Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Immunother Cancer ; 11(2)2023 02.
Article in English | MEDLINE | ID: mdl-36854569

ABSTRACT

BACKGROUND: Approximately one-third of diffuse large B cell lymphoma (DLBCL) patients exhibit co-expression of MYC and BCL2 (double-expressor lymphoma, DEL) and have a dismal prognosis. Targeted inhibition of the anti-apoptotic protein BCL2 with venetoclax (ABT-199) has been approved in multiple B-cell malignancies and is currently being investigated in clinical trials for DLBCL. Whether BCL2 anti-apoptotic function represents a multifaceted vulnerability for DEL-DLBCL, affecting both lymphoma B cells and T cells within the tumor microenvironment, remains to be elucidated. METHODS: Here, we present novel genetically engineered mice that preclinically recapitulate DEL-DLBCL lymphomagenesis, and evaluate their sensitivity ex vivo and in vivo to the promising combination of venetoclax with anti-CD20-based standard immunotherapy. RESULTS: Venetoclax treatment demonstrated specific killing of MYC+/BCL2+ lymphoma cells by licensing their intrinsically primed apoptosis, and showed previously unrecognized immunomodulatory activity by specifically enriching antigen-activated effector CD8 T cells infiltrating the tumors. Whereas DEL-DLBCL mice were refractory to venetoclax alone, inhibition of BCL2 significantly extended overall survival of mice that were simultaneously treated with a murine surrogate for anti-CD20 rituximab. CONCLUSIONS: These results suggest that the combination of anti-CD20-based immunotherapy and BCL2 inhibition leads to cooperative immunomodulatory effects and improved preclinical responses, which may offer promising therapeutic opportunities for DEL-DLBCL patients.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic , Immunotherapy , Lymphoma, Large B-Cell, Diffuse , Animals , Mice , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Disease Models, Animal , Immunotherapy/methods , Lymphoma, Large B-Cell, Diffuse/drug therapy , Proto-Oncogene Proteins c-bcl-2 , Tumor Microenvironment , Proto-Oncogene Proteins c-myc
2.
Leukemia ; 35(5): 1438-1450, 2021 05.
Article in English | MEDLINE | ID: mdl-33597729

ABSTRACT

Multiple myeloma (MM) is an incurable disease, whose clinical heterogeneity makes its management challenging, highlighting the need for biological features to guide improved therapies. Deregulation of specific long non-coding RNAs (lncRNAs) has been shown in MM, nevertheless, the complete lncRNA transcriptome has not yet been elucidated. In this work, we identified 40,511 novel lncRNAs in MM samples. lncRNAs accounted for 82% of the MM transcriptome and were more heterogeneously expressed than coding genes. A total of 10,351 overexpressed and 9,535 downregulated lncRNAs were identified in MM patients when compared with normal bone-marrow plasma cells. Transcriptional dynamics study of lncRNAs in the context of normal B-cell maturation revealed 989 lncRNAs with exclusive expression in MM, among which 89 showed de novo epigenomic activation. Knockdown studies on one of these lncRNAs, SMILO (specific myeloma intergenic long non-coding RNA), resulted in reduced proliferation and induction of apoptosis of MM cells, and activation of the interferon pathway. We also showed that the expression of lncRNAs, together with clinical and genetic risk alterations, stratified MM patients into several progression-free survival and overall survival groups. In summary, our global analysis of the lncRNAs transcriptome reveals the presence of specific lncRNAs associated with the biological and clinical behavior of the disease.


Subject(s)
Multiple Myeloma/genetics , RNA, Long Noncoding/genetics , Transcriptome/genetics , Apoptosis/genetics , Cell Proliferation/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Progression-Free Survival
3.
Blood Adv ; 4(5): 893-905, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32150608

ABSTRACT

Intraclonal subpopulations of circulating chronic lymphocytic leukemia (CLL) cells with different proliferative histories and reciprocal surface expression of CXCR4 and CD5 have been observed in the peripheral blood of CLL patients and named proliferative (PF), intermediate (IF), and resting (RF) cellular fractions. Here, we found that these intraclonal circulating fractions share persistent DNA methylation signatures largely associated with the mutation status of the immunoglobulin heavy chain locus (IGHV) and their origins from distinct stages of differentiation of antigen-experienced B cells. Increased leukemic birth rate, however, showed a very limited impact on DNA methylation of circulating CLL fractions independent of IGHV mutation status. Additionally, DNA methylation heterogeneity increased as leukemic cells advanced from PF to RF in the peripheral blood. This frequently co-occurred with heterochromatin hypomethylation and hypermethylation of Polycomb-repressed regions in the PF, suggesting accumulation of longevity-associated epigenetic features in recently born cells. On the other hand, transcriptional differences between paired intraclonal fractions confirmed their proliferative experience and further supported a linear advancement from PF to RF in the peripheral blood. Several of these differentially expressed genes showed unique associations with clinical outcome not evident in the bulk clone, supporting the pathological and therapeutic relevance of studying intraclonal CLL fractions. We conclude that independent methylation and transcriptional landscapes reflect both preexisting cell-of-origin fingerprints and more recently acquired hallmarks associated with the life cycle of circulating CLL cells.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , B-Lymphocytes , DNA Methylation , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics
4.
World Allergy Organ J ; 12(8): 100047, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31384359

ABSTRACT

BACKGROUND: Small non-coding RNAs (snRNAs) develop important functions related to epigenetic regulation. YRNAs are snRNAs involved in the initiation of DNA replication and RNA stability that regulate gene expression. They have been related to autoimmune, cancer and inflammatory diseases but never before to allergy. In this work we described for the first time in allergic patients the differential expression profile of YRNAs, their regulatory mechanisms and their potential as new diagnostic and therapeutic targets. METHODS: From a previous whole RNAseq study in B cells of allergic patients, differential expression profiles of coding and non-coding transcripts were obtained. To select the most differentially expressed non coding transcripts, fold change and p-values were analyzed. A validation of the expression differences detected was developed in an independent cohort of 304 individuals, 208 allergic patients and 96 controls by using qPCR. Potential binding and retrotransponibility capacity were characterized by in silico structural analysis. Using a novel bioinformatics approach, RNA targets identification, functional enrichment and network analyses were performed. RESULTS: We found that almost 70% of overexpressed non-coding transcripts in allergic patients corresponded to YRNAs. From the three more differentially overexpressed candidates, increased expression was independently confirmed in the peripheral blood of allergic patients. Structural analysis suggested a protein binding capacity decrease and an increase in retrotransponibility. Studies of RNA targets allowed the identification of sequences related to the immune mechanisms underlying allergy. CONCLUSIONS: Overexpression of YRNAs is observed for the first time in allergic patients. Structural and functional information points to their implication on regulatory mechanisms of the disease.

5.
Blood ; 133(22): 2401-2412, 2019 05 30.
Article in English | MEDLINE | ID: mdl-30975638

ABSTRACT

Refractory or relapsed diffuse large B-cell lymphoma (DLBCL) often associates with the activated B-cell-like (ABC) subtype and genetic alterations that drive constitutive NF-κB activation and impair B-cell terminal differentiation. Here, we show that DNA damage response by p53 is a central mechanism suppressing the pathogenic cooperation of IKK2ca-enforced canonical NF-κB and impaired differentiation resulting from Blimp1 loss in ABC-DLBCL lymphomagenesis. We provide evidences that the interplay between these genetic alterations and the tumor microenvironment select for additional molecular addictions that promote lymphoma progression, including aberrant coexpression of FOXP1 and the B-cell mutagenic enzyme activation-induced deaminase, and immune evasion through major histocompatibility complex class II downregulation, PD-L1 upregulation, and T-cell exhaustion. Consistently, PD-1 blockade cooperated with anti-CD20-mediated B-cell cytotoxicity, promoting extended T-cell reactivation and antitumor specificity that improved long-term overall survival in mice. Our data support a pathogenic cooperation among NF-κB-driven prosurvival, genetic instability, and immune evasion mechanisms in DLBCL and provide preclinical proof of concept for including PD-1/PD-L1 blockade in combinatorial immunotherapy for ABC-DLBCL.


Subject(s)
B-Lymphocytes/immunology , B7-H1 Antigen/immunology , Gene Expression Regulation, Neoplastic , Lymphocyte Activation , Lymphoma, Large B-Cell, Diffuse/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Escape , Tumor Suppressor Protein p53/immunology , Animals , B-Lymphocytes/pathology , B7-H1 Antigen/genetics , Female , Humans , Immunotherapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Male , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Suppressor Protein p53/genetics
6.
Haematologica ; 104(8): 1572-1579, 2019 08.
Article in English | MEDLINE | ID: mdl-30655376

ABSTRACT

In this study we interrogated the DNA methylome of myelofibrosis patients using high-density DNA methylation arrays. We detected 35,215 differentially methylated CpG, corresponding to 10,253 genes, between myelofibrosis patients and healthy controls. These changes were present both in primary and secondary myelofibrosis, which showed no differences between them. Remarkably, most differentially methylated CpG were located outside gene promoter regions and showed significant association with enhancer regions. This aberrant enhancer hypermethylation was negatively correlated with the expression of 27 genes in the myelofibrosis cohort. Of these, we focused on the ZFP36L1 gene and validated its decreased expression and enhancer DNA hypermethylation in an independent cohort of patients and myeloid cell-lines. In vitro reporter assay and 5'-azacitidine treatment confirmed the functional relevance of hyper-methylation of ZFP36L1 enhancer. Furthermore, in vitro rescue of ZFP36L1 expression had an impact on cell proliferation and induced apoptosis in SET-2 cell line indicating a possible role of ZFP36L1 as a tumor suppressor gene in myelofibrosis. Collectively, we describe the DNA methylation profile of myelofibrosis, identifying extensive changes in enhancer elements and revealing ZFP36L1 as a novel candidate tumor suppressor gene.


Subject(s)
Butyrate Response Factor 1/genetics , DNA Methylation , Enhancer Elements, Genetic/genetics , Epigenomics/methods , Primary Myelofibrosis/genetics , Apoptosis/drug effects , Butyrate Response Factor 1/metabolism , Butyrate Response Factor 1/pharmacology , Case-Control Studies , Cell Line , Cell Proliferation/drug effects , Epigenesis, Genetic , Genes, Tumor Suppressor , Humans
7.
Nat Genet ; 47(7): 746-56, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26053498

ABSTRACT

We analyzed the DNA methylome of ten subpopulations spanning the entire B cell differentiation program by whole-genome bisulfite sequencing and high-density microarrays. We observed that non-CpG methylation disappeared upon B cell commitment, whereas CpG methylation changed extensively during B cell maturation, showing an accumulative pattern and affecting around 30% of all measured CpG sites. Early differentiation stages mainly displayed enhancer demethylation, which was associated with upregulation of key B cell transcription factors and affected multiple genes involved in B cell biology. Late differentiation stages, in contrast, showed extensive demethylation of heterochromatin and methylation gain at Polycomb-repressed areas, and genes with apparent functional impact in B cells were not affected. This signature, which has previously been linked to aging and cancer, was particularly widespread in mature cells with an extended lifespan. Comparing B cell neoplasms with their normal counterparts, we determined that they frequently acquire methylation changes in regions already undergoing dynamic methylation during normal B cell differentiation.


Subject(s)
B-Lymphocytes/physiology , DNA Methylation , Epigenesis, Genetic/immunology , Base Sequence , Cell Differentiation , Cells, Cultured , CpG Islands , Gene Expression Regulation, Leukemic , Genome, Human , Humans , Leukemia, B-Cell/genetics , Sequence Analysis, DNA
8.
Genome Res ; 25(4): 478-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25644835

ABSTRACT

While analyzing the DNA methylome of multiple myeloma (MM), a plasma cell neoplasm, by whole-genome bisulfite sequencing and high-density arrays, we observed a highly heterogeneous pattern globally characterized by regional DNA hypermethylation embedded in extensive hypomethylation. In contrast to the widely reported DNA hypermethylation of promoter-associated CpG islands (CGIs) in cancer, hypermethylated sites in MM, as opposed to normal plasma cells, were located outside CpG islands and were unexpectedly associated with intronic enhancer regions defined in normal B cells and plasma cells. Both RNA-seq and in vitro reporter assays indicated that enhancer hypermethylation is globally associated with down-regulation of its host genes. ChIP-seq and DNase-seq further revealed that DNA hypermethylation in these regions is related to enhancer decommissioning. Hypermethylated enhancer regions overlapped with binding sites of B cell-specific transcription factors (TFs) and the degree of enhancer methylation inversely correlated with expression levels of these TFs in MM. Furthermore, hypermethylated regions in MM were methylated in stem cells and gradually became demethylated during normal B-cell differentiation, suggesting that MM cells either reacquire epigenetic features of undifferentiated cells or maintain an epigenetic signature of a putative myeloma stem cell progenitor. Overall, we have identified DNA hypermethylation of developmentally regulated enhancers as a new type of epigenetic modification associated with the pathogenesis of MM.


Subject(s)
DNA Methylation/genetics , Enhancer Elements, Genetic/genetics , Multiple Myeloma/genetics , Neoplastic Stem Cells/cytology , Plasma Cells/cytology , Cell Differentiation/genetics , Cell Line, Tumor , CpG Islands/genetics , DNA, Neoplasm/genetics , Down-Regulation/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Promoter Regions, Genetic , Transcription Factors/biosynthesis , Transcription Factors/genetics
10.
Haematologica ; 98(9): 1414-20, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23716560

ABSTRACT

Most DNA methylation studies in classic Philadelphia-negative myeloproliferative neoplasms have been performed on a gene-by-gene basis. Therefore, a more comprehensive methylation profiling is needed to study the implications of this epigenetic marker in myeloproliferative neoplasms. Here, we have analyzed 71 chronic (24 polycythemia vera, 23 essential thrombocythemia and 24 primary myelofibrosis) and 13 transformed myeloproliferative neoplasms using genome-wide DNA methylation arrays. The three types of chronic Philadelphia-negative myeloproliferative neoplasms showed a similar aberrant DNA methylation pattern when compared to control samples. Differentially methylated regions were enriched in a gene network centered on the NF-κB pathway, indicating that they may be involved in the pathogenesis of these diseases. In the case of transformed myeloproliferative neoplasms, we detected an increased number of differentially methylated regions with respect to chronic myeloproliferative neoplasms. Interestingly, these genes were enriched in a list of differentially methylated regions in primary acute myeloid leukemia and in a gene network centered around the IFN pathway. Our results suggest that alterations in the DNA methylation landscape play an important role in the pathogenesis and leukemic transformation of myeloproliferative neoplasms. The therapeutic modulation of epigenetically-deregulated pathways may allow us to design targeted therapies for these patients.


Subject(s)
DNA Methylation/genetics , Gene Expression Profiling/methods , Gene Regulatory Networks/genetics , Polycythemia Vera/genetics , Primary Myelofibrosis/genetics , Chronic Disease , Humans , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Polycythemia Vera/diagnosis , Primary Myelofibrosis/diagnosis
11.
Epigenetics ; 6(9): 1131-7, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21975512

ABSTRACT

Although there is no doubt about the influence of the genetic background in the onset of the allergic diseases, Epigenome-Wide Association Studies are needed to elucidate the possible relationship between allergic diseases and epigenomic dysregulation. In this study we aimed to analyze the epigenetic patterns, in terms of DNA methylation, of three well-characterized populations of house dust mite allergic subjects, aspirin-intolerant asthmatics and controls. As a first, genome-wide phase, we used the HELP assay to study the methylation patterns in CD19 (+) B lymphocytes in these populations, and found that there are reproducible epigenetic differences at limited numbers of loci distinguishing the groups, corroborated by bisulphite MassArray in a second validation phase of an expanded 40 subject group. These validated epigenetic changes occur at loci characterized as important for the immune response. One such locus is a new candidate gene, CYP26A1, which shows differential methylation patterns and expression levels between groups. Our results suggest that epigenomic dysregulation may contribute to the susceptibility to allergic diseases, showing for the first time differences in DNA methylation between allergic and non-allergic healthy subjects, both globally and at specific loci. These observations indicate that the epigenome may offer new pathophysiological insights and therapeutic targets in atopic diseases.


Subject(s)
Asthma/genetics , B-Lymphocytes/pathology , Epigenesis, Genetic , Pyroglyphidae/immunology , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, CD19/metabolism , Aspirin/adverse effects , Aspirin/pharmacology , Asthma/immunology , Asthma/physiopathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Case-Control Studies , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , DNA Methylation , Disease Susceptibility/immunology , Flow Cytometry , Genetic Loci , Humans , Oligonucleotide Array Sequence Analysis/methods , Promoter Regions, Genetic , Retinoic Acid 4-Hydroxylase
12.
Genome Res ; 21(11): 1833-40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21957152

ABSTRACT

Heterochromatin is believed to be associated with increased levels of cytosine methylation. With the recent availability of genome-wide, high-resolution molecular data reflecting chromatin organization and methylation, such relationships can be explored systematically. As well-defined surrogates for heterochromatin, we tested the relationship between DNA replication timing and DNase hypersensitivity with cytosine methylation in two human cell types, unexpectedly finding the later-replicating, more heterochromatic regions to be less methylated than early replicating regions. When we integrated gene-expression data into the study, we found that regions of increased gene expression were earlier replicating, as previously identified, and that transcription-targeted cytosine methylation in gene bodies contributes to the positive correlation with early replication. A self-organizing map (SOM) approach was able to identify genomic regions with early replication and increased methylation, but lacking annotated transcripts, loci missed in simple two variable analyses, possibly encoding unrecognized intergenic transcripts. We conclude that the relationship of cytosine methylation with heterochromatin is not simple and depends on whether the genomic context is tandemly repetitive sequences often found near centromeres, which are known to be heterochromatic and methylated, or the remaining majority of the genome, where cytosine methylation is targeted preferentially to the transcriptionally active, euchromatic compartment of the genome.


Subject(s)
Cytosine/metabolism , DNA Methylation , DNA Replication , Genome, Human , Heterochromatin/metabolism , Cell Line , Gene Expression Profiling , Gene Expression Regulation , Humans , Time Factors , Transcription, Genetic
13.
Mol Cell Biol ; 30(17): 4159-74, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20584988

ABSTRACT

Previous studies have shown that tumor progression in the transgenic adenocarcinoma of mouse prostate (TRAMP) model is characterized by global DNA hypomethylation initiated during early-stage disease and locus-specific DNA hypermethylation occurring predominantly in late-stage disease. Here, we utilized Dnmt1 hypomorphic alleles to examine the role of Dnmt1 in normal prostate development and in prostate cancer in TRAMP. Prostate tissue morphology and differentiation status was normal in Dnmt1 hypomorphic mice, despite global DNA hypomethylation. TRAMP; Dnmt1 hypomorphic mice also displayed global DNA hypomethylation, but were characterized by altered tumor phenotype. Specifically, TRAMP; Dnmt1 hypomorphic mice exhibited slightly increased tumor incidence and significantly increased pathological progression at early ages and, conversely, displayed slightly decreased tumor incidence and significantly decreased pathological progression at advanced ages. Remarkably, hypomorphic Dnmt1 expression abrogated local and distant site macrometastases. Thus, Dnmt1 has tumor suppressor activity in early-stage prostate cancer, and oncogenic activity in late stage prostate cancer and metastasis. Consistent with the biological phenotype, epigenomic studies revealed that TRAMP; Dnmt1 hypomorphic mice show dramatically reduced CpG island and promoter DNA hypermethylation in late-stage primary tumors compared to control mice. Taken together, the data reveal a crucial role for Dnmt1 in prostate cancer and suggest that Dnmt1-targeted interventions may have utility specifically for advanced and/or metastatic prostate cancer.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , Alleles , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/genetics , Prostatic Neoplasms/secondary
14.
Front Biosci (Schol Ed) ; 2(3): 815-24, 2010 06 01.
Article in English | MEDLINE | ID: mdl-20515827

ABSTRACT

Several studies have proven the important influence that environmental exposures have in the individual's susceptibility to suffer allergy and other related diseases, mostly during embryonic or early life. Although the relationship between the environment and allergic diseases had been previously reported, one interesting attempt to describe this relationship was Strachan's hygiene hypothesis, proposed almost two decades ago. Since then, several studies have identified new environmental factors related to an increased risk of allergic disease. In this context, epigenetic modifications appear as a possible link between the environment and genome, providing a plausible mechanism for explaining how the recent changes in lifestyle can modify gene expression, and thus, lead to a disease state. Here, we will focus on the environmental modifiers that have been described and the possible role of epigenetic modifications.


Subject(s)
Epigenesis, Genetic , Hypersensitivity/genetics , Animals , DNA Methylation , Diet/adverse effects , Environment , Female , Genetic Predisposition to Disease , Humans , Hypersensitivity/etiology , Hypersensitivity/immunology , Hypersensitivity, Immediate/etiology , Hypersensitivity, Immediate/genetics , Hypersensitivity, Immediate/immunology , Life Style , Male , Pregnancy , Risk Factors , Sex Characteristics , Th1 Cells/immunology , Th2 Cells/immunology , Tobacco Smoke Pollution/adverse effects
15.
Genome Biol ; 11(4): R36, 2010.
Article in English | MEDLINE | ID: mdl-20359321

ABSTRACT

Using the type III restriction-modification enzyme EcoP15I, we isolated sequences flanking sites digested by the methylation-sensitive HpaII enzyme or its methylation-insensitive MspI isoschizomer for massively parallel sequencing. A novel data transformation allows us to normalise HpaII by MspI counts, resulting in more accurate quantification of methylation at >1.8 million loci in the human genome. This HELP-tagging assay is not sensitive to sequence polymorphism or base composition and allows exploration of both CG-rich and depleted genomic contexts.


Subject(s)
Cytosine/metabolism , Sequence Analysis, DNA , Base Sequence , Cells, Cultured , Cytosine/chemistry , DNA Methylation , DNA-Cytosine Methylases/chemistry , DNA-Cytosine Methylases/metabolism , Genome, Human , Humans , Polymorphism, Genetic , Site-Specific DNA-Methyltransferase (Adenine-Specific)/chemistry , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism
16.
Cancer Prev Res (Phila) ; 2(12): 1065-75, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19934341

ABSTRACT

Green tea polyphenols (GTP) have been reported to inhibit DNA methylation in cultured cells. Here, we tested whether oral consumption of GTPs affects normal or cancer-specific DNA methylation in vivo, using mice. Wild-type (WT) and transgenic adenocarcinoma of mouse prostate (TRAMP) mice were given 0.3% GTPs in drinking water beginning at 4 weeks of age. To monitor DNA methylation, we measured 5-methyl-deoxycytidine (5mdC) levels, methylation of the B1 repetitive element, and methylation of the Mage-a8 gene. Each of these parameters were unchanged in prostate, gut, and liver from WT mice at both 12 and 24 weeks of age, with the single exception of a decrease of 5mdC in the liver at 12 weeks. In GTP-treated TRAMP mice, 5mdC levels and the methylation status of four loci hypermethylated during tumor progression were unaltered in TRAMP prostates at 12 or 24 weeks. Quite surprisingly, GTP treatment did not inhibit tumor progression in TRAMP mice, although known pharmacodynamic markers of GTPs were altered in both WT and TRAMP prostates. We also administered 0.1%, 0.3%, or 0.6% GTPs to TRAMP mice for 12 weeks and measured 5mdC levels and methylation of B1 and Mage-a8 in prostate, gut, and liver tissues. No dose-dependent alterations in DNA methylation status were observed. Genome-wide DNA methylation profiling using the HpaII tiny fragment enrichment by ligation-mediated PCR assay also revealed no significant hypomethylating effect of GTP. These data indicate that oral administration of GTPs does not affect normal or cancer-specific DNA methylation in the murine prostate.


Subject(s)
Adenocarcinoma/genetics , DNA Methylation , Flavonoids/pharmacology , Phenols/pharmacology , Prostate/pathology , Prostatic Neoplasms/genetics , Tea , Adenocarcinoma/pathology , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/physiology , Flavonoids/pharmacokinetics , Guanosine Triphosphate/metabolism , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenols/pharmacokinetics , Polyphenols , Prostate/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...