期刊论文详细信息
Clinical Epigenetics
Expression profiling of DNA methylation-mediated epigenetic gene-silencing factors in breast cancer
Samir Kumar Patra1  Madhumita Rakshit1  Nibedita Pradhan1  Sandip Kumar Rath1  Sabnam Parbin1  Arunima Shilpi1  Moonmoon Deb1  Dipta Sengupta1  Swayamsiddha Kar1 
[1] Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
关键词: Cancer;    Gene silencing;    Methyl-CpG-binding domain proteins;    DNA methyltransferases;    DNA methylation;    Epigenetics;   
Others  :  1092815
DOI  :  10.1186/1868-7083-6-20
 received in 2014-05-16, accepted in 2014-09-29,  发布年份 2014
PDF
【 摘 要 】

Background

DNA methylation mediates gene silencing primarily by inducing repressive chromatin architecture via a common theme of interaction involving methyl-CpG binding (MBD) proteins, histone modifying enzymes and chromatin remodelling complexes. Hence, targeted inhibition of MBD protein function is now considered a potential therapeutic alternative for thwarting DNA hypermethylation prompted neoplastic progress. We have analyzed the gene and protein expression level of the principal factors responsible for gene silencing, that is, DNMT and MBD proteins in MCF-7 and MDA-MB-231 breast cancer cell lines after treatment with various epigenetic drugs.

Results

Our study reveals that the epigenetic modulators affect the expression levels at both transcript and protein levels as well as encourage growth arrest and apoptosis in MCF-7 and MDA-MB-231 cells. AZA, TSA, SFN, and SAM inhibit cell growth in MCF-7 and MDA-MB-231 cell lines in a dose-dependent manner, that is, with increasing concentrations of drugs the cell viability gradually decreases. All the epigenetic modulators promote apoptotic cell death, as is evident form increased chromatin condensation which is a distinct characteristic of apoptotic cells. From FACS analysis, it is also clear that these drugs induce G2-M arrest and apoptosis in breast cancer cells. Further, transcript and protein level expression of MBDs and DNMTs is also affected - after treatment with epigenetic drugs; the level of transcripts/mRNA of MBDs and DNMTs has consistently increased in general. The increase in level of gene expression is substantiated at the protein level also where treated cells show higher expression of DNMT1, DNMT3A, DNMT3B, and MBD proteins in comparison to untreated cells. In case of tissue samples, the expression of different DNMTs is tissue stage-specific. DNMT1 exhibits significantly higher expression in the metastatic stage, whereas, DNMT3A and DNMT3B have higher expression in the primary stage in comparison to the metastatic samples.

Conclusion

The epigenetic modulators AZA, TSA, SFN, and SAM may provide opportunities for cancer prevention by regulating the components of epigenetic gene-silencing machinery especially DNMTs and MBDs.

【 授权许可】

   
2014 Kar et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20150130152947456.pdf 1957KB PDF download
Figure 7. 144KB Image download
Figure 6. 169KB Image download
Figure 5. 152KB Image download
Figure 4. 75KB Image download
Figure 3. 122KB Image download
Figure 2. 76KB Image download
Figure 1. 106KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

Figure 6.

Figure 7.

【 参考文献 】
  • [1]Jaenisch R, Bird A: Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 2003, 33:245-254.
  • [2]Klose RJ, Bird AP: Genomic DNA methylation: the mark and its mediators. Trends Biochem Sci 2006, 31:89-97.
  • [3]Patra SK, Deb M, Patra A: Molecular marks for epigenetic identification of developmental and cancer stem cells. Clin Epigenet 2011, 2:27-53.
  • [4]Kar S, Deb M, Sengupta D, Shilpi A, Parbin S, Torrisani J, Pradhan S, Patra SK: An insight into the various regulatory mechanisms modulating Human DNA Methyltransferase 1 stability and function. Epigenetics 2012, 7:994-1007.
  • [5]Jones PA, Liang G: Rethinking how DNA methylation patterns are maintained. Nat Rev Gen 2009, 10:805-811.
  • [6]Denis H, Ndlovu MN, Fuks F: Regulation of mammalian methyltransferase: a route to new mechanisms. EMBO Rep 2011, 12:647-656.
  • [7]Weber M, Schubeler D: Genomic patterns of DNA methylation: targets and function of an epigenetic mark. Curr Opin Cell Biol 2007, 19:273-280.
  • [8]Jeltsch A: Reading and writing DNA methylation. Nat Struct Mol Biol 2008, 15:1003-1004.
  • [9]Wade PA: Methyl CpG binding proteins: coupling chromatin architecture to gene regulation. Oncogene 2001, 20:3166-3173.
  • [10]Deb M, Kar S, Sengupta D, Shilpi A, Parbin S, Rath SK, Londhe V, Patra SK: Chromatin dynamics: H3K4 methylation and H3 variant replacement during development and in cancer. Cell Mol Life Sci 2014, 71:3439-3463.
  • [11]Ballestar E, Esteller M: The impact of chromatin in human cancer: linking DNA methylation to gene silencing. Carcinogenesis 2002, 23:1103-1109.
  • [12]Fatemi M, Wade PA: MBD family proteins: reading the epigenetic code. J Cell Sci 2006, 119:3033-3037.
  • [13]Dhasarathy A, Wade PA: The MBD protein family-reading an epigenetic mark? Mutat Res 2008, 647:39-43.
  • [14]Bogdanovic O, Veenstra GJ: DNA methylation and methyl-CpG binding proteins: developmental requirements and function. Chromosoma 2009, 118:549-565.
  • [15]Sasai N, Defossez PA: Many paths to one goal? The proteins that recognize methylated DNA in eukaryotes. Int J Dev Biol 2009, 53:323-334.
  • [16]Wade PA: Methyl CpG-binding protiens and transcriptional repression. Bioessays 2001, 23:1131-1137.
  • [17]Patra SK, Patra A, Zhao H, Carroll P, Dahiya R: Methyl-CpG-DNA binding proteins in human prostate cancer: expression of CXXC sequence containing MBD1 and repression of MBD2 and MeCP2. Biochem Biophys Res Commun 2003, 302:759-766.
  • [18]Clouaire T, Stancheva I: Methyl-CpG binding proteins: specialized transcriptional repressors or structural components of chromatin? Cell Mol Life Sci 2008, 65:1509-1522.
  • [19]Vaissiere T, Sawan C, Herceg Z: Epigenetic interplay between histone modifications and DNA methylation in gene silencing. Mutat Res 2008, 659:40-48.
  • [20]Baylin SB: DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol 2005, 2:S4-S11.
  • [21]Esteller M: Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet 2007, 16:50-59.
  • [22]Strathdee G, Brown R: Aberrant DNA methylation in cancer: potential clinical interventions. Expert Rev Mol Med 2002, 4:1-17.
  • [23]Das PM, Singal R: DNA methylation and cancer. J Clin Oncol 2004, 22:4632-4642.
  • [24]Luczak MW, Jagodzinski PP: The role of DNA methylation in cancer development. Folia Histochem Cytobiol 2006, 44:143-154.
  • [25]McCabe MT, Brandes JC, Vertino PM: Cancer DNA methylation: molecular mechanisms and clinical implications. Clin Cancer Res 2009, 15:3927-3937.
  • [26]Lopez-Serra L, Esteller M: Proteins that bind methylated DNA and human cancer: reading the wrong words. Br J Cancer 2008, 98:1881-1885.
  • [27]Lopez-Serra L, Ballestar E, Fraga MF, Alaminos M, Setien F, Esteller M: A profile of methyl-CpG binding domain protein occupancy of hypermethylated promoter CpG islands of tumor suppressor genes in human cancer. Cancer Res 2006, 66:8342-8346.
  • [28]Parry L, Clarke AR: The roles of the methyl-CpG binding proteins in cancer. Genes Cancer 2011, 2:618-630.
  • [29]Sansom OJ, Maddison K, Clarke AR: Mechanisms of disease: methyl-binding domain proteins as potential therapeutic targets in cancer. Nat Clin Pract Oncol 2007, 4:305-315.
  • [30]Ballestar E, Paz MF, Valle L, Wei S, Fraga MF, Espada J, Cigudosa JC, Huang TH, Esteller M: Methyl-CpG binding proteins identify novel sites of epigenetic inactivation in human cancer. EMBO J 2003, 22:6335-6345.
  • [31]Lopez-Serra L, Ballestar E, Ropero S, Setien F, Billard LM, Fraga MF, Lopez-Nieva P, Alaminos M, Guerrero D, Dante R, Esteller M: Unmasking of epigenetically silenced candidate tumor suppressor genes by removal of Methyl-CpG binding domain proteins. Oncogene 2008, 27:3556-3566.
  • [32]Egger G, Liang G, Aparicio A, Jones PA: Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004, 429:457-463.
  • [33]Rodenhiser D, Mann M: Epigenetics and human disease: translating basic biology into clinical applications. Can Med Assoc J 2006, 174:341-348.
  • [34]Tone S, Sugimoto K, Tanda K, Suda T, Uehira K, Kanouchi H, Samejima K, Minatogawa Y, Earnshaw WC: Three distinct stages of apoptotic nuclear condensation revealed by time-lapse imaging, biochemical and electron microscopy analysis of cell-free apoptosis. Exp Cell Res 2007, 313:3635-3644.
  • [35]Hsu A, Wong CP, Yu Z, Williams DE, Dashwood RH, Ho E: Promoter de-methylation of cyclin D2 by sulforaphane in prostate cancer cells. Clin Epigenetics 2011, 3:3. BioMed Central Full Text
  • [36]Jones PA, Baylin SB: The epigenomics of cancer. Cell 2007, 128:683-692.
  • [37]Vilain A, Vogt N, Dutrillaux B, Malfoy B: DNA methylation and chromosome instability in breast cancer cell lines. FEBS Lett 1999, 460:231-234.
  • [38]Mirza S, Sharma G, Parshad R, Gupta SD, Pandya P, Ralhan R: Expression of DNA methyltransferases in breast cancer patients and to analyze the effect of naturalcompounds on DNA methyltransferases and associated proteins. J Breast Cancer 2013, 16:23-31.
  • [39]Schmittgen TD, Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 2008, 3:1101-1108.
  • [40]Patra A, Deb M, Dahiya R, Patra SK: 5-Aza-2'-deoxycytidine stress response and apoptosis in prostate cancer. Clin Epigenetics 2011, 2:339-348.
  • [41]Patra SK, Patra A, Dahiya R: Histone deacetylase and DNA methyltransferase in human prostate cancer. Biochem Biophys Res Commun 2001, 287:705-713.
  • [42]Patra SK, Patra A, Zhao H, Dahiya R: DNA methyltransferase and demethylase in human prostate cancer. Mol Carcinog 2002, 33:163-171.
  • [43]Patra SK, Bettuzzi S: Epigenetic DNA-(Cytosine-5-Carbon) Modifications: 5-Aza-2'-Deoxycytidine and DNA-Demethylation. Biochem Mosc 2009, 74:613-619.
  文献评价指标  
  下载次数:10次 浏览次数:6次