Skip to main content

Advertisement

Log in

Antitumor activity of SAHA, a novel histone deacetylase inhibitor, against murine B cell lymphoma A20 cells in vitro and in vivo

  • Research Article
  • Published:
Tumor Biology

Abstract

Suberoylanilide hydroxamic acid (SAHA; vorinostat), the second generation of histone deacetylase (HDAC) inhibitor, has been approved for the treatment of cutaneous manifestations of cutaneous T cell lymphoma (CTCL). It has also shown its anticancer activity over a large range of other hematological and solid malignancies, but few studies have been reported in B cell lymphoma. In this study, we aimed to investigate the antitumor activity of SAHA on murine B cell lymphoma cell line A20 cells. We treated A20 cells with different concentrations of SAHA. The effect of SAHA on the proliferation of A20 cells was studied by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium (MTT) assay in vitro; the anti-proliferation activity in vivo was evaluated by proliferating cell nuclear antigen (PCNA) of xenograft tumor tissues through immunocytochemical staining. Apoptosis were detected by Hoechst 33258 staining and Annexin V/propidium iodide (PI) double-labeled cytometry in vitro. The effect of SAHA on cell cycle of A20 cells was studied by a propidium iodide method. Autophagic cell death induced by SAHA was confirmed by transmission electron microscopy (TEM). Angiogenesis marker (CD31) was measured by immunocytochemical staining to investigate the anti-angiogenic effect of SAHA. Western blot was used to detect the expression of signaling pathway factors (phospho-AKT, phospho-ERK, AKT, ERK, Nur77, HIF-1α, and VEGF). Our results showed that SAHA inhibited the proliferation of A20 cells in a time- and dose-dependent manner, induced cell apoptosis and G0/G1 phase arrest of cell cycle, promoted autophagic cell death, and suppressed tumor progress in NCI-A20 cells nude mice xenograft model in vivo. SAHA decreased the activation of AKT (phospho-AKT: p-AKT) and ERK1/2 (phospho-ERK: p-ERK) proteins and inhibited the expression of pro-angiogenic factors (VEGF and HIF-1α), downregulated its downstream signaling factor (Nur77), which might be contributed to the antitumor mechanisms of SAHA.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Li Q, Burgess R, Zhang Z. All roads lead to chromatin: multiple pathways for histone deposition. Biochim Biophys Acta. 2012;1819:238–46.

    Article  CAS  PubMed  Google Scholar 

  2. Groth A, Rocha W, Verreault A, Almouzni G. Chromatin challenges during DNA replication and repair. Cell. 2007;128:721–33.

    Article  CAS  PubMed  Google Scholar 

  3. Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705.

    Article  CAS  PubMed  Google Scholar 

  4. Nguyen HN, Kim JH, Jeong CY, Hong SW, Lee H. Inhibition of histone deacetylation alters arabidopsis root growth in response to auxin via pin1 degradation. Plant Cell Rep. 2013;32:1625–36.

    Article  CAS  PubMed  Google Scholar 

  5. Ververis K, Hiong A, Karagiannis TC, Licciardi PV. Histone deacetylase inhibitors (hdacis): multitargeted anticancer agents. Biologics. 2013;7:47–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Chen WL, Sheu JR, Hsiao CJ, Hsiao SH, Chung CL, Hsiao G. Histone deacetylase inhibitor impairs plasminogen activator inhibitor-1 expression via inhibiting tnf-alpha-activated mapk/ap-1 signaling cascade. Biomed Res Int. 2014;2014:231012.

    PubMed  PubMed Central  Google Scholar 

  7. Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA approval summary: vorinostat for treatment of advanced primary cutaneous t-cell lymphoma. Oncologist. 2007;12:1247–52.

    Article  CAS  PubMed  Google Scholar 

  8. Garcia-Manero G, Yang H, Bueso-Ramos C, Ferrajoli A, Cortes J, Wierda WG, et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood. 2008;111:1060–6.

    Article  CAS  PubMed  Google Scholar 

  9. Mazumder A, Vesole DH, Jagannath S. Vorinostat plus bortezomib for the treatment of relapsed/refractory multiple myeloma: a case series illustrating utility in clinical practice. Clin Lymphoma Myeloma Leuk. 2010;10:149–51.

    Article  CAS  PubMed  Google Scholar 

  10. Karelia N, Desai D, Hengst JA, Amin S, Rudrabhatla SV, Yun J. Selenium-containing analogs of SAHA induce cytotoxicity in lung cancer cells. Bioorg Med Chem Lett. 2010;20:6816–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hurwitz JL, Stasik I, Kerr EM, Holohan C, Redmond KM, McLaughlin KM, et al. Vorinostat/saha-induced apoptosis in malignant mesothelioma is flip/caspase 8-dependent and hr23b-independent. Eur J Cancer. 2012;48:1096–107.

    Article  CAS  PubMed  Google Scholar 

  12. Siegel D, Hussein M, Belani C, Robert F, Galanis E, Richon VM, et al. Vorinostat in solid and hematologic malignancies. J Hematol Oncol. 2009;2:31.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Lee JH, Choy ML, Ngo L, Foster SS, Marks PA. Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc Natl Acad Sci U S A. 2010;107:14639–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Xu Y. Regulation of p53 responses by post-translational modifications. Cell Death Differ. 2003;10:400–3.

    Article  CAS  PubMed  Google Scholar 

  15. Zhang Y, Adachi M, Kawamura R, Imai K. Bmf is a possible mediator in histone deacetylase inhibitors fk228 and cbha-induced apoptosis. Cell Death Differ. 2006;13:129–40.

    Article  CAS  PubMed  Google Scholar 

  16. Zhao Y, Tan J, Zhuang L, Jiang X, Liu ET, Yu Q. Inhibitors of histone deacetylases target the rb-e2f1 pathway for apoptosis induction through activation of proapoptotic protein bim. Proc Natl Acad Sci U S A. 2005;102:16090–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Liang D, Kong X, Sang N. Effects of histone deacetylase inhibitors on hif-1. Cell Cycle. 2006;5:2430–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Deroanne CF, Bonjean K, Servotte S, Devy L, Colige A, Clausse N, et al. Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene. 2002;21:427–36.

    Article  CAS  PubMed  Google Scholar 

  19. Ungerstedt JS, Sowa Y, Xu WS, Shao Y, Dokmanovic M, Perez G, et al. Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc Natl Acad Sci U S A. 2005;102:673–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Marks PA. Discovery and development of SAHA as an anticancer agent. Oncogene. 2007;26:1351–6.

    Article  CAS  PubMed  Google Scholar 

  21. Lai JP, Sandhu DS, Shire AM, Roberts LR. The tumor suppressor function of human sulfatase 1 (sulf1) in carcinogenesis. J Gastrointest Cancer. 2008;39:149–58.

    Article  CAS  PubMed  Google Scholar 

  22. Ilieva H, Nagano I, Murakami T, Shiote M, Shoji M, Abe K. Sustained induction of survival p-akt and p-erk signals after transient hypoxia in mice spinal cord with g93a mutant human sod1 protein. J Neurol Sci. 2003;215:57–62.

    Article  CAS  PubMed  Google Scholar 

  23. Datta SR, Brunet A, Greenberg ME. Cellular survival: a play in three Akts. Genes Dev. 1999;13:2905–27.

    Article  CAS  PubMed  Google Scholar 

  24. Srinivasan R, Zabuawala T, Huang H, Zhang J, Gulati P, Fernandez S, et al. Erk1 and erk2 regulate endothelial cell proliferation and migration during mouse embryonic angiogenesis. PLoS One. 2009;4:e8283.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Huynh N, Liu KH, Baldwin GS, He H. P21-activated kinase 1 stimulates colon cancer cell growth and migration/invasion via erk- and akt-dependent pathways. Biochim Biophys Acta. 1803;2010:1106–13.

    Google Scholar 

  26. Chen CS, Weng SC, Tseng PH, Lin HP. Histone acetylation-independent effect of histone deacetylase inhibitors on akt through the reshuffling of protein phosphatase 1 complexes. J Biol Chem. 2005;280:38879–87.

    Article  CAS  PubMed  Google Scholar 

  27. Chen N, Karantza-Wadsworth V. Role and regulation of autophagy in cancer. Biochimica Biophys Acta (BBA) - Mol Cell Res. 2009;1793:1516–23.

    Article  CAS  Google Scholar 

  28. Chen N, Debnath J. Autophagy and tumorigenesis. FEBS Lett. 2010;584:1427–35.

    Article  CAS  PubMed  Google Scholar 

  29. Kobayashi S, Liang Q. Autophagy and mitophagy in diabetic cardiomyopathy. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2015;1852:252–61.

    Article  CAS  Google Scholar 

  30. Lee M-S. Role of islet β cell autophagy in the pathogenesis of diabetes. Trends Endocrinol Metab. 2014;25:620–7.

    Article  CAS  PubMed  Google Scholar 

  31. Vidal RL, Matus S, Bargsted L, Hetz C. Targeting autophagy in neurodegenerative diseases. Trends Pharmacol Sci. 2014;35:583–91.

    Article  CAS  PubMed  Google Scholar 

  32. Ni B-B, Li B, Yang Y-H, Chen J-W, Chen K, Jiang S-D, et al. The effect of transforming growth factor β1 on the crosstalk between autophagy and apoptosis in the annulus fibrosus cells under serum deprivation. Cytokine. 2014;70:87–96.

    Article  CAS  PubMed  Google Scholar 

  33. Zhou X, Münger K. Expression of the human papillomavirus type 16 e7 oncoprotein induces an autophagy-related process and sensitizes normal human keratinocytes to cell death in response to growth factor deprivation. Virology. 2009;385:192–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhang Q, Zen K. Chapter 21—hypoxia-induced autophagy promotes tumor cell survival. In: Hayat MA, editor. Autophagy: cancer, other pathologies, inflammation, immunity, infection, and aging. Amsterdam: Academic; 2014. p. 305–17.

    Chapter  Google Scholar 

  35. Liang N, Jia L, Liu Y, Liang B, Kong D, Yan M, et al. Atm pathway is essential for ionizing radiation-induced autophagy. Cell Signal. 2013;25:2530–9.

    Article  CAS  PubMed  Google Scholar 

  36. Gewirtz DA, Hilliker ML, Wilson EN. Promotion of autophagy as a mechanism for radiation sensitization of breast tumor cells. Radiother Oncol. 2009;92:323–8.

    Article  CAS  PubMed  Google Scholar 

  37. Hashimoto D, Bläuer M, Sand J, Hirota M, Laukkarinen J. The role of autophagy in pancreatic cancer cells (panc-1) treated with anticancer drugs and inhibitors of autophagy. Pancreatology. 2013;13:e32–3.

    Google Scholar 

  38. Shao Y, Gao Z, Marks PA, Jiang X. Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci U S A. 2004;101:18030–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Carew JS, Giles FJ, Nawrocki ST. Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett. 2008;269:7–17.

    Article  CAS  PubMed  Google Scholar 

  40. Richter-Landsberg C, Leyk J. Inclusion body formation, macroautophagy, and the role of hdac6 in neurodegeneration. Acta Neuropathol. 2013;126:793–807.

    Article  CAS  PubMed  Google Scholar 

  41. Oh M, Choi I-K, Kwon HJ. Inhibition of histone deacetylase1 induces autophagy. Biochem Biophys Res Commun. 2008;369:1179–83.

    Article  CAS  PubMed  Google Scholar 

  42. Li J, Liu R, Lei Y, Wang K, Lau QC, Xie N, et al. Proteomic analysis revealed association of aberrant ros signaling with suberoylanilide hydroxamic acid-induced autophagy in jurkat t-leukemia cells. Autophagy. 2010;6:711–24.

    Article  CAS  PubMed  Google Scholar 

  43. Deepak AV, Salimath BP. Antiangiogenic and proapoptotic activity of a novel glycoprotein from U. indica is mediated by nf-kappab and caspase activated DNase in ascites tumor model. Biochimie. 2006;88:297–307.

    Article  CAS  PubMed  Google Scholar 

  44. Marme D. Tumor angiogenesis: the pivotal role of vascular endothelial growth factor. World J Urol. 1996;14:166–74.

    Article  CAS  PubMed  Google Scholar 

  45. Sumpio BE, Yun S, Cordova AC, Haga M, Zhang J, Koh Y, et al. Mapks (erk1/2, p38) and akt can be phosphorylated by shear stress independently of platelet endothelial cell adhesion molecule-1 (cd31) in vascular endothelial cells. J Biol Chem. 2005;280:11185–91.

    Article  CAS  PubMed  Google Scholar 

  46. Khattab AZ, Ahmed MI, Fouad MA, Essa WA. Significance of p53 and cd31 in astrogliomas. Med Oncol. 2009;26:86–92.

    Article  CAS  PubMed  Google Scholar 

  47. Martinez-Gonzalez J, Badimon L. The nr4a subfamily of nuclear receptors: new early genes regulated by growth factors in vascular cells. Cardiovasc Res. 2005;65:609–18.

    Article  CAS  PubMed  Google Scholar 

  48. Ismail H, Mofarrahi M, Echavarria R, Harel S, Verdin E, Lim HW, et al. Angiopoietin-1 and vascular endothelial growth factor regulation of leukocyte adhesion to endothelial cells: role of nuclear receptor-77. Arterioscler Thromb Vasc Biol. 2012;32:1707–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bras A, Albar JP, Leonardo E, de Buitrago GG, Martinez AC. Ceramide-induced cell death is independent of the fas/fas ligand pathway and is prevented by nur77 overexpression in a20 b cells. Cell Death Differ. 2000;7:262–71.

    Article  CAS  PubMed  Google Scholar 

  50. Zeng H, Qin L, Zhao D, Tan X, Manseau EJ, Van Hoang M, et al. Orphan nuclear receptor tr3/nur77 regulates vegf-a-induced angiogenesis through its transcriptional activity. J Exp Med. 2006;203:719–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Deutsch AJ, Rinner B, Wenzl K, Pichler M, Troppan K, Steinbauer E, et al. Nr4a1-mediated apoptosis suppresses lymphomagenesis and is associated with a favorable cancer-specific survival in patients with aggressive b-cell lymphomas. Blood. 2014;123:2367–77.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

Thanks are expressed to the Laboratory of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, and for the laboratory of Electron Microscopy of Tongji Medical College for offering relevant experimental facilities and technical support.

This work was supported by the Science Foundation of Hubei Health Department (No. JX6B08), Wujieping Medical Foundation (No.320.6750.13277), and National Nature Science Foundation of China (No. 81472707).

Conflicts of interest

None

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hongli Liu.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, B., Yu, D., Liu, J. et al. Antitumor activity of SAHA, a novel histone deacetylase inhibitor, against murine B cell lymphoma A20 cells in vitro and in vivo. Tumor Biol. 36, 5051–5061 (2015). https://doi.org/10.1007/s13277-015-3156-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13277-015-3156-1

Keywords

Navigation