Effects of valproic acid and pioglitazone on cell cycle progression and proliferation of T-cell acute lymphoblastic leukemia Jurkat cells

Document Type : Original Article

Authors

1 Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran

2 Department of Molecular Medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran

3 Infectious Diseases Research Center and Laboratory Science Research Center, Golestan University of Medical Sciences, Gorgan, Iran

4 Joint, Bone, and Connective tissue Research Center (JBCRC), Golestan University of Medical Sciences, Gorgan, Iran

5 Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran

Abstract

Objective(s): T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignant tumor. Administration of chemical compounds influencing apoptosis and T cell development has been discussed as promising novel therapeutic strategies. Valproic acid (VPA) as a recently emerged anti-neoplastic histone deacetylase (HDAC) inhibitor and pioglitazone (PGZ) as a high-affinity peroxisome proliferator-activated receptor-gamma (PPARγ) agonist have been shown to induce apoptosis and cell cycle arrest in different studies. Here, we aimed to investigate the underlying molecular mechanisms involved in anti-proliferative effects of these compounds on human Jurkat cells.
Materials and Methods: Treated cells were evaluated for cell cycle progression and apoptosis using flowcytometry and MTT viability assay. Real-time RT-PCR was carried out to measure the alterations in key genes associated with cell death and cell cycle arrest.
Results: Our findings illustrated that both VPA and PGZ can inhibit Jurkat E6.1 cells in vitro after   24 hr; however, PGZ 400 μM presents the most anti-proliferative effect. Interestingly, treated cells have been arrested in G2/M with deregulated cell division cycle 25A (Cdc25A) phosphatase and cyclin-dependent kinase inhibitor 1B (CDKN1B or p27) expression. Expression of cyclin D1 gene was inhibited when DNA synthesis entry was declined. Cell cycle deregulation in PGZ and VPA-exposed cells generated an increase in the proportion of aneuploid cell population, which has not reported before.
Conclusion: These findings define that anti-proliferative effects of PGZ and VPA on Jurkat cell line are mediated by cell cycle deregulation. Thus, we suggest PGZ and VPA may relieve potential therapeutic application against apoptosis-resistant malignancies.

Keywords


1.Ge J, Liu Y, Li Q, Guo X, Gu L, Ma ZG, et al. Resveratrol induces apoptosis and autophagy in T-cell acute lymphoblastic leukemia cells by inhibiting Akt/mTOR and activating p38-MAPK. Biomed Environ Sci 2013; 26:902-911.
2. Pui CH, Carroll WL, Meshinchi S, Arceci RJ. Biology, risk stratification, and therapy of pediatric acute leukemias: an update. J Clin Oncol 2011; 29:551-565.
3.Gao M, Gao L, Tao Y, Hou J, Yang G, Wu X, et al. Proteasome inhibitor carfilzomib interacts synergistically with histone deacetylase inhibitor vorinostat in Jurkat T-leukemia cells. Acta Biochim Biophys Sin 2014; 46:484-491.
4. Conter V, Bartram CR, Valsecchi MG, Schrauder A, Panzer-Grümayer R, Möricke A, et al. Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood 2010; 115:3206-3214.
5. Belayachi L, Aceves-Luquero C, Merghoub N, Bakri Y, de Mattos SF, Amzazi S, et al. Retama monosperma n-hexane extract induces cell cycle arrest and extrinsic pathway-dependent apoptosis in Jurkat cells. BMC Complement Altern Med 2014; 14:38.
6. Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell 2012; 150:12-27.
7. Robert C, Rassool FV. HDAC inhibitors: roles of DNA damage and repair. Adv Cancer Res 2012; 116:87-129.
8. Bollino D, Balan I, Aurelian L. Valproic acid induces neuronal cell death through a novel calpain‐dependent necroptosis pathway. J Neurochem 2015; 133:174-186.
9. Lamparter C, Winn LM. Tissue-specific effects of valproic acid on DNA repair genes and apoptosis in postimplantation mouse embryos. Toxicol Sci 2014; 141:59-67.
10. Mu Q, Ma Q, Lu S, Zhang T, Yu M, Huang X, et al. 10058-F4, a c-Myc inhibitor, markedly increases valproic acid-induced cell death in Jurkat and CCRF-CEM T-lymphoblastic leukemia cells. Oncol Lett 2014; 8:1355-1359.
11. Chateauvieux S, Morceau F, Dicato M, Diederich M. Molecular and therapeutic potential and toxicity of valproic acid. Bio Med Res Int 2010; 2010 . pii: 479364.
12.Wagner JM, Hackanson B, Lübbert M, Jung M. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin Epigenetics 2010; 1:117-136.
13. Miao M, Du B, Hu R, Yang Y, Yang W, Liao AJ, et al. [Effect of valproic acid sodium on proliferation and apoptosis of acute T-lymphoblastic leukemia Jurkat cells]. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2013; 21:343-346.
14. Yoon JY, Ishdorj G, Graham BA, Johnston JB, Gibson SB. Valproic acid enhances fludarabine-induced apoptosis mediated by ROS and involving decreased AKT and ATM activation in B-cell-lymphoid neoplastic cells. Apoptosis 2014; 19:191-200.
15. Rosenson RS, Wright RS, Farkouh M, Plutzky J. Modulating peroxisome proliferator–activated receptors for therapeutic benefit? Biology, clinical experience, and future prospects. Am Heart J 2012; 164:672-680.
16. da Rocha Junior LF, Dantas AT, Duarte ÂL, de Melo Rego MJ, Pitta Ida R, Pitta MG. PPAR agonists in adaptive immunity: what do immune disorders and their models have to tell us?. PPAR res. 2013;2013: 519724.
17. Cavender MA, Lincoff AM. Therapeutic potential of aleglitazar, a new dual PPAR-α/γ agonist. Am J Cardiovasc Drugs 2010; 10:209-216.
18. Tennis MA, Van Scoyk MM, Freeman SV, Vandervest KM, Nemenoff RA, Winn RA. Sprouty-4 inhibits transformed cell growth, migration and invasion, and epithelial-mesenchymal transition, and is regulated by Wnt7A through PPARγ in non–small cell lung cancer. Mol Cancer Res 2010; 8:833-843.
19. Rashid-Kolvear F, Taboski MA, Nguyen J, Wang DY, Harrington LA, Done SJ. Troglitazone suppresses telomerase activity independently of PPARγ in estrogen-receptor negative breast cancer cells. BMC Cancer 2010; 10:390.
20. Barros AL, Aguiar JS, Araújo LC, Peixoto CA, de Medeiros PL, Catanho MTJ, et al. Synergistic anticancer effects of valproic acid, atorvastatin and pioglitazone in human malignant and murine cells. Afr J Pharm Pharmacol 2014; 8:31-39.
21. Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications–a review. Nutr J 2014; 13:1.
22. Ninomiya I, Yamazaki K, Oyama K, Hayashi H, Tajima H, Kitagawa H, et al. Pioglitazone inhibits the proliferation and metastasis of human pancreatic cancer cells. Oncol Lett 2014; 8:2709-2714.
23. Youssef J, Badr M. Peroxisome proliferator‐activated receptors and cancer: challenges and opportunities. Br J Pharmacol 2011; 164:68-82.
24.Wu CH, Cheng SM, Chu KM, Yang SP, Lai JH, Cheng CC. Pioglitazone, a PPAR-γ agonist, downregulates cytokine production and AP-1 DNA-binding protein in human peripheral blood T cells–potential implication in the treatment of atherosclerotic disease. Acta Cardiol Sin 2009; 25:127-133.
25. Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 2013; 140:3079-3093.
26.Borghese L, Dolezalova D, Opitz T, Haupt S, Leinhaas A, Steinfarz B, et al. Inhibition of notch signaling in human embryonic stem cell–derived neural stem cells delays G1/S phase transition and accelerates neuronal differentiation in vitro and in vivo. Stem cells 2010; 28:955-964.
27.Neganova I, Vilella F, Atkinson SP, Lloret M, Passos JF, von Zglinicki T, et al. An important role for CDK2 in G1 to S checkpoint activation and DNA damage response in human embryonic stem cells. Stem Cells 2011; 29:651-659.
28.Dietrich S, Hullein J, Lee SC, Hutter B, Gonzalez D, Jayne S, et al. Recurrent CDKN1B (p27) mutations in hairy cell leukemia. Blood 2015; 126:1005-1008.
29.Bhattacharya K, Samanta SK, Tripathi R, Mallick A, Chandra S, Pal BC, et al. Apoptotic effects of mahanine on human leukemic cells are mediated through crosstalk between Apo-1/Fas signaling and the Bid protein and via mitochondrial pathways. Biochem Pharm 2010; 79:361-372.
30. Yagi Y, Fushida S, Harada S, Kinoshita J, Makino I, Oyama K, et al. Effects of valproic acid on the cell cycle and apoptosis through acetylation of histone and tubulin in a scirrhous gastric cancer cell line. J Exp Clin Cancer Res 2010; 29:1.
31. Suzuki T, Higgins P, Crawford D. Control selection for RNA quantitation. Biotechniques 2000; 29:332-337.
32. van Meerloo J, Kaspers GJ, Cloos J. Cell sensitivity assays: the MTT assay. Methods Mol Biol 2011; 731:237-245.
33.Yazdani Y, Sadeghi H, Alimohammadian M, Andalib A, Moazen F, Rezaei A. Iran J Pharm Res 2011; 10:559-568.
34. Yazdani Y, Keyhanvar N, Kalhor HR, Rezaei A. Functional analyses of recombinant mouse hepcidin-1 in cell culture and animal model. Biotechnol Lett 2013; 35:1191-1197.
35. Blaheta RA, Nau H, Michaelis M. Valproate and valproate-analogues: potent tools to fight against cancer. Curr Med Chem 2002; 9:1417-1433.
36. Chateauvieux S, Morceau F, Dicato M, Diederich M. Molecular and therapeutic potential and toxicity of valproic acid. J Biomed Biotechnol 2010; 2010:479364.
37.Tan J, Cang S, Ma Y, Petrillo RL, Liu D. Novel histone deacetylase inhibitors in clinical trials as anti-cancer agents. J Hematol Oncol 2010; 3:212-225.
38. Quintas-Cardama A, Santos F, Garcia-Manero G. Histone deacetylase inhibitors for the treatment of myelodysplastic syndrome and acute myeloid leukemia. Leukemia 2011; 25:226-235.
39. Cervera E, Candelaria M, López-Navarro O, Labardini J, Gonzalez-Fierro A, Taja-Chayeb L, et al. Epigenetic therapy with hydralazine and magnesium valproate reverses imatinib resistance in patients with chronic myeloid leukemia. Clin Lymphoma Myeloma Leuk 2012; 12:207-212.
40. Mithraprabhu S, Grigoriadis G, Khong T, Spencer A. Deactylase inhibition in myeloproliferative neoplasms. Investigational new drugs. 2010; 28:50-57.
41.Van Vlierberghe P, Ferrando A. The molecular basis of T cell acute lymphoblastic leukemia. J Clin Invest 2012; 122:3398-3406.
42. Ververis K, Hiong A, Karagiannis TC, Licciardi PV. Histone deacetylase inhibitors (HDACIs): multitargeted anticancer agents. Biologics 2013; 7:47-60.
43. Robert T, Vanoli F, Chiolo I, Shubassi G, Bernstein KA, Rothstein R, et al. HDACs link the DNA damage response, processing of double-strand breaks and autophagy. Nature 2011; 471:74-79.
44. Saiki M, Hatta Y, Yamazaki T, Itoh T, Enomoto Y, Takeuchi J, et al. Pioglitazone inhibits the growth of human leukemia cell lines and primary leukemia cells while sparing normal hematopoietic stem cells. Int J Oncol 2006; 29:437-443.
45. Burton JD, Reichel H, Castillo ME, Blumenthal RD. Growth inhibition of hematologic and epithelial cancer cell lines by both PPARγ antagonist and agonist drugs. Cancer Res 2005; 65:156-156.
46. Sikka S, Chen L, Sethi G, Kumar AP. Targeting PPARγ signaling cascade for the prevention and treatment of prostate cancer. PPAR Res 2012; 2012: 968040.
47. Wu CW, Farrell GC, Yu J. Functional role of peroxisome‐proliferator‐activated receptor γ in hepatocellular carcinoma. J Gastroenterol Hepatol 2012; 27:1665-1669.
48. Koga H, Selvendiran K, Sivakumar R, Yoshida T, Torimura T, Ueno T, et al. PPARγ potentiates anticancer effects of gemcitabine on human pancreatic cancer cells. Int J Oncol 2012; 40:679-685.
49. Koziel R, Szczepanowska J, Magalska A, Piwocka K, Duszynski J, Zablocki K. Ciprofloxacin inhibits proliferation and promotes generation of aneuploidy in Jurkat cells. J Physiol Pharmacol 2010; 61:233.
50. Alzoubi K, Khabour O, Hussain N, Al-Azzam S, Mhaidat N. Evaluation of vitamin B12 effects on DNA damage induced by pioglitazone. Mutat Res Genet Toxicol Environ Mutagenesis 2012; 748:48-51.