Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Genome Biol ; 24(1): 264, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38012713

ABSTRACT

BACKGROUND: Common diseases manifest differentially between patients, but the genetic origin of this variation remains unclear. To explore possible involvement of gene transcriptional-variation, we produce a DNA methylation-oriented, driver-gene-wide dataset of regulatory elements in human glioblastomas and study their effect on inter-patient gene expression variation. RESULTS: In 175 of 177 analyzed gene regulatory domains, transcriptional enhancers and silencers are intermixed. Under experimental conditions, DNA methylation induces enhancers to alter their enhancing effects or convert into silencers, while silencers are affected inversely. High-resolution mapping of the association between DNA methylation and gene expression in intact genomes reveals methylation-related regulatory units (average size = 915.1 base-pairs). Upon increased methylation of these units, their target-genes either increased or decreased in expression. Gene-enhancing and silencing units constitute cis-regulatory networks of genes. Mathematical modeling of the networks highlights indicative methylation sites, which signified the effect of key regulatory units, and add up to make the overall transcriptional effect of the network. Methylation variation in these sites effectively describe inter-patient expression variation and, compared with DNA sequence-alterations, appears as a major contributor of gene-expression variation among glioblastoma patients. CONCLUSIONS: We describe complex cis-regulatory networks, which determine gene expression by summing the effects of positive and negative transcriptional inputs. In these networks, DNA methylation induces both enhancing and silencing effects, depending on the context. The revealed mechanism sheds light on the regulatory role of DNA methylation, explains inter-individual gene-expression variation, and opens the way for monitoring the driving forces behind deferential courses of cancer and other diseases.


Subject(s)
DNA Methylation , Regulatory Sequences, Nucleic Acid , Humans , Gene Expression Regulation , Mutation
2.
Genome Biol ; 17(1): 145, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27386949

ABSTRACT

BACKGROUND: Chronic inflammation has been recognized as one of the hallmarks of cancer. We recently showed that parainflammation, a unique variant of inflammation between homeostasis and chronic inflammation, strongly promotes mouse gut tumorigenesis upon p53 loss. Here we explore the prevalence of parainflammation in human cancer and determine its relationship to certain molecular and clinical parameters affecting treatment and prognosis. RESULTS: We generated a transcriptome signature to identify parainflammation in many primary human tumors and carcinoma cell lines as distinct from their normal tissue counterparts and the tumor microenvironment and show that parainflammation-positive tumors are enriched for p53 mutations and associated with poor prognosis. Non-steroidal anti-inflammatory drug (NSAID) treatment suppresses parainflammation in both murine and human cancers, possibly explaining a protective effect of NSAIDs against cancer. CONCLUSIONS: We conclude that parainflammation, a low-grade form of inflammation, is widely prevalent in human cancer, particularly in cancer types commonly harboring p53 mutations. Our data suggest that parainflammation may be a driver for p53 mutagenesis and a guide for cancer prevention by NSAID treatment.


Subject(s)
Carcinogenesis/genetics , Inflammation/genetics , Neoplasms/genetics , Transcriptome/genetics , Tumor Suppressor Protein p53/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Genome, Human , Humans , Inflammation/complications , Inflammation/drug therapy , Mice , Mutation , Neoplasms/complications , Neoplasms/drug therapy
3.
PLoS Genet ; 12(2): e1005840, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26886256

ABSTRACT

Cancers often display gene expression profiles resembling those of undifferentiated cells. The mechanisms controlling these expression programs have yet to be identified. Exploring transcriptional enhancers throughout hematopoietic cell development and derived cancers, we uncovered a novel class of regulatory epigenetic mutations. These epimutations are particularly enriched in a group of enhancers, designated ES-specific enhancers (ESSEs) of the hematopoietic cell lineage. We found that hematopoietic ESSEs are prone to DNA methylation changes, indicative of their chromatin activity states. Strikingly, ESSE methylation is associated with gene transcriptional activity in cancer. Methylated ESSEs are hypermethylated in cancer relative to normal somatic cells and co-localized with silenced genes, whereas unmethylated ESSEs tend to be hypomethylated in cancer and associated with reactivated genes. Constitutive or hematopoietic stem cell-specific enhancers do not show these trends, suggesting selective reactivation of ESSEs in cancer. Further analyses of a hypomethylated ESSE downstream to the VEGFA gene revealed a novel regulatory circuit affecting VEGFA transcript levels across cancers and patients. We suggest that the discovered enhancer sites provide a framework for reactivation of ES genes in cancer.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Human Embryonic Stem Cells/metabolism , Neoplasms/genetics , Cell Line, Tumor , Cell Lineage/genetics , DNA Methylation/genetics , Hematopoiesis/genetics , Humans , Neoplasms/pathology , Polycomb-Group Proteins/metabolism , Reproducibility of Results , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Clin Epigenetics ; 7: 35, 2015.
Article in English | MEDLINE | ID: mdl-25829970

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (T2D) is highly prevalent in Middle-Eastern and North African Arab populations, but the molecular basis for this susceptibility is unknown. Altered DNA methylation levels were reported in insulin-secreting and responding tissues, but whether methylation in accessible tissues such as peripheral blood is associated with T2D risk remains an open question. Age-related alteration of DNA methylation level was reported in certain methylation sites, but no association with T2D has been shown. Here we report on a population-based study of 929 men and women representing the East Jerusalem Palestinian (EJP) Arab population and compare with the findings among Israeli Ashkenazi Jews. This is the first reported epigenetic study of an Arab population with a characteristic high prevalence of T2D. RESULTS: We found that DNA methylation of a prespecified regulatory site in peripheral blood leukocytes (PBLs) is associated with impaired glucose metabolism and T2D independent of sex, body mass index, and white blood cell composition. This CpG site (Chr16: 53,809,231-2; hg19) is located in a region within an intron of the FTO gene, suspected to serve as a tissue-specific enhancer. The association between PBL hypomethylation and T2D varied by age, revealing differential patterns of methylation aging in healthy and diabetic individuals and between ethnic groups: T2D patients displayed prematurely low methylation levels, and this hypomethylation was greater and occurred earlier in life among Palestinian Arabs than Ashkenazi Jews. CONCLUSIONS: Our study suggests that premature DNA methylation aging is associated with increased risk of T2D. These findings should stimulate the search for more such sites and may pave the way to improved T2D risk prediction within and between human populations.

5.
Bioessays ; 36(2): 184-90, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24277586

ABSTRACT

Paradoxically, DNA sequence polymorphisms in cancer risk loci rarely correlate with the expression of cancer genes. Therefore, the molecular mechanism underlying an individual's susceptibility to cancer has remained largely unknown. However, recent evaluations of the correlations between DNA methylation and gene expression levels across healthy and cancerous genomes have revealed enrichment of disease-related DNA methylation variations within disease-associated risk loci. Moreover, it appears that transcriptional enhancers embedded in cancer risk loci often contain DNA methylation sites that closely define the expression of prominent cancer genes, despite the lack of significant correlations between gene expression levels and the surrounding disease-associated polymorphic sequences. We suggest that DNA methylation variations may obscure the effect of co-residing risk sequence alleles. Analysis of enhancer methylation data may help to reveal the regulatory circuits underlying predisposition to cancers and other common diseases.


Subject(s)
DNA Methylation/genetics , Enhancer Elements, Genetic/genetics , Neoplasms/genetics , Animals , Genetic Predisposition to Disease/genetics , Humans , Regulatory Sequences, Nucleic Acid/genetics
6.
Epigenetics Chromatin ; 6(1): 36, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24279375

ABSTRACT

BACKGROUND: Altered DNA methylation patterns represent an attractive mechanism for understanding the phenotypic changes associated with human aging. Several studies have described global and complex age-related methylation changes, but their structural and functional significance has remained largely unclear. RESULTS: We have used transcriptome sequencing to characterize age-related gene expression changes in the human epidermis. The results revealed a significant set of 75 differentially expressed genes with a strong functional relationship to skin homeostasis. We then used whole-genome bisulfite sequencing to identify age-related methylation changes at single-base resolution. Data analysis revealed no global aberrations, but rather highly localized methylation changes, particularly in promoter and enhancer regions that were associated with altered transcriptional activity. CONCLUSIONS: Our results suggest that the core developmental program of human skin is stably maintained through the aging process and that aging is associated with a limited destabilization of the epigenome at gene regulatory elements.

8.
Genome Biol ; 14(3): R21, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23497655

ABSTRACT

BACKGROUND: Abnormal epigenetic marking is well documented in gene promoters of cancer cells, but the study of distal regulatory siteshas lagged behind.We performed a systematic analysis of DNA methylation sites connected with gene expression profilesacross normal and cancerous human genomes. RESULTS: Utilizing methylation and expression data in 58 cell types, we developed a model for methylation-expression relationships in gene promoters and extrapolated it to the genome. We mapped numerous sites at which DNA methylation was associated with expression of distal genes. These sites bind transcription factors in a methylation-dependent manner, and carry the chromatin marks of a particular class of transcriptional enhancers. In contrast to the traditional model of one enhancer site per cell type, we found that single enhancer sites may define gradients of expression levels across many different cell types. Strikingly, the identified sites were drastically altered in cancers: hypomethylated enhancer sites associated with upregulation of cancer-related genes and hypermethylated sites with downregulation. Moreover, the association between enhancer methylation and gene deregulation in cancerwas significantly stronger than the association of promoter methylationwith gene deregulation. CONCLUSIONS: Methylation of distal regulatory sites is closely related to gene expression levels across the genome. Single enhancers may modulate ranges of cell-specific transcription levels, from constantlyopen promoters. In contrast to the remote relationships between promoter methylation and gene dysregulation in cancer, altered methylation of enhancer sites is closely related to gene expression profiles of transformed cells.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Regulatory Sequences, Nucleic Acid/genetics , CpG Islands/genetics , Enhancer Elements, Genetic , Gene Expression Profiling , Humans , Promoter Regions, Genetic , Reproducibility of Results , Transcription, Genetic
9.
Hum Mol Genet ; 21(2): 371-83, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21994764

ABSTRACT

Inter-individual DNA methylation variations were frequently hypothesized to alter individual susceptibility to Type 2 Diabetes Mellitus (T2DM). Sequence-influenced methylations were described in T2DM-associated genomic regions, but evidence for direct, sequence-independent association with disease risk is missing. Here, we explore disease-contributing DNA methylation through a stepwise study design: first, a pool-based, genome-scale screen among 1169 case and control individuals revealed an excess of differentially methylated sites in genomic regions that were previously associated with T2DM through genetic studies. Next, in-depth analyses were performed at selected top-ranking regions. A CpG site in the first intron of the FTO gene showed small (3.35%) but significant (P = 0.000021) hypomethylation of cases relative to controls. The effect was independent of the sequence polymorphism in the region and persists among individuals carrying the sequence-risk alleles. The odds of belonging to the T2DM group increased by 6.1% for every 1% decrease in methylation (OR = 1.061, 95% CI: 1.032-1.090), the odds ratio for decrease of 1 standard deviation of methylation (adjusted to gender) was 1.5856 (95% CI: 1.2824-1.9606) and the sensitivity (area under the curve = 0.638, 95% CI: 0.586-0.690; males = 0.675, females = 0.609) was better than that of the strongest known sequence variant. Furthermore, a prospective study in an independent population cohort revealed significant hypomethylation of young individuals that later progressed to T2DM, relative to the individuals who stayed healthy. Further genomic analysis revealed co-localization with gene enhancers and with binding sites for methylation-sensitive transcriptional regulators. The data showed that low methylation level at the analyzed sites is an early marker of T2DM and suggests a novel mechanism by which early-onset, inter-individual methylation variation at isolated non-promoter genomic sites predisposes to T2DM.


Subject(s)
DNA Methylation , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Adult , Aged , Case-Control Studies , CpG Islands , Diabetes Mellitus, Type 2/blood , Female , Humans , Male , Middle Aged
10.
Hum Mol Genet ; 20(4): 670-80, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21112978

ABSTRACT

Understanding how the epigenetic blueprint of the genome shapes human phenotypes requires systematic evaluation of the complex interplay between gene activity and the different layers of the epigenome. Utilizing microarray-based techniques, we explored the relationships between DNA methylation, DNA replication timing and gene expression levels across a variety of human tissues and cell lines. The analyses revealed unequal methylation levels among early- and late-replicating fractions of the genome: late-replicating DNA was hypomethylated compared with early-replicating DNA. Moreover, late-replicating regions were gradually demethylated with cell divisions, whereas the methylation of early-replicating regions was better maintained. As active genes concentrate at early-replicating regions, they are overall hypermethylated relative to inactive genes. Accordingly, we show that the previously reported positive correlation between gene-body methylation (methylation of the transcribed portion of genes) and gene expression is restricted to proliferative tissues and cell lines, whereas in tissues containing few proliferating cells, active and inactive genes have similar methylation levels. We further show that active gene bodies are hypermethylated not only compared with inactive gene bodies, but also compared with their flanking sequences. This specific hypermethylation of the active gene bodies is severely disrupted in cells of an immunodeficiency, centromeric region instability, facial anomalies (ICF) syndrome patient bearing mutated DNA methyltransferase 3B (DNMT3B). Our data show that a high methylation level is preferentially maintained in active gene bodies through independent cellular processes. Rather than serving as a distinctive mark between active and inactive genes, gene-body methylation appears to serve a vital, currently unknown function in active genes.


Subject(s)
DNA Methylation/genetics , DNA Replication , Gene Expression Regulation , Immunologic Deficiency Syndromes/genetics , Cell Line, Transformed , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenomics , Genome-Wide Association Study , High-Throughput Screening Assays , Humans , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , DNA Methyltransferase 3B
11.
Epigenetics Chromatin ; 3(1): 11, 2010 May 24.
Article in English | MEDLINE | ID: mdl-20497546

ABSTRACT

BACKGROUND: Epigenetic polymorphisms are a potential source of human diversity, but their frequency and relationship to genetic polymorphisms are unclear. DNA methylation, an epigenetic mark that is a covalent modification of the DNA itself, plays an important role in the regulation of gene expression. Most studies of DNA methylation in mammalian cells have focused on CpG methylation present in CpG islands (areas of concentrated CpGs often found near promoters), but there are also interesting patterns of CpG methylation found outside of CpG islands. RESULTS: We compared DNA methylation patterns on both alleles between many pairs (and larger groups) of related and unrelated individuals. Direct observation and simulation experiments revealed that around 10% of common single nucleotide polymorphisms (SNPs) reside in regions with differences in the propensity for local DNA methylation between the two alleles. We further showed that for the most common form of SNP, a polymorphism at a CpG dinucleotide, the presence of the CpG at the SNP positively affected local DNA methylation in cis. CONCLUSIONS: Taken together with the known effect of DNA methylation on mutation rate, our results suggest an interesting interdependence between genetics and epigenetics underlying diversity in the human genome.

12.
Science ; 315(5815): 1141-3, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17322062

ABSTRACT

Differential DNA methylation is important for the epigenetic regulation of gene expression. Allele-specific methylation of the inactive X chromosome has been demonstrated at promoter CpG islands, but the overall pattern of methylation on the active X(Xa) and inactive X (Xi) chromosomes is unknown. We performed allele-specific analysis of more than 1000 informative loci along the human X chromosome. The Xa displays more than two times as much allele-specific methylation as Xi. This methylation is concentrated at gene bodies, affecting multiple neighboring CpGs. Before X inactivation, all of these Xa gene body-methylated sites are biallelically methylated. Thus, a bipartite methylation-demethylation program results in Xa-specific hypomethylation at gene promoters and hypermethylation at gene bodies. These results suggest a relationship between global methylation and expression potentiality.


Subject(s)
Chromosomes, Human, X/genetics , DNA Methylation , Alleles , Cell Line , Chromosomes, Human, X/metabolism , CpG Islands , Embryonic Stem Cells , Epigenesis, Genetic , Female , Gene Expression Regulation , Gene Silencing , Heterozygote , Humans , Male , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , X Chromosome Inactivation
13.
Mol Biol Cell ; 14(9): 3730-40, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12972560

ABSTRACT

Human HT2-19 cells with a conditional cdk1 mutation stop dividing upon cdk1 inactivation and undergo multiple rounds of endoreplication. We show herein that major cell cycle events remain synchronized in these endoreplicating cells. DNA replication alternates with gap phases and cell cycle-specific cyclin E expression is maintained. Centrosomes duplicate in synchrony with chromosome replication, giving rise to polyploid cells with multiple centrosomes. Centrosome migration, a typical prophase event, also takes place in endoreplicating cells. The timing of these events is unaffected by cdk1 inactivation compared with normally dividing cells. Nuclear lamina breakdown, in contrast, previously shown to be dependent on cdk1, does not take place in endoreplicating HT2-19 cells. Moreover, breakdown of all other major components of the nuclear lamina, like the inner nuclear membrane proteins and nuclear pore complexes, seems also to depend on cdk1. Interestingly, the APC/C ubiquitin ligase is activated in these endoreplicating cells by fzr but not by fzy. The oscillations of interphase events are thus independent of cdk1 and of mitosis but may depend on APC/Cfzr activity.


Subject(s)
CDC2 Protein Kinase/physiology , Cell Cycle Proteins/metabolism , Centrosome/physiology , Chromosomes/physiology , Interphase/physiology , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cdc20 Proteins , Cdh1 Proteins , Cells, Cultured , Cyclin A/metabolism , Cyclin A2 , Cyclin B/metabolism , Cyclin B1 , Cyclin E/metabolism , Humans , Mitosis , Mutation , Nuclear Lamina/physiology , Nuclear Pore/physiology , Time Factors , Ubiquitin-Protein Ligase Complexes
14.
Curr Biol ; 12(24): R849-51, 2002 Dec 23.
Article in English | MEDLINE | ID: mdl-12498705

ABSTRACT

Thousands of neurons expressing a given mouse odorant receptor project to a few glomeruli in the olfactory bulb. New observations on mice expressing small odorant receptor transgenes provide support for the idea that interdependence is involved in the maturation of this remarkable convergence.


Subject(s)
Axons/physiology , Mice, Transgenic , Olfactory Receptor Neurons/physiology , Receptors, Odorant/physiology , Animals , Gene Expression Regulation , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Receptors, Odorant/genetics , tau Proteins/genetics , tau Proteins/metabolism
15.
Cancer Cell ; 1(1): 89-97, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12086891

ABSTRACT

Oncogene amplification is an important process in human tumorigenesis, but its underlying mechanism is currently unknown. Cytogenetic analysis indicates that amplification of drug-selected genes in rodent cells is driven by recurrent breaks within chromosomal common fragile sites (CFSs), via the breakage-fusion-bridge (BFB) mechanism. Here we show that BFB cycles drive the intrachromosomal amplification of the MET oncogene in a human gastric carcinoma. Our molecular evidence includes a "ladder-like" structure and inverted repeat organization of the MET amplicons. Furthermore, we show that the breakpoints, setting the centromeric amplicon boundaries, are within the CFS FRA7G region. Upon replication stress, this region showed perturbed chromatin organization, predisposing it to breakage. Thus, in vivo induction of CFSs can play an important role in human oncogenesis.


Subject(s)
Chromosome Fragility/genetics , Gene Amplification/genetics , Oncogenes/genetics , Proto-Oncogene Proteins c-met/genetics , Stomach Neoplasms/genetics , Centromere/physiology , Chromatin/genetics , Chromosome Aberrations , Chromosome Disorders , Chromosome Fragile Sites , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Proto-Oncogene Proteins c-met/metabolism , Repetitive Sequences, Nucleic Acid , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...