Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Epigenetics ; 14(2): 185-197, 2019 02.
Article in English | MEDLINE | ID: mdl-30764732

ABSTRACT

The POTE gene family consists of 14 homologous genes localized to autosomal pericentromeres, and a sub-set of POTEs are cancer-testis antigen (CTA) genes. POTEs are over-expressed in epithelial ovarian cancer (EOC), including the high-grade serous subtype (HGSC), and expression of individual POTEs correlates with chemoresistance and reduced survival in HGSC. The mechanisms driving POTE overexpression in EOC and other cancers is unknown. Here, we investigated the role of epigenetics in regulating POTE expression, with a focus on DNA hypomethylation. Consistent with their pericentromeric localization, Pan-POTE expression in EOC correlated with expression of the pericentromeric repeat NBL2, which was not the case for non-pericentromeric CTAs. POTE genomic regions contain LINE-1 (L1) sequences, and Pan-POTE expression correlated with both global and POTE-specific L1 hypomethylation in EOC. Analysis of individual POTEs using RNA-seq and DNA methylome data from fallopian tube epithelia (FTE) and HGSC revealed that POTEs C, E, and F have increased expression in HGSC in conjunction with DNA hypomethylation at 5' promoter or enhancer regions. Moreover, POTEs C/E/F showed additional increased expression in recurrent HGSC in conjunction with 5' hypomethylation, using patient-matched samples. Experiments using decitabine treatment and DNMT knockout cell lines verified a functional contribution of DNA methylation to POTE repression, and epigenetic drug combinations targeting histone deacetylases (HDACs) and histone methyltransferases (HMTs) in combination with decitabine further increased POTE expression. In summary, several alterations of the cancer epigenome, including pericentromeric activation, global and locus-specific L1 hypomethylation, and locus-specific 5' CpG hypomethylation, converge to promote POTE expression in ovarian cancer.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , DNA Methylation , Ovarian Neoplasms/genetics , Proteins/genetics , Up-Regulation , Cell Line, Tumor , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Long Interspersed Nucleotide Elements , Promoter Regions, Genetic
2.
Sci Rep ; 8(1): 17136, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30459449

ABSTRACT

The POTE family includes 14 genes in three phylogenetic groups. We determined POTE mRNA expression in normal tissues, epithelial ovarian and high-grade serous ovarian cancer (EOC, HGSC), and pan-cancer, and determined the relationship of POTE expression to ovarian cancer clinicopathology. Groups 1 & 2 POTEs showed testis-specific expression in normal tissues, consistent with assignment as cancer-testis antigens (CTAs), while Group 3 POTEs were expressed in several normal tissues, indicating they are not CTAs. Pan-POTE and individual POTEs showed significantly elevated expression in EOC and HGSC compared to normal controls. Pan-POTE correlated with increased stage, grade, and the HGSC subtype. Select individual POTEs showed increased expression in recurrent HGSC, and POTEE specifically associated with reduced HGSC OS. Consistent with tumors, EOC cell lines had significantly elevated Pan-POTE compared to OSE and FTE cells. Notably, Group 1 & 2 POTEs (POTEs A/B/B2/C/D), Group 3 POTE-actin genes (POTEs E/F/I/J/KP), and other Group 3 POTEs (POTEs G/H/M) show within-group correlated expression, and pan-cancer analyses of tumors and cell lines confirmed this relationship. Based on their restricted expression in normal tissues and increased expression and association with poor prognosis in ovarian cancer, POTEs are potential oncogenes and therapeutic targets in this malignancy.


Subject(s)
Antigens, Neoplasm/genetics , Ovarian Neoplasms/genetics , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Multigene Family , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Survival Analysis
3.
Oncotarget ; 7(11): 12840-56, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26883197

ABSTRACT

Cancer testis antigens (CTAs) are promising cancer associated antigens in solid tumors, but in acute myeloid leukemia, dense promoter methylation silences their expression. Leukemia cell lines exposed to HMAs induce expression of CTAs. We hypothesized that AML patients treated with standard of care decitabine (20mg/m2 per day for 10 days) would demonstrate induced expression of CTAs. Peripheral blood blasts serially isolated from AML patients treated with decitabine were evaluated for CTA gene expression and demethylation. Induction of NY-ESO-1 and MAGEA3/A6, were observed following decitabine. Re-expression of NY-ESO-1 and MAGEA3/A6 was associated with both promoter specific and global (LINE-1) hypomethylation. NY-ESO-1 and MAGEA3/A6 mRNA levels were increased irrespective of clinical response, suggesting that these antigens might be applicable even in patients who are not responsive to HMA therapy. Circulating blasts harvested after decitabine demonstrate induced NY-ESO-1 expression sufficient to activate NY-ESO-1 specific CD8+ T-cells. Induction of CTA expression sufficient for recognition by T-cells occurs in AML patients receiving decitabine. Vaccination against NY-ESO-1 in this patient population is feasible.


Subject(s)
Antigens, Neoplasm/biosynthesis , Antineoplastic Agents/therapeutic use , Azacitidine/analogs & derivatives , Leukemia, Myeloid, Acute/drug therapy , Membrane Proteins/biosynthesis , Aged , Aged, 80 and over , Azacitidine/therapeutic use , Decitabine , Female , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged
4.
Epigenetics ; 10(3): 237-46, 2015.
Article in English | MEDLINE | ID: mdl-25793777

ABSTRACT

We aimed to determine the effect of SGI-110 on methylation and expression of the cancer testis antigens (CTAs) NY-ESO-1 and MAGE-A in epithelial ovarian cancer (EOC) cells in vitro and in vivo and to establish the impact of SGI-110 on expression of major histocompatibility (MHC) class I and Intracellular Adhesion Molecule 1 (ICAM-1) on EOC cells, and on recognition of EOC cells by NY-ESO-1-specific CD8+ T-cells. We also tested the impact of combined SGI-110 and NY-ESO-1-specific CD8+ T-cells on tumor growth and/or murine survival in a xenograft setting. EOC cells were treated with SGI-110 in vitro at various concentrations and as tumor xenografts with 3 distinct dose schedules. Effects on global methylation (using LINE-1), NY-ESO-1 and MAGE-A methylation, mRNA, and protein expression were determined and compared to controls. SGI-110 treated EOC cells were evaluated for expression of immune-modulatory genes using flow cytometry, and were co-cultured with NY-ESO-1 specific T-cell clones to determine immune recognition. In vivo administration of SGI-110 and CD8+ T-cells was performed to determine anti-tumor effects on EOC xenografts. SGI-110 treatment induced hypomethylation and CTA gene expression in a dose dependent manner both in vitro and in vivo, at levels generally superior to azacitidine or decitabine. SGI-110 enhanced the expression of MHC I and ICAM-1, and enhanced recognition of EOC cells by NY-ESO-1-specific CD8+ T-cells. Sequential SGI-110 and antigen-specific CD8+ cell treatment restricted EOC tumor growth and enhanced survival in a xenograft setting. SGI-110 is an effective hypomethylating agent and immune modulator and, thus, an attractive candidate for combination with CTA-directed vaccines in EOC.


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Methyltransferases/antagonists & inhibitors , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Antigens, Neoplasm/genetics , Azacitidine/pharmacology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Heterografts , Humans , Mice, SCID , Neoplasm Transplantation , T-Lymphocytes
6.
Leuk Res ; 38(11): 1332-41, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25260825

ABSTRACT

The mechanism of clinical action for the FDA approved hypomethylating drugs azacitidine and decitabine remains unresolved and in this context the potential immunomodulatory effect of these agents on leukemic cells is an area of active investigation. Induced expression of methylated Cancer Testis Antigen (CTA) genes has been demonstrated in leukemic cell lines following exposure to hypomethylating drugs in vitro. SGI-110 is a novel hypomethylating dinucleotide with prolonged in vivo exposure and clinical activity in patients with MDS and AML. We demonstrate that this agent, like decitabine, produces robust re-expression of the CTAs NY-ESO-1 and MAGE-A, both in vitro and in leukemia-bearing AML xenografts. Upregulation of these genes in vitro was sufficient to induce cytotoxicity by HLA-compatible CD8+ T-cells specific for NY-ESO-1, a well-recognized and immunogenic CTA. Additionally, exposure to SGI-110 enhances MHC class I and co-stimulatory molecule expression, potentially contributing to recognition of CTAs. SGI-110, like the parent compound decitabine, induces expression of CTAs and might modulate immune recognition of myeloid malignancy.


Subject(s)
Azacitidine/analogs & derivatives , Immunologic Factors/pharmacology , Leukemia, Myeloid, Acute/pathology , Animals , Azacitidine/pharmacology , Cell Line, Tumor , DNA Methylation , Female , Humans , Mice , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
7.
Cancer Immunol Res ; 2(1): 37-49, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24535937

ABSTRACT

The cancer-testis/cancer-germline antigen NY-ESO-1 is a vaccine target in epithelial ovarian cancer (EOC), but its limited expression is a barrier to vaccine efficacy. As NY-ESO-1 is regulated by DNA methylation, we hypothesized that DNA methyltransferase (DNMT) inhibitors may augment NY-ESO-1 vaccine therapy. In agreement, global DNA hypomethylation in EOC was associated with the presence of circulating antibodies to NY-ESO-1. Pre-clinical studies using EOC cell lines showed that decitabine treatment enhanced both NY-ESO-1 expression and NY-ESO-1-specific CTL-mediated responses. Based on these observations, we performed a phase I dose-escalation trial of decitabine, as an addition to NY-ESO-1 vaccine and doxorubicin liposome (doxorubicin) chemotherapy, in 12 patients with relapsed EOC. The regimen was safe, with limited and clinically manageable toxicities. Both global and promoter-specific DNA hypomethylation occurred in blood and circulating DNAs, the latter of which may reflect tumor cell responses. Increased NY-ESO-1 serum antibodies and T cell responses were observed in the majority of patients, and antibody spreading to additional tumor antigens was also observed. Finally, disease stabilization or partial clinical response occurred in 6/10 evaluable patients. Based on these encouraging results, evaluation of similar combinatorial chemo-immunotherapy regimens in EOC and other tumor types is warranted.


Subject(s)
Antigens, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer Vaccines/immunology , Epigenesis, Genetic/drug effects , Membrane Proteins/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Adult , Aged , Antigens, Neoplasm/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Cancer Vaccines/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , DNA Methylation , Decitabine , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Female , Gene Expression , Humans , Immunity, Humoral , Immunotherapy, Active , Long Interspersed Nucleotide Elements , Membrane Proteins/genetics , Middle Aged , Neoplasm Recurrence, Local , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Polyethylene Glycols/administration & dosage , T-Lymphocyte Subsets/immunology , Treatment Outcome
8.
Epigenetics ; 8(8): 849-63, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23839233

ABSTRACT

MAGEA11 is a cancer germline (CG) antigen and androgen receptor co-activator. Its expression in cancers other than prostate, and its mechanism of activation, has not been reported. In silico analyses reveal that MAGEA11 is frequently expressed in human cancers, is increased during tumor progression, and correlates with poor prognosis and survival. In prostate and epithelial ovarian cancers (EOC), MAGEA11 expression was associated with promoter and global DNA hypomethylation, and with activation of other CG genes. Pharmacological or genetic inhibition of DNA methyltransferases (DNMTs) and/or histone deacetylases (HDACs) activated MAGEA11 in a cell line specific manner. MAGEA11 promoter activity was directly repressed by DNA methylation, and partially depended on Sp1, as pharmacological or genetic targeting of Sp1 reduced MAGEA11 promoter activity and endogenous gene expression. Importantly, DNA methylation regulated nucleosome occupancy specifically at the -1 positioned nucleosome of MAGEA11. Methylation of a single Ets site near the transcriptional start site (TSS) correlated with -1 nucleosome occupancy and, by itself, strongly repressed MAGEA11 promoter activity. Thus, DNA methylation regulates nucleosome occupancy at MAGEA11, and this appears to function cooperatively with sequence-specific transcription factors to regulate gene expression. MAGEA11 regulation is highly instructive for understanding mechanisms regulating CG antigen genes in human cancer.


Subject(s)
Antigens, Neoplasm/genetics , DNA Methylation/genetics , Neoplasm Proteins/genetics , Neoplasms, Glandular and Epithelial/genetics , Nucleosomes/genetics , Ovarian Neoplasms/genetics , Prostatic Hyperplasia/genetics , Prostatic Neoplasms/genetics , Antigens, Neoplasm/metabolism , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Gene Expression Regulation , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Neoplasm Proteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Nucleosomes/metabolism , Ovarian Neoplasms/metabolism , Promoter Regions, Genetic , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , Sp1 Transcription Factor/metabolism
9.
Clin Epigenetics ; 2(2): 349-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-22704347

ABSTRACT

DNA methylation plays an integral role in development and aging through epigenetic regulation of genome function. DNA methyltransferase 1 (Dnmt1) is the most prevalent DNA methyltransferase that maintains genomic methylation stability. To further elucidate the function of Dnmt1 in aging and age-related diseases, we exploited the Dnmt1+/- mouse model to investigate how Dnmt1 haploinsufficiency impacts the aging process by assessing the changes of several major aging phenotypes. We confirmed that Dnmt1 haploinsufficiency indeed decreases DNA methylation as a result of reduced Dnmt1 expression. To assess the effect of Dnmt1 haploinsufficiency on general body composition, we performed dual-energy X-ray absorptiometry analysis and showed that reduced Dnmt1 activity decreased bone mineral density and body weight, but with no significant impact on mortality or body fat content. Using behavioral tests, we demonstrated that Dnmt1 haploinsufficiency impairs learning and memory functions in an age-dependent manner. Taken together, our findings point to the interesting likelihood that reduced genomic methylation activity adversely affects the healthy aging process without altering survival and mortality. Our studies demonstrated that cognitive functions of the central nervous system are modulated by Dnmt1 activity and genomic methylation, highlighting the significance of the original epigenetic hypothesis underlying memory coding and function.

10.
Cancer Immun ; 10: 6, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20649179

ABSTRACT

Expression of the cancer-germline (CG) (or cancer-testis) antigen gene BORIS/CTCFL has been proposed to mediate activation of CG antigen genes in cancer. Consistent with this idea, we have observed that BORIS is frequently expressed in ovarian cancer, often in conjunction with other CG genes. Here we assessed the role of BORIS in CG antigen gene regulation and DNA methylation using normal and cancerous ovarian cell lines, and the CG genes MAGE-A1, NY-ESO-1, and XAGE-1 as models. Adenoviral vectored BORIS was expressed at robust levels and exhibited predominant nuclear localization in ovarian cells. However, BORIS expression in immortalized ovarian surface epithelial cells or ovarian cancer cell lines did not induce CG antigen gene expression or lead to CG antigen promoter DNA hypomethylation. BORIS overexpression also did not alter global DNA methylation, as assessed by genomic 5-methyl-deoxycytidine levels and LINE-1 methylation. We used decitabine to further assess the role of BORIS in CG gene activation and found that decitabine treatment induced BORIS and other CG genes with similar kinetics, suggesting that BORIS induction does not account for the induction of other CG genes by decitabine in ovarian cancer cells. In agreement, siRNA knockdown of BORIS did not block decitabine-mediated induction of CG genes or DNA hypomethylation in ovarian cancer cells treated with this agent. We conclude that BORIS is insufficient for CG antigen gene expression and DNA hypomethylation in ovarian cell lines, and that additional factors are likely required for CG antigen expression in ovarian cancer.


Subject(s)
Antigens, Neoplasm/biosynthesis , DNA Methylation , DNA-Binding Proteins/biosynthesis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Decitabine , Epigenesis, Genetic , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Organ Specificity , Promoter Regions, Genetic , Transcription Initiation Site
11.
J Biol Chem ; 285(6): 4110-4121, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19946145

ABSTRACT

Although DNA methylation is critical for proper embryonic and tissue-specific development, how different DNA methyltransferases affect tissue-specific development and their targets remains unknown. We address this issue in zebrafish through antisense-based morpholino knockdown of Dnmt3 and Dnmt1. Our data reveal that Dnmt3 is required for proper neurogenesis, and its absence results in profound defects in brain and retina. Interestingly, other organs such as intestine remain unaffected suggesting tissue-specific requirements of Dnmt3. Further, comparison of Dnmt1 knockdown phenotypes with those of Dnmt3 suggested that these two families have distinct functions. Consistent with this idea, Dnmt1 failed to complement Dnmt3 deficiency, and Dnmt3 failed to complement Dnmt1 deficiency. Downstream of Dnmt3 we identify a neurogenesis regulator, lef1, as a Dnmt3-specific target gene that is demethylated and up-regulated in dnmt3 morphants. Knockdown of lef1 rescued neurogenesis defects resulting from Dnmt3 absence. Mechanistically, we show cooperation between Dnmt3 and an H3K9 methyltransferase G9a in regulating lef1. Further, like Dnmt1-Suv39h1 cooperativity, Dnmt3 and G9a seemed to function together for tissue-specific development. G9a knockdown, but not Suv39h1 loss, phenocopied dnmt3 morphants and G9a overexpression provided a striking rescue of dnmt3 morphant phenotypes, whereas Suv39h1 overexpression failed, supporting the notion of specific DNMT-histone methyltransferase networks. Consistent with this model, H3K9me3 levels on the lef1 promoter were reduced in both dnmt3 and g9a morphants, and its knockdown rescued neurogenesis defects in g9a morphants. We propose a model wherein specific DNMT-histone methyltransferase networks are utilized to silence critical regulators of cell fate in a tissue-specific manner.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Histone-Lysine N-Methyltransferase/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Brain/cytology , Brain/embryology , Brain/metabolism , Chromatin Immunoprecipitation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/metabolism , In Situ Hybridization , Models, Biological , Neurogenesis/genetics , Retina/cytology , Retina/embryology , Retina/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
12.
Carcinogenesis ; 31(3): 376-81, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20008439

ABSTRACT

Breast cancer, the most common malignancy in women, emerges through a multistep process, encompassing the progressive sequential evolution of morphologically distinct stages from a normal cell to hyperplasia (with and without atypia), carcinoma in situ, invasive carcinoma and metastasis. The success of treatment of breast cancer could be greatly improved by the detection at early stages of cancer. In the present study, we investigated the underlying molecular mechanisms involved in breast carcinogenesis in Augustus and Copenhagen-Irish female rats, a cross between the ACI strains, induced by continuous exposure to 17beta-estradiol. The results of our study demonstrate that early stages of estrogen-induced breast carcinogenesis are characterized by altered global DNA methylation, aberrant expression of proteins responsible for the proper maintenance of DNA methylation pattern and epigenetic silencing of the critical Rassf1a (Ras-association domain family 1, isoform A) tumor suppressor gene. Interestingly, transcriptional repression of the Rassf1a gene in mammary glands during early stages of breast carcinogenesis was associated with an increase in trimethylation of histones H3 lysine 9 and H3 lysine 27 and de novo CpG island methylation and at the Rassf1a promoter and first exon. In conclusion, we demonstrate that epigenetic alterations precede formation of preneoplastic lesions indicating the significance of epigenetic events in induction of oncogenic pathways in early stages of carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Estradiol/toxicity , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Mammary Neoplasms, Experimental/genetics , Animals , CpG Islands , DNA Methylation , Exons/genetics , Female , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Hyperplasia , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/chemically induced , Neoplasm Proteins/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Promoter Regions, Genetic/genetics , Random Allocation , Rats , Rats, Inbred ACI , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
13.
Carcinogenesis ; 30(11): 1889-97, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19584139

ABSTRACT

BACKGROUND: Global DNA hypomethylation may result in chromosomal instability and oncogene activation, and as a surrogate of systemic methylation activity, may be associated with breast cancer risk. METHODS: Samples and data were obtained from women with incident early-stage breast cancer (I-IIIa) and women who were cancer free, frequency matched on age and race. In preliminary analyses, genomic methylation of leukocyte DNA was determined by measuring 5-methyldeoxycytosine (5-mdC), as well as methylation analysis of the LINE-1-repetitive DNA element. Further analyses used only 5-mdC levels. Logistic regression models were used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for risk of breast cancer in relation to amounts of methylation. RESULTS: In a subset of samples tested (n = 37), 5-mdC level was not correlated with LINE-1 methylation. 5-mdC level in leukocyte DNA was significantly lower in breast cancer cases than healthy controls (P = 0.001), but no significant case-control differences were observed with LINE-1 methylation (P = 0.176). In the entire data set, we noted significant differences in 5-mdC levels in leukocytes between cases (n = 176) and controls (n = 173); P value < 0.001. Compared with women in the highest 5-mdC tertile (T3), women in the second (T2; OR = 1.49, 95% CI = 0.84-2.65) and lowest tertile (T1; OR = 2.86, 95% CI = 1.65-4.94) had higher risk of breast cancer (P for trend < or = 0.001). Among controls only and cases and controls combined, only alcohol intake was found to be inversely associated with methylation levels. CONCLUSION: These findings suggest that leukocyte DNA hypomethylation is independently associated with development of breast cancer.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , 5-Methylcytosine/analogs & derivatives , Adult , Aged , Alcohol Drinking , Breast Neoplasms/blood , Breast Neoplasms/pathology , Confidence Intervals , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Dietary Supplements , Female , Humans , Leukocytes/metabolism , Middle Aged , Neoplasm Staging , Odds Ratio , Repetitive Sequences, Nucleic Acid , Risk , Risk Factors , Smoking
14.
Mol Cancer Res ; 7(6): 851-62, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19531572

ABSTRACT

The H3K9me2 histone methyltransferases G9a and GLP repress Mage-a class cancer germ-line (CG) antigen gene expression in murine embryonic stem (ES) cells, but the role of these enzymes in CG antigen gene regulation in human cancer cells is unknown. Here we show that whereas independent or dual knockdown of G9a and GLP in human cancer cells leads to reduced global and CG antigen promoter-associated H3K9me2 levels, it does not activate CG antigen gene expression. Moreover, CG antigen gene repression is maintained following pharmacologic targeting of G9a or treatment of G9a knockdown cells with the histone deacetylase inhibitor trichostatin A. However, G9a knockdown cells display increased sensitivity to CG antigen gene activation mediated by the DNA methyltransferase inhibitor decitabine. To account for these findings, we examined DNA methylation at CG antigen gene promoters in both cell types. We found robust DNA hypomethylation in G9a/GLP targeted murine ES cells but a lack of DNA methylation changes in G9a/GLP targeted human cancer cells; intriguingly, this distinction also extended to markers of global DNA methylation. These data reveal that G9a/GLP is required for DNA methylation of CG antigen genes and genomic DNA in murine ES cells, but not human cancer cells, and implicate DNA methylation status as the key epigenetic mechanism involved in CG antigen gene repression.


Subject(s)
Antigens, Neoplasm/metabolism , Embryonic Stem Cells/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Animals , Antigens, Neoplasm/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line , Cell Line, Tumor , DNA Methylation , Decitabine , Embryonic Stem Cells/immunology , Gene Expression/drug effects , Gene Expression Regulation , Histocompatibility Antigens/genetics , Histone Deacetylase Inhibitors , Histone Deacetylases/pharmacology , Histone-Lysine N-Methyltransferase/genetics , Humans , Hydroxamic Acids/pharmacology , Mice , Mice, Knockout
15.
Mol Cancer Res ; 7(4): 523-35, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19372581

ABSTRACT

Melanoma antigen gene protein-A11 (MAGE-11) of the MAGE family of cancer germ-line antigens increases androgen receptor (AR) transcriptional activity through its interaction with the AR NH(2)-terminal FXXLF motif. The present study investigated the regulatory mechanisms that control MAGE-11 expression during androgen deprivation therapy and prostate cancer progression. Studies include the CWR22 xenograft model of human prostate cancer, clinical specimens of benign and malignant prostate, and prostate cancer cell lines. MAGE-11 mRNA levels increased 100- to 1,500-fold during androgen deprivation therapy and prostate cancer progression, with highest levels in the castration-recurrent CWR22 xenograft and clinical specimens of castration-recurrent prostate cancer. Pyrosequencing of genomic DNA from prostate cancer specimens and cell lines indicated the increase in MAGE-11 resulted from DNA hypomethylation of a CpG island in the 5' promoter of the MAGE-11 gene. Sodium bisulfite sequencing of genomic DNA from benign and malignant prostate tumors and prostate cancer cell lines revealed DNA hypomethylation at individual CpG sites at the transcription start site were most critical for MAGE-11 expression. Cyclic AMP (cAMP) also increased MAGE-11 expression and AR transcriptional activity in prostate cancer cell lines. However, cAMP did not alter DNA methylation of the promoter and its effects were inhibited by extensive DNA methylation in the MAGE-11 promoter region. Increased expression of the AR coregulator MAGE-11 through promoter DNA hypomethylation and cAMP provides a novel mechanism for increased AR signaling in castration-recurrent prostate cancer.


Subject(s)
Antigens, Neoplasm/genetics , Cyclic AMP/pharmacology , DNA Methylation , Gene Expression Regulation/physiology , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Adult , Aged , Androgens/pharmacology , Animals , Castration , CpG Islands/genetics , Humans , Immunoenzyme Techniques , Male , Mice , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Promoter Regions, Genetic , Prostatectomy , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transplantation, Heterologous
16.
Cell ; 135(7): 1201-12, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109892

ABSTRACT

Evidence for active DNA demethylation in vertebrates is accumulating, but the mechanisms and enzymes remain unclear. Using zebrafish embryos we provide evidence for 5-methylcytosine (5-meC) removal in vivo via the coupling of a 5-meC deaminase (AID, which converts 5-meC to thymine) and a G:T mismatch-specific thymine glycosylase (Mbd4). The injection of methylated DNA into embryos induced a potent DNA demethylation activity, which was attenuated by depletion of AID or the non enzymatic factor Gadd45. Remarkably, overexpression of the deaminase/glycosylase pair AID/Mbd4 in vivo caused demethylation of the bulk genome and injected methylated DNA fragments, likely involving a G:T intermediate. Furthermore, AID or Mbd4 knockdown caused the remethylation of a set of common genes. Finally, Gadd45 promoted demethylation and enhanced functional interactions between deaminase/glycosylase pairs. Our results provide evidence for a coupled mechanism of 5-meC demethylation, whereby AID deaminates 5-meC, followed by thymine base excision by Mbd4, promoted by Gadd45.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Glycosylases/metabolism , DNA Methylation , Intracellular Signaling Peptides and Proteins/metabolism , Thymine DNA Glycosylase/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Line , Cytidine Deaminase/metabolism , Embryo, Nonmammalian/metabolism , Humans , Neuropeptides/metabolism , Up-Regulation , GADD45 Proteins
17.
Cancer Res ; 68(22): 9358-66, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010910

ABSTRACT

While the therapeutic activity of the deoxycytidine analogue decitabine is thought to reflect its ability to reactivate methylation-silenced genes, this agent is also known to trigger p53-dependent DNA damage responses. Here, we report that p53-inducible ribonucleotide reductase (p53R2/RRM2B) is a robust transcriptional target of decitabine. In cancer cells, decitabine treatment induces p53R2 mRNA expression, protein expression, and promoter activity in a p53-dependent manner. The mechanism of p53R2 gene induction by decitabine does not seem to be promoter DNA hypomethylation, as the p53R2 5' CpG island is hypomethylated before treatment. Small interfering RNA (siRNA) targeting of DNA methyltransferase 1 (DNMT1) in wild-type p53 cells leads to genomic DNA hypomethylation but does not induce p53R2, suggesting that DNMT/DNA adduct formation is the molecular trigger for p53R2 induction. Consistent with this idea, only nucleoside-based DNMT inhibitors that form covalent DNA adducts induce p53R2 expression. siRNA targeting of p53R2 reduces the extent of cell cycle arrest following decitabine treatment, supporting a functional role for p53R2 in decitabine-mediated cellular responses. To determine the clinical relevance of p53R2 induction, we measured p53R2 expression in bone marrow samples from 15 myelodysplastic syndrome/acute myelogenous leukemia (MDS/AML) patients undergoing decitabine therapy. p53R2 mRNA and protein were induced in 7 of 13 (54%) and 6 of 9 (67%) patients analyzed, respectively, despite a lack of methylation changes in the p53R2 promoter. Most notably, there was a significant association (P = 0.0047) between p53R2 mRNA induction and clinical response in MDS/AML. These data establish p53R2 as a novel hypomethylation-independent decitabine gene target associated with clinical response.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Cell Cycle Proteins/genetics , DNA Methylation , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Ribonucleotide Reductases/genetics , Azacitidine/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Decitabine , G1 Phase , Humans , Ribonucleotide Reductases/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology
18.
Brain Res ; 1237: 25-34, 2008 Oct 27.
Article in English | MEDLINE | ID: mdl-18694733

ABSTRACT

The maintenance of the cellular epigenomic landscape, which depends on the status of the one-carbon metabolic pathway, is essential for normal central nervous system development and function. In the present study, we examined the epigenetic alterations in the brains of Fisher 344 rats induced by the long-term administration of a diet lacking of essential one-carbon nutrients, methionine, choline, and folic acid. The results demonstrated that feeding a folate/methyl-deficient diet causes global DNA hypermethylation as indicated by an increase of genomic 5-methyl-2'-deoxycytidine (5mdC) content and more importantly, by an increase of methylation within unmethylated CpG-rich DNA domains. Interestingly, these epigenetic changes were opposite to those observed in the livers of the same folate/methyl-deficient rats. The hypermethylation changes were associated with an increased protein expression of de novo DNA methyltransferase DNMT3a and methyl-CpG-binding protein 2. Additionally, the gene expression profiling identified 33 significantly up- or down-regulated genes (fold change > or =1.5 and p< or =0.05) in the brains of rats fed a folate/methyl-deficient diet for 36 weeks. Interestingly, we detected an up-regulation of regulatory factor X, 3 (Rfx3) gene, a sequence-specific DNA-binding protein, that mediates the transcriptional activation of silenced by methylation genes, which may be an adaptive protective brain response to hypermethylation. Together, these data suggest that the proper maintenance of the epigenomic landscape in normal brain depends on the adequate supply of essential nutrients involved in the metabolism of methyl groups.


Subject(s)
Brain/metabolism , Diet, Reducing/adverse effects , Epigenesis, Genetic/physiology , Folic Acid Deficiency/etiology , Folic Acid Deficiency/pathology , Animals , DNA Modification Methylases/classification , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Homocysteine/metabolism , Male , Methionine/deficiency , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Inbred F344 , Regulatory Factor X Transcription Factors , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Mol Cancer Res ; 6(8): 1365-74, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18667590

ABSTRACT

We analyzed DNA methyltransferase (Dnmt) protein expression and DNA methylation patterns during four progressive stages of prostate cancer in the transgenic adenocarcinoma of mouse prostate (TRAMP) model, including prostatic intraepithelial neoplasia, well-differentiated tumors, early poorly differentiated tumors, and late poorly differentiated tumors. Dnmt1, Dnmt3a, and Dnmt3b protein expression were increased in all stages; however, after normalization to cyclin A to account for cell cycle regulation, Dnmt proteins remained overexpressed in prostatic intraepithelial neoplasia and well-differentiated tumors, but not in poorly differentiated tumors. Restriction landmark genomic scanning analysis of locus-specific methylation revealed a high incidence of hypermethylation only in poorly differentiated (early and late) tumors. Several genes identified by restriction landmark genomic scanning showed hypermethylation of downstream regions correlating with mRNA overexpression, including p16INK4a, p19ARF, and Cacna1a. Parallel gene expression and DNA methylation analyses suggests that gene overexpression precedes downstream hypermethylation during prostate tumor progression. In contrast to gene hypermethylation, genomic DNA hypomethylation, including hypomethylation of repetitive elements and loss of genomic 5-methyldeoxycytidine, occurred in both early and late stages of prostate cancer. DNA hypermethylation and DNA hypomethylation did not correlate in TRAMP, and Dnmt protein expression did not correlate with either variable, with the exception of a borderline significant association between Dnmt1 expression and DNA hypermethylation. In summary, our data reveal the relative timing of and relationship between key alterations of the DNA methylation pathway occurring during prostate tumor progression in an in vivo model system.


Subject(s)
Adenocarcinoma/enzymology , Adenocarcinoma/pathology , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Disease Models, Animal , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Adenocarcinoma/genetics , Animals , Calcium Channels, N-Type , Calcium Channels, P-Type/genetics , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/genetics , Calcium Channels, Q-Type/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Genome/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Staging , Prostatic Neoplasms/genetics
20.
Carcinogenesis ; 29(3): 638-46, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18204080

ABSTRACT

Genotoxic carcinogens, including 2-acetylaminofluorene (2-AAF), in addition to exerting their genotoxic effects, often cause a variety of non-genotoxic alterations in cells. It is believed that these non-genotoxic effects may be indispensable events in tumorigenesis; however, there is insufficient knowledge to clarify the role of carcinogens in both the genetic and epigenetic changes in premalignant tissues and a lack of conclusive information on the link between epigenetic alterations and carcinogenic exposure. In the current study, we investigated whether or not the mechanism of 2-AAF-induced hepatocarcinogenesis consists of both genotoxic (genetic) and non-genotoxic (epigenetic) alterations. Male and female Sprague-Dawley rats were fed NIH-31 diet containing 0.02% of 2-AAF for 6, 12, 18 or 24 weeks. The levels of DNA adducts obtained from 2-AAF in liver and kidney tissues were assessed by high-performance liquid chromatography combined with electrospray tandem mass spectrometry (HPLC-ES-MS/MS). N-(Deoxyguanosine-8-yl)-2-aminofluorene was the major adduct detected at all time points in both tissues. Global DNA methylation in the livers and kidneys, as determined by an HpaII-based cytosine extension assay and by HPLC-ES-MS/MS, did not change over the 24-week period. In the livers of male rats, there was a progressive decrease of global and long interspersed nucleotide element-1-associated histone H4 lysine 20 trimethylation, as well as hypermethylation of the p16(INK4A) gene. These epigenetic changes were not observed in the livers of female rats or the kidneys of both sexes. Importantly, morphological evidence of formation and progression of neoplastic process was observed in the liver of male rats only. In conclusion, we have demonstrated that exposure of rats to genotoxic hepatocarcinogen 2-AAF, in addition to formation of 2-AAF-specific DNA lesions, resulted in substantial alterations in cellular epigenetic status.


Subject(s)
2-Acetylaminofluorene/toxicity , Carcinogens/toxicity , Epigenesis, Genetic , Liver Neoplasms, Experimental/genetics , Precancerous Conditions/genetics , Animals , Chromatography, High Pressure Liquid , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Adducts/metabolism , Female , Immunohistochemistry , Kidney/metabolism , Liver/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Male , Polymerase Chain Reaction , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...