The depletion of DNA methyltransferase-1 and the epigenetic effects of 5-aza-2’deoxycytidine (decitabine) are differentially regulated by cell cycle progression

5-Aza-2'-deoxycytidine (decitabine) is a drug targeting the epigenetic abnormalities of tumors. The basis for its limited efficacy in solid tumors is unresolved, but may relate to their indolent growth, their p53 genotype or both. We report that the primary molecular mechanism of decitabine—depletion of DNA methyltransferase-1 following its “suicide” inactivation—is not absolutely associated with cell cycle progression in HCT 116 colon cancer cells, but is associated with their p53 genotype. Control experiments affirmed that the secondary molecular effects of decitabine on global and promoter-specific CpG methylation and MAGE-A1 mRNA expression were S-phase dependent, as expected. Secondary changes in CpG methylation occurred only in growing cells ~24–48 h after decitabine treatment; these epigenetic changes coincided with p53 accumulation, an index of DNA damage. Conversely, primary depletion of DNA methyltransferase-1 began immediately after a single exposure to 300 nM decitabine and it progressed to completion within ~8 h, even in confluent cells arrested in G1 and G2/M. Our results suggest that DNA repair and remodeling activity in arrested, confluent cells may be sufficient to support the primary molecular action of decitabine, while its secondary, epigenetic effects require cell cycle progression through S-phase.

[1]  Pan‐Chyr Yang,et al.  Dysregulation of p53/Sp1 control leads to DNA methyltransferase-1 overexpression in lung cancer. , 2010, Cancer research.

[2]  B. Ramsahoye,et al.  Targeting of 5-aza-2′-deoxycytidine residues by chromatin-associated DNMT1 induces proteasomal degradation of the free enzyme , 2010, Nucleic acids research.

[3]  J. Issa,et al.  Targeting DNA Methylation , 2009, Clinical Cancer Research.

[4]  I. Wistuba,et al.  Decitabine Effect on Tumor Global DNA Methylation and Other Parameters in a Phase I Trial in Refractory Solid Tumors and Lymphomas , 2009, Clinical Cancer Research.

[5]  David A Jones,et al.  p53-inducible ribonucleotide reductase (p53R2/RRM2B) is a DNA hypomethylation-independent decitabine gene target that correlates with clinical response in myelodysplastic syndrome/acute myelogenous leukemia. , 2008, Cancer research.

[6]  Andreas Schätzlein,et al.  Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. , 2007, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[7]  H. Leonhardt,et al.  Dynamics of Dnmt1 interaction with the replication machinery and its role in postreplicative maintenance of DNA methylation , 2007, Nucleic acids research.

[8]  W. Deppert,et al.  Transcription-independent pro-apoptotic functions of p53. , 2005, Current opinion in cell biology.

[9]  M. Ljungman Activation of DNA damage signaling. , 2005, Mutation research.

[10]  H. Leonhardt,et al.  Recruitment of DNA methyltransferase I to DNA repair sites. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[11]  David A Jones,et al.  Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[12]  K. Ghoshal,et al.  5-Aza-Deoxycytidine Induces Selective Degradation of DNA Methyltransferase 1 by a Proteasomal Pathway That Requires the KEN Box, Bromo-Adjacent Homology Domain, and Nuclear Localization Signal , 2005, Molecular and Cellular Biology.

[13]  S. Pradhan,et al.  Human maintenance DNA (cytosine-5)-methyltransferase and p53 modulate expression of p53-repressed promoters. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[14]  H. Leonhardt,et al.  Replication‐independent chromatin loading of Dnmt1 during G2 and M phases , 2004, EMBO reports.

[15]  Albert Jeltsch,et al.  The Dnmt1 DNA-(cytosine-C5)-methyltransferase Methylates DNA Processively with High Preference for Hemimethylated Target Sites* , 2004, Journal of Biological Chemistry.

[16]  H. Komori,et al.  Differential regulation of expression of the mammalian DNA repair genes by growth stimulation , 2004, Oncogene.

[17]  Jorge Cortes,et al.  Phase 1 study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2'-deoxycytidine (decitabine) in hematopoietic malignancies. , 2004, Blood.

[18]  W. Schulz,et al.  Decrease of DNA methyltransferase 1 expression relative to cell proliferation in transitional cell carcinoma , 2003, International journal of cancer.

[19]  P. Laird,et al.  Phase I trial of continuous infusion 5-aza-2′-deoxycytidine , 2003, Cancer Chemotherapy and Pharmacology.

[20]  M. O’Reilly,et al.  Growth arrest in G1 protects against oxygen‐induced DNA damage and cell death , 2002, Journal of cellular physiology.

[21]  David A Jones,et al.  Reactivating the expression of methylation silenced genes in human cancer , 2002, Oncogene.

[22]  Peter A. Jones,et al.  Cell division is required for de novo methylation of CpG islands in bladder cancer cells. , 2002, Cancer research.

[23]  Y. Seo,et al.  p53 regulation of DNA excision repair pathways. , 2002, Mutagenesis.

[24]  Keith D Robertson,et al.  DNA methylation, methyltransferases, and cancer , 2001, Oncogene.

[25]  D. Jones,et al.  Activation of the p53 DNA damage response pathway after inhibition of DNA methyltransferase by 5-aza-2'-deoxycytidine. , 2001, Molecular pharmacology.

[26]  Samuel H. Wilson,et al.  A role for p53 in base excision repair , 2001, The EMBO journal.

[27]  F. Ishikawa,et al.  MBD2‐MBD3 complex binds to hemi‐methylated DNA and forms a complex containing DNMT1 at the replication foci in late S phase , 2000, Genes to cells : devoted to molecular & cellular mechanisms.

[28]  K. Robertson,et al.  Differential mRNA expression of the human DNA methyltransferases (DNMTs) 1, 3a and 3b during the G(0)/G(1) to S phase transition in normal and tumor cells. , 2000, Nucleic acids research.

[29]  M. Lübbert,et al.  Low-dose 5-aza-2'-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase II study in elderly patients. , 2000, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[30]  Peter A. Jones,et al.  Roles of Cell Division and Gene Transcription in the Methylation of CpG Islands , 1999, Molecular and Cellular Biology.

[31]  K. Kinzler,et al.  Disruption of p53 in human cancer cells alters the responses to therapeutic agents. , 1999, The Journal of clinical investigation.

[32]  G. Wahl,et al.  ES cells do not activate p53-dependent stress responses and undergo p53-independent apoptosis in response to DNA damage , 1998, Current Biology.

[33]  R. Jaenisch,et al.  Toxicity of 5-aza-2'-deoxycytidine to mammalian cells is mediated primarily by covalent trapping of DNA methyltransferase rather than DNA demethylation. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[34]  D. Santi,et al.  Covalent bond formation between a DNA-cytosine methyltransferase and DNA containing 5-azacytosine. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[35]  R. Snyder Inhibitors of ribonucleotide reductase alter DNA repair in human fibroblasts through specific depletion of purine deoxynucleotide triphosphates , 1984, Cell Biology and Toxicology.

[36]  J. Christman,et al.  Inhibition of DNA methyltransferase and induction of Friend erythroleukemia cell differentiation by 5-azacytidine and 5-aza-2'-deoxycytidine. , 1982, The Journal of biological chemistry.

[37]  Peter A. Jones,et al.  Epigenetics in cancer. , 2010, Carcinogenesis.

[38]  Matthew Tudor,et al.  Loss of genomic methylation causes p53-dependent apoptosis and epigenetic deregulation , 2001, Nature Genetics.

[39]  S. Peller Clinical implications of p53: effect on prognosis, tumor progression and chemotherapy response. , 1998, Seminars in cancer biology.