Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
J Vis Exp ; (151)2019 09 16.
Article in English | MEDLINE | ID: mdl-31566610

ABSTRACT

Fragile X syndrome (FXS) and associated disorders are caused by expansion of the cytosine-guanine-guanine (CGG) trinucleotide repeat in the 5' untranslated region (UTR) of the Fragile X mental retardation-1 (FMR1) gene promoter. Conventionally, capillary electrophoresis fragment analysis on a genetic analyzer is used for the sizing of the CGG repeats of FMR1, but additional Southern blot analysis is required for exact measurement when the repeat number is higher than 200. Here, we present an accurate and robust polymerase chain reaction (PCR)-based method for quantification of the CGG repeats of FMR1. The first step of this test is PCR amplification of the repeat sequences in the 5'UTR of the FMR1 promoter using a Fragile X PCR kit, followed by purification of the PCR products and fragment sizing on a microfluidic capillary electrophoresis instrument, and subsequent interpretation of the number of CGG repeats by referencing standards with known repeats using the analysis software. This PCR-based assay is reproducible and capable of identifying the full range of CGG repeats of FMR1 promoters, including those with a repeat number of more than 200 (classified as full mutation), 55 to 200 (premutation), 46 to 54 (intermediate), and 10 to 45 (normal). It is a cost-effective method that facilitates classification of the FXS and Fragile X-associated disorders with robustness and rapid reporting time.


Subject(s)
Cytosine/physiology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Guanine/physiology , Polymerase Chain Reaction/methods , Trinucleotide Repeats/genetics , Blotting, Southern/methods , Female , Fragile X Syndrome/diagnosis , Genetic Testing/methods , Humans , Male , Mutation/genetics
2.
Gene ; 570(1): 17-24, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26115768

ABSTRACT

The recent discovery of 5-hydroxymethylcytosine (5hmC), an epigenetic modifier and oxidation product of 5-methylcytosine (5mC), has broadened the scope and understanding of neural development and neurodegenerative diseases. By virtue of their functional groups, 5mC and 5hmC exert opposite effects on gene expression; the former is generally associated with gene silencing whereas the latter is mainly involved in up-regulation of gene expression affecting the cellular processes such as differentiation, development, and aging. Although DNA methylation plays an important role in normal neural development and neuroprotection, an altered pathway due to complex interaction with environmental and genetic factors may cause severe neurodegeneration. The levels of 5hmC in brain increase progressively from birth until death, while in patients with neurodegenerative disorders, the levels are found to be highly compromised. This article discusses the recent developments in the area of hydroxymethylation, with particular emphasis on the role of 5hmC in neurodegenerative diseases including Alzheimer's disease, Parkinson's diseases and Huntington's disease. We have also included recent findings on the role of 5hmC in brain tumors (gliomas). Despite compelling evidence on the involvement of 5hmC in neurodegeneration, it is yet to be established whether this epigenetic molecule is the cause or the effect of the onset and progression of neurodegenerative diseases.


Subject(s)
Cytosine/analogs & derivatives , Neurodegenerative Diseases/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Cytosine/physiology , DNA Methylation , Epigenesis, Genetic , Humans
3.
Pharmacol Ther ; 152: 42-53, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25956466

ABSTRACT

Aberrant DNA methylation is a hallmark of many cancers, including the myeloid malignancies acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). The discovery of TET-mediated demethylation of 5-methylcytosine (5mC) and technological advancements in next-generation sequencing have permitted the examination of other cytosine modifications, namely 5-hydroxymethylcytosine (5hmC), in these myeloid malignancies on a genome-wide scale. Due to the prominence of mutations in epigenetic modifiers that can influence cytosine modifications in these disorders, including IDH1/2, TET2, and DNMT3A, many recent studies have evaluated the relative levels, distribution, and functional consequences of cytosine modifications in leukemic cells. Furthermore, several therapies are being used to treat AML and MDS that target various proteins within the cytosine modification pathway in an effort to revert the abnormal epigenetic patterns that contribute to the diseases. In this review, we provide an overview of cytosine modifications and selected technologies currently used to distinguish and analyze these epigenetic marks in the genome. Then, we discuss the role of mutant enzymes, including DNMT3A, TET2, IDH1/2, and the transcription factor, WT1, in disrupting normal patterns of 5mC and 5hmC in AML and MDS. Finally, we describe several therapies, both standard, front-line treatments and new drugs in clinical trials, aimed at inhibiting the proteins that ultimately lead to aberrant cytosine modifications in these diseases.


Subject(s)
Cytosine/physiology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Animals , DNA Methylation/physiology , Epigenomics/methods , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Mutation/physiology
4.
Cell Rep ; 9(5): 1841-1855, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25482556

ABSTRACT

Somatic mutations in IDH1/IDH2 and TET2 result in impaired TET2-mediated conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). The observation that WT1 inactivating mutations anticorrelate with TET2/IDH1/IDH2 mutations in acute myeloid leukemia (AML) led us to hypothesize that WT1 mutations may impact TET2 function. WT1 mutant AML patients have reduced 5hmC levels similar to TET2/IDH1/IDH2 mutant AML. These mutations are characterized by convergent, site-specific alterations in DNA hydroxymethylation, which drive differential gene expression more than alterations in DNA promoter methylation. WT1 overexpression increases global levels of 5hmC, and WT1 silencing reduced 5hmC levels. WT1 physically interacts with TET2 and TET3, and WT1 loss of function results in a similar hematopoietic differentiation phenotype as observed with TET2 deficiency. These data provide a role for WT1 in regulating DNA hydroxymethylation and suggest that TET2 IDH1/IDH2 and WT1 mutations define an AML subtype defined by dysregulated DNA hydroxymethylation.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/physiology , WT1 Proteins/genetics , 5-Methylcytosine/analogs & derivatives , Animals , Cell Differentiation , Cytosine/analogs & derivatives , Cytosine/physiology , Dioxygenases , Enhancer Elements, Genetic , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/metabolism , Mice, Knockout , Mutation , Promoter Regions, Genetic , Protein Binding , Sequence Analysis, DNA
5.
Genome Biol ; 15(9): 456, 2014 Sep 24.
Article in English | MEDLINE | ID: mdl-25248841

ABSTRACT

Current methods for genomic mapping of 5-hydroxymethylcytosine (5hmC) have been limited by either costly sequencing depth, high DNA input, or lack of single-base resolution. We present an approach called Reduced Representation 5-Hydroxymethylcytosine Profiling (RRHP) to map 5hmC sites at single-base resolution by exploiting the use of beta-glucosyltransferase to inhibit enzymatic digestion at the junction where adapters are ligated to a genomic library. Therefore, only library fragments presenting glucosylated 5hmC residues at the junction are sequenced. RRHP can detect sites with low 5hmC abundance, and when combined with RRBS data, 5-methylcytosine and 5-hydroxymethylcytosine can be compared at a specific site.


Subject(s)
Cytosine/analogs & derivatives , 5-Methylcytosine/analogs & derivatives , Cytosine/physiology , DNA Methylation , Gene Library , High-Throughput Nucleotide Sequencing , Humans , Liver Neoplasms/genetics , Molecular Sequence Annotation , Sequence Analysis, DNA
6.
Exp Hematol ; 42(8): 609-17, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24993071

ABSTRACT

DNA methylation is a well-studied epigenetic modification essential for efficient cellular differentiation. Aberrant DNA methylation patterns are a characteristic feature of cancer, including myeloid malignancies such as acute myeloid leukemia. Recurrent mutations in DNA-modifying enzymes were identified in acute myeloid leukemia and linked to distinct DNA methylation signatures. In addition, discovery of Tet enzymes provided new mechanisms for the reversal of DNA methylation. Advances in base-resolution profiling of DNA methylation have enabled a more comprehensive understanding of the methylome landscape in the genome. This review will summarize and discuss the key questions in the function of DNA methylation in the hematopoietic system, including where and how DNA methylation regulates diverse biological processes in the genome as elucidated by recent studies.


Subject(s)
DNA Methylation , Hematopoietic Stem Cells/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , CpG Islands , Cytosine/analogs & derivatives , Cytosine/physiology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Enhancer Elements, Genetic , Hematopoiesis , Mice , Promoter Regions, Genetic , Repetitive Sequences, Nucleic Acid
7.
Neurobiol Aging ; 35(6): 1334-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24387984

ABSTRACT

DNA methylation (5-methylcytosine [5mC]) is one of several epigenetic markers altered in Alzheimer's disease (AD) brain. More recently, attention has been given to DNA hydroxymethylation (5-hydroxymethylcytosine [5hmC]), the oxidized form of 5mC. Whereas 5mC is generally associated with the inhibition of gene expression, 5hmC has been associated with increased gene expression and is involved in cellular processes such as differentiation, development, and aging. Recent findings point toward a role for 5hmC in the development of diseases including AD, potentially opening new pathways for treating AD through correcting methylation and hydroxymethylation alterations. In the present study, levels of 5mC and 5hmC were investigated in the human middle frontal gyrus (MFG) and middle temporal gyrus (MTG) by immunohistochemistry. Immunoreactivity for 5mC and 5hmC were significantly increased in AD MFG (N = 13) and MTG (N = 29) compared with age-matched controls (MFG, N = 13 and MTG, N = 29). Global levels of 5mC and 5hmC positively correlated with each other and with markers of AD including amyloid beta, tau, and ubiquitin loads. Our results showed a global hypermethylation in the AD brain and revealed that levels of 5hmC were also significantly increased in AD MFG and MTG with no apparent influence of gender, age, postmortem delay, or tissue storage time. Using double-fluorescent immunolabeling, we found that in control and AD brains, levels of 5mC and 5hmC were low in astrocytes and microglia but were elevated in neurons. In addition, our colocalization study showed that within the same nuclei, 5mC and 5hmC mostly do not coexist. The present study clearly demonstrates the involvement of 5mC and 5hmC in AD emphasizing the need for future studies determining the exact time frame of these epigenetic changes during the progression of AD pathology.


Subject(s)
5-Methylcytosine/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cytosine/analogs & derivatives , DNA Methylation , Genetic Markers , 5-Methylcytosine/metabolism , Aged , Aged, 80 and over , Astrocytes/metabolism , Cytosine/metabolism , Cytosine/physiology , DNA Methylation/drug effects , DNA Methylation/genetics , Disease Progression , Epigenesis, Genetic/genetics , Female , Gene Expression/genetics , Humans , Male , Microglia/metabolism , Middle Aged , Neurons/metabolism , Parahippocampal Gyrus/metabolism
8.
Med Oncol ; 30(2): 504, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23430443

ABSTRACT

GATA3 is an enriched transcription factor in mammary epithelium. To date, there has been no study on the relationship between microsatellites in the GATA3 gene and breast cancer risk. In this study, we investigated the existence of polymorphisms in the cytosine-thymine (CT) dinucleotide repeat in intron 3 of the GATA3 gene and its association with breast cancer risk. A case-control study of 206 breast cancer patients and 262 controls was conducted in Iranian women. Several different CT repeat alleles of GATA3 were detected in both the patients and controls. The frequencies of 17 and 18 alleles in patients were significantly lower than controls. Our findings demonstrate that women who carry 17-CT (OR = 0.5; p = 0.003) or 18-CT (OR = 0.41, p = 0.02) alleles of GATA3 gene are at lower risk of developing breast cancer. The highest protection against breast cancer was observed with heterozygotes of 16/17 repeats (OR = 0.12, p = 0.02). Also, the presence of the 17-CT allele has a positive relation with estrogen receptor expression. However, we found that the allelic length of GATA3 polymorphisms had no significant effect on the age onset or grade of the disease, as well as the expression of progesterone receptors and HER2.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Dinucleotide Repeats/genetics , GATA3 Transcription Factor/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/diagnosis , Breast Neoplasms/epidemiology , Case-Control Studies , Cytosine/physiology , Female , Humans , Iran/epidemiology , Middle Aged , Population Surveillance/methods , Risk Factors , Thymine/physiology
9.
Sheng Li Ke Xue Jin Zhan ; 43(3): 164-70, 2012 Jun.
Article in Chinese | MEDLINE | ID: mdl-23019917

ABSTRACT

Recently, 5-hydroxymethyl cytosine (5-hmC) has been discovered as a naturally existed component of normal mammalian genomic DNAs, and it is generally accepted as the sixth base in the genome. This review will introduce the recent advances in the researches on 5-hmC of its formation, tissue-specific distribution, the roles in cell differentiation and gene expression regulation, and the connections as a epigenetic marker with diseases, such as various cancers. We also summarized the current development of the methodologies to detect methylated or hydroxymetholated cytosines of cells at the genomic levels.


Subject(s)
Cytosine/analogs & derivatives , DNA/genetics , Epigenesis, Genetic/genetics , Genome/genetics , 5-Methylcytosine/analogs & derivatives , Animals , Biomarkers , Cell Differentiation/physiology , Cytosine/chemistry , Cytosine/physiology , Gene Expression Regulation , Humans , Methylation
10.
Curr Alzheimer Res ; 9(5): 545-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22272626

ABSTRACT

Epigenetic modifications have been proposed to underlie age-related dysfunction and associated disorders. 5- hydroxymethylcytosine (5-hmC) is a newly described epigenetic modification. It is generated by the oxidation of 5- methylcytosine (5-mC) by the ten-eleven translocation (TET) family of enzymes. Various studies have shown that 5-hmC is present in high levels in the brain. Its lower affinity to methyl-binding proteins as compared to 5-mC suggests that it might have a different role in the regulation of gene expression, while it is also implicated in the DNA demethylation process. Interestingly, various widely used methods for DNA methylation detection fail to discriminate between 5-hmC and 5-mC, while numerous specific techniques are currently being developed. Recent studies have indicated an increase of 5-hmC with age in the mouse brain as well as an age- and gene-expression-level-related enrichment of 5-hmC in genes implicated in neurodegeneration. These findings suggest that 5-hmC may play an important role in the etiology and course of age-related neurodegenerative disorders. The present perspective summarizes the current knowledge on 5-hmC, discusses methodological challenges related to its detection, and suggests future strategies for examining the functional role of this epigenetic modification and its possible implication in aging and Alzheimer's disease.


Subject(s)
Aging/physiology , Alzheimer Disease/metabolism , Cytosine/analogs & derivatives , DNA Methylation/physiology , Translocation, Genetic/physiology , 5-Methylcytosine/analogs & derivatives , Alzheimer Disease/physiopathology , Animals , Cytosine/physiology , Disease Models, Animal , Epigenesis, Genetic , Humans
11.
Mol Syst Biol ; 7: 562, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22186736

ABSTRACT

The discovery of the Ten-Eleven-Translocation (TET) oxygenases that catalyze the hydroxylation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) has triggered an avalanche of studies aiming to resolve the role of 5hmC in gene regulation if any. Hitherto, TET1 is reported to bind to CpG-island (CGI) and bivalent promoters in mouse embryonic stem cells, whereas binding at DNAseI hypersensitive sites (HS) had escaped previous analysis. Significant enrichment/accumulation of 5hmC but not 5mC can indeed be detected at bivalent promoters and at DNaseI-HS. Surprisingly, however, 5hmC is not detected or present at very low levels at CGI promoters notwithstanding the presence of TET1. Our meta-analysis of DNA methylation profiling points to potential issues with regard to the various methodologies that are part of the toolbox used to detect 5mC and 5hmC. Discrepancies between published studies and technical limitations prevent an unambiguous assignment of 5hmC as a 'true' epigenetic mark, that is, read and interpreted by other factors and/or as a transiently accumulating intermediary product of the conversion of 5mC to unmodified cytosines.


Subject(s)
Cytosine/analogs & derivatives , DNA Methylation , Gene Expression Regulation , 5-Methylcytosine/physiology , Animals , Cell Line , Chromatin , Cluster Analysis , Cytosine/physiology , Embryonic Stem Cells , Epigenesis, Genetic , Humans , Mice , Oxygenases , Transcription, Genetic , Translocation, Genetic
12.
Semin Cell Dev Biol ; 21(2): 238-46, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19892028

ABSTRACT

Symmetrical methylation of cytosine residues at CpG dinucleotides of the DNA molecule is a central epigenetic and heritable hallmark of the genome. This epigenetic modification of DNA is directly associated with a closed molecular conformation of the chromatin fibre which is, in turn, intrinsically linked to an inactive transcriptional status. Thus, DNA methylation is a major determinant of the functional outcome of the nucleus. Equally important, DNA methylation is also involved in the large-scale maintenance of the nuclear architecture, which is required for proper nuclear function. Densely DNA methylated regions tend to occupy large and discrete regions of the genome and can act as referential structural blocks for building up the whole functional organization of the nucleus. In this context, interpreting the three-dimensional pattern of DNA methylation is crucial to our understanding of the dynamic biology of genomes.


Subject(s)
Cell Nucleus/physiology , DNA Methylation/physiology , Cell Nucleus/genetics , Cytosine/physiology , Epigenesis, Genetic , Humans
13.
Mol Pharmacol ; 75(2): 342-54, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19029285

ABSTRACT

The epigenetic down-regulation of genes is emerging as a possible underlying mechanism of the GABAergic neuron dysfunction in schizophrenia. For example, evidence has been presented to show that the promoters associated with reelin and GAD67 are down-regulated as a consequence of DNA methyltransferase (DNMT)-mediated hypermethylation. Using neuronal progenitor cells to study this regulation, we have previously demonstrated that DNMT inhibitors coordinately increase reelin and GAD67 mRNAs. Here, we report that another group of epigenetic drugs, histone deacetylase (HDAC) inhibitors, activate these two genes with dose and time dependence comparable with that of DNMT inhibitors. In parallel, both groups of drugs decrease DNMT1, DNMT3A, and DNMT3B protein levels and reduce DNMT enzyme activity. Furthermore, induction of the reelin and GAD67 mRNAs is accompanied by the dissociation of repressor complexes containing all three DNMTs, MeCP2, and HDAC1 from the corresponding promoters and by increased local histone acetylation. Our data imply that drug-induced promoter demethylation is relevant for maximal activation of reelin and GAD67 transcription. The results suggest that HDAC and DNMT inhibitors activate reelin and GAD67 expression through similar mechanisms. Both classes of drugs attenuate, directly or indirectly, the enzymatic and transcriptional repressor activities of DNMTs and HDACs. These data provide a mechanistic rationale for the use of epigenetic drugs, individually or in combination, as a potential novel therapeutic strategy to alleviate deficits associated with schizophrenia.


Subject(s)
Glutamate Decarboxylase/metabolism , Promoter Regions, Genetic/drug effects , Benzamides/pharmacology , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Cytosine/physiology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/physiology , DNA Methyltransferase 3A , Doxorubicin , Epigenesis, Genetic , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression/drug effects , Glutamate Decarboxylase/genetics , Histone Deacetylase Inhibitors , Humans , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/genetics , Pyridines/pharmacology , RNA, Messenger , Reelin Protein , Repressor Proteins/genetics , Repressor Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , DNA Methyltransferase 3B
14.
Pharmacogenomics ; 8(8): 985-96, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17716232

ABSTRACT

Several studies indicate that low thymidylate synthase (TS) protein levels in tumor and normal tissues of colorectal cancer patients are associated with better clinical response to fluorouracil-based chemotherapy and higher risk of toxicity. However, no correlation or even reverse correlation has also been reported. These conflicting results may be partly due to the methodological limitations of the immunohistochemical techniques generally used to quantify thymidylate synthase expression. In this sense, a genetic approach aiming at determining the influence of the TS gene polymorphisms on clinical outcome seems more appealing. So far three polymorphisms have been identified and studied in the TYMS gene: the variable number of 28-bp tandem repeats (2R or 3R) in the 5 UTR; the G>C substitution at the 12th nucleotide in the second repeat of the 3R allele (3RG>3RC) and the 6-bp deletion in the 3 UTR (+6bp/-6bp 3 UTR). In vitro studies indicate that each of these polymorphisms can influence thymidylate synthase expression. In particular, the G>C SNP, which alters the E-box sequence binding an upstream stimulatory factor (USF-1), seems more important than the variable number of tandem repeats in determining TS gene expression in that the 3RC allele has a reduced translational activity compared with the 3RG allele, while showing the same activity as the 2R allele. In contrast with the in vitro findings, the clinical studies in colorectal patients failed to find a consistent relationship between the G>C polymorphism and clinical outcome measures (response, survival or toxicity). This discrepancy may be due to methodological heterogeneities amongst the studies, including genotyping in normal or tumor tissues, loss of heterozygosity in tumor cells not evaluated, variable doses and schedules of fluorouracil-based therapy, and variable tumor stage. The complexity of TYMS gene regulation, and the possibility that other polymorphisms may contribute to fluorouracil response, call for further studies before TYMS genotyping can be used in clinical practice to select colorectal cancer patients who are most likely to benefit from chemotherapy.


Subject(s)
Colorectal Neoplasms/genetics , Cytosine/physiology , Guanine/physiology , Polymorphism, Single Nucleotide/genetics , Thymidylate Synthase/genetics , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Humans
15.
J Hypertens ; 24(6): 1115-24, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16685212

ABSTRACT

CONTEXT: RGS2 (regulators of G-protein signaling) is a negative regulator of Galphaq protein signaling, which mediates the action of several vasoconstrictors. RGS2-deficient mouse line exhibits a hypertensive phenotype and a prolonged response to vasoconstrictors. OBJECTIVE: To compare RGS2 expression in peripheral blood mononuclear cells (PBMs) and cultured fibroblasts from normotensive subjects and hypertensive patients. METHODS: PBMs were isolated from 100 controls and 150 essential hypertensives. Additionally, fibroblasts were isolated from skin biopsy of 11 normotensives and 12 hypertensives and cultured up to the third passage. Quantitative mRNA and protein RGS2 expression were performed by real-time quantitative reverse transcriptase-polymerase chain reaction and by immunoblotting, respectively. Free Ca measurement was performed in monolayers of 24-h serum-deprived cells, using FURA-2 AM. Phosphorylation of the extracellular signal-regulated kinases ERK1/2 was measured by immunoblotting. Polymorphism (C1114G) in the 3' untranslated region of the RGS2 gene was investigated by direct sequencing and real-time polymerase chain reaction (PCR). RESULTS: RGS2 mRNA expression was significantly lower in PBM and in fibroblasts from hypertensives, in comparison to normotensives. C1114G polymorphism was associated with RGS2 expression, with the lowest values in GG hypertensives. The 1114G allele frequency was increased in hypertensives compared with normotensives. Angiotensin II-stimulated intracellular Ca increase and ERK1/2 phosphorylation were higher in fibroblasts from hypertensive patients compared with control subjects, and in those with the G allele, independently of the blood pressure status. The angiotensin II-stimulated Ca mobilization and ERK1/2 phosphorylation were negatively correlated with RGS2 mRNA expression. CONCLUSION: Low expression of RGS2 contributes to increased G-protein-coupled signaling in hypertensive patients. The allele G is associated with low RGS2 expression and blood pressure increase in humans.


Subject(s)
Angiotensin II/physiology , Calcium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypertension/metabolism , RGS Proteins/metabolism , Adult , Cells, Cultured , Cytosine/physiology , Female , Fibroblasts/metabolism , Guanine/physiology , Humans , Hypertension/genetics , Intracellular Fluid/metabolism , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Phosphorylation , Polymorphism, Single Nucleotide , RGS Proteins/genetics
16.
Proc Natl Acad Sci U S A ; 101(39): 14051-6, 2004 Sep 28.
Article in English | MEDLINE | ID: mdl-15381779

ABSTRACT

AlkB repairs 1-alkyladenine and 3-methylcytosine lesions in DNA by directly reversing the base damage. Although repair studies with randomly alkylated substrates have been performed, the miscoding nature of these and related individually alkylated bases and the suppression of mutagenesis by AlkB within cells have not yet been explored. Here, we address the miscoding potential of 1-methyldeoxyadenosine (m1A), 3-methyldeoxycytidine (m3C), 3-ethyldeoxycytidine (e3C), 1-methyldeoxyguanosine (m1G), and 3-methyldeoxythymidine (m3T) by synthesizing single-stranded vectors containing each alkylated base, followed by vector passage through Escherichia coli. In SOS(-), AlkB-deficient cells, m1A was only 1% mutagenic; however, m3C and e3C were 30% mutagenic, rising to 70% in SOS(+) cells. In contrast, the mutagenicity of m1G and m3T in AlkB(-) cells dropped slightly when SOS polymerases were expressed (m1G from 80% to 66% and m3T from 60% to 53%). Mutagenicity was abrogated for m1A, m3C, and e3C in wild-type (AlkB(+)) cells, whereas m3T mutagenicity was only partially reduced. Remarkably, m1G mutagenicity was also eliminated in AlkB(+) cells, establishing it as a natural AlkB substrate. All lesions were blocks to replication in AlkB-deficient cells. The m1A, m3C, and e3C blockades were completely removed in wild-type cells; the m1G blockade was partially removed and that for m3T was unaffected by the presence of AlkB. All lesions demonstrated enhanced bypass when SOS polymerases were induced. This work provides direct evidence that AlkB suppresses both genotoxicity and mutagenesis by physiologically realistic low doses of 1-alkylpurine and 3-alkylpyrimidine DNA damage in vivo.


Subject(s)
Adenine/analogs & derivatives , DNA Damage/physiology , DNA Repair/physiology , Escherichia coli Proteins/physiology , Guanine/analogs & derivatives , Mutagenesis/physiology , Nucleotides/physiology , Thymine/analogs & derivatives , Adenine/physiology , Base Sequence , Cytosine/analogs & derivatives , Cytosine/physiology , DNA Adducts/genetics , DNA Adducts/metabolism , DNA Methylation , DNA Replication/physiology , DNA, Bacterial/physiology , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Escherichia coli/enzymology , Genetic Techniques , Guanine/physiology , SOS Response, Genetics/physiology , Thymine/physiology
17.
Toxicol Sci ; 79(1): 178-88, 2004 May.
Article in English | MEDLINE | ID: mdl-15103049

ABSTRACT

DNA methylation is an epigenetic mechanism regulating patterns of gene expression. Our goal was to see if the assessment of DNA methylation might be a useful tool, when used in conjunction with initial, basic in vitro tests, to provide a more informative preliminary appraisal of the toxic potential of chemicals to prioritize them for further evaluation. We sought to give better indications of a compound's toxic potential and its possible mechanism of action at an earlier time and, thereby, contribute to a rational approach of an overall reduction in testing by making improved early decisions. Global and GC-rich patterns of DNA methylation were evaluated along with more traditional cytolethality measurements, e.g., cytolethality and genotoxicity assessments, on rat hepatoma (H4IIE) cells. The relative toxic potential of model compounds camptothecin, 5-fluorouracil, rotenone, and staurosporine was estimated by employing DNA methylation assessments combined with our cytolethality data plus genotoxicity information gleaned from the literature. The overall contribution of the methylation assessment was threefold; it (1) strengthened a ranking based on genotoxicity; (2) provided an indication that a compound might be more potentially problematic than what cytolethality and genotoxicity assessments alone would indicate; and (3) suggested that compounds, particularly nongenotoxins, that are more potent regarding their ability to alter methylation, especially at noncytolethal concentrations, may be more potentially toxic. Altered methylation per se is not proof of toxicity; this needs to be viewed as a component of an evaluation.


Subject(s)
DNA Methylation/drug effects , Drug Evaluation, Preclinical/methods , Mutagenicity Tests/methods , Animals , Azacitidine/metabolism , Azacitidine/pharmacology , Base Composition/drug effects , Base Composition/genetics , Cell Line, Tumor , Cytosine/chemistry , Cytosine/physiology , Dose-Response Relationship, Drug , Fluorouracil/metabolism , Fluorouracil/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Guanine/chemistry , Guanine/physiology , Mutagenicity Tests/classification , Polymerase Chain Reaction/methods , Rats , Rotenone/metabolism , Rotenone/pharmacology , Staurosporine/metabolism , Staurosporine/pharmacology
19.
Physiol Genomics ; 14(2): 161-6, 2003 Jul 07.
Article in English | MEDLINE | ID: mdl-12783984

ABSTRACT

The associations of the C34T polymorphism of the adenosine monophosphate deaminase 1 (AMPD1) gene with cardiorespiratory phenotypes were tested during cycling exercise at absolute and relative power outputs progressing to exhaustion before and after endurance training for 20 wk in the HERITAGE Family Study cohort (n = 779). Since no blacks were mutant homozygotes (TT), only whites were considered for analysis (400 normal homozygotes, CC; 97 heterozygotes, CT; and 6 TT). For sedentary state, cycling at the absolute power output of 50 W resulted in a higher rating of perceived exertion in TT (P < 0.0001). At the relative intensity of 60% of Vo(2 max), stroke volume was lower in TT (P < 0.05). Maximal values for power output, systolic blood pressure, heart rate, Vco(2), and respiratory exchange ratio were lower in TT (P < 0.05). The cardiorespiratory training response at 50 W and at 60% of Vo(2 max) was similar across C34T-AMPD1 genotypes. However, the maximal values for ventilation, Vo(2), and Vco(2) during exercise increased less in TT (P < 0.01). The results indicate that subjects with the TT genotype at the C34T AMPD1 gene have diminished exercise capacity and cardiorespiratory responses to exercise in the sedentary state. Furthermore, the training response of ventilatory phenotypes during maximal exercise is more limited in TT.


Subject(s)
AMP Deaminase/genetics , Cardiovascular Physiological Phenomena , Cytosine/physiology , Physical Exertion/physiology , Polymorphism, Genetic/genetics , Respiratory Physiological Phenomena , Thymine/physiology , Adolescent , Adult , Aged , Alleles , Bicycling/physiology , Cohort Studies , Female , Genetics, Population , Genotype , Humans , Male , Middle Aged , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiology , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Polymorphism, Genetic/physiology
20.
BMC Mol Biol ; 4: 3, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12659659

ABSTRACT

BACKGROUND: Expansion of an unstable (CGG)n repeat to over 200 triplets within the promoter region of the human FMR1 gene leads to extensive local methylation and transcription silencing, resulting in the loss of FMRP protein and the development of the clinical features of fragile X syndrome. The causative link between (CGG)n expansion, methylation and gene silencing is unknown, although gene silencing is associated with extensive changes to local chromatin architecture. RESULTS: In order to determine the direct effects of increased repeat length on gene transcription in a chromatin context, we have examined the influence of FMR1 (CGG)n repeats upon transcription from the HSV thymidine kinase promoter in the Xenopus laevis oocyte. We observe a reduction in mRNA production directly associated with increasing repeat length, with a 90% reduction in mRNA production from arrays over 100 repeats in length. Using a kinetic approach, we show that this transcriptional repression is concomitant with chromatin maturation and, using in vitro transcription, we show that chromatin formation is a fundamental part of the repressive pathway mediated by (CGG)n repeats. Using Trichostatin A, a histone deacetylase inhibitor, we show reactivation of the silenced promoter. CONCLUSIONS: Thus, isolated fragile X associated (CGG)n repeat arrays can exert a modifying and transcriptionally repressive influence over adjacent promoters and this repressive phenomenon is, in part, mediated by histone deacetylation.


Subject(s)
Cytosine/physiology , Gene Silencing/physiology , Genetic Linkage/genetics , Guanine/physiology , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , Transcription, Genetic/physiology , Trinucleotide Repeat Expansion/physiology , Animals , Chromatin/genetics , Chromatin/metabolism , Chromatin/physiology , Cytosine/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fragile X Syndrome/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Silencing/drug effects , Genes, Reporter/genetics , Genes, Viral/genetics , Guanine/metabolism , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Histone Deacetylase Inhibitors , Histone Deacetylases/physiology , Humans , Hydroxamic Acids/pharmacology , Oocytes , Plasmids/genetics , Thymidine Kinase/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Trinucleotide Repeat Expansion/genetics , Viral Structural Proteins/genetics , Xenopus laevis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...