Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Sci Rep ; 13(1): 6745, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37185990

RESUMO

Enhancers are important cis-regulatory elements controlling cell-type specific expression patterns of genes. Furthermore, combinations of enhancers and minimal promoters are utilized to construct small, artificial promoters for gene delivery vectors. Large-scale functional screening methodology to construct genomic maps of enhancer activities has been successfully established in cultured cell lines, however, not yet applied to terminally differentiated cells and tissues in a living animal. Here, we transposed the Self-Transcribing Active Regulatory Region Sequencing (STARR-seq) technique to the mouse brain using adeno-associated-viruses (AAV) for the delivery of a highly complex screening library tiling entire genomic regions and covering in total 3 Mb of the mouse genome. We identified 483 sequences with enhancer activity, including sequences that were not predicted by DNA accessibility or histone marks. Characterizing the expression patterns of fluorescent reporters controlled by nine candidate sequences, we observed differential expression patterns also in sparse cell types. Together, our study provides an entry point for the unbiased study of enhancer activities in organisms during health and disease.


Assuntos
Elementos Facilitadores Genéticos , Genômica , Animais , Camundongos , Genômica/métodos , Mapeamento Cromossômico/métodos , Regiões Promotoras Genéticas , Encéfalo
2.
Cell Rep ; 41(3): 111503, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36261000

RESUMO

Concurrent mutation of a RAS oncogene and the tumor suppressor p53 is common in tumorigenesis, and inflammation can promote RAS-driven tumorigenesis without the need to mutate p53. Here, we show, using a well-established mutant RAS and an inflammation-driven mouse skin tumor model, that loss of the p53 inhibitor iASPP facilitates tumorigenesis. Specifically, iASPP regulates expression of a subset of p63 and AP1 targets, including genes involved in skin differentiation and inflammation, suggesting that loss of iASPP in keratinocytes supports a tumor-promoting inflammatory microenvironment. Mechanistically, JNK-mediated phosphorylation regulates iASPP function and inhibits iASPP binding with AP1 components, such as JUND, via PXXP/SH3 domain-mediated interaction. Our results uncover a JNK-iASPP-AP1 regulatory axis that is crucial for tissue homeostasis. We show that iASPP is a tumor suppressor and an AP1 coregulator.


Assuntos
Proteínas Repressoras , Proteína Supressora de Tumor p53 , Animais , Camundongos , Transformação Celular Neoplásica/genética , Inflamação/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Repressoras/metabolismo , Microambiente Tumoral , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , MAP Quinase Quinase 4/metabolismo , Fator de Transcrição AP-1/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-31616643

RESUMO

Background: The failure to translate preclinical results to the clinical setting is the rule, not the exception. One reason that is frequently overlooked is whether the animal model reproduces distinctive features of human disease. Another is the reproducibility of the method used to measure treatment effects in preclinical studies. Left ventricular (LV) function improvement is the most common endpoint in preclinical cardiovascular disease studies, while echocardiography is the most frequently used method to evaluate LV function. In this work, we conducted a robust echocardiographic evaluation of LV size and function in dogs chronically infected by Trypanosoma cruzi. Methods and Results: Echocardiography was performed blindly by two distinct observers in mongrel dogs before and between 6 and 9 months post infection. Parameters analyzed included end-systolic volume (ESV), end-diastolic volume (EDV), ejection fraction (EF), and fractional shortening (FS). We observed a significant LVEF and FS reduction in infected animals compared to controls, with no significant variation in volumes. However, the effect of chronic infection in systolic function was quite variable, with EF ranging from 17 to 66%. Using the cut-off value of EF ≤ 40%, established for dilated cardiomyopathy (DCM) in dogs, only 28% of the infected dogs were affected by the chronic infection. Conclusions: The canine model of CCC mimics human disease, reproducing the percentage of individuals that develop heart failure during the chronic infection. It is thus mandatory to establish inclusion criteria in the experimental design of canine preclinical studies to account for the variable effect that chronic infection has on systolic function.


Assuntos
Cardiomiopatia Chagásica/diagnóstico por imagem , Ecocardiografia/métodos , Ventrículos do Coração/diagnóstico por imagem , Animais , Modelos Animais de Doenças , Cães , Reprodutibilidade dos Testes , Função Ventricular
4.
Mol Cell ; 74(6): 1148-1163.e7, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31005419

RESUMO

Self-renewal and pluripotency of the embryonic stem cell (ESC) state are established and maintained by multiple regulatory networks that comprise transcription factors and epigenetic regulators. While much has been learned regarding transcription factors, the function of epigenetic regulators in these networks is less well defined. We conducted a CRISPR-Cas9-mediated loss-of-function genetic screen that identified two epigenetic regulators, TAF5L and TAF6L, components or co-activators of the GNAT-HAT complexes for the mouse ESC (mESC) state. Detailed molecular studies demonstrate that TAF5L/TAF6L transcriptionally activate c-Myc and Oct4 and their corresponding MYC and CORE regulatory networks. Besides, TAF5L/TAF6L predominantly regulate their target genes through H3K9ac deposition and c-MYC recruitment that eventually activate the MYC regulatory network for self-renewal of mESCs. Thus, our findings uncover a role of TAF5L/TAF6L in directing the MYC regulatory network that orchestrates gene expression programs to control self-renewal for the maintenance of mESC state.


Assuntos
Células-Tronco Embrionárias/metabolismo , Redes Reguladoras de Genes , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Animais , Sistemas CRISPR-Cas , Ciclo Celular/genética , Proliferação de Células , Reprogramação Celular , Embrião de Mamíferos , Células-Tronco Embrionárias/citologia , Epigênese Genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Edição de Genes , Regulação da Expressão Gênica , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Cultura Primária de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Fatores Associados à Proteína de Ligação a TATA/metabolismo
5.
Cell Rep ; 25(12): 3490-3503.e4, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30566872

RESUMO

Transcription factor p63 is a key regulator of epidermal keratinocyte proliferation and differentiation. Mutations in the p63 DNA-binding domain are associated with ectrodactyly, ectodermal dysplasia, and cleft lip/palate (EEC) syndrome. However, the underlying molecular mechanism of these mutations remains unclear. Here, we characterized the transcriptome and epigenome of p63 mutant keratinocytes derived from EEC patients. The transcriptome of p63 mutant keratinocytes deviated from the normal epidermal cell identity. Epigenomic analyses showed an altered enhancer landscape in p63 mutant keratinocytes contributed by loss of p63-bound active enhancers and unexpected gain of enhancers. The gained enhancers were frequently bound by deregulated transcription factors such as RUNX1. Reversing RUNX1 overexpression partially rescued deregulated gene expression and the altered enhancer landscape. Our findings identify a disease mechanism whereby mutant p63 rewires the enhancer landscape and affects epidermal cell identity, consolidating the pivotal role of p63 in controlling the enhancer landscape of epidermal keratinocytes.


Assuntos
Elementos Facilitadores Genéticos/genética , Células Epidérmicas/citologia , Células Epidérmicas/metabolismo , Mutação/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Sequência de Aminoácidos , Diferenciação Celular/genética , Cromatina/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Modelos Biológicos , Ligação Proteica , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transcriptoma/genética , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo
6.
Sci Rep ; 8(1): 11685, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-30076349

RESUMO

Tissue culture medium routinely contains fetal bovine serum (FBS). Here we show that culturing human hepatoma cells in their native, adult serum (human serum, HS) results in the restoration of key morphological and metabolic features of normal liver cells. When moved to HS, these cells show differential transcription of 22-32% of the genes, stop proliferating, and assume a hepatocyte-like morphology. Metabolic analysis shows that the Warburg-like metabolic profile, typical for FBS-cultured cells, is replaced by a diverse metabolic profile consistent with in vivo hepatocytes, including the formation of large lipid and glycogen stores, increased glycogenesis, increased beta-oxidation and ketogenesis, and decreased glycolysis. Finally, organ-specific functions are restored, including xenobiotics degradation and secretion of bile, VLDL and albumin. Thus, organ-specific functions are not necessarily lost in cell cultures, but might be merely suppressed in FBS. The effect of serum is often overseen in cell culture and we provide a detailed study in the changes that occur and provide insight in some of the serum components that may play a role in the establishment of the differentiated phenotype.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Diferenciação Celular , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Soro/metabolismo , Adulto , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/ultraestrutura , Forma Celular , Sistema Enzimático do Citocromo P-450/metabolismo , Citoesqueleto/metabolismo , Regulação Neoplásica da Expressão Gênica , Glicólise , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/ultraestrutura , Redes e Vias Metabólicas , Análise de Componente Principal , Células Tumorais Cultivadas , Xenobióticos/metabolismo
7.
Cereb Cortex ; 28(3): 833-851, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28108491

RESUMO

The prefrontal cortex (PFC) is one of the latest brain regions to mature, which allows the acquisition of complex cognitive abilities through experience. To unravel the underlying gene expression changes during postnatal development, we performed RNA-sequencing (RNA-seq) in the rat medial PFC (mPFC) at five developmental time points from infancy to adulthood, and analyzed the differential expression of protein-coding genes, long intergenic noncoding RNAs (lincRNAs), and alternative exons. We showed that most expression changes occur in infancy, and that the number of differentially expressed genes reduces toward adulthood. We observed 137 differentially expressed lincRNAs and 796 genes showing alternative exon usage during postnatal development. Importantly, we detected a genetic switch from neuronal network establishment in infancy to maintenance of neural networks in adulthood based on gene expression dynamics, involving changes in protein-coding and lincRNA gene expression as well as alternative exon usage. Our gene expression datasets provide insights into the multifaceted transcriptional regulation of the developing PFC. They can be used to study the basic developmental processes of the mPFC and to understand the mechanisms of neurodevelopmental and neuropsychiatric disorders. Our study provides an important contribution to the ongoing efforts to complete the "brain map", and to the understanding of PFC development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Fatores Etários , Animais , Animais Recém-Nascidos , Perfilação da Expressão Gênica , Ontologia Genética , Estudo de Associação Genômica Ampla , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Ratos , Ratos Wistar
8.
Methods Mol Biol ; 1704: 433-450, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29277877

RESUMO

Since the pioneering studies of Thomas Hunt Morgan and coworkers at the dawn of the twentieth century, Drosophila melanogaster and its sister species have tremendously contributed to unveil the rules underlying animal genetics, development, behavior, evolution, and human disease. Recent advances in DNA sequencing technologies launched Drosophila into the post-genomic era and paved the way for unprecedented comparative genomics investigations. The complete sequencing and systematic comparison of the genomes from 12 Drosophila species represents a milestone achievement in modern biology, which allowed a plethora of different studies ranging from the annotation of known and novel genomic features to the evolution of chromosomes and, ultimately, of entire genomes. Despite the efforts of countless laboratories worldwide, the vast amount of data that were produced over the past 15 years is far from being fully explored.In this chapter, we will review some of the bioinformatic approaches that were developed to interrogate the genomes of the 12 Drosophila species. Setting off from alignments of the entire genomic sequences, the degree of conservation can be separately evaluated for every region of the genome, providing already first hints about elements that are under purifying selection and therefore likely functional. Furthermore, the careful analysis of repeated sequences sheds light on the evolutionary dynamics of transposons, an enigmatic and fascinating class of mobile elements housed in the genomes of animals and plants. Comparative genomics also aids in the computational identification of the transcriptionally active part of the genome, first and foremost of protein-coding loci, but also of transcribed nevertheless apparently noncoding regions, which were once considered "junk" DNA. Eventually, the synergy between functional and comparative genomics also facilitates in silico and in vivo studies on cis-acting regulatory elements, like transcription factor binding sites, that due to the high degree of sequence variability usually impose increased challenges for bioinformatics approaches.


Assuntos
Drosophila/genética , Evolução Molecular , Genoma de Inseto , Genômica/métodos , Algoritmos , Animais , Biologia Computacional , Anotação de Sequência Molecular , Software
9.
Methods Mol Biol ; 1704: 451-472, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29277878

RESUMO

Genomes can be compared at different levels of divergence, either between species or within species. Within species genomes can be compared between different subpopulations, such as human subpopulations from different continents. Investigating the genomic differences between different human subpopulations is important when studying complex diseases that are affected by many genetic variants, as the variants involved can differ between populations. The 1000 Genomes Project collected genome-scale variation data for 2504 human individuals from 26 different populations, enabling a systematic comparison of variation between human subpopulations. In this chapter, we present step-by-step a basic protocol for the identification of population-specific variants employing the 1000 Genomes data. These variants are subsequently further investigated for those that affect the proteome or RNA splice sites, to investigate potentially biologically relevant differences between the populations.


Assuntos
Genética Populacional , Genoma Humano , Genômica/métodos , Polimorfismo de Nucleotídeo Único , Projeto Genoma Humano , Humanos , Fenótipo
10.
Am J Hum Genet ; 100(5): 737-750, 2017 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-28457472

RESUMO

Keratolytic winter erythema (KWE) is a rare autosomal-dominant skin disorder characterized by recurrent episodes of palmoplantar erythema and epidermal peeling. KWE was previously mapped to 8p23.1-p22 (KWE critical region) in South African families. Using targeted resequencing of the KWE critical region in five South African families and SNP array and whole-genome sequencing in two Norwegian families, we identified two overlapping tandem duplications of 7.67 kb (South Africans) and 15.93 kb (Norwegians). The duplications segregated with the disease and were located upstream of CTSB, a gene encoding cathepsin B, a cysteine protease involved in keratinocyte homeostasis. Included in the 2.62 kb overlapping region of these duplications is an enhancer element that is active in epidermal keratinocytes. The activity of this enhancer correlated with CTSB expression in normal differentiating keratinocytes and other cell lines, but not with FDFT1 or NEIL2 expression. Gene expression (qPCR) analysis and immunohistochemistry of the palmar epidermis demonstrated significantly increased expression of CTSB, as well as stronger staining of cathepsin B in the stratum granulosum of affected individuals than in that of control individuals. Analysis of higher-order chromatin structure data and RNA polymerase II ChIA-PET data from MCF-7 cells did not suggest remote effects of the enhancer. In conclusion, KWE in South African and Norwegian families is caused by tandem duplications in a non-coding genomic region containing an active enhancer element for CTSB, resulting in upregulation of this gene in affected individuals.


Assuntos
Catepsina B/metabolismo , Elementos Facilitadores Genéticos , Eritema/genética , Duplicação Gênica , Regulação da Expressão Gênica , Ceratose/genética , Dermatopatias Genéticas/genética , Estudos de Casos e Controles , Catepsina B/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 8/genética , Variações do Número de Cópias de DNA , DNA Glicosilases/genética , DNA Glicosilases/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Epiderme/metabolismo , Epigenômica , Eritema/epidemiologia , Feminino , Marcadores Genéticos , Humanos , Queratinócitos/metabolismo , Ceratose/epidemiologia , Células MCF-7 , Masculino , Noruega/epidemiologia , Linhagem , Dermatopatias Genéticas/epidemiologia , África do Sul/epidemiologia
11.
Dis Model Mech ; 10(2): 105-118, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28067622

RESUMO

A consanguineous family from Pakistan was ascertained to have a novel deafness-dystonia syndrome with motor regression, ichthyosis-like features and signs of sensory neuropathy. By applying a combined strategy of linkage analysis and whole-exome sequencing in the presented family, a homozygous nonsense mutation, c.4G>T (p.Glu2*), in FITM2 was identified. FITM2 and its paralog FITM1 constitute an evolutionary conserved protein family involved in partitioning of triglycerides into cellular lipid droplets. Despite the role of FITM2 in neutral lipid storage and metabolism, no indications for lipodystrophy were observed in the affected individuals. In order to obtain independent evidence for the involvement of FITM2 in the human pathology, downregulation of the single Fitm ortholog, CG10671, in Drosophila melanogaster was pursued using RNA interference. Characteristics of the syndrome, including progressive locomotor impairment, hearing loss and disturbed sensory functions, were recapitulated in Drosophila, which supports the causative nature of the FITM2 mutation. Mutation-based genetic counseling can now be provided to the family and insight is obtained into the potential impact of genetic variation in FITM2.


Assuntos
Surdocegueira/genética , Proteínas de Drosophila/genética , Distonia/genética , Ictiose/genética , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Atividade Motora , Mutação/genética , Atrofia Óptica/genética , Células Receptoras Sensoriais/patologia , Adiposidade , Animais , Audiometria de Tons Puros , Sequência de Bases , Criança , Códon sem Sentido/genética , Surdocegueira/sangue , Surdocegueira/fisiopatologia , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Distonia/sangue , Distonia/fisiopatologia , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Perda Auditiva/genética , Homozigoto , Humanos , Ictiose/complicações , Ictiose/fisiopatologia , Deficiência Intelectual/sangue , Deficiência Intelectual/fisiopatologia , Gotículas Lipídicas/metabolismo , Fígado/metabolismo , Locomoção , Masculino , Proteínas de Membrana/metabolismo , Atrofia Óptica/sangue , Atrofia Óptica/fisiopatologia , Linhagem , Sequenciamento do Exoma , Adulto Jovem
12.
Sci Rep ; 6: 32406, 2016 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-27617755

RESUMO

Recent advances in the cost-efficiency of sequencing technologies enabled the combined DNA- and RNA-sequencing of human individuals at the population-scale, making genome-wide investigations of the inter-individual genetic impact on gene expression viable. Employing mRNA-sequencing data from the Geuvadis Project and genome sequencing data from the 1000 Genomes Project we show that the computational analysis of DNA sequences around splice sites and poly-A signals is able to explain several observations in the phenotype data. In contrast to widespread assessments of statistically significant associations between DNA polymorphisms and quantitative traits, we developed a computational tool to pinpoint the molecular mechanisms by which genetic markers drive variation in RNA-processing, cataloguing and classifying alleles that change the affinity of core RNA elements to their recognizing factors. The in silico models we employ further suggest RNA editing can moonlight as a splicing-modulator, albeit less frequently than genomic sequence diversity. Beyond existing annotations, we demonstrate that the ultra-high resolution of RNA-Seq combined from 462 individuals also provides evidence for thousands of bona fide novel elements of RNA processing-alternative splice sites, introns, and cleavage sites-which are often rare and lowly expressed but in other characteristics similar to their annotated counterparts.


Assuntos
Processamento Alternativo , Variação Genética , Genoma Humano , Edição de RNA , Sítios de Splice de RNA , RNA Mensageiro/genética , Alelos , Sequência de Bases , Éxons , Frequência do Gene , Humanos , Íntrons , Poliadenilação , RNA Mensageiro/metabolismo , Análise de Sequência de RNA
13.
BMC Genomics ; 17: 252, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-27004515

RESUMO

BACKGROUND: The CCTC-binding factor (CTCF) protein is involved in genome organization, including mediating three-dimensional chromatin interactions. Human patient lymphocytes with mutations in a single copy of the CTCF gene have reduced expression of enhancer-associated genes involved in response to stimuli. We hypothesize that CTCF interactions stabilize enhancer-promoter chromatin interaction domains, facilitating increased expression of genes in response to stimuli. Here we systematically investigate this model using computational analyses. RESULTS: We use CTCF ChIA-PET data from the ENCODE project to show that CTCF-associated chromatin loops have a tendency to enclose regions of enhancer-regulated stimulus responsive genes, insulating them from neighboring regions of constitutively expressed housekeeping genes. To facilitate cell type-specific CTCF loop identification, we develop an algorithm to predict CTCF loops from ChIP-seq data alone by exploiting the CTCF motif directionality in loop anchors. We apply this algorithm to a hundred ENCODE cell line datasets, confirming the universality of our observations as well as identifying a general distinction between primary and immortal cells in loop-enclosed gene content. Finally, we combine the existing evidence to propose a model for the formation of CTCF loops in which partner sites are brought together by chromatin template reeling through stationary RNA polymerases, consistent with the transcription factory hypothesis. CONCLUSIONS: We provide computational evidence that CTCF-mediated chromatin interactions enclose domains of stimulus responsive enhancer-regulated genes, insulating them from nearby housekeeping genes.


Assuntos
Cromatina/química , Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , Proteínas Repressoras/química , Fator de Ligação a CCCTC , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica , Humanos , Células K562 , Células MCF-7 , Proteínas Repressoras/genética
14.
Hum Genet ; 135(1): 41-59, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26561393

RESUMO

Orofacial clefts (OFCs) represent a large fraction of human birth defects and are one of the most common phenotypes affected by large copy number variants (CNVs). Due to the limited number of CNV patients in individual centers, CNV analyses of a large number of OFC patients are challenging. The present study analyzed 249 genomic deletions and 226 duplications from a cohort of 312 OFC patients reported in two publicly accessible databases of chromosome imbalance and phenotype in humans, DECIPHER and ECARUCA. Genomic regions deleted or duplicated in multiple patients were identified, and genes in these overlapping CNVs were prioritized based on the number of genes encompassed by the region and gene expression in embryonic mouse palate. Our analyses of these overlapping CNVs identified two genes known to be causative for human OFCs, SATB2 and MEIS2, and 12 genes (DGCR6, FGF2, FRZB, LETM1, MAPK3, SPRY1, THBS1, TSHZ1, TTC28, TULP4, WHSC1, WHSC2) that are associated with OFC or orofacial development. Additionally, we report 34 deleted and 24 duplicated genes that have not previously been associated with OFCs but are associated with the BMP, MAPK and RAC1 pathways. Statistical analyses show that the high number of overlapping CNVs is not due to random occurrence. The identified genes are not located in highly variable genomic regions in healthy populations and are significantly enriched for genes that are involved in orofacial development. In summary, we report a CNV analysis pipeline of a large cohort of OFC patients and identify novel candidate OFC genes.


Assuntos
Fenda Labial/genética , Fissura Palatina/genética , Variações do Número de Cópias de DNA , Face/anormalidades , Predisposição Genética para Doença , Humanos , Fenótipo
15.
Genom Data ; 5: 159-63, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26484246

RESUMO

The transcription factor p63 is a key regulator in epidermal keratinocyte proliferation and differentiation. However, the role of p63 in gene regulation during these processes is not well understood. To investigate this, we recently generated genome-wide profiles of gene expression, p63 binding sites and active regulatory regions with the H3K27ac histone mark (Kouwenhoven et al., 2015). We showed that only a subset of p63 binding sites are active in keratinocytes, and that differentiation-associated gene expression dynamics correlate with the activity of p63 binding sites rather than with their occurrence per se. Here we describe in detail the generation and processing of the ChIP-seq and RNA-seq datasets used in this study. These data sets are deposited in the Gene Expression Omnibus (GEO) repository under the accession number GSE59827.

16.
EMBO Rep ; 16(7): 863-78, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26034101

RESUMO

The transcription factor p63 plays a pivotal role in keratinocyte proliferation and differentiation in the epidermis. However, how p63 regulates epidermal genes during differentiation is not yet clear. Using epigenome profiling of differentiating human primary epidermal keratinocytes, we characterized a catalog of dynamically regulated genes and p63-bound regulatory elements that are relevant for epithelial development and related diseases. p63-bound regulatory elements occur as single or clustered enhancers, and remarkably, only a subset is active as defined by the co-presence of the active enhancer mark histone modification H3K27ac in epidermal keratinocytes. We show that the dynamics of gene expression correlates with the activity of p63-bound enhancers rather than with p63 binding itself. The activity of p63-bound enhancers is likely determined by other transcription factors that cooperate with p63. Our data show that inactive p63-bound enhancers in epidermal keratinocytes may be active during the development of other epithelial-related structures such as limbs and suggest that p63 bookmarks genomic loci during the commitment of the epithelial lineage and regulates genes through temporal- and spatial-specific active enhancers.


Assuntos
Diferenciação Celular , Elementos Facilitadores Genéticos , Células Epidérmicas , Regulação da Expressão Gênica , Queratinócitos/citologia , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Linhagem da Célula , Loci Gênicos , Humanos , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
17.
Mol Genet Genomic Med ; 2(1): 44-57, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24498628

RESUMO

Current single-locus-based analyses and candidate disease gene prediction methodologies used in genome-wide association studies (GWAS) do not capitalize on the wealth of the underlying genetic data, nor functional data available from molecular biology. Here, we analyzed GWAS data from the Wellcome Trust Case Control Consortium (WTCCC) on coronary artery disease (CAD). Gentrepid uses a multiple-locus-based approach, drawing on protein pathway- or domain-based data to make predictions. Known disease genes may be used as additional information (seeded method) or predictions can be based entirely on GWAS single nucleotide polymorphisms (SNPs) (ab initio method). We looked in detail at specific predictions made by Gentrepid for CAD and compared these with known genetic data and the scientific literature. Gentrepid was able to extract known disease genes from the candidate search space and predict plausible novel disease genes from both known and novel WTCCC-implicated loci. The disease gene candidates are consistent with known biological information. The results demonstrate that this computational approach is feasible and a valuable discovery tool for geneticists.

18.
PLoS Genet ; 9(10): e1003911, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204314

RESUMO

Intellectual Disability (ID) disorders, defined by an IQ below 70, are genetically and phenotypically highly heterogeneous. Identification of common molecular pathways underlying these disorders is crucial for understanding the molecular basis of cognition and for the development of therapeutic intervention strategies. To systematically establish their functional connectivity, we used transgenic RNAi to target 270 ID gene orthologs in the Drosophila eye. Assessment of neuronal function in behavioral and electrophysiological assays and multiparametric morphological analysis identified phenotypes associated with knockdown of 180 ID gene orthologs. Most of these genotype-phenotype associations were novel. For example, we uncovered 16 genes that are required for basal neurotransmission and have not previously been implicated in this process in any system or organism. ID gene orthologs with morphological eye phenotypes, in contrast to genes without phenotypes, are relatively highly expressed in the human nervous system and are enriched for neuronal functions, suggesting that eye phenotyping can distinguish different classes of ID genes. Indeed, grouping genes by Drosophila phenotype uncovered 26 connected functional modules. Novel links between ID genes successfully predicted that MYCN, PIGV and UPF3B regulate synapse development. Drosophila phenotype groups show, in addition to ID, significant phenotypic similarity also in humans, indicating that functional modules are conserved. The combined data indicate that ID disorders, despite their extreme genetic diversity, are caused by disruption of a limited number of highly connected functional modules.


Assuntos
Olho/metabolismo , Deficiência Intelectual/genética , Redes e Vias Metabólicas/genética , Sinapses/genética , Animais , Animais Geneticamente Modificados , Drosophila/genética , Olho/crescimento & desenvolvimento , Técnicas de Silenciamento de Genes , Variação Genética , Humanos , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Neurônios/metabolismo , Fenótipo , Interferência de RNA , Sinapses/metabolismo
19.
BMC Bioinformatics ; 14: 249, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23947436

RESUMO

BACKGROUND: Candidate disease gene prediction is a rapidly developing area of bioinformatics research with the potential to deliver great benefits to human health. As experimental studies detecting associations between genetic intervals and disease proliferate, better bioinformatic techniques that can expand and exploit the data are required. DESCRIPTION: Gentrepid is a web resource which predicts and prioritizes candidate disease genes for both Mendelian and complex diseases. The system can take input from linkage analysis of single genetic intervals or multiple marker loci from genome-wide association studies. The underlying database of the Gentrepid tool sources data from numerous gene and protein resources, taking advantage of the wealth of biological information available. Using known disease gene information from OMIM, the system predicts and prioritizes disease gene candidates that participate in the same protein pathways or share similar protein domains. Alternatively, using an ab initio approach, the system can detect enrichment of these protein annotations without prior knowledge of the phenotype. CONCLUSIONS: The system aims to integrate the wealth of protein information currently available with known and novel phenotype/genotype information to acquire knowledge of biological mechanisms underpinning disease. We have updated the system to facilitate analysis of GWAS data and the study of complex diseases. Application of the system to GWAS data on hypertension using the ICBP data is provided as an example. An interesting prediction is a ZIP transporter additional to the one found by the ICBP analysis. The webserver URL is https://www.gentrepid.org/.


Assuntos
Biologia Computacional/métodos , Bases de Dados Genéticas , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Internet , Humanos , Fenótipo
20.
Am J Hum Genet ; 93(1): 124-31, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23746550

RESUMO

An increasing number of genes involved in chromatin structure and epigenetic regulation has been implicated in a variety of developmental disorders, often including intellectual disability. By trio exome sequencing and subsequent mutational screening we now identified two de novo frameshift mutations and one de novo missense mutation in CTCF in individuals with intellectual disability, microcephaly, and growth retardation. Furthermore, an individual with a larger deletion including CTCF was identified. CTCF (CCCTC-binding factor) is one of the most important chromatin organizers in vertebrates and is involved in various chromatin regulation processes such as higher order of chromatin organization, enhancer function, and maintenance of three-dimensional chromatin structure. Transcriptome analyses in all three individuals with point mutations revealed deregulation of genes involved in signal transduction and emphasized the role of CTCF in enhancer-driven expression of genes. Our findings indicate that haploinsufficiency of CTCF affects genomic interaction of enhancers and their regulated gene promoters that drive developmental processes and cognition.


Assuntos
Mutação da Fase de Leitura , Deficiência Intelectual/genética , Mutação de Sentido Incorreto , Proteínas Repressoras/genética , Adolescente , Fator de Ligação a CCCTC , Criança , Pré-Escolar , Cromatina/genética , Cromatina/metabolismo , Análise Mutacional de DNA , Elementos Facilitadores Genéticos , Exoma , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genoma Humano , Haploinsuficiência , Humanos , Masculino , Microcefalia/genética , Mutação Puntual , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA