Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Nat Commun ; 11(1): 5238, 2020 10 16.
Article in English | MEDLINE | ID: mdl-33067439

ABSTRACT

In heterozygous genomes, allele-specific measurements can reveal biologically significant differences in DNA methylation between homologous alleles associated with local changes in genetic sequence. Current approaches for detecting such events from whole-genome bisulfite sequencing (WGBS) data perform statistically independent marginal analysis at individual cytosine-phosphate-guanine (CpG) sites, thus ignoring correlations in the methylation state, or carry-out a joint statistical analysis of methylation patterns at four CpG sites producing unreliable statistical evidence. Here, we employ the one-dimensional Ising model of statistical physics and develop a method for detecting allele-specific methylation (ASM) events within segments of DNA containing clusters of linked single-nucleotide polymorphisms (SNPs), called haplotypes. Comparisons with existing approaches using simulated and real WGBS data show that our method provides an improved fit to data, especially when considering large haplotypes. Importantly, the method employs robust hypothesis testing for detecting statistically significant imbalances in mean methylation level and methylation entropy, as well as for identifying haplotypes for which the genetic variant carries significant information about the methylation state. As such, our ASM analysis approach can potentially lead to biological discoveries with important implications for the genetics of complex human diseases.


Subject(s)
DNA Methylation , Disease/genetics , Alleles , CpG Islands , Haplotypes , Humans , Polymorphism, Single Nucleotide , Species Specificity , Whole Genome Sequencing
2.
J Intern Med ; 276(1): 5-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24635672

ABSTRACT

The aim of this review is to summarize an evolution of thinking about the epigenetic basis of human cancer, from the earliest studies of altered DNA methylation in cancer to the modern comprehensive epigenomic era. Converging data from epigenetic studies of primary cancers and from experimental studies of chromatin in development and epithelial-mesenchymal transition suggest a role for epigenetic stochasticity as a driving force of cancer, with Darwinian selection of tumour cells at the expense of the host. This increased epigenetic stochasticity appears to be mediated by large-scale changes in DNA methylation and chromatin in domains associated with the nuclear lamina. The implications for diagnosis include the potential to identify stochastically disrupted progenitor cells years before cancer develops, and to target drugs to epigenetic drivers of gene expression instability rather than to mean effects per se.


Subject(s)
Epigenesis, Genetic/genetics , Neoplasms/genetics , DNA Methylation/genetics , Epigenomics/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Stochastic Processes
3.
Mol Psychiatry ; 19(8): 862-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23999529

ABSTRACT

Autism spectrum disorders (ASD) are increasingly common neurodevelopmental disorders defined clinically by a triad of features including impairment in social interaction, impairment in communication in social situations and restricted and repetitive patterns of behavior and interests, with considerable phenotypic heterogeneity among individuals. Although heritability estimates for ASD are high, conventional genetic-based efforts to identify genes involved in ASD have yielded only few reproducible candidate genes that account for only a small proportion of ASDs. There is mounting evidence to suggest environmental and epigenetic factors play a stronger role in the etiology of ASD than previously thought. To begin to understand the contribution of epigenetics to ASD, we have examined DNA methylation (DNAm) in a pilot study of postmortem brain tissue from 19 autism cases and 21 unrelated controls, among three brain regions including dorsolateral prefrontal cortex, temporal cortex and cerebellum. We measured over 485,000 CpG loci across a diverse set of functionally relevant genomic regions using the Infinium HumanMethylation450 BeadChip and identified four genome-wide significant differentially methylated regions (DMRs) using a bump hunting approach and a permutation-based multiple testing correction method. We replicated 3/4 DMRs identified in our genome-wide screen in a different set of samples and across different brain regions. The DMRs identified in this study represent suggestive evidence for commonly altered methylation sites in ASD and provide several promising new candidate genes.


Subject(s)
Autistic Disorder/genetics , Cerebellum/metabolism , DNA Methylation/genetics , Genetic Predisposition to Disease/genetics , Prefrontal Cortex/metabolism , Temporal Lobe/metabolism , Case-Control Studies , Epigenesis, Genetic/genetics , Female , Humans , Male , Pilot Projects
4.
Nature ; 467(7313): 285-90, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20644535

ABSTRACT

Somatic cell nuclear transfer and transcription-factor-based reprogramming revert adult cells to an embryonic state, and yield pluripotent stem cells that can generate all tissues. Through different mechanisms and kinetics, these two reprogramming methods reset genomic methylation, an epigenetic modification of DNA that influences gene expression, leading us to hypothesize that the resulting pluripotent stem cells might have different properties. Here we observe that low-passage induced pluripotent stem cells (iPSCs) derived by factor-based reprogramming of adult murine tissues harbour residual DNA methylation signatures characteristic of their somatic tissue of origin, which favours their differentiation along lineages related to the donor cell, while restricting alternative cell fates. Such an 'epigenetic memory' of the donor tissue could be reset by differentiation and serial reprogramming, or by treatment of iPSCs with chromatin-modifying drugs. In contrast, the differentiation and methylation of nuclear-transfer-derived pluripotent stem cells were more similar to classical embryonic stem cells than were iPSCs. Our data indicate that nuclear transfer is more effective at establishing the ground state of pluripotency than factor-based reprogramming, which can leave an epigenetic memory of the tissue of origin that may influence efforts at directed differentiation for applications in disease modelling or treatment.


Subject(s)
Epigenesis, Genetic , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cellular Reprogramming/genetics , DNA Methylation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Nuclear Transfer Techniques , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Biophys J ; 96(4): L32-4, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19217843

ABSTRACT

Methylation of cytosine is a covalent modification of DNA that can be used to silence genes, orchestrating a myriad of biological processes including cancer. We have discovered that a synthetic nanopore in a membrane comparable in thickness to a protein binding site can be used to detect methylation. We observe a voltage threshold for permeation of methylated DNA through a <2 nm diameter pore, which we attribute to the stretching transition; this can differ by >1 V/20 nm depending on the methylation level, but not the DNA sequence.


Subject(s)
DNA Methylation , DNA/chemistry , DNA/metabolism , Membranes, Artificial , Algorithms , Base Sequence , Electrophoresis, Agar Gel , Genes, BRCA1 , Insulin-Like Growth Factor II/genetics , Permeability , Polymerase Chain Reaction , Protein Conformation
6.
Article in English | MEDLINE | ID: mdl-16869770

ABSTRACT

In over 20 years since the discovery of altered methylation in cancer, many epigenetic alterations have been found in human cancer, including global and specific gene hypomethylation, hypermethylation, altered chromatin marks, and loss of genomic imprinting. Cancer epigenetics has been limited by questions of cause and effect, since epigenetic changes can arise secondary to the cancer process and its associated widespread changes in gene expression. Furthermore, mutations in the DNA methylation machinery have not been observed in tumors, whereas they have been for chromatin modification. To address the issue of human cancer etiology, we have taken a genetic approach to cancer epigenetics. One line of investigation has been on the disorder Beckwith-Wiedemann syndrome (BWS). We have found that loss of imprinting (LOI) of the autocrine growth factor gene IGF2 and of the untranslated antisense RNA LIT1, within the K(V)LQT1 gene, account for most cases of BWS, and that cancer risk is specifically associated with LOI of IGF2. Wilms' tumors, both in BWS and in the general population, involve LOI leading to an expansion of nephrogenic precursor cells. We have also developed an animal model for the role of LOI of IGF2 in cancer, showing that it cooperates with Apc mutations to increase cancer frequency, consistent with human data suggesting a severalfold increased cancer risk for this common epigenetic variant in the adult population. These data suggest that a major component of cancer risk involves epigenetic changes in normal cells that increase the probability of cancer after genetic mutation. They suggest a model of cancer prevention that involves the epigenetic analysis of normal cells for risk stratification and cancer prevention strategies.


Subject(s)
Epigenesis, Genetic , Neoplasms/genetics , Animals , Beckwith-Wiedemann Syndrome/genetics , Colorectal Neoplasms/genetics , Genomic Imprinting , Humans , Insulin-Like Growth Factor II/genetics , Kidney Neoplasms/genetics , Mice , Models, Genetic , Mutation , RNA, Long Noncoding , RNA, Untranslated/genetics , Wilms Tumor/genetics
8.
Gene Ther ; 10(6): 513-22, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12621455

ABSTRACT

Epigenetic control of transcription is essential for mammalian development and its deregulation causes human disease. For example, loss of proper imprinting control at the IGF2-H19 domain is a hallmark of cancer and Beckwith-Wiedemann syndrome, with no targeted therapeutic approaches available. To address this deficiency, we engineered zinc-finger transcription proteins (ZFPs) that specifically activate or repress the IGF2 and H19 genes in a domain-dependent manner. Importantly, we used these ZFPs successfully to reactivate the transcriptionally silent IGF2 and H19 alleles, thus overriding the natural mechanism of imprinting and validating an entirely novel avenue for 'transcription therapy' of human disease.


Subject(s)
Genetic Therapy/methods , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Neoplasms/therapy , Zinc Fingers , Beckwith-Wiedemann Syndrome/therapy , Female , Gene Expression Regulation , Gene Targeting/methods , Genes, Tumor Suppressor , Genetic Engineering , Humans , Kidney Neoplasms/therapy , Male , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Wilms Tumor/therapy
9.
J Natl Cancer Inst ; 93(22): 1698-703, 2001 Nov 21.
Article in English | MEDLINE | ID: mdl-11717330

ABSTRACT

BACKGROUND: Loss of imprinting (LOI) of the insulin-like growth factor-II (IGF2) gene, an epigenetic alteration associated with expression of the normally silent maternal allele, was observed first in Wilms tumor. Although LOI has subsequently been detected in most adult tumors, the biologic role of LOI in cancer remains obscure. We analyzed the imprinting status of Wilms tumors with respect to pathologic subtype, stage, and patient's age at diagnosis and examined the expression of genes potentially affected by LOI. METHODS: Of 60 Wilms tumors examined, 25 were informative for an ApaI polymorphism in the IGF2 gene, allowing analysis of allele-specific gene expression, and could be classified by pathologic subtype. Gene expression was measured quantitatively by real-time polymerase chain reaction, and pathologic analysis was blinded for genetic status. All statistical tests were two-sided. RESULTS: We observed LOI of IGF2 in nine (90%) of 10 Wilms tumors classified as having a pathologic subtype associated with a later stage of renal development and in only one (6.7%) of 15 Wilms tumors with a pathologic subtype associated with an earlier stage of renal development (P< .001). LOI was associated with a 2.2-fold increase (95% confidence interval [CI] = 1.6-fold to 3.1-fold) in IGF2 expression (P< .001). Children whose Wilms tumors displayed LOI of IGF2 were statistically significantly older at diagnosis (median = 65 months; interquartile range [IQR] = 47-83 months) than children whose tumors displayed normal imprinting (median = 24 months; IQR = 13-35 months; P< .001). CONCLUSIONS: These data demonstrate a clear relationship between LOI and altered expression of IGF2 in Wilms tumors and provide a molecular basis for understanding the divergent pathogenesis of this cancer. Analysis of LOI could provide a valuable molecular tool for the classification of Wilms tumor.


Subject(s)
Gene Expression Regulation, Neoplastic , Genomic Imprinting/genetics , Insulin-Like Growth Factor II/genetics , Wilms Tumor/classification , Wilms Tumor/genetics , Age of Onset , Child , Child, Preschool , DNA Mutational Analysis , Genes, Wilms Tumor , Humans , Infant , Kidney/cytology , Kidney/metabolism , Loss of Heterozygosity/genetics , Models, Biological , Polymerase Chain Reaction , Wilms Tumor/pathology
10.
Cancer Res ; 61(13): 4947-50, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431321

ABSTRACT

Loss of imprinting (LOI) is the most common molecular abnormality in Wilms' tumor (WT), other embryonal cancers, and most other tumor types. LOI in WT involves activation of the normally silent maternal allele of the insulin-like growth factor-II (IGF2) gene, silencing of the normally active maternal allele of the H19 gene, and aberrant methylation of a differentially methylated region (DMR) upstream of the maternal copy of H19. Recently, the transcription factor CTCF, which binds to the H19 DMR, has been implicated in the maintenance of H19 and IGF2 imprinting. Here, we show that mutations in the CTCF gene or in the H19 DMR do not occur at significant frequency in WT, nor is there transcriptional silencing of CTCF. We also confirm that methylation of the H19 DMR in WT with LOI includes the CTCF core consensus site. However, some WTs with normal imprinting of IGF2 also show aberrant methylation of CTCF binding sites, indicating that methylation of these sites is necessary but not sufficient for LOI in WT.


Subject(s)
DNA Methylation , DNA-Binding Proteins/genetics , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Repressor Proteins , Transcription Factors/genetics , Wilms Tumor/genetics , Alleles , Base Sequence , Binding Sites , CCCTC-Binding Factor , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mutation , RNA, Long Noncoding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
13.
J Clin Invest ; 106(12): 1447-55, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11120752

ABSTRACT

The KvLQT1 gene encodes a voltage-gated potassium channel. Mutations in KvLQT1 underlie the dominantly transmitted Ward-Romano long QT syndrome, which causes cardiac arrhythmia, and the recessively transmitted Jervell and Lange-Nielsen syndrome, which causes both cardiac arrhythmia and congenital deafness. KvLQT1 is also disrupted by balanced germline chromosomal rearrangements in patients with Beckwith-Wiedemann syndrome (BWS), which causes prenatal overgrowth and cancer. Because of the diverse human disorders and organ systems affected by this gene, we developed an animal model by inactivating the murine Kvlqt1. No electrocardiographic abnormalities were observed. However, homozygous mice exhibited complete deafness, as well as circular movement and repetitive falling, suggesting imbalance. Histochemical study revealed severe anatomic disruption of the cochlear and vestibular end organs, suggesting that Kvlqt1 is essential for normal development of the inner ear. Surprisingly, homozygous mice also displayed threefold enlargement by weight of the stomach resulting from mucous neck cell hyperplasia. Finally, there were no features of BWS, suggesting that Kvlqt1 is not responsible for BWS.


Subject(s)
Deafness/genetics , Hyperplasia/genetics , Long QT Syndrome/genetics , Potassium Channels, Voltage-Gated , Potassium Channels/deficiency , Potassium Channels/metabolism , Stomach/pathology , Animals , Brain Stem/physiology , Cochlea/pathology , Cochlea/physiopathology , Deafness/physiopathology , Disease Models, Animal , Ear, Inner/pathology , Ear, Inner/physiopathology , Electrocardiography , Evoked Potentials, Auditory, Brain Stem , Female , Histocytochemistry , Humans , Hyperplasia/pathology , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Locomotion/physiology , Male , Mice , Mice, Knockout , Mutation/genetics , Organ Size , Phenotype , Potassium Channels/genetics
14.
Genome Res ; 10(11): 1697-710, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11076855

ABSTRACT

A major barrier to conceptual advances in understanding the mechanisms and regulation of imprinting of a genomic region is our relatively poor understanding of the overall organization of genes and of the potentially important cis-acting regulatory sequences that lie in the nonexonic segments that make up 97% of the genome. Interspecies sequence comparison offers an effective approach to identify sequence from conserved functional elements. In this article we describe the successful use of this approach in comparing a approximately 1-Mb imprinted genomic domain on mouse chromosome 7 to its orthologous region on human 11p15.5. Within the region, we identified 112 exons of known genes as well as a novel gene identified uniquely in the mouse region, termed Msuit, that was found to be imprinted. In addition to these coding elements, we identified 33 CpG islands and 49 orthologous nonexonic, nonisland sequences that met our criteria as being conserved, and making up 4.1% of the total sequence. These conserved noncoding sequence elements were generally clustered near imprinted genes and the majority were between Igf2 and H19 or within Kvlqt1. Finally, the location of CpG islands provided evidence that suggested a two-island rule for imprinted genes. This study provides the first global view of the architecture of an entire imprinted domain and provides candidate sequence elements for subsequent functional analyses.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Genomic Imprinting/genetics , Sequence Analysis, DNA , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Chromosomes, Artificial, Bacterial/genetics , Conserved Sequence , Contig Mapping/methods , CpG Islands/genetics , DNA, Complementary/analysis , Female , Humans , Insulin-Like Growth Factor II/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Proteins/genetics , RNA, Long Noncoding , RNA, Messenger/analysis , RNA, Untranslated/genetics , Sequence Analysis, DNA/methods , Species Specificity
16.
Nat Genet ; 25(4): 375-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10932175

ABSTRACT

We have developed a simple, quantitative assay for measurement of allele ratios that circumvents the problem of heteroduplex formation skewing the results of restriction endonuclease digestion of PCR products. This assay, 'hot-stop PCR', involves addition of a radiolabelled PCR primer at the final cycle. We applied the assay to analysis of loss of imprinting (LOI) of the insulin-like growth factor II gene (IGF2) in tumours.


Subject(s)
Alleles , Polymerase Chain Reaction/methods , DNA/genetics , DNA Primers , Genomic Imprinting , Humans , Insulin-Like Growth Factor II/genetics , Phosphorus Radioisotopes
17.
Gene ; 250(1-2): 171-80, 2000 May 30.
Article in English | MEDLINE | ID: mdl-10854790

ABSTRACT

The human BARX2 gene encodes a homeodomain-containing protein of 254 amino acids, which binds optimally to the DNA consensus sequence YYTAATGRTTTTY. BARX2 is highly expressed in adult salivary gland and is expressed at lower levels in other tissues, including mammary gland, kidney, and placenta. The BARX2 gene consists of four exons, and is located on human chromosome 11q25. This chromosomal location is within the minimal deletion region for Jacobsen syndrome, a syndrome including craniosynostosis and other developmental abnormalities. This chromosomal location, along with the reported expression of murine barx2 in craniofacial development, suggests that BARX2 may be causally involved in the craniofacial abnormalities in Jacobsen syndrome.


Subject(s)
Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Amino Acid Sequence , Base Sequence , Binding, Competitive , Chromosome Deletion , Chromosome Mapping , Chromosomes, Human, Pair 11/genetics , Cloning, Molecular/methods , Contig Mapping , Craniofacial Abnormalities/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Oligonucleotides/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tumor Cells, Cultured
19.
Am J Hum Genet ; 66(5): 1473-84, 2000 May.
Article in English | MEDLINE | ID: mdl-10762538

ABSTRACT

The genetics of Beckwith-Wiedemann syndrome (BWS) is complex and is thought to involve multiple genes. It is known that three regions on chromosome 11p15 (BWSCR1, BWSCR2, and BWSCR3) may play a role in the development of BWS. BWSCR2 is defined by two BWS breakpoints. Here we describe the cloning and sequence analysis of 73 kb containing BWSCR2. Within this region, we detected a novel zinc-finger gene, ZNF215. We show that two of its five alternatively spliced transcripts are disrupted by both BWSCR2 breakpoints. Parts of the 3' end of these splice forms are transcribed from the antisense strand of a second zinc-finger gene, ZNF214. We show that ZNF215 is imprinted in a tissue-specific manner.


Subject(s)
Beckwith-Wiedemann Syndrome/genetics , Chromosome Breakage/genetics , Chromosomes, Human, Pair 11/genetics , DNA-Binding Proteins/genetics , Genomic Imprinting/genetics , Zinc Fingers , Alleles , Alternative Splicing/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Cell Line , Cloning, Molecular , Contig Mapping , DNA Mutational Analysis , DNA-Binding Proteins/chemistry , Female , Fetus/metabolism , Humans , Male , Molecular Sequence Data , Organ Specificity , Polymorphism, Single-Stranded Conformational , RNA, Antisense/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics
20.
Cancer Res ; 59(16): 3889-92, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10463576

ABSTRACT

Numerous observations link the loss of imprinting of insulin-like growth factor 2 (IGF2) and an overdosage of this growth factor gene with cancer, in general, and with Wilms' tumorigenesis, in particular. It is not known, however, if loss of imprinting correlates with specific stages of neoplasia or if allelic expression patterns vary within the tumor. By applying an allele-specific in situ hybridization technique to formalin-fixed thin sections, we show that the parental IGF2 alleles can be differentially expressed, not only in Wilms' tumors, but also in nephrogenic rests (which represent premalignant lesions) of Wilms' tumor patients. Moreover, a subpopulation of mesenchymal cells, which surrounds tumor nodules, expresses IGF2 biallelically irrespective of the imprinted state of IGF2 within the tumor. These data show that Wilms' tumorigenesis involves epigenetic heterogeneity as visualized by variable allelic IGF2 expression patterns.


Subject(s)
Alleles , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/genetics , Kidney Neoplasms/genetics , Wilms Tumor/genetics , Genetic Variation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...