Effect of Valproic Acid on Proliferation and Apoptosis of Colon Cancer HT 29 Cell Line

Masumeh Sanaei, Fraidoon Kavoosi, Shekoufeh Atashpour

Abstract


To understand the scientific work below, you should also research what is Valproic Acid and for that go to https://qualitycustomessays.com and read only selected and quality facts and explanations. The structure of chromatin is made up of DNA, histones, and non-histone proteins with the basic repeating unit, nucleosome. The structure of chromatin can be reversibly modified in several ways including histone acetylation. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) are two groups of enzymes that involved in acetylation and regulate gene expression. Histone deacetylases have a key role in the remodeling of chromatin and the epigenetic regulation of gene expression. HDAC inhibitors can inhibit HDACs and affect gene expression. HDAC inhibitor valproic acid (VPA) has antitumor activities against certain cancers that make it an attractive drug for treating cancer. The aim of the present study was to analyze the effect of VPA on proliferation and apoptosisin of colon cancer HT 29 cell line. Materials and Methods: MTT assay and flow cytometry assay were used to evaluate proliferative and apoptotic effects of VPA. Results: VPA inhibited the growth of HT 29 cells and induced apoptosis significantly with a time- and dose-dependent manner. Discussion: Our finding clearly indicated that VPA has a significant inhibitory and apoptotic effects. Conclusion: VPA can significantly inhibit the growth and induce apoptosis in the HT 29 cell line.


Full Text:

PDF

References


Marks P, Rifkind RA, Richon VM, Breslow R, Miller T, Kelly WK. Histone deacetylases and cancer: Causes and therapies. Nat Rev Cancer. 2001; 1: 194–202.

Lehrmann H, Pritchard LL, Harel-Bellan A. Histone acetyltransferases and deacetylases in the control of cell proliferation and differentiation. Adv Cancer Res. 2002; 86: 41–65.

Allfrey VG, Faulkner R, Mirsky AE. Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc Natl Acad Sci USA. 1964; 51: 786–794.

Zhang Y, Reinberg D. Transcription regulation by histone methylation: Interplay between different covalent modifications of the core histone tails. Genes Dev. 2001; 15:2343–2360

Spotswood HT, Turner BM. An increasingly complex code. J Clin Invest. 2002; 110:577–582.

Fischle W, Wang Y, Allis CD. Binary switches and modification cassettes in histone biology and beyond. Nature. 2003; 425:475–479

Gray SG, Teh BT. Histone acetylation/deacetylation and cancer: an "open" and "shut" case? Curr Mol Med 2001; 1:401-29.

De Ruijter AJ, Van Gennip AH, Caron HN, et al. Histone deacetylases: characterisation of the classical HDAC family. Biochemical Journal. 2003; 15: 370 (3) 737-749

Gregory PD, Wagner K, Horz W. Histone acetylation and chromatin remodeling. Exp Cell Res 2001; 265:195-202.

Gray SG, Ekstrom TJ. The human histone deacetylase family. Exp Cell Res 2001; 262:75-83.

Jessica E. Bolden, Melissa J. Peart & Ricky W. Johnstone. Anticancer activities of histone deacetylase inhibitors. Nature Reviews Drug Discovery. 2006; 5: 769-784

Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 2006; 5:769 -84.

de Ruijter AJ, van Gennip AH, Caron HN, et al. Histone deacetylases (HDACs): characterization of the classical HDAC family. BiochemJ 2003; 370: 737-49.

Gao L, Cueto MA, Asselbergs F, Atadja P. Cloning and functional characterization of HDAC11, a novel member of the human histone deacetylase family. J Biol Chem 2002; 277: 25748 -55.

Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov 2006; 5:37-50.

Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 2006; 6:38-51

FinninMS, DonigianJR, Cohen A, et al. Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors. Nature 1999; 401:188-93.

NielsenTK, HildmannC, Dickmanns A, et al. Crystal structure of a bacterial class 2 histone deacetylase homologue. J Mol Biol 2005; 354:107-20.

Sternson SM, Wong JC, Grozinger CM, Schreiber SL. Synthesis of 7200 small molecules based on a substructural analysis of the histone deacetylase inhibitors trichostatin and trapoxin. Org Lett 2001; 3: 4239- 42.

Marks PA, Rifkind RA, Richon VM, Breslow R, Kelly WK. Histone deacetylases and cancer: causes and therapies. Nature Reviews Cancer 2001; 1:194-202

Wang C, Fu M, Mani S, Wadler S, Senderowicz AM, Pestell RG. Histone acetylation and the cell-cycle in cancer. Front Biosci 2001; 6: 610-29.

De Ruijter, A. J., van Gennip, A. H., Caron, H. N., Kemp, S., and van Kuilenburg, A. B. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem. J., 2003; 370: 737–749

Yoshida, M., Kijima, M., Akita, M., and Beppu, T. Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A. J. Biol. Chem., 1990; 265: 17174–17179

Wharton, W., Savell, J., Cress, W. D., et al. Inhibition of mitogenesis in Balb/c-3T3 cells by trichostatin A–multiple alterations in the induction and activation of cyclin-cyclin-dependent kinase complexes. J. Biol. Chem. 2000; 275: 33981–33987

Yoshida, M., Hoshikawa, Y., Koseki, K., Mori, K., and Beppu, T. Structural specificity for biological activity of trichostatin A, a specific inhibitor of mammalian cell cycle with potent differentiation-inducing activity in Friend leukemia cells. J. Antibiot. 1990; 43: 1101–1106

Vigushin, D. M., Ali, S., Pace, P. E., Mirsaidi, N., Ito, K., Adcock, I., and Coombes, R. C. Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo. Clin. Cancer Res. 2001; 7: 971–976 27. Yoshida, M., Horinouchi, S., and Beppu, T. Trichostatin A and trapoxin: novel chemical probes for the role of histone acetylation in chromatin structure and function. Bioessays. 1995; 17: 423–430

Kwon, H. J., Owa, T., Hassig, C. A., Shimada, J., and Schreiber, S. L. Depudecin induces morphological reversion of transformed fibroblast via the inhibition of histone deacetylase. Proc. Natl. Acad. Sci. USA. 1998; 95: 3356–3361

Marks, P. A., Richon, V. M., and Rifkind, R. A. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J. Natl. Cancer Inst. 2000; 92: 1210–1216

Zhou, Q., Melkoumian, Z. K., Lucktong, A., et al. Rapid induction of histone hyperacetylation and cellular differentiation in human breast tumor cell lines following deg- radation of histone deacetylase-1. J. Biol. Chem.2000; 27: 35256–35263

Kosugi, H., Towatari, M., Hatano, S., et al. Histone deacetylase inhibitors are the potent inducer/enhancer of differentiation in acute myeloid leukemia: a new approach to antileukemia therapy. Leukemia (Baltimore). 1999; 13: 1316–1324

Cinatl J, Jr., Kotchetkov R, Blaheta R, et al. Induction of differentiation and suppression of malignant phenotype of human neuroblastoma BE(2)-C cells by valproic acid: enhancement by combination with interferon-a. Int J Oncol 2002; 20:97–106.

Yuan PX, Huang LD, Jiang YM, et al. The mood stabilizer valproic acid activates mitogen-activated protein kinases and promotes neurite growth. J Biol Chem 2001; 276: 31674–83.

Bacon CL, Gallagher HC, Haughey JC, Regan CM. Antiproliferative action of valproate is associated with aberrant expression and nuclear translocation of cyclin D3 during the C6 glioma G1 phase. J Neurochem 2002; 83:12–9.

Kamitani H, Taniura S, Watanabe K, et al. Histone acetylation may suppress human glioma cell proliferation when p21 WAF/Cip1 and gelsolin are induced. Neuro-oncol 2002; 4:95–101.

Kawagoe R, Kawagoe H, Sano K. Valproic acid induces apoptosis in human leukemia cells by stimulating both caspase-dependent and -independent apoptotic signaling pathways. Leuk Res 2002; 26:495–502.

Fischkoff SA, Walter E, Jr. Induction of neutrophilic differentiation of human promyelocytic leukemic cells by branched-chain carboxylic acid anticonvulsant drugs. J Biol Response Modif 1984; 3:132–7.

Olsen CM, Meussen-Elholm ET, Roste LS, Tauboll E. Antiepileptic drugs inhibit cell growth in the human breast cancer cell line MCF7. Mol Cell Endocrinol 2004; 213:173–9.

Thelen P, Schweyer S, Hemmerlein B, et al. Expressional changes after histone deacetylase inhibition by valproic acid in LNCaP human prostate cancer cells. Int J Oncol 2004; 24:25–31.

Masatoshi Shoji, Itasu Ninomiya, Isamu Makino et al. Valproic acid, a histone deacetylase inhibitor, enhances radiosensitivity in esophageal squamous cell carcinoma. International journal of oncology. 2012; 40: 2140-2146

Marks PA, Miller T, Richon VM. Histone deacetylases. Curr Opin Pharmacol 2003; 3: 344–51.

Richon VM, O’Brien JP. Histone deacetylase inhibitors: a new class of potential therapeutic agents for cancer treatment. Clin Cancer Res 2002; 8:662–4.

Gottlicher M, Minucci S, Zhu P, et al. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J 2001; 20:6969–78.

Loscher W: Basic pharmacology of valproate: a review after 35 years of clinical use for the treatment of epilepsy. CNS Drugs 2002, 16:669-669.

Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS: Histone

deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem 2001, 276:36734-36741.

Kramer OH, Zhu P, Ostendorff HP, et al. The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2. EMBO J 2003,

:3411-3420.

Xiao-Nan Li, Qin Shu, Jack Men-Feng Su et al. Valproic acid induces growth arrest, apoptosis, and senescence in medulloblastomas by increasing histone hyperacetylation and regulating expression of p21Cip1, CDK4, and CMYC. Mol Cancer Ther 2005; 4(12):1912–22

David Yu Greenblatt, Max A. Cayo, Joel T. Adler, et al. Valproic Acid Activates Notch1 Signaling and Induces Apoptosis in Medullary Thyroid Cancer Cells.

Ann Surg. 2008; 247(6): 1036–1040.

Christopher S. Platta, B.S., David Yü Greenblatt, M.D., Muthusamy Kunnimalaiyaan. Valproic Acid Induces Notch1 Signaling in Small Cell Lung Cancer Cells. Journal of Surgical Research. 2008; 148 (1): 31–37

Yasumichi Yagi, Sachio Fushida, Shinichi Harada 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. Journal of Experimental & Clinical Cancer Research 2010, 29:149: 1-11

Stockhausen MT, Sjolund J, Manetopoulos C, et al. Effects of the histone deacetylase inhibitor valproic acid on Notch signalling in human neuroblastoma cells. Br J Cancer 2005; 92:751–759.

Marks PA, Richon VM, Miller T, Kelly WK. Histone deacetylase inhibitors. Adv Cancer Res. 2004; 91: 137–168.

Piekarz R, Bates S. A review of depsipeptide and other histone deacetylase inhibitors in clinical trials. Curr Pharm Des. 2004; 10:2289–2298.

Chien-Lun Chen, Jennifer Sung, Michael Cohen et al. Valproic Acid Inhibits Invasiveness in Bladder Cancer but Not in Prostate Cancer Cells. The journal of pharmacology and experimental therapeutics. 2006; 319 (2): 533-543

Alessandra Valentini1, Paolo Gravina, Giorgio Federici1 et al. Valproic Acid Induces Apoptosis, p16INK4A Upregulation and Sensitization to Chemotherapy in Human Melanoma Cells. Cancer Biology & Therapy. Cancer Biology & Therapy. 2016; 6:2, 185-191.


Refbacks

  • There are currently no refbacks.


Creative Commons License
This work is licensed under a Creative Commons Attribution 3.0 License.

Copyright ©2022 Academic Journals Center

To make sure that you can receive messages from us, please add the 'academicjournalscenter.org' domain to your e-mail 'safe list'. If you do not receive e-mail in your 'inbox', check your 'bulk mail' or 'junk mail' folders.