DOI QR코드

DOI QR Code

Effect of p16 on glucocorticoid response in a B-cell lymphoblast cell line

  • Kim, Sun-Young (Department of Pediatrics, College of Medicine, Hanyang University) ;
  • Lee, Kyung-Yil (Department of Pediatrics, College of Medicine, The Catholic University of Korea) ;
  • Jeong, Dae-Chul (Department of Pediatrics, College of Medicine, The Catholic University of Korea) ;
  • Kim, Hak-Ki (Department of Pediatrics, College of Medicine, The Catholic University of Korea)
  • 투고 : 2010.02.02
  • 심사 : 2010.05.18
  • 발행 : 2010.07.15

초록

Purpose: It has been suggested that p16 has a role in glucocorticoid (GC)-related apoptosis in leukemic cells, but the exact mechanisms have yet to be clarified. We evaluated the relationship between the GC response and p16 expression in a lymphoma cell line. Methods: We used p16 siRNA transfection to construct p16-inactivated cells by using the B-cell lymphoblast cell line NC-37. We compared glucocorticoid receptor (GR) expression, apoptosis, and cell viability between control (p16+NC-37) and p16 siRNA-transfected (p16-NC-37) cells after a single dose of dexamethasone (DX). Results: In both groups, there was a significant increase in cytoplasmic GR expression, which tended to be higher for p16+NC-37 cells than for p16- NC37 cells at all times, and the difference at 18 h was significant (P<0.05). Similar patterns of early apoptosis were observed in both groups, and late apoptosis occurred at higher levels at 18 h when the GR had already been downregulated ($P$<0.05). Cell viability decreased in both groups but the degree of reduction was more severe in p16+NC-37 cells after 18 h ($P$<0.05). Conclusion: These results suggest a relationship between GR expression and cell cycle inhibition, in which the absence of p16 leads to reduced cell sensitivity to DX.

키워드

참고문헌

  1. Biondi A, Valsecchi MG, Seriu T, D'Aniello E, Willemse MJ, Fasching K, et al. Molecular detection of minimal residual disease is a strong predictive factor of relapse in childhood B-lineage acute lymphoblastic leukemia with medium risk features. A case control study of the International BFM Study Group. Leukemia 2000; 4:1939-43.
  2. Myoumoto A, Nakatani K, Koshimizu TA, Matsubara H, Adachi S, Tsujimoto G. Glucocorticoid-induced granzyme A expression can be used as a marker of glucocorticoid sensitivity for acute lymphoblastic leukemia therapy. J Hum Genet 2007;52:328-33. https://doi.org/10.1007/s10038-007-0119-4
  3. Tutor O, Diaz MA, Ramirez M, Algara P, Madero L, Martinez P. Loss of heterozygosity of p16 correlates with minimal residual disease at the end of the induction therapy in non-high risk childhood B-cell precursor acute lymphoblastic leukemia. Leuk Res 2002;26:817-20. https://doi.org/10.1016/S0145-2126(02)00020-6
  4. Frankfurt O, Rosen ST. Mechanisms of glucocorticoid-induced apoptosis in hematologic malignancies: updates. Curr Opin Oncol 2004;16:553-63. https://doi.org/10.1097/01.cco.0000142072.22226.09
  5. Renner K, Ausserlechner MJ, R. Kofler R. A conceptual view on glucocorticoid-induced apoptosis, cell cycle arrest and glucocorticoid resistance in lymphoblastic leukemia. Curr Mol Med 2003;3:707-17. https://doi.org/10.2174/1566524033479357
  6. Jenkins BD, Pullen CB, Darimont BD. Novel glucocorticoid receptor coactivator effector mechanisms. Trends Endocrinol Metab 2001;12:22-6. https://doi.org/10.1016/S1043-2760(00)00336-2
  7. Bladh LG, Liden J, Dahlman-Wright K, Reimers M, Nilsson S, Okret S. Identification of endogenous glucocorticoid repressed genes differentially regulated by a glucocorticoid receptor mutant able to separate between nuclear factor-kappaB and activator protein-1 repression. Mol Pharmacol 2005;67:815-26.
  8. Guo SX, Taki T, Ohnishi H, Piao HY, Tabuchi K, Bessho F, et al. Hypermethylation of p16 and p15 genes and RB protein expression in acute leukemia. Leuk Res 2000;24:39-46. https://doi.org/10.1016/S0145-2126(99)00158-7
  9. Chim CS, Wong AS, Kwong YL. Epigenetic inactivation of the CIP/KIP cell-cycle control pathway in acute leukemias. Am J Hematol 2005;80:282-7. https://doi.org/10.1002/ajh.20503
  10. Chim CS, Wong KY, Loong F, Lam WW, Srivastava G. Frequent epigenetic inactivation of Rb1 in addition to p15 and p16 in mantle cell and follicular lymphoma. Hum Pathol 2007;38:1849-57. https://doi.org/10.1016/j.humpath.2007.05.009
  11. Drexler HG. Review of alterations of the cyclin-dependent kinase inhibitor INK4 family genes p15, p16, p18 and p19 in human leukemia-lymphoma cells. Leukemia 1998;12:845-59. https://doi.org/10.1038/sj.leu.2401043
  12. Suga Y, Miyajima K, Oikawa T, Maeda J, Usuda J, Kajiwara N, et al. Quantitative p16 and ESR1 methylation in the peripheral blood of patients with non-small cell lung cancer. Oncol Rep 2008;20:1137-42.
  13. de Snoo FA, Bishop DT, Bergman W, van Leeuwen I, van der Drift C, van Nieuwpoort FA, et al. Increased risk of cancer other than melanoma in CDKN2A founder mutation (p16-Leiden)-positive melanoma families. Clin Cancer Res 2008;14:7151-7. https://doi.org/10.1158/1078-0432.CCR-08-0403
  14. Ausserlechner MJ, Obexer P, Wiegers GJ, Hartmann BL, Geley S, Kofler R. The cell cycle inhibitor p16 (INK4A) sensitizes lymphoblastic leukemia cells to apoptosis by physiologic glucocorticoid levels. J Biol Chem 2001;276:10984-9. https://doi.org/10.1074/jbc.M008188200
  15. Holleman A, den Boer ML, Kazemier KM, Janka-Schaub GE, Pieters R. Resistance to different classes of drugs is associated with impaired apoptosis in childhood acute lymphoblastic leukemia. Blood 2003;102:4541-6. https://doi.org/10.1182/blood-2002-11-3612
  16. Ogawa S, Hirano N, Sato N, Takahashi T, Hangaishi A, Tanaka K, et al. Homozygous loss of the cyclin-dependent kinase 4-inhibitor (p16) gene in human leukemias. Blood 1994;84:2431-5.
  17. Cayuela JM, Madani A, Sanhes L, Stern MH, Sigaux F. Multiple tumor-suppressor gene 1 inactivation is the most frequent genetic alteration in T-cell acute lymphoblastic leukemia. Blood 1996;87:2180-6.
  18. Ohnishi H, Kawamura M, Ida K, Sheng XM, Hanada R, Nobori T, et al. Homozygous deletions of p16/MTS1 gene are frequent but mutations are infrequent in childhood T-cell acute lymphoblastic leukemia. Blood 1995;86:1269-75.
  19. Gombart AF, Morosetti R, Miller CW, Said JW, Koeffler HP. Deletions of the cyclin-dependent kinase inhibitor genes p16INK4A and p15INK4B in non-Hodgkin's lymphomas. Blood 1995;86:1534-9.
  20. Stranks G, Height SE, Mitchell P, Jadayel D, Yuille MA, De Lord C, et al. Deletions and rearrangement of CDKN2 in lymphoid malignancy. Blood 1995;85:893-901.
  21. Uchida T, Watanabe T, Kinoshita T, Murate T, Saito H, Hotta T. Mutational analysis of the CDKN2 (MTS1/p16ink4A) gene in primary B-cell lymphomas. Blood 1995;86:2724-31.
  22. Koduru PR, Zariwala M, Soni M, Gong JZ, Xiong Y, Broome JD. Deletion of cyclin-dependent kinase 4 inhibitor genes P15 and P16 in non-Hodgkin's lymphoma. Blood 1995;86:2900-5.
  23. Dreyling MH, Bohlander SK, Le Beau MM, Olopade OI. Refined mapping of genomic rearrangements involving the short arm of chromosome 9 in acute lymphoblastic leukemias and other hematologic malignancies. Blood 1995;86:1931-8.
  24. Serrano M, Lee H, Chin L, Cordon-Cardo C, Beach D, DePinho RA. Role of the INK4a locus in tumor suppression and cell mortality. Cell 1996;85:27-37. https://doi.org/10.1016/S0092-8674(00)81079-X
  25. Okret S, Poellinger L, Dong Y, Gustafsson JA. Down-regulation of glucocorticoid receptor mRNA by glucocorticoid hormones and recognition by the receptor of a specific binding sequence within a receptor cDNA clone. Proc Natl Acad Sci USA 1986;83:5899-903. https://doi.org/10.1073/pnas.83.16.5899
  26. Rosewicz S, McDonald AR, Maddux BA, Goldfine ID, Miesfeld RL, Logsdon CD. Mechanism of glucocorticoid receptor down-regulation by glucocorticoids. J Biol Chem 1988;263:2581-4.
  27. Hoeck W, Rusconi S, Groner B. Down-regulation and phosphorylation of glucocorticoid receptors in cultured cells. Investigations with a monospecific antiserum against a bacterially expressed receptor fragment. J Biol Chem 1989;264:14396-402.