A Role for DNA Methylation in Regulating the Growth Suppressor PMEPA1 Gene in Prostate Cancer

A cascade of epigenetic events contributes to the selective growth advantage of cancer cells during tumor progression. PMEPA1 gene is an androgen-inducible negative regulator of cell growth in the prostate epithelium. During prostate cancer progression PMEPA1 gene transcription is reduced or lost prompting us to investigate the role of epigenetic events in this process. In LAPC4 cells harboring wild type androgen receptor decitabine (5-aza-2’-deoxycitidine) treatment resulted in increased expression of PMEPA1 along with other androgen-inducible genes suggesting a role for DNA methylation in the repression of androgenic cell growth control signals in prostate cancer. In contrast, mutant androgen receptor expressing LNCaP cells were deficient in this response. Therefore, decitabine-induced expression of cell growth controlling genes such as NKX3.1 or PMEPA1 underlines the clinical applicability of decitabine in prostate tumors harboring wild type androgen receptor. Further analysis of DNA methylation within the PMEPA1 promoter downstream sequences suggests that methylation of SP1 binding sites may also contribute to the repression of PMEPA1 gene.

[1]  A. D. De Marzo,et al.  GSTP1 CpG island hypermethylation as a molecular biomarker for prostate cancer , 2004, Journal of cellular biochemistry.

[2]  R. Vessella,et al.  Molecular determinants of resistance to antiandrogen therapy , 2004, Nature Medicine.

[3]  T. Guthrie,et al.  Prostate cancer. , 2020, American family physician.

[4]  S. Abdulkadir,et al.  Notch Signaling and ERK Activation Are Important for the Osteomimetic Properties of Prostate Cancer Bone Metastatic Cell Lines* , 2004, Journal of Biological Chemistry.

[5]  C. Bieberich,et al.  Prostate-specific and Androgen-dependent Expression of a Novel Homeobox Gene* , 1996, The Journal of Biological Chemistry.

[6]  J. Christman,et al.  5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy , 2002, Oncogene.

[7]  J. Clements,et al.  Characterization of a novel gene, STAG1/PMEPA1, upregulated in renal cell carcinoma and other solid tumors , 2001, Molecular carcinogenesis.

[8]  Simon A. Williams,et al.  Does PSA play a role as a promoting agent during the initiation and/or progression of prostate cancer? , 2007, The Prostate.

[9]  Jeanne Kowalski,et al.  Hypermethylation of CpG Islands in Primary and Metastatic Human Prostate Cancer , 2004, Cancer Research.

[10]  Weihua Ni,et al.  OAZ Regulates Bone Morphogenetic Protein Signaling through Smad6 Activation* , 2006, Journal of Biological Chemistry.

[11]  S. Srivastava,et al.  Androgen receptor mutation (T877A) promotes prostate cancer cell growth and cell survival , 2006, Oncogene.

[12]  David A Jones,et al.  Limited gene activation in tumor and normal epithelial cells treated with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine. , 2004, Molecular pharmacology.

[13]  Wei Zhang,et al.  PMEPA1, an androgen-regulated NEDD4-binding protein, exhibits cell growth inhibitory function and decreased expression during prostate cancer progression. , 2003, Cancer research.

[14]  P. Nguyen,et al.  The epigenome as a molecular marker and target , 2005, Cancer.

[15]  S. Arya,et al.  The LNCaP cell line--a new model for studies on human prostatic carcinoma. , 1980, Progress in clinical and biological research.

[16]  Kirsten L. Greene,et al.  CpG methylation at promoter site −140 inactivates TGFβ2 receptor gene in prostate cancer , 2005, Cancer.

[17]  Manel Esteller,et al.  The necessity of a human epigenome project. , 2006, Carcinogenesis.

[18]  R. Singal,et al.  Cytosine methylation represses glutathione S-transferase P1 (GSTP1) gene expression in human prostate cancer cells. , 2001, Cancer research.

[19]  A. Hata,et al.  Poly(ADP-ribose) polymerase 1 interacts with OAZ and regulates BMP-target genes. , 2003, Biochemical and biophysical research communications.

[20]  W. Schulz,et al.  Epigenetics of prostate cancer: beyond DNA methylation , 2006, Journal of cellular and molecular medicine.

[21]  S. Baylin,et al.  Epigenetic gene silencing in cancer – a mechanism for early oncogenic pathway addiction? , 2006, Nature Reviews Cancer.

[22]  Matthias Frisch,et al.  Androgen receptor binding sites identified by a GREF_GATA model. , 2005, Journal of molecular biology.

[23]  J. Lutterbaugh,et al.  PMEPA1, a transforming growth factor-beta-induced marker of terminal colonocyte differentiation whose expression is maintained in primary and metastatic colon cancer. , 2003, Cancer research.

[24]  Ximing J. Yang,et al.  Insensitivity to transforming growth factor-beta results from promoter methylation of cognate receptors in human prostate cancer cells (LNCaP). , 2005, Molecular endocrinology.

[25]  John K Field,et al.  Quantitative high-throughput analysis of DNA methylation patterns by base-specific cleavage and mass spectrometry. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[26]  Yusuke Nakamura,et al.  Identification of STAG1 as a key mediator of a p53-dependent apoptotic pathway , 2004, Oncogene.

[27]  Thomas Werner,et al.  MatInspector and beyond: promoter analysis based on transcription factor binding sites , 2005, Bioinform..

[28]  J W Moul,et al.  A novel androgen-regulated gene, PMEPA1, located on chromosome 20q13 exhibits high level expression in prostate. , 2000, Genomics.