DOI QR코드

DOI QR Code

Trichostatin A-induced Apoptosis is Mediated by Krüppel-like Factor 4 in Ovarian and Lung Cancer

  • Zohre, Sadeghi (Hematology and Oncology Research Center) ;
  • Kazem, Nejati-Koshki (Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences) ;
  • Abolfazl, Akbarzadeh (Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences) ;
  • Mohammad, Rahmati-Yamchi (Hematology and Oncology Research Center) ;
  • Aliakbar, Movassaghpour (Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences) ;
  • Effat, Alizadeh (Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences) ;
  • Zahra, Davoudi (Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences) ;
  • Hassan, Dariushnejad (Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences) ;
  • Nosratollah, Zarghami (Hematology and Oncology Research Center)
  • 발행 : 2014.08.30

초록

Background: The istone deacetylase (HDAC) inhibitor trichostatin A (TSA) is known to mediate the regulation of gene expression and antiproliferation activity in cancer cells. Kr$\ddot{u}$ppel-like factor 4 (klf4) is a zinc finger-containing transcription factor of the SP/KLF family, that is expressed in a variety of tissues and regulates cell proliferation, differentiation, tumorigenesis, and apoptosis. It may either either function as a tumor suppressor or an oncogene depending on genetic context of tumors. Aims: In this study, we tested the possibility that TSA may increase klf4 expression and cancer cell growth inhibition and apoptosis in SKOV-3 and A549 cells. Materials and Methods: The cytotoxicity of TSA was determined using the MTT assay test, while klf4 gene expression was assessed by real time PCR andto ability of TSA to induce apoptosis using a Vybrant Apoptosis Assay kit. Results: Our results showed that TSA exerted dose and time dependent cytotoxicity effect on SKOV-3 and A549 cells. Moreover TSA up-regulated klf4 expression. Flow cytometric analysis demonstrated that apoptosis was increased after TSA treatment. Conclusions: Taken together, this study showed that TSA increased klf4 expression in SKOV3 and A549 cell lines, consequently, klf4 may played a tumor-suppressor role by increasing both cell growth inhibition and apoptosis. This study sheds light on the details of molecular mechanisms of HDACI-induced cell cycle arrest and apoptosis.

키워드

참고문헌

  1. Ai W, Liu Y, Langlois M, Wang TC (2004). Kruppel-like factor 4 (KLF4) represses histidine decarboxylase gene expression through an upstream Sp1 site and downstream gastrin responsive elements. J Biol Chem, 279, 8684-93. https://doi.org/10.1074/jbc.M308278200
  2. Akbarzadeh A, Ghasemali S, Nejati K, et al (2013). Study of inhibitory effect of $\beta$-cyclodextrin-helenalin complex on HTERT gene expression in T47D breast cancer cell line by real time quantitative PCR (q-PCR). Asian Pac J Cancer Prev, 14, 6949-53. https://doi.org/10.7314/APJCP.2013.14.11.6949
  3. Akbarzadeh A, Hosseininasab S, Davaran S, et al (2014). Synthesis, characterization, and In vitro studies of PLGAPEG nanoparticles for oral Insulin delivery. Chem Biol Drug Des, 3, 1-9.
  4. Akbarzadeh A, Mikaeili H, Zarghami N, et al (2012). Preparation and in-vitro evaluation of doxorubicin-loaded Fe3O4 magnetic nanoparticles modified with biocompatible copolymer. Int J Nanomedicine, 7, 1-16. https://doi.org/10.2217/nnm.11.171
  5. Akbarzadeh A, Nejati-Koshki K, Mahmoudi Soghrati M, et al (2013). In vitro studies of NIPAAM-MAA-VP copolymercoated magnetic nanoparticles for controlled anticancer drug release. JEAS, 3, 108-15. https://doi.org/10.4236/jeas.2013.34013
  6. Akbarzadeh A, Omidfar K, Ahmadin A, et al (2014). An electrochemical immunosensor for digoxin using core-shell gold coated magnetic nanoparticles as labels. Mol Biol Rep, 41, 1659-68. https://doi.org/10.1007/s11033-013-3014-4
  7. Akbarzadeh A, Pourhassan-Moghaddam M, Rahmati-Yamchi M, et al (2013). Protein detection through different platforms of immuno-loop-mediated isothermal amplification. Nanoscale Res Lett, 8, 485-95. https://doi.org/10.1186/1556-276X-8-485
  8. Akbarzadeh A, Rezaei A, Nejati-Koshki K, et al (2014). Synthesis and physicochemical characterization of biodegradable star-shaped poly lactide-co-glycolide- $\beta$ -cyclodextrin copolymer nanoparticles containing albumin. J Adv Nanoparticles, 3, 1-9. https://doi.org/10.4236/anp.2014.31001
  9. Akbarzadeh A, Rezaei-Sadabady R, Zarghami N, et al (2013). Studies of the relationship between structure and antioxidant activity in interesting systems, including tyrosol, hydroxytyrosol derivatives indicated by quantum chemical calculations. Soft, 2, 13-8. https://doi.org/10.4236/soft.2013.23004
  10. Akbarzadeh A, Samiei M, Davaran S, et al (2012). Magnetic nanoparticles: preparation, physical properties, and applications in biomedicine. Nanoscale Res Lett, 7, 14-26. https://doi.org/10.1186/1556-276X-7-14
  11. Akbarzadeh A, Samiei M, Joo SW, et al (2012). Synthesis, characterization and in vitro studies of doxorubicin-loaded magnetic nanoparticles grafted to smart copolymers on A549 lung cancer cell line. J Nanobiotechnol, 10, 46-52. https://doi.org/10.1186/1477-3155-10-46
  12. Akbarzadeh A, Zarghami N, Mikaeili H, et al (2012). Synthesis, characterization and in vitro evaluation of novel polymercoated magnetic nanoparticles for controlled delivery of doxorubicin. Nanotechnol Sci Appl, 5, 13-25.
  13. Aksoy I, Giudice V, Delahaye E, et al (2014). Klf4 and Klf5 differentially inhibit mesoderm and endoderm differentiation in embryonic stem cells. Nat Commun, 5, 1-15.
  14. Bellarosa D, Bressan A, Bigioni M, et al (2012). SAHA/ Vorinostat induces the expression of the CD137 receptor/ ligand system and enhances apoptosis mediated by soluble CD137 receptor in a human breast cancer cell line. Int J Oncol, 41, 1486-94.
  15. Butler LM, Agus DB, Scher HI, et al (2000). Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res, 60, 165-70.
  16. Butler LM, Webb Y, Agus DB, et al (2001). Inhibition of transformed cell growth and induction of cellular differentiation by pyroxamide, an inhibitor of histone deacetylase. Clin Cancer Res, 7, 962-70.
  17. Chen X, Erika MW, Gao SY, Yang VW (2003). Transcriptional profiling of Kruppel-like factor 4 reveals a function in cell cycle regulation and epithelial differentiation. J Mol Biol, 326, 665-77. https://doi.org/10.1016/S0022-2836(02)01449-3
  18. Chen Z, Clark S, Birkeland M, et al (2002). Induction and superinduction of growth arrest and DNA damage gene 45 (GADD45) alpha and beta messenger RNAs by histone deacetylase inhibitors trichostatin A (TSA) and butyrate in SW620 human colon carcinoma cells. Cancer Lett, 188, 127-40. https://doi.org/10.1016/S0304-3835(02)00322-1
  19. Chen ZY, Shie J, Tseng C (2000). Up-regulation of gut-enriched kruppel-like factor by interferon-gamma in human colon carcinoma cells. FEBS Lett, 477, 67-72. https://doi.org/10.1016/S0014-5793(00)01764-6
  20. Chen ZY, Rex S, Tseng CC (2004). Kruppel-like factor 4 is transactivated by butyrate in colon cancer cells. J Nutr, 134, 792-8.
  21. Cheng DD, Yang QC, Zhang ZH, Yang CX, Liu YW (2012). Antitumor activity of histone deacetylase inhibitor trichostatin a in osteosarcoma cells. Asian Pac J Cancer Prev, 13, 1395-9. https://doi.org/10.7314/APJCP.2012.13.4.1395
  22. Choi BJ, Cho YG, Song JW, et al (2006). Altered expression of the KLF4 in colorectal cancers. Pathology Res Pract, 202, 585-9. https://doi.org/10.1016/j.prp.2006.05.001
  23. Choudhary C, Kumer C, Gnad F, et al (2009). Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science, 325, 834-40. https://doi.org/10.1126/science.1175371
  24. Cress WD, Seto E (2000). Histone Deacetylases, Transcriptional Control, and Cancer. J Cell Physiol, 184, 1-16. https://doi.org/10.1002/(SICI)1097-4652(200007)184:1<1::AID-JCP1>3.0.CO;2-7
  25. Ebrahimnezhad Z, Zarghami N, Keyhani M, et al (2013). Inhibition of hTERT gene expression by silibinin-loaded PLGA-PEG-Fe3O4 in T47D breast cancer cell line. Bioimpacts, 3, 67-74.
  26. Feng L, Pan M, Sun J, et al (2013). Histone deacetylase 3 inhibits expression of PUMA in gastric cancer cells. J Mol Med, 91, 49-58. https://doi.org/10.1007/s00109-012-0932-x
  27. Francisco R, Perez-Perarnau A, Cortes C, et al (2012). Histone deacetylase inhibition induces apoptosis and autophagy in human neuroblastoma cells. Cancer Lett, 1, 42-52.
  28. Ghasemali S, Nejati-Koshki K, Tafsiri E, et al (2013). Inhibitory effects of $\beta$-cyclodextrin-helenalin complexes on H-TERT gene expression in the T47D breast cancer cell line - results of real time quantitative PCR. Asian Pac J Cancer Prev, 14, 6949-53. https://doi.org/10.7314/APJCP.2013.14.11.6949
  29. Glaser KB, Staver MJ, Waring JF, et al (2003). Gene expression profiling of multiple histone deacetylase (HDAC) inhibitors: defining a common gene set produced by HDAC inhibition in T24 and MD acarcinoma cell lines. Mol Cancer Ther, 2, 151-63.
  30. Glaser KB (2007). HDAC inhibitor: clinical update and mechanism-based potential. Biochem Pharmacol, 74, 659-71. https://doi.org/10.1016/j.bcp.2007.04.007
  31. Herold C, Ganslmayer M, Ocker M, et al (2002). The histonedeacetylase inhibitor trichostatin a blocks proliferation and triggers apoptotic programs in hepatoma cells. J Hepatol, 36, 233-40.
  32. Horing E, Podlech O, Silkenstedt B, et al (2013). The histone deacetylase inhibitor trichostatin a promotes apoptosis and antitumor immunity in glioblastoma cells. Anticancer Res, 33, 1351-60.
  33. Liu T, Kulijaca S, Tee A, Marshall GM (2006). Histone deacetylase inhibitors: multifunctional anticancer agents. Cancer Treat Rev, 32, 157-65. https://doi.org/10.1016/j.ctrv.2005.12.006
  34. Marks PA, Richard A, Rifkind RA (2000). Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Nati Cancer Inst, 92, 1210-6. https://doi.org/10.1093/jnci/92.15.1210
  35. Mees S, Nemunai J, Senzer N (2009). Transcription factors: their potential as targets for anindividualized therapeutic approach to cancer. Cancer Gene Ther, 16, 103-12. https://doi.org/10.1038/cgt.2008.73
  36. Mollazade M, Nejati-Koshki K, Akbarzadeh A, et al (2013). PAMAM dendrimers augment inhibitory effects of curcumin on cancer cell proliferation: possible inhibition of telomerase. Asian Pac J Cancer Prev, 14, 6925-8. https://doi.org/10.7314/APJCP.2013.14.11.6925
  37. Nejati-Koshki K, Zarghami N, Pourhassan-Moghaddam M, et al (2012). Inhibition of leptin gene expression and secretion by silibinin: possible role of estrogen receptors. Cytotechnology, 64, 719-26. https://doi.org/10.1007/s10616-012-9452-3
  38. Nejati-Koshki K, Akbarzadeh A, Pourhasan-Moghaddam M, et al (2013). Inhibition of leptin and leptin receptor gene expression by silibinin-curcumin combination. Asian Pac J Cancer Prev, 14, 6595-9. https://doi.org/10.7314/APJCP.2013.14.11.6595
  39. Nasiri M, Zarghami N, Koshki KN, et al (2013). Curcumin and silibinin inhibit telomerase expression in T47D human breast cancer cells. Asian Pac J Cancer Prev,14, 3449-53. https://doi.org/10.7314/APJCP.2013.14.6.3449
  40. Ohnishi S, Ohnami S, Laub F, et al (2003). Downregulation and growth inhibitory effect of epithelial-type Kruppel-like transcription factor KLF4, but not KLF5, in bladder cancer. Biochem Biophys Res Commun, 308, 251-6. https://doi.org/10.1016/S0006-291X(03)01356-1
  41. Owen GI, Richer JK, Tung L, Takimoto G, Horwitz KB (1998). Progesterone regulates transcription of the p21(WAF1) cyclin- dependent kinase inhibitor gene through Sp1 and CBP/p300. J Biol Chem, 273, 10696-701. https://doi.org/10.1074/jbc.273.17.10696
  42. Peiffer L, Poll-Wolbeck SJ, Flamme H, et al (2014). Trichostatin A effectively induces apoptosis in chronic lymphocytic leukemia cells via inhibition of Wnt signaling and histone deacetylation. J Cancer Res Clin Oncol, [Epub ahead of print]
  43. Piacentini P, Conadelli M, Costanzo C, et al (2006). Trichostatin A enhances the response of chemotherapeutic agents in inhibiting pancreatic cancer cell proliferation. Virchows Arch, 448, 797-804. https://doi.org/10.1007/s00428-006-0173-x
  44. Platta CS, Greenblatt DY, Kunnimalaiyaan M, Chen H (2007). The HDAC inhibitor trichostatin a inhibits growth of small cell lung cancer cells. J Surg Res, 142, 219-26. https://doi.org/10.1016/j.jss.2006.12.555
  45. Pourhassan-Moghaddam M, Rahmati-Yamchi M, Akbarzadeh A, et al (2013). Protein detection through different platforms of immuno-loop-mediated isothermal amplification. Nanoscale Res Lett, 8, 485. https://doi.org/10.1186/1556-276X-8-485
  46. Rasheed WK, Johnstone RW, Prince HM (2007). Histone deacetylase inhibitors in cancer therapy. Expert Opin Investig Drugs, 16, 659-78. https://doi.org/10.1517/13543784.16.5.659
  47. Sadat Tabatabaei Mirakabad F, Nejati-Koshki K, Akbarzadeh A, et al (2014). PLGA-based nanoparticles as cancer drug delivery systems. Asian Pac J Cancer Prev, 15, 517-35. https://doi.org/10.7314/APJCP.2014.15.2.517
  48. Sawa H, Murakami H, Ohshima Y, et al (2001). Histone deacetylase inhibitors such as sodium butyrate and trichostatin A induce apoptosis through an increase of the bcl-2-related protein Bad. Brain Tumor Pathol, 18, 109-14. https://doi.org/10.1007/BF02479423
  49. Shan Z, Feng-Nian R, Jie G, Ting Z (2012). Effects of valproic acid on proliferation, apoptosis, angiogenesis and metastasis of ovarian cancer in vitro and in vivo. Asian Pac J Cancer Prev, 13, 3977-82. https://doi.org/10.7314/APJCP.2012.13.8.3977
  50. Shie JL, Chen ZY, Fu M, Pestell RG, Tseng CC (2000). Gut enriched kruppel-like factor represses cyclin D1 promoter activity through Sp1 motif. Nucleic Acids Res, 28, 2969-76. https://doi.org/10.1093/nar/28.15.2969
  51. Shields JM, Christy RJ, Yang VW (2009). Identification and characterization of a gene encoding a gut-enriched Kruppellike factor expressed during growth arrest. J Biol Chem, 271, 20009-17.
  52. Sonnemann J HI, Sigler M, Palani CD, et al (2008). Histone deacetylase inhibitors and aspirin interact synergistically to induce cell death in ovarian cancer cells. Oncol Rep, 20, 219-24.
  53. Van LC, Verdin E, Verdin E (1996). Expression of a small fraction of cellular genes is changed in response to histone hyperacetylation, Gene Expr, 5, 245-53.
  54. Vigushin DM, Ali S, Pace PE, et al (2001). Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo. Clin Cancer Res, 7, 971-76.
  55. Wang F, Qi Y, Li X, et al (2013). HDAC inhibitor trichostatin A suppresses esophageal squamous cell carcinoma metastasis through HADC2 reduced MMP-2/9. Clin Invest Med, 36, 87-94.
  56. Wang J, Place RF, Huang V, et al (2010). Prognostic value and function of KLF4 in prostate cancer: RNAa and vector mediated overexpression identify KLF4 as an inhibitor of tumor cell growth and migration. Cancer Res, 70, 10182-91. https://doi.org/10.1158/0008-5472.CAN-10-2414
  57. Wei D, Gong W, Kanai M, et al (2005). Drastic down-regulation of Kruppel-like factor 4 expression is critical in human gastric cancer development and progression. Cancer Res, 65, 2746-54. https://doi.org/10.1158/0008-5472.CAN-04-3619
  58. Wei D, Kanai M, Jia Z, Le X, Xie K (2008). Kruppel-like Factor 4 induces p27Kip1 expression in and suppresses the growth and metastasis of human pancreatic cancer cells. Cancer Res, 68, 4631-9. https://doi.org/10.1158/0008-5472.CAN-07-5953
  59. Weichert W, Roske A, Gekeler V, et al (2008). Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer, 98, 604-10. https://doi.org/10.1038/sj.bjc.6604199
  60. Wilson AJ, Holson E, Wagner F, et al (2011). The DNA damage mark pH2AX differentiates the cytotoxic effects of small molecule HDAC inhibitors in ovarian cancer cells. Cancer Biol Ther, 12, 484-93. https://doi.org/10.4161/cbt.12.6.15956
  61. Wu ZQ, Zhang R, Chao C, Zhang JF, Zhang YQ (2007). Histone deacetylase inhibitor trichostatin A induced caspaseindependent apoptosis in human gastric cancer cell. Chin Med J, 120, 2112-8.
  62. Yoon HS, Chen X, Yang VW (2003). Kruppel-like factor 4 mediates p53-dependent G1/S cell cycle arrest in response to DNA damage. J Biol Chem, 278, 2101-5. https://doi.org/10.1074/jbc.M211027200
  63. Yoon HS, Yang VW (2004). Requirement of Kruppel-like factor 4 in preventing entry into mitosis following DNA damage. J Biol Chem, 279, 5035-41.
  64. Yoon O, Roh J (2012). Downregulation of KLF4 and the Bcl-2/ Bax ratio in advanced epithelial ovarian cancer. Oncol Lett, 4, 1033-6.
  65. Yori JL, Seachrist DD, Johnson E, et al (2011). Kruppel-like factor 4 inhibits tumorigenic progression and metastasis in a mouse model of breast cancer. Neoplasia, 13, 601-10. https://doi.org/10.1593/neo.11260
  66. You BR, Park WH (2013). Trichostatin A induces apoptotic cell death of HeLa cells in a Bcl-2 and oxidative stress-dependent manner. Int J Oncol, 42, 359-66.
  67. Yu F, Li J, Chen H, et al (2011). Kruppel-like factor 4 (KLF4) is required for maintenance of breast cancer stem cells and for cell migration and invasion. Oncogene, 30, 2161-72. https://doi.org/10.1038/onc.2010.591
  68. Zhang QC, Jiang SJ, Zhang S, Ma XB (2012). Histone deacetylase inhibitor trichostatin A enhances anti-tumor effects of docetaxel or erlotinib in A549 Cell Line. Asian Pac J Cancer Prev, 13, 3471-6. https://doi.org/10.7314/APJCP.2012.13.7.3471
  69. Zhang S, Zhang QC, Jiang SJ, Chin SJ (2013). Effect of trichostatin A and paclitaxel on the proliferation and apoptosis of lung adenocarcinoma cells. Chin Med J, 126, 129-34.
  70. Zhang W, Geiman DE, Shields JM (2000). The gut enriched Kruppel-like factor (Kruppel-like factor 4) mediates the transactivating effect of p53 on the p21WAF1/Cip1 promoter. J Biol Chem, 275, 18391-8. https://doi.org/10.1074/jbc.C000062200

피인용 문헌

  1. Effect of inhibition proliferation in human lung adenocarcinoma A549 cells by cytokine-induced killer cells vol.6, pp.4, 2015, https://doi.org/10.1111/1759-7714.12205
  2. Upregulation of MiR-122 via Trichostatin A Treatments in Hepatocyte-like Cells Derived from Mesenchymal Stem Cells vol.87, pp.2, 2016, https://doi.org/10.1111/cbdd.12664
  3. Magnetic nanoparticles: Applications in gene delivery and gene therapy pp.2169-141X, 2015, https://doi.org/10.3109/21691401.2015.1014093
  4. Comparison, synthesis and evaluation of anticancer drug-loaded polymeric nanoparticles on breast cancer cell lines pp.2169-141X, 2015, https://doi.org/10.3109/21691401.2015.1008510
  5. Current methods for synthesis of magnetic nanoparticles vol.44, pp.2, 2016, https://doi.org/10.3109/21691401.2014.982802
  6. Study of the Cytotoxic and Bactericidal Effects of Sila-substituted Thioalkyne and Mercapto-thione Compounds based on 1,2,3-Triazole Scaffold vol.121, pp.5, 2017, https://doi.org/10.1111/bcpt.12822
  7. Development of Emu oil-loaded PCL/collagen bioactive nanofibers for proliferation and stemness preservation of human adipose-derived stem cells: possible application in regenerative medicine vol.43, pp.12, 2017, https://doi.org/10.1080/03639045.2017.1357731
  8. An overview on different strategies for the stemness maintenance of MSCs vol.45, pp.7, 2017, https://doi.org/10.1080/21691401.2016.1246452
  9. Trichostatin A Enhances the Apoptotic Potential of Palladium Nanoparticles in Human Cervical Cancer Cells vol.17, pp.8, 2016, https://doi.org/10.3390/ijms17081354
  10. MiR-326/Sp1/KLF3: A novel regulatory axis in lung cancer progression pp.09607722, 2018, https://doi.org/10.1111/cpr.12551
  11. FOXC1 silencing promotes A549 cell apoptosis through inhibiting the PI3K/AKT/hedgehog/Gli2 signaling pathway vol.8, pp.59, 2018, https://doi.org/10.1039/C8RA06041J