68
Views
4
CrossRef citations to date
0
Altmetric
Special Report

Pharmacoepigenetic aspects of gene polymorphism on drug therapies: effects of DNA methylation on drug response

&
Pages 55-65 | Published online: 10 Jan 2014

References

  • Klose RJ, Bird AP. Genomic DNA methylation: the mark and its mediators. Trends Biochem. Sci.31, 89–97 (2006).
  • Ballestar E, Esteller M. The impact of chromatin in human cancer: linking DNA methylation to gene silencing. Carcinogenesis23, 1103–1109 (2002).
  • Georgel PT. Role of chromatin/epigenetic modifications on DNA accessibility. Drug News Perspect.20, 549–556 (2007).
  • Galm O, Esteller M. Beyond genetics – the emerging role of epigenetic changes in hematopoietic malignancies. Int. J. Hematol.80, 120–127 (2004).
  • Baylin SB, Esteller M, Rountree MR et al. Aberrant patterns of DNA methylation, chromatin formation and gene expression in cancer. Hum. Mol. Genet.10, 687–692 (2001).
  • Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat. Rev. Genet.8, 253–262 (2007).
  • Rountree MR, Bachman KE, Herman JG et al. DNA methylation, chromatin inheritance, and cancer. Oncogene20, 3156–3165 (2001).
  • Tate PH, Bird AP. Effects of DNA methylation on DNA-binding proteins and gene expression. Curr. Opin. Genet. Dev.3, 226–231 (1993).
  • Stirzaker C, Song JZ, Davidson B et al. Transcriptional gene silencing promotes DNA hypermethylation through a sequential change in chromatin modifications in cancer cells. Cancer Res.64, 3871–3877 (2004).
  • Keshet I, Lieman-Hurwitz J, Cedar H. DNA methylation affects the formation of active chromatin. Cell44, 535–543 (1986).
  • Padjen K, Ratnam S, Storb U. DNA methylation precedes chromatin modifications under the influence of the strain-specific modifier Ssm1. Mol. Cell Biol.25, 4782–4791 (2005).
  • Caiafa P, Zampieri M. DNA methylation and chromatin structure: the puzzling CpG islands. J. Cell. Biochem.94, 257–265 (2005).
  • Ogino S, Kawasaki T, Nosho K et al. LINE-1 hypomethylation is inversely associated with microsatellite instability and CpG island methylator phenotype in colorectal cancer. Int. J. Cancer122, 2767–2773 (2008).
  • Kawasaki T, Nosho K, Ohnishi M et al. IGFBP3 promoter methylation in colorectal cancer: relationship with microsatellite instability, CpG island methylator phenotype, and p53. Neoplasia9, 1091–1098 (2007).
  • Imai K, Yamamoto H. Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis29, 673–680 (2008).
  • Jass JR. Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology50, 113–130 (2007).
  • Raja P, Sanville BC, Buchmann RC et al. Viral genome methylation as an epigenetic defense against geminiviruses. J. Virol.82(18), 8997–9007 (2008).
  • Doerfler W. De novo methylation, long-term promoter silencing, methylation patterns in the human genome, and consequences of foreign DNA insertion. Curr. Top. Microbiol. Immunol.301, 125–175 (2006).
  • Hochstein N, Muiznieks I, Mangel L et al. Epigenetic status of an adenovirus type 12 transgenome upon long-term cultivation in hamster cells. J. Virol.81, 5349–5361 (2007).
  • Sutter D, Westphal M, Doerfler W. Patterns of integration of viral DNA sequences in the genomes of adenovirus type 12-transformed hamster cells. Cell14, 569–585 (1978).
  • Miranda TB, Jones PA. DNA methylation: the nuts and bolts of repression. J. Cell Physiol.213, 384–390 (2007).
  • Matouk CC, Marsden PA. Epigenetic regulation of vascular endothelial gene expression. Circ. Res.102, 873–887 (2008).
  • Jones PA, Takai D. The role of DNA methylation in mammalian epigenetics. Science293, 1068–1070 (2001).
  • Larsen F, Gundersen G, Lopez R et al. CpG islands as gene markers in the human genome. Genomics13, 1095–1107 (1992).
  • Bird AP. DNA methylation and the frequency of CpG in animal DNA. Nucleic Acids Res.8, 1499–1504 (1980).
  • Takai D, Jones PA. Comprehensive analysis of CpG islands in human chromosomes 21 and 22. Proc. Natl Acad. Sci. USA99, 3740–3745 (2002).
  • Gardiner-Garden M, Frommer M. CpG islands in vertebrate genomes. J. Mol. Biol.196, 261–282 (1987).
  • Li E, Bestor TH, Jaenisch R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell69, 915–926 (1992).
  • Bestor TH. The DNA methyltransferases of mammals. Hum. Mol. Genet.9, 2395–2402 (2000).
  • Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int. J. Cancer123, 8–13 (2008).
  • Mund C, Brueckner B, Lyko F. Reactivation of epigenetically silenced genes by DNA methyltransferase inhibitors: basic concepts and clinical applications. Epigenetics1, 7–13 (2006).
  • Ptak C, Petronis A. Epigenetics and complex disease: from etiology to new therapeutics. Annu. Rev. Pharmacol. Toxicol.48, 257–276 (2008).
  • Garcia-Manero G. Demethylating agents in myeloid malignancies. Curr. Opin. Oncol.20, 705–710 (2008).
  • Atallah E, Kantarjian H, Garcia-Manero G. The role of decitabine in the treatment of myelodysplastic syndromes. Expert Opin. Pharmacother.8, 65–73 (2007).
  • Silverman LR, Demakos EP, Peterson BL et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J. Clin. Oncol.20, 2429–2440 (2002).
  • Momparler RL. Pharmacology of 5-aza-2´-deoxycytidine (decitabine). Semin. Hematol.42, S9–S16 (2005).
  • Cheng JC, Yoo CB, Weisenberger DJ et al. Preferential response of cancer cells to zebularine. Cancer Cell6, 151–158 (2004).
  • Cheng JC, Matsen CB, Gonzales FA et al. Inhibition of DNA methylation and reactivation of silenced genes by zebularine. J. Natl Cancer Inst.95, 399–409 (2003).
  • Holleran JL, Parise RA, Joseph E et al. Plasma pharmacokinetics, oral bioavailability, and interspecies scaling of the DNA methyltransferase inhibitor, zebularine. Clin. Cancer Res.11, 3862–3868 (2005).
  • Liu H, Xue ZT, Sjogren HO et al. Low dose Zebularine treatment enhances immunogenicity of tumor cells. Cancer Lett.257, 107–115 (2007).
  • Lyko F, Brown R. DNA methyltransferase inhibitors and the development of epigenetic cancer therapies. J. Natl Cancer Inst.97, 1498–1506 (2005).
  • de Vos D. Epigenetic drugs: a longstanding story. Semin. Oncol.32, 437–442 (2005).
  • Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. Nat. Rev. Drug Discov.5, 37–50 (2006).
  • Brueckner B, Lyko F. DNA methyltransferase inhibitors: old and new drugs for an epigenetic cancer therapy. Trends Pharmacol. Sci.25, 551–554 (2004).
  • Jiang YH, Bressler J, Beaudet AL. Epigenetics and human disease. Annu. Rev. Genomics Hum. Genet.5, 479–510 (2004).
  • Bateson P, Barker D, Clutton-Brock T et al. Developmental plasticity and human health. Nature430, 419–421 (2004).
  • Petronis A, Gottesman II, Kan P et al. Monozygotic twins exhibit numerous epigenetic differences: clues to twin discordance? Schizophr. Bull.29, 169–178 (2003).
  • Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat. Genet.33(Suppl.), 245–254 (2003).
  • Poulsen P, Esteller M, Vaag A et al. The epigenetic basis of twin discordance in age-related diseases. Pediatr. Res.61, R38–R42 (2007).
  • Fraga MF, Ballestar E, Paz MF et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl Acad. Sci. USA102, 10604–10609 (2005).
  • Vaiserman AM. Epigenetic engineering and its possible role in anti-aging intervention. Rejuvenation Res.11, 39–42 (2008).
  • Cobiac L. Epigenomics and nutrition. Forum Nutr.60, 31–41 (2007).
  • Gallou-Kabani C, Vige A, Gross MS et al. Nutri-epigenomics: lifelong remodelling of our epigenomes by nutritional and metabolic factors and beyond. Clin. Chem. Lab. Med.45, 321–327 (2007).
  • Junien C. Impact of diets and nutrients/drugs on early epigenetic programming. J. Inherit. Metab. Dis.29, 359–365 (2006).
  • Dolinoy DC, Huang D, Jirtle RL. Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proc. Natl Acad. Sci. USA104, 13056–13061 (2007).
  • Pennisi E. Environmental epigenomics meeting. Food, tobacco, and future generations. Science310, 1760–1761 (2005).
  • Mathers JC. Early nutrition: impact on epigenetics. Forum Nutr.60, 42–48 (2007).
  • Cooney CA, Dave AA, Wolff GL. Maternal methyl supplements in mice affect epigenetic variation and DNA methylation of offspring. J. Nutr.132, S2393–S2400 (2002).
  • Wolff GL, Kodell RL, Moore SR et al. Maternal epigenetics and methyl supplements affect agouti gene expression in Avy/a mice. FASEB J.12, 949–957 (1998).
  • Dolinoy DC, Das R, Weidman JR et al. Metastable epialleles, imprinting, and the fetal origins of adult diseases. Pediatr. Res.61, R30–R37 (2007).
  • Kelly TL, Trasler JM. Reproductive epigenetics. Clin. Genet.65, 247–260 (2004).
  • Chen Z, Karaplis AC, Ackerman SL et al. Mice deficient in methylenetetrahydrofolate reductase exhibit hyperhomocysteinemia and decreased methylation capacity, with neuropathology and aortic lipid deposition. Hum. Mol. Genet.10, 433–443 (2001).
  • Powers HJ. Interaction among folate, riboflavin, genotype, and cancer, with reference to colorectal and cervical cancer. J. Nutr.135, S2960–S2966 (2005).
  • Friso S, Choi SW. Gene-nutrient interactions in one-carbon metabolism. Curr. Drug Metab.6, 37–46 (2005).
  • Schwerdtle T, Walter I, Mackiw I et al. Induction of oxidative DNA damage by arsenite and its trivalent and pentavalent methylated metabolites in cultured human cells and isolated DNA. Carcinogenesis24, 967–974 (2003).
  • Hei TK, Liu SX, Waldren C. Mutagenicity of arsenic in mammalian cells: role of reactive oxygen species. Proc. Natl Acad. Sci. USA95, 8103–8107 (1998).
  • Lynn S, Lai HT, Gurr JR et al. Arsenite retards DNA break rejoining by inhibiting DNA ligation. Mutagenesis12, 353–358 (1997).
  • Eblin KE, Bowen ME, Cromey DW et al. Arsenite and monomethylarsonous acid generate oxidative stress response in human bladder cell culture. Toxicol. Appl. Pharmacol.217, 7–14 (2006).
  • Wang TC, Jan KY, Wang AS et al. Trivalent arsenicals induce lipid peroxidation, protein carbonylation, and oxidative DNA damage in human urothelial cells. Mutat. Res.615, 75–86 (2007).
  • Marsit CJ, Karagas MR, Danaee H et al. Carcinogen exposure and gene promoter hypermethylation in bladder cancer. Carcinogenesis27, 112–116 (2006).
  • Jensen TJ, Novak P, Eblin KE et al. Epigenetic remodeling during arsenical-induced malignant transformation. Carcinogenesis29(8), 1500–1508 (2008).
  • Chen H, Li S, Liu J et al. Chronic inorganic arsenic exposure induces hepatic global and individual gene hypomethylation: implications for arsenic hepatocarcinogenesis. Carcinogenesis25, 1779–1786 (2004).
  • Zhao CQ, Young MR, Diwan BA et al. Association of arsenic-induced malignant transformation with DNA hypomethylation and aberrant gene expression. Proc. Natl Acad. Sci. USA94, 10907–10912 (1997).
  • Aposhian HV. Enzymatic methylation of arsenic species and other new approaches to arsenic toxicity. Annu. Rev. Pharmacol. Toxicol.37, 397–419 (1997).
  • Liu J, Benbrahim-Tallaa L, Qian X et al. Further studies on aberrant gene expression associated with arsenic-induced malignant transformation in rat liver TRL1215 cells. Toxicol. Appl. Pharmacol.216, 407–415 (2006).
  • Lee YW, Klein CB, Kargacin B et al. Carcinogenic nickel silences gene expression by chromatin condensation and DNA methylation: a new model for epigenetic carcinogens. Mol. Cell Biol.15, 2547–2557 (1995).
  • Weaver IC, Meaney MJ, Szyf M. Maternal care effects on the hippocampal transcriptome and anxiety-mediated behaviors in the offspring that are reversible in adulthood. Proc. Natl Acad. Sci. USA103, 3480–3485 (2006).
  • Shukla SD, Velazquez J, French SW et al. Emerging role of epigenetics in the actions of alcohol. Alcohol Clin. Exp. Res.32(9), 1525–1534 (2008).
  • Shukla SD, Aroor AR. Epigenetic effects of ethanol on liver and gastrointestinal injury. World J. Gastroenterol.12, 5265–5271 (2006).
  • Bonsch D, Lenz B, Fiszer R et al. Lowered DNA methyltransferase (DNMT-3b) mRNA expression is associated with genomic DNA hypermethylation in patients with chronic alcoholism. J. Neural Transm.113, 1299–1304 (2006).
  • Nielsen DA, Yuferov V, Hamon S et al. Increased OPRM1 DNA methylation in lymphocytes of methadone-maintained former heroin addicts. Neuropsychopharmacology (2008) (Epub ahead of print).
  • Zhang Y, Wang D, Johnson AD et al. Allelic expression imbalance of human µ opioid receptor (OPRM1) caused by variant A118G. J. Biol. Chem.280, 32618–32624 (2005).
  • Ingelman-Sundberg M, Sim SC, Gomez A et al. Influence of cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and clinical aspects. Pharmacol. Ther.116, 496–526 (2007).
  • Tokizane T, Shiina H, Igawa M et al. Cytochrome P450 1B1 is overexpressed and regulated by hypomethylation in prostate cancer. Clin. Cancer Res.11, 5793–5801 (2005).
  • Gomez A, Karlgren M, Edler D et al. Expression of CYP2W1 in colon tumors: regulation by gene methylation. Pharmacogenomics8, 1315–1325 (2007).
  • Karlgren M, Gomez A, Stark K et al. Tumor-specific expression of the novel cytochrome P450 enzyme, CYP2W1. Biochem. Biophys. Res. Commun.341, 451–458 (2006).
  • Anttila S, Hakkola J, Tuominen P et al. Methylation of cytochrome P4501A1 promoter in the lung is associated with tobacco smoking. Cancer Res.63, 8623–8628 (2003).
  • Vidal DO, Lopes LF, Valera ET. Drug resistance and methylation in myelodysplastic syndrome. Curr. Pharm. Biotechnol.8, 77–81 (2007).
  • Chekhun VF, Kulik GI, Yurchenko OV et al. Role of DNA hypomethylation in the development of the resistance to doxorubicin in human MCF-7 breast adenocarcinoma cells. Cancer Lett.231, 87–93 (2006).
  • Baker EK, Johnstone RW, Zalcberg JR et al. Epigenetic changes to the MDR1 locus in response to chemotherapeutic drugs. Oncogene24, 8061–8075 (2005).
  • Kuwano M, Uchiumi T, Hayakawa H et al. The basic and clinical implications of ABC transporters, Y-box-binding protein-1 (YB-1) and angiogenesis-related factors in human malignancies. Cancer Sci.94, 9–14 (2003).
  • Cheetham S, Tang MJ, Mesak F et al. SPARC promoter hypermethylation in colorectal cancers can be reversed by 5-aza-2´deoxycytidine to increase SPARC expression and improve therapy response. Br. J. Cancer98, 1810–1819 (2008).
  • Tang MJ, Tai IT. A novel interaction between procaspase 8 and SPARC enhances apoptosis and potentiates chemotherapy sensitivity in colorectal cancers. J. Biol. Chem.282, 34457–34467 (2007).
  • Muerkoster SS, Werbing V, Koch D et al. Role of myofibroblasts in innate chemoresistance of pancreatic carcinoma-epigenetic downregulation of caspases. Int. J. Cancer123(8), 1751–1760 (2008).
  • Missiaglia E, Donadelli M, Palmieri M et al. Growth delay of human pancreatic cancer cells by methylase inhibitor 5-aza-2´-deoxycytidine treatment is associated with activation of the interferon signalling pathway. Oncogene24, 199–211 (2005).
  • Stoklsa T, Golab J. Prospects for p53-based cancer therapy. Acta Biochim. Pol.52, 321–328 (2005).
  • Wiman KG. Restoration of wild-type p53 function in human tumors: strategies for efficient cancer therapy. Adv. Cancer Res.97, 321–338 (2007).
  • Selivanova G, Wiman KG. Reactivation of mutant p53: molecular mechanisms and therapeutic potential. Oncogene26, 2243–2254 (2007).
  • Pogribny IP, James SJ. Reduction of p53 gene expression in human primary hepatocellular carcinoma is associated with promoter region methylation without coding region mutation. Cancer Lett.176, 169–174 (2002).
  • Bunz F, Hwang PM, Torrance C et al. Disruption of p53 in human cancer cells alters the responses to therapeutic agents. J. Clin. Invest.104, 263–269 (1999).
  • Lopez-Serra L, Esteller M. Proteins that bind methylated DNA and human cancer: reading the wrong words. Br. J. Cancer98, 1881–1885 (2008).
  • Hendrich B, Bird A. Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol. Cell Biol.18, 6538–6547 (1998).
  • Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat. Rev. Cancer6, 38–51 (2006).
  • Insinga A, Monestiroli S, Ronzoni S et al. Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat. Med.11, 71–76 (2005).
  • Butler LM, Zhou X, Xu WS et al. The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc. Natl Acad. Sci. USA99, 11700–11705 (2002).
  • Schnekenburger M, Peng L, Puga A. HDAC1 bound to the Cyp1a1 promoter blocks histone acetylation associated with Ah receptor-mediated trans-activation. Biochim. Biophys. Acta1769, 569–578 (2007).
  • Henkens T, Papeleu P, Elaut G et al. Trichostatin A, a critical factor in maintaining the functional differentiation of primary cultured rat hepatocytes. Toxicol. Appl. Pharmacol.218, 64–71 (2007).
  • Jin B, Ryu DY. Regulation of CYP1A2 by histone deacetylase inhibitors in mouse hepatocytes. J. Biochem. Mol. Toxicol.18, 131–132 (2004).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.