pnas.0400053101.
    1. Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando SM, Iacobelli S, et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med. 2013;369:111–21. doi: 10.1056/NEJMoa1300874.
    1. Au WY, Kumana CR, Lee HK, Lin SY, Liu H, Yeung DY, et al. Oral arsenic trioxide-based maintenance regimens for first complete remission of acute promyelocytic leukemia: a 10-year follow-up study. Blood. 2011;118:6535–43. doi: 10.1182/blood-2011-05-354530.
    1. Zhang XW, Yan XJ, Zhou ZR, Yang FF, Wu ZY, Sun HB, et al. Arsenic trioxide controls the fate of the PML-RARalpha oncoprotein by directly binding PML. Science. 2010;328:240–3. doi: 10.1126/science.1183424.
    1. Tatham MH, Geoffroy MC, Shen L, Plechanovova A, Hattersley N, Jaffray EG, et al. RNF4 is a poly-SUMO-specific E3 ubiquitin ligase required for arsenic-induced PML degradation. Nat Cell Biol. 2008;10:538–46. doi: 10.1038/ncb1716.
    1. Jeanne M, Lallemand-Breitenbach V, Ferhi O, Koken M, Le Bras M, Duffort S, et al. PML/RARA oxidation and arsenic binding initiate the antileukemia response of As2O3. Cancer Cell. 2010;18:88–98. doi: 10.1016/j.ccr.2010.06.003.
    1. Chen YC, Lin-Shiau SY, Lin JK. Involvement of reactive oxygen species and caspase 3 activation in arsenite-induced apoptosis. J Cell Physiol. 1998;177:324–33. doi: 10.1002/(SICI)1097-4652(199811)177:2<324::AID-JCP14>;2-9.
    1. Larochette N, Decaudin D, Jacotot E, Brenner C, Marzo I, Susin SA, et al. Arsenite induces apoptosis via a direct effect on the mitochondrial permeability transition pore. Exp Cell Res. 1999;249:413–21. doi: 10.1006/excr.1999.4519.
    1. Li H, Wang Y, Xu W, Dong L, Guo Y, Bi K, et al. Arsenic trioxide inhibits DNA methyltransferase and restores TMS1 gene expression in K562 cells. Acta Haematol. 2015;133:18–25. doi: 10.1159/000362683.
    1. Tsukaguchi H, Weremowicz S, Morton CC, Hediger MA. Functional and molecular characterization of the human neutral solute channel aquaporin-9. Am J Physiol. 1999;277:F685–96.
    1. Leung J, Pang A, Yuen WH, Kwong YL, Tse EW. Relationship of expression of aquaglyceroporin 9 with arsenic uptake and sensitivity in leukemia cells. Blood. 2007;109:740–6. doi: 10.1182/blood-2006-04-019588.
    1. Bhattacharjee H, Carbrey J, Rosen BP, Mukhopadhyay R. Drug uptake and pharmacological modulation of drug sensitivity in leukemia by AQP9. Biochem Biophys Res Commun. 2004;322:836–41. doi: 10.1016/j.bbrc.2004.08.002.
    1. Lubbert M, Suciu S, Baila L, Ruter BH, Platzbecker U, Giagounidis A, et al. Low-dose decitabine versus best supportive care in elderly patients with intermediate- or high-risk myelodysplastic syndrome (MDS) ineligible for intensive chemotherapy: final results of the randomized phase III study of the European Organisation for Research and Treatment of Cancer Leukemia Group and the German MDS Study Group. J Clin Oncol. 2011;29:1987–96. doi: 10.1200/JCO.2010.30.9245.
    1. Cashen AF, Schiller GJ, O’Donnell MR, DiPersio JF. Multicenter, phase II study of decitabine for the first-line treatment of older patients with acute myeloid leukemia. J Clin Oncol. 2010;28:556–61. doi: 10.1200/JCO.2009.23.9178.
    1. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Gattermann N, Germing U, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol. 2010;28:562–9. doi: 10.1200/JCO.2009.23.8329.
    1. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10:223–32. doi: 10.1016/S1470-2045(09)70003-8.
    1. Peng CY, Jiang J, Zheng HT, Liu XS. Growth-inhibiting effects of arsenic trioxide plus epigenetic therapeutic agents on leukemia cell lines. Leuk Lymphoma. 2010;51:297–303. doi: 10.3109/10428190903486212.
    1. Welch JS, Klco JM, Gao F, Procknow E, Uy GL, Stockerl-Goldstein KE, et al. Combination decitabine, arsenic trioxide, and ascorbic acid for the treatment of myelodysplastic syndrome and acute myeloid leukemia: a phase I study. Am J Hematol. 2011;86:796–800. doi: 10.1002/ajh.22092.
    1. Pang RW, Lee TK, Man K, Poon RT, Fan ST, Kwong YL, et al. PIN1 expression contributes to hepatic carcinogenesis. J Pathol. 2006;210:19–25. doi: 10.1002/path.2024.
    1. Au WY, Cheung GT, Yuen TW, Kumana CR, Kwong YL. Successful treatment of relapsed acute promyelocytic leukemia in a patient receiving continuous ambulatory peritoneal dialysis with oral arsenic trioxide. Arch Intern Med. 2005;165:1067–8. doi: 10.1001/archinte.165.9.1067.
    1. Ishibashi K, Kuwahara M, Gu Y, Tanaka Y, Marumo F, Sasaki S. Cloning and functional expression of a new aquaporin (AQP9) abundantly expressed in the peripheral leukocytes permeable to water and urea, but not to glycerol. Biochem Biophys Res Commun. 1998;244:268–74. doi: 10.1006/bbrc.1998.8252.
    1. Carbrey JM, Song L, Zhou Y, Yoshinaga M, Rojek A, Wang Y, et al. Reduced arsenic clearance and increased toxicity in aquaglyceroporin-9-null mice. Proc Natl Acad Sci U S A. 2009;106:15956–60. doi: 10.1073/pnas.0908108106.
    1. Wang L, Zhou GB, Liu P, Song JH, Liang Y, Yan XJ, et al. Dissection of mechanisms of Chinese medicinal formula Realgar-Indigo naturalis as an effective treatment for promyelocytic leukemia. Proc Natl Acad Sci U S A. 2008;105:4826–31. doi: 10.1073/pnas.0712365105.
    1. Mendel DB, Crabtree GR. HNF-1, a member of a novel class of dimerizing homeodomain proteins. J Biol Chem. 1991;266:677–80.
    1. Costa RH, Kalinichenko VV, Holterman AX, Wang X. Transcription factors in liver development, differentiation, and regeneration. Hepatology. 2003;38:1331–47. doi: 10.1016/j.hep.2003.09.034.
    1. Kikuchi R, Yagi S, Kusuhara H, Imai S, Sugiyama Y, Shiota K. Genome-wide analysis of epigenetic signatures for kidney-specific transporters. Kidney Int. 2010;78:569–77. doi: 10.1038/ki.2010.176.
    1. Pastoret A, Marcos R, Sampayo-Reyes A, Saucedo-Cardenas O, Lozano-Garza GH, Hernandez A. Inhibition of hepatocyte nuclear factor 1 and 4 alpha (HNF1alpha and HNF4alpha) as a mechanism of arsenic carcinogenesis. Arch Toxicol. 2013;87:1001–12. doi: 10.1007/s00204-012-0948-6.

Source: PubMed

3
Předplatit