Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 33(2): 184-196, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36577521

RESUMEN

Short tandem repeats (STRs) contribute significantly to genetic diversity in humans, including disease-causing variation. Although the effect of STR variation on gene expression has been extensively assessed, their impact on epigenetics has been poorly studied and limited to specific genomic regions. Here, we investigated the hypothesis that some STRs act as independent regulators of local DNA methylation in the human genome and modify risk of common human traits. To address these questions, we first analyzed two independent data sets comprising PCR-free whole-genome sequencing (WGS) and genome-wide DNA methylation levels derived from whole-blood samples in 245 (discovery cohort) and 484 individuals (replication cohort). Using genotypes for 131,635 polymorphic STRs derived from WGS using HipSTR, we identified 11,870 STRs that associated with DNA methylation levels (mSTRs) of 11,774 CpGs (Bonferroni P < 0.001) in our discovery cohort, with 90% successfully replicating in our second cohort. Subsequently, through fine-mapping using CAVIAR we defined 585 of these mSTRs as the likely causal variants underlying the observed associations (fm-mSTRs) and linked a fraction of these to previously reported genome-wide association study signals, providing insights into the mechanisms underlying complex human traits. Furthermore, by integrating gene expression data, we observed that 12.5% of the tested fm-mSTRs also modulate expression levels of nearby genes, reinforcing their regulatory potential. Overall, our findings expand the catalog of functional sequence variants that affect genome regulation, highlighting the importance of incorporating STRs in future genetic association analysis and epigenetics data for the interpretation of trait-associated variants.


Asunto(s)
Metilación de ADN , Estudio de Asociación del Genoma Completo , Humanos , Repeticiones de Microsatélite , Genoma Humano , Genotipo
2.
Am J Hum Genet ; 109(6): 1065-1076, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35609568

RESUMEN

The human genome contains tens of thousands of large tandem repeats and hundreds of genes that show common and highly variable copy-number changes. Due to their large size and repetitive nature, these variable number tandem repeats (VNTRs) and multicopy genes are generally recalcitrant to standard genotyping approaches and, as a result, this class of variation is poorly characterized. However, several recent studies have demonstrated that copy-number variation of VNTRs can modify local gene expression, epigenetics, and human traits, indicating that many have a functional role. Here, using read depth from whole-genome sequencing to profile copy number, we report results of a phenome-wide association study (PheWAS) of VNTRs and multicopy genes in a discovery cohort of ∼35,000 samples, identifying 32 traits associated with copy number of 38 VNTRs and multicopy genes at 1% FDR. We replicated many of these signals in an independent cohort and observed that VNTRs showing trait associations were significantly enriched for expression QTLs with nearby genes, providing strong support for our results. Fine-mapping studies indicated that in the majority (∼90%) of cases, the VNTRs and multicopy genes we identified represent the causal variants underlying the observed associations. Furthermore, several lie in regions where prior SNV-based GWASs have failed to identify any significant associations with these traits. Our study indicates that copy number of VNTRs and multicopy genes contributes to diverse human traits and suggests that complex structural variants potentially explain some of the so-called "missing heritability" of SNV-based GWASs.


Asunto(s)
Variaciones en el Número de Copia de ADN , Repeticiones de Minisatélite , Variaciones en el Número de Copia de ADN/genética , Genoma Humano , Estudio de Asociación del Genoma Completo , Humanos , Repeticiones de Minisatélite/genética , Fenotipo
3.
Am J Hum Genet ; 108(5): 809-824, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33794196

RESUMEN

Variable number tandem repeats (VNTRs) are composed of large tandemly repeated motifs, many of which are highly polymorphic in copy number. However, because of their large size and repetitive nature, they remain poorly studied. To investigate the regulatory potential of VNTRs, we used read-depth data from Illumina whole-genome sequencing to perform association analysis between copy number of ∼70,000 VNTRs (motif size ≥ 10 bp) with both gene expression (404 samples in 48 tissues) and DNA methylation (235 samples in peripheral blood), identifying thousands of VNTRs that are associated with local gene expression (eVNTRs) and DNA methylation levels (mVNTRs). Using an independent cohort, we validated 73%-80% of signals observed in the two discovery cohorts, while allelic analysis of VNTR length and CpG methylation in 30 Oxford Nanopore genomes gave additional support for mVNTR loci, thus providing robust evidence to support that these represent genuine associations. Further, conditional analysis indicated that many eVNTRs and mVNTRs act as QTLs independently of other local variation. We also observed strong enrichments of eVNTRs and mVNTRs for regulatory features such as enhancers and promoters. Using the Human Genome Diversity Panel, we define sets of VNTRs that show highly divergent copy numbers among human populations and show that these are enriched for regulatory effects and preferentially associate with genes that have been linked with human phenotypes through GWASs. Our study provides strong evidence supporting functional variation at thousands of VNTRs and defines candidate sets of VNTRs, copy number variation of which potentially plays a role in numerous human phenotypes.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Metilación de ADN , Regulación de la Expresión Génica , Repeticiones de Minisatélite/genética , Sitios de Carácter Cuantitativo/genética , Adolescente , Adulto , Algoritmos , Niño , Preescolar , Cromosomas Humanos X/genética , Estudios de Cohortes , Islas de CpG/genética , Elementos de Facilitación Genéticos/genética , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Fenotipo , Regiones Promotoras Genéticas/genética , Adulto Joven
4.
BMC Genomics ; 24(1): 226, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37127568

RESUMEN

Open reading frames (ORFs) with fewer than 100 codons are generally not annotated in genomes, although bona fide genes of that size are known. Newer biochemical studies have suggested that thousands of small protein-coding ORFs (smORFs) may exist in the human genome, but the true number and the biological significance of the micropeptides they encode remain uncertain. Here, we used a comparative genomics approach to identify high-confidence smORFs that are likely protein-coding. We identified 3,326 high-confidence smORFs using constraint within human populations and evolutionary conservation as additional lines of evidence. Next, we validated that, as a group, our high-confidence smORFs are conserved at the amino-acid level rather than merely residing in highly conserved non-coding regions. Finally, we found that high-confidence smORFs are enriched among disease-associated variants from GWAS. Overall, our results highlight that smORF-encoded peptides likely have important functional roles in human disease.


Asunto(s)
Péptidos , Proteínas , Humanos , Sistemas de Lectura Abierta , Proteínas/genética , Péptidos/genética , Genoma Humano , Micropéptidos
5.
Am J Hum Genet ; 107(4): 654-669, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32937144

RESUMEN

There is growing recognition that epivariations, most often recognized as promoter hypermethylation events that lead to gene silencing, are associated with a number of human diseases. However, little information exists on the prevalence and distribution of rare epigenetic variation in the human population. In order to address this, we performed a survey of methylation profiles from 23,116 individuals using the Illumina 450k array. Using a robust outlier approach, we identified 4,452 unique autosomal epivariations, including potentially inactivating promoter methylation events at 384 genes linked to human disease. For example, we observed promoter hypermethylation of BRCA1 and LDLR at population frequencies of ∼1 in 3,000 and ∼1 in 6,000, respectively, suggesting that epivariations may underlie a fraction of human disease which would be missed by purely sequence-based approaches. Using expression data, we confirmed that many epivariations are associated with outlier gene expression. Analysis of variation data and monozygous twin pairs suggests that approximately two-thirds of epivariations segregate in the population secondary to underlying sequence mutations, while one-third are likely sporadic events that occur post-zygotically. We identified 25 loci where rare hypermethylation coincided with the presence of an unstable CGG tandem repeat, validated the presence of CGG expansions at several loci, and identified the putative molecular defect underlying most of the known folate-sensitive fragile sites in the genome. Our study provides a catalog of rare epigenetic changes in the human genome, gives insight into the underlying origins and consequences of epivariations, and identifies many hypermethylated CGG repeat expansions.


Asunto(s)
Proteína BRCA1/genética , Epigénesis Genética , Enfermedades Genéticas Congénitas/genética , Genoma Humano , Receptores de LDL/genética , Expansión de Repetición de Trinucleótido , Proteína BRCA1/metabolismo , Metilación de ADN , Femenino , Ácido Fólico/metabolismo , Silenciador del Gen , Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/patología , Sitios Genéticos , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Regiones Promotoras Genéticas , Receptores de LDL/metabolismo , Gemelos Monocigóticos
6.
Am J Hum Genet ; 107(3): 555-563, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32758449

RESUMEN

Helsmoortel-Van der Aa syndrome (HVDAS) is a neurodevelopmental condition associated with intellectual disability/developmental delay, autism spectrum disorder, and multiple medical comorbidities. HVDAS is caused by mutations in activity-dependent neuroprotective protein (ADNP). A recent study identified genome-wide DNA methylation changes in 22 individuals with HVDAS, adding to the group of neurodevelopmental disorders with an epigenetic signature. This methylation signature segregated those with HVDAS into two groups based on the location of the mutations. Here, we conducted an independent study on 24 individuals with HVDAS and replicated the existence of the two mutation-dependent episignatures. To probe whether the two distinct episignatures correlate with clinical outcomes, we used deep behavioral and neurobiological data from two prospective cohorts of individuals with a genetic diagnosis of HVDAS. We found limited phenotypic differences between the two HVDAS-affected groups and no evidence that individuals with more widespread methylation changes are more severely affected. Moreover, in spite of the methylation changes, we observed no profound alterations in the blood transcriptome of individuals with HVDAS. Our data warrant caution in harnessing methylation signatures in HVDAS as a tool for clinical stratification, at least with regard to behavioral phenotypes.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas de Homeodominio/genética , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Trastorno del Espectro Autista/patología , Niño , Metilación de ADN/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Epigénesis Genética/genética , Femenino , Humanos , Discapacidad Intelectual/patología , Masculino , Mutación/genética , Trastornos del Neurodesarrollo/patología , Fenotipo , Transcriptoma/genética
7.
PLoS Genet ; 16(11): e1009189, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33216750

RESUMEN

Although DNA methylation is the best characterized epigenetic mark, the mechanism by which it is targeted to specific regions in the genome remains unclear. Recent studies have revealed that local DNA methylation profiles might be dictated by cis-regulatory DNA sequences that mainly operate via DNA-binding factors. Consistent with this finding, we have recently shown that disruption of CTCF-binding sites by rare single nucleotide variants (SNVs) can underlie cis-linked DNA methylation changes in patients with congenital anomalies. These data raise the hypothesis that rare genetic variation at transcription factor binding sites (TFBSs) might contribute to local DNA methylation patterning. In this work, by combining blood genome-wide DNA methylation profiles, whole genome sequencing-derived SNVs from 247 unrelated individuals along with 133 predicted TFBS motifs derived from ENCODE ChIP-Seq data, we observed an association between the disruption of binding sites for multiple TFs by rare SNVs and extreme DNA methylation values at both local and, to a lesser extent, distant CpGs. While the majority of these changes affected only single CpGs, 24% were associated with multiple outlier CpGs within ±1kb of the disrupted TFBS. Interestingly, disruption of functionally constrained sites within TF motifs lead to larger DNA methylation changes at nearby CpG sites. Altogether, these findings suggest that rare SNVs at TFBS negatively influence TF-DNA binding, which can lead to an altered local DNA methylation profile. Furthermore, subsequent integration of DNA methylation and RNA-Seq profiles from cardiac tissues enabled us to observe an association between rare SNV-directed DNA methylation and outlier expression of nearby genes. In conclusion, our findings not only provide insights into the effect of rare genetic variation at TFBS on shaping local DNA methylation and its consequences on genome regulation, but also provide a rationale to incorporate DNA methylation data to interpret the functional role of rare variants.


Asunto(s)
Islas de CpG/genética , Metilación de ADN , Epigénesis Genética , Genoma Humano/genética , Factores de Transcripción/metabolismo , Adolescente , Adulto , Sitios de Unión/genética , Niño , Preescolar , Secuenciación de Inmunoprecipitación de Cromatina , Estudios de Cohortes , Femenino , Cardiopatías Congénitas/sangre , Cardiopatías Congénitas/genética , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Secuenciación Completa del Genoma , Adulto Joven
8.
Bioinformatics ; 36(3): 922-924, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31397844

RESUMEN

SUMMARY: While next-generation sequencing (NGS) has dramatically increased the availability of genomic data, phased genome assembly and structural variant (SV) analyses are limited by NGS read lengths. Long-read sequencing from Pacific Biosciences and NGS barcoding from 10x Genomics hold the potential for far more comprehensive views of individual genomes. Here, we present MsPAC, a tool that combines both technologies to partition reads, assemble haplotypes (via existing software) and convert assemblies into high-quality, phased SV predictions. MsPAC represents a framework for haplotype-resolved SV calls that moves one step closer to fully resolved, diploid genomes. AVAILABILITY AND IMPLEMENTATION: https://github.com/oscarlr/MsPAC. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Genoma , Haplotipos , Análisis de Secuencia de ADN , Programas Informáticos
9.
PLoS Genet ; 14(10): e1007707, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30273333

RESUMEN

While population studies have resulted in detailed maps of genetic variation in humans, to date there are few robust maps of epigenetic variation. We identified sites containing clusters of CpGs with high inter-individual epigenetic variation, termed Variably Methylated Regions (VMRs) in five purified cell types. We observed that VMRs occur preferentially at enhancers and 3' UTRs. While the majority of VMRs have high heritability, a subset of VMRs within the genome show highly correlated variation in trans, forming co-regulated networks that have low heritability, differ between cell types and are enriched for specific transcription factor binding sites and biological pathways of functional relevance to each tissue. For example, in T cells we defined a network of 95 co-regulated VMRs enriched for genes with roles in T-cell activation; in fibroblasts a network of 34 co-regulated VMRs comprising all four HOX gene clusters enriched for control of tissue growth; and in neurons a network of 18 VMRs enriched for roles in synaptic signaling. By culturing genetically-identical fibroblasts under varying environmental conditions, we experimentally demonstrated that some VMR networks are responsive to the environment, with methylation levels at these loci changing in a coordinated fashion in trans dependent on cellular growth. Intriguingly these environmentally-responsive VMRs showed a strong enrichment for imprinted loci (p<10-80), suggesting that these are particularly sensitive to environmental conditions. Our study provides a detailed map of common epigenetic variation in the human genome, showing that both genetic and environmental causes underlie this variation.


Asunto(s)
Metilación de ADN , Redes Reguladoras de Genes , Genoma Humano , Técnicas de Cultivo de Célula , Islas de CpG/genética , Epigénesis Genética , Epigenómica/métodos , Fibroblastos/fisiología , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Interacción Gen-Ambiente , Variación Genética , Humanos , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Secuencias Reguladoras de Ácidos Nucleicos
10.
J Allergy Clin Immunol ; 145(4): 1219-1230, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31838046

RESUMEN

BACKGROUND: Unexpected allergic reactions to peanut are the most common cause of fatal food-related anaphylaxis. Mechanisms underlying the variable severity of peanut-allergic reactions remain unclear. OBJECTIVES: We sought to expand mechanistic understanding of reaction severity in peanut allergy. METHODS: We performed an integrated transcriptomic and epigenomic study of peanut-allergic children as they reacted in vivo during double-blind, placebo-controlled peanut challenges. We integrated whole-blood transcriptome and CD4+ T-cell epigenome profiles to identify molecular signatures of reaction severity (ie, how severely a peanut-allergic child reacts when exposed to peanut). A threshold-weighted reaction severity score was calculated for each subject based on symptoms experienced during peanut challenge and the eliciting dose. Through linear mixed effects modeling, network construction, and causal mediation analysis, we identified genes, CpGs, and their interactions that mediate reaction severity. Findings were replicated in an independent cohort. RESULTS: We identified 318 genes with changes in expression during the course of reaction associated with reaction severity, and 203 CpG sites with differential DNA methylation associated with reaction severity. After replicating these findings in an independent cohort, we constructed interaction networks with the identified peanut severity genes and CpGs. These analyses and leukocyte deconvolution highlighted neutrophil-mediated immunity. We identified NFKBIA and ARG1 as hubs in the networks and 3 groups of interacting key node CpGs and peanut severity genes encompassing immune response, chemotaxis, and regulation of macroautophagy. In addition, we found that gene expression of PHACTR1 and ZNF121 causally mediates the association between methylation at corresponding CpGs and reaction severity, suggesting that methylation may serve as an anchor upon which gene expression modulates reaction severity. CONCLUSIONS: Our findings enhance current mechanistic understanding of the genetic and epigenetic architecture of reaction severity in peanut allergy.


Asunto(s)
Anafilaxia/genética , Linfocitos T CD4-Positivos/fisiología , Hipersensibilidad al Cacahuete/genética , Adolescente , Alérgenos/inmunología , Arachis/inmunología , Niño , Estudios de Cohortes , Metilación de ADN , Progresión de la Enfermedad , Epigénesis Genética , Femenino , Redes Reguladoras de Genes , Humanos , Inmunidad/genética , Inmunización , Masculino , Transcriptoma
11.
Hum Mutat ; 41(4): 800-806, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31898844

RESUMEN

The mechanisms underlying de novo insertion/deletion (indel) genesis, such as polymerase slippage, have been hypothesized but not well characterized in the human genome. We implemented two methodological improvements, which were leveraged to dissect indel mutagenesis. We assigned de novo variants to parent-of-origin (i.e., phasing) with low-coverage long-read whole-genome sequencing, achieving better phasing compared to short-read sequencing (medians of 84% and 23%, respectively). We then wrote an application programming interface to classify indels into three subtypes according to sequence context. Across three cohorts with different phasing methods (Ntrios = 540, all cohorts), we observed that one de novo indel subtype, change in copy count (CCC), was significantly correlated with father's (p = 7.1 × 10-4 ) but not mother's (p = .45) age at conception. We replicated this effect in three cohorts without de novo phasing (ppaternal = 1.9 × 10-9 , pmaternal = .61; Ntrios = 3,391, all cohorts). Although this is consistent with polymerase slippage during spermatogenesis, the percentage of variance explained by paternal age was low, and we did not observe an association with replication timing. These results suggest that spermatogenesis-specific events have a minor role in CCC indel mutagenesis, one not observed for other indel subtypes nor for maternal age in general. These results have implications for indel modeling in evolution and disease.


Asunto(s)
Biología Computacional/métodos , Genoma Humano , Genómica/métodos , Mutación INDEL , Programas Informáticos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Polimorfismo de Nucleótido Simple
12.
Brain ; 142(3): 647-661, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30698680

RESUMEN

Cell-permeable formulations of metabolites, such as fumaric acid esters, have been used as highly effective immunomodulators in patients with multiple sclerosis and yet their mechanism of action remains elusive. Since fumaric acid esters are metabolites, and cell metabolism is highly intertwined with the epigenetic regulation of gene expression, we investigated whether this metabolic-epigenetic interplay could be leveraged for therapeutic purposes. To this end we recruited 47 treatment-naïve and 35 fumaric acid ester-treated patients with multiple sclerosis, as well as 16 glatiramer acetate-treated patients as a non-metabolite treatment control. Here we identify a significant immunomodulatory effect of fumaric acid esters on the expression of the brain-homing chemokine receptor CCR6 in CD4 and CD8 T cells of patients with multiple sclerosis, which include T helper-17 and T cytotoxic-17 cells. We report differences in DNA methylation of CD4 T cells isolated from untreated and treated patients with multiple sclerosis, using the Illumina EPIC 850K BeadChip. We first demonstrate that Krebs cycle intermediates, such as fumaric acid esters, have a significantly higher impact on epigenome-wide DNA methylation changes in CD4 T cells compared to amino-acid polymers such as glatiramer acetate. We then define a fumaric acid ester treatment-specific hypermethylation effect on microRNA MIR-21, which is critical for the differentiation of T helper-17 cells. This hypermethylation effect was attributed to the subpopulation of T helper-17 cells using a decomposition analysis and was further validated in an independent prospective cohort of seven patients before and after treatment with fumaric acid esters. In vitro treatment of CD4 and CD8 T cells with fumaric acid esters supported a direct and dose-dependent effect on DNA methylation at the MIR-21 promoter. Finally, the upregulation of miR-21 transcripts and CCR6 expression was inhibited if CD4 or CD8 T cells stimulated under T helper-17 or T cytotoxic-17 polarizing conditions were treated with fumaric acid esters in vitro. These data collectively define a direct link between fumaric acid ester treatment and hypermethylation of the MIR-21 locus in both CD4 and CD8 T cells and suggest that the immunomodulatory effect of fumaric acid esters in multiple sclerosis is at least in part due to the epigenetic regulation of the brain-homing CCR6+ CD4 and CD8 T cells.


Asunto(s)
Fumaratos/metabolismo , Esclerosis Múltiple/metabolismo , Adulto , Encéfalo/inmunología , Encéfalo/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Metilación de ADN/genética , Epigénesis Genética/genética , Femenino , Fumaratos/farmacología , Regulación de la Expresión Génica/genética , Acetato de Glatiramer/uso terapéutico , Humanos , Factores Inmunológicos/uso terapéutico , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Esclerosis Múltiple/fisiopatología , Estudios Prospectivos , Linfocitos T/metabolismo , Linfocitos T/fisiología
13.
BMC Biol ; 17(1): 50, 2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31234833

RESUMEN

BACKGROUND: Identification of imprinted genes, demonstrating a consistent preference towards the paternal or maternal allelic expression, is important for the understanding of gene expression regulation during embryonic development and of the molecular basis of developmental disorders with a parent-of-origin effect. Combining allelic analysis of RNA-Seq data with phased genotypes in family trios provides a powerful method to detect parent-of-origin biases in gene expression. RESULTS: We report findings in 296 family trios from two large studies: 165 lymphoblastoid cell lines from the 1000 Genomes Project and 131 blood samples from the Genome of the Netherlands (GoNL) participants. Based on parental haplotypes, we identified > 2.8 million transcribed heterozygous SNVs phased for parental origin and developed a robust statistical framework for measuring allelic expression. We identified a total of 45 imprinted genes and one imprinted unannotated transcript, including multiple imprinted transcripts showing incomplete parental expression bias that was located adjacent to strongly imprinted genes. For example, PXDC1, a gene which lies adjacent to the paternally expressed gene FAM50B, shows a 2:1 paternal expression bias. Other imprinted genes had promoter regions that coincide with sites of parentally biased DNA methylation identified in the blood from uniparental disomy (UPD) samples, thus providing independent validation of our results. Using the stranded nature of the RNA-Seq data in lymphoblastoid cell lines, we identified multiple loci with overlapping sense/antisense transcripts, of which one is expressed paternally and the other maternally. Using a sliding window approach, we searched for imprinted expression across the entire genome, identifying a novel imprinted putative lncRNA in 13q21.2. Overall, we identified 7 transcripts showing parental bias in gene expression which were not reported in 4 other recent RNA-Seq studies of imprinting. CONCLUSIONS: Our methods and data provide a robust and high-resolution map of imprinted gene expression in the human genome.


Asunto(s)
Alelos , Expresión Génica/genética , Impresión Genómica/genética , Haplotipos/genética , Análisis Químico de la Sangre , Línea Celular , Humanos , Análisis de Secuencia de ARN
14.
Hum Mutat ; 40(7): 952-961, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30900359

RESUMEN

While many studies have led to the identification of rare sequence variants linked with susceptibility to autism and schizophrenia, the contribution of rare epigenetic variations (epivariations) in these disorders remains largely unexplored. Previously we presented evidence that epivariations occur relatively frequently in the human genome, and likely contribute to a subset of congenital and neurodevelopmental disorders through the disruption of dosage-sensitive genes. Here we extend this approach, studying methylation profiles from 297 samples with autism and 767 cases with schizophrenia, identifying 84 and 268 rare epivariations in these two cohorts, respectively, that were absent from 4,860 population controls. We observed multiple features associated with these epivariations that support their pathogenic relevance, including (a) a significant enrichment for epivariations in schizophrenic individuals at genes previously linked with schizophrenia, (b) increased brain expression of genes associated with epivariations found in autism cases compared with controls, (c) in autism families, a significant excess of epivariations found specifically in affected versus unaffected sibs, (d) Gene Ontology terms linked with epivariations found in autism, including "D1 dopamine receptor binding." Our study provides additional evidence that rare epivariations likely contribute to the mutational spectra underlying neurodevelopmental disorders.


Asunto(s)
Trastorno Autístico/genética , Metilación de ADN , Esquizofrenia/genética , Estudios de Casos y Controles , Epigénesis Genética , Femenino , Ontología de Genes , Predisposición Genética a la Enfermedad , Humanos , Masculino
15.
Am J Hum Genet ; 99(3): 555-566, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27569549

RESUMEN

Genomic imprinting is a mechanism in which gene expression varies depending on parental origin. Imprinting occurs through differential epigenetic marks on the two parental alleles, with most imprinted loci marked by the presence of differentially methylated regions (DMRs). To identify sites of parental epigenetic bias, here we have profiled DNA methylation patterns in a cohort of 57 individuals with uniparental disomy (UPD) for 19 different chromosomes, defining imprinted DMRs as sites where the maternal and paternal methylation levels diverge significantly from the biparental mean. Using this approach we identified 77 DMRs, including nearly all those described in previous studies, in addition to 34 DMRs not previously reported. These include a DMR at TUBGCP5 within the recurrent 15q11.2 microdeletion region, suggesting potential parent-of-origin effects associated with this genomic disorder. We also observed a modest parental bias in DNA methylation levels at every CpG analyzed across ∼1.9 Mb of the 15q11-q13 Prader-Willi/Angelman syndrome region, demonstrating that the influence of imprinting is not limited to individual regulatory elements such as CpG islands, but can extend across entire chromosomal domains. Using RNA-seq data, we detected signatures consistent with imprinted expression associated with nine novel DMRs. Finally, using a population sample of 4,004 blood methylomes, we define patterns of epigenetic variation at DMRs, identifying rare individuals with global gain or loss of methylation across multiple imprinted loci. Our data provide a detailed map of parental epigenetic bias in the human genome, providing insights into potential parent-of-origin effects.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética/genética , Genoma Humano/genética , Padres , Disomía Uniparental/genética , Alelos , Síndrome de Angelman/genética , Aberraciones Cromosómicas , Cromosomas Humanos/genética , Cromosomas Humanos Par 15/genética , Estudios de Cohortes , Islas de CpG/genética , Femenino , Impresión Genómica/genética , Humanos , Discapacidad Intelectual/genética , Cariotipo , Masculino , Proteínas Asociadas a Microtúbulos/genética , Síndrome de Prader-Willi/genética , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN
16.
Hum Mutat ; 39(6): 870-881, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29527824

RESUMEN

Multiple tools have been developed to identify copy number variants (CNVs) from whole exome (WES) and whole genome sequencing (WGS) data. Current tools such as XHMM for WES and CNVnator for WGS identify CNVs based on changes in read depth. For WGS, other methods to identify CNVs include utilizing discordant read pairs and split reads and genome-wide local assembly with tools such as Lumpy and SvABA, respectively. Here, we introduce a new method to identify deletion CNVs from WES and WGS trio data based on the clustering of Mendelian errors (MEs). Using our Mendelian Error Method (MEM), we identified 127 deletions (inherited and de novo) in 2,601 WES trios from the Pediatric Cardiac Genomics Consortium, with a validation rate of 88% by digital droplet PCR. MEM identified additional de novo deletions compared with XHMM, and a significant enrichment of 15q11.2 deletions compared with controls. In addition, MEM identified eight cases of uniparental disomy, sample switches, and DNA contamination. We applied MEM to WGS data from the Genome In A Bottle Ashkenazi trio and identified deletions with 97% specificity. MEM provides a robust, computationally inexpensive method for identifying deletions, and an orthogonal approach for verifying deletions called by other tools.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Análisis Mutacional de ADN/métodos , Genoma Humano/genética , Eliminación de Secuencia/genética , Mapeo Cromosómico , Exoma/genética , Femenino , Cardiopatías Congénitas/genética , Humanos , Masculino , Secuenciación del Exoma , Secuenciación Completa del Genoma
17.
Nucleic Acids Res ; 44(8): 3750-62, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27060133

RESUMEN

Despite representing an important source of genetic variation, tandem repeats (TRs) remain poorly studied due to technical difficulties. We hypothesized that TRs can operate as expression (eQTLs) and methylation (mQTLs) quantitative trait loci. To test this we analyzed the effect of variation at 4849 promoter-associated TRs, genotyped in 120 individuals, on neighboring gene expression and DNA methylation. Polymorphic promoter TRs were associated with increased variance in local gene expression and DNA methylation, suggesting functional consequences related to TR variation. We identified >100 TRs associated with expression/methylation levels of adjacent genes. These potential eQTL/mQTL TRs were enriched for overlaps with transcription factor binding and DNaseI hypersensitivity sites, providing a rationale for their effects. Moreover, we showed that most TR variants are poorly tagged by nearby single nucleotide polymorphisms (SNPs) markers, indicating that many functional TR variants are not effectively assayed by SNP-based approaches. Our study assigns biological significance to TR variations in the human genome, and suggests that a significant fraction of TR variations exert functional effects via alterations of local gene expression or epigenetics. We conclude that targeted studies that focus on genotyping TR variants are required to fully ascertain functional variation in the genome.


Asunto(s)
Metilación de ADN , Regulación de la Expresión Génica , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Secuencias Repetidas en Tándem , Técnicas de Genotipaje , Humanos , Desequilibrio de Ligamiento , Sitios de Carácter Cuantitativo , Análisis de Secuencia de ADN
18.
PLoS Genet ; 11(12): e1005661, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26658498

RESUMEN

A fundamental initiative for evolutionary biologists is to understand the molecular basis underlying phenotypic diversity. A long-standing hypothesis states that species-specific traits may be explained by differences in gene regulation rather than differences at the protein level. Over the past few years, evolutionary studies have shifted from mere sequence comparisons to integrative analyses in which gene regulation is key to understanding species evolution. DNA methylation is an important epigenetic modification involved in the regulation of numerous biological processes. Nevertheless, the evolution of the human methylome and the processes driving such changes are poorly understood. Here, we review the close interplay between Cytosine-phosphate-Guanine (CpG) methylation and the underlying genome sequence, as well as its evolutionary impact. We also summarize the latest advances in the field, revisiting the main literature on human and nonhuman primates. We hope to encourage the scientific community to address the many challenges posed by the field of comparative epigenomics.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética , Evolución Molecular , Genoma Humano , Animales , Islas de CpG/genética , Regulación de la Expresión Génica , Humanos , Primates/genética
19.
PLoS Genet ; 11(3): e1005062, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25803843

RESUMEN

The trisomy of human chromosome 21 (Hsa21), which causes Down syndrome (DS), is the most common viable human aneuploidy. In contrast to trisomy, the complete monosomy (M21) of Hsa21 is lethal, and only partial monosomy or mosaic monosomy of Hsa21 is seen. Both conditions lead to variable physiological abnormalities with constant intellectual disability, locomotor deficits, and altered muscle tone. To search for dosage-sensitive genes involved in DS and M21 phenotypes, we created two new mouse models: the Ts3Yah carrying a tandem duplication and the Ms3Yah carrying a deletion of the Hspa13-App interval syntenic with 21q11.2-q21.3. Here we report that the trisomy and the monosomy of this region alter locomotion, muscle strength, mass, and energetic balance. The expression profiling of skeletal muscles revealed global changes in the regulation of genes implicated in energetic metabolism, mitochondrial activity, and biogenesis. These genes are downregulated in Ts3Yah mice and upregulated in Ms3Yah mice. The shift in skeletal muscle metabolism correlates with a change in mitochondrial proliferation without an alteration in the respiratory function. However, the reactive oxygen species (ROS) production from mitochondrial complex I decreased in Ms3Yah mice, while the membrane permeability of Ts3Yah mitochondria slightly increased. Thus, we demonstrated how the Hspa13-App interval controls metabolic and mitochondrial phenotypes in muscles certainly as a consequence of change in dose of Gabpa, Nrip1, and Atp5j. Our results indicate that the copy number variation in the Hspa13-App region has a peripheral impact on locomotor activity by altering muscle function.


Asunto(s)
Síndrome de Down/genética , Monosomía/genética , Actividad Motora/genética , Fuerza Muscular/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Cromosomas Humanos Par 21/genética , Modelos Animales de Enfermedad , Síndrome de Down/fisiopatología , Metabolismo Energético/genética , Factor de Transcripción de la Proteína de Unión a GA/genética , Humanos , Ratones , Mitocondrias Musculares/genética , Mitocondrias Musculares/patología , ATPasas de Translocación de Protón Mitocondriales/genética , Monosomía/fisiopatología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Proteínas Nucleares/genética , Proteína de Interacción con Receptores Nucleares 1
20.
Annu Rev Genomics Hum Genet ; 15: 215-244, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24773319

RESUMEN

Chromosomal abnormalities, including microdeletions and microduplications, have long been associated with abnormal developmental outcomes. Early discoveries relied on a common clinical presentation and the ability to detect chromosomal abnormalities by standard karyotype analysis or specific assays such as fluorescence in situ hybridization. Over the past decade, the development of novel genomic technologies has allowed more comprehensive, unbiased discovery of microdeletions and microduplications throughout the human genome. The ability to quickly interrogate large cohorts using chromosome microarrays and, more recently, next-generation sequencing has led to the rapid discovery of novel microdeletions and microduplications associated with disease, including very rare but clinically significant rearrangements. In addition, the observation that some microdeletions are associated with risk for several neurodevelopmental disorders contributes to our understanding of shared genetic susceptibility for such disorders. Here, we review current knowledge of microdeletion/duplication syndromes, with a particular focus on recurrent rearrangement syndromes.


Asunto(s)
Aberraciones Cromosómicas , Deleción Cromosómica , Duplicación Cromosómica/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Genoma Humano , Humanos , Cariotipificación , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA