Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Clin Exp Ophthalmol ; 51(4): 300-312, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36882936

RESUMO

BACKGROUND: With the promise of gene replacement therapy, eligible males and females with X-linked inherited retinal dystrophy (XL-IRD) should be identified. METHODS: Retrospective observational cohort study to establish the phenotypic and genotypic spectrum of XL-IRD within New Zealand (NZ). Thirty-two probands, including 9 females, with molecularly proven XL-IRD due to RP2 or RPGR mutations, and 72 family members, of which 43 were affected, were identified from the NZ IRD Database. Comprehensive ophthalmic phenotyping, familial cosegregation, genotyping, and bioinformatics were undertaken. Main outcome measures were: RP2 and RPGR pathogenic variant spectrum, phenotype in males and females (symptoms, age of onset, visual acuity, refraction, electrophysiology, autofluorescence, retinal appearance), and genotype-phenotype correlation. RESULTS: For 32 families, 26 unique pathogenic variants were identified; in RP2 (n = 6, 21.9% of all families), RPGR exons 1-14 (n = 10, 43.75%), and RPGR-ORF15 (n = 10, 34.3%). Three RP2 and 8 RPGR exons 1-14 variants are novel, rare, and cosegregate. Thirty-one percent of carrier females were significantly affected, with 18.5% of families initially classified as autosomal dominant. Of five Polynesian families, 80% had novel disease-causing variants. One Maori family showed keratoconus segregating with an ORF15 variant. CONCLUSIONS: Significant disease was present in 31% of genetically proven female carriers, often leading to an erroneous presumption of the inheritance pattern. Pathogenic variants in 44% of the families were in exon 1-14 of RPGR, more frequent than usually described, which may inform the gene testing algorithm. Proving cosegregation in families for novel variants and identifying affected females and males translates to optimised clinical care and potential for gene therapy.


Assuntos
Proteínas do Olho , Proteínas de Ligação ao GTP , Doenças Genéticas Ligadas ao Cromossomo X , Proteínas de Membrana , Distrofias Retinianas , Retinose Pigmentar , Feminino , Humanos , Masculino , Análise Mutacional de DNA , Proteínas do Olho/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Genótipo , Proteínas de Ligação ao GTP/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Mutação , Linhagem , Fenótipo , Retinose Pigmentar/genética , Estudos Retrospectivos , Nova Zelândia
2.
Clin Exp Ophthalmol ; 45(9): 901-910, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28488341

RESUMO

IMPORTANCE: This study identifies unique genetic variation observed in a cohort of Maori and Polynesian patients with rod-cone retinal dystrophies using a targeted next-generation sequencing retinal disease gene panel. BACKGROUND: With over 250 retinal disease genes identified, genetic diagnosis is still only possible in 60-70% of individuals and even less within unique ethnic groups. DESIGN: Prospective genetic testing in patients with rod-cone retinal dystrophies identified from the New Zealand Inherited Retinal Disease Database, PARTICIPANTS: Sixteen patients of Maori and Polynesian ancestry. METHODS: Next-generation sequencing of a targeted retinal gene panel. Sanger sequencing for a novel PDE6B mutation in subsequent Maori patients. MAIN OUTCOME MEASURES: Genetic diagnosis, genotype-phenotype correlation. RESULTS: Thirteen unique pathogenic variants were identified in 9 of 16 (56.25%) patients in 10 different genes. A definitive genetic diagnosis was made in 7/16 patients (43.7%). Six changes were novel and not in public databases of human variation. In four patients, a homozygous, novel pathogenic variant (c.2197G > C, p.(Ala 733Pro)) in PDE6B was identified and also present in a further five similarly affected Maori patients. CONCLUSIONS AND RELEVANCE: Over half of the Maori and Polynesian patients with inherited rod-cone diseases have no pathogenic variant(s) detected with a targeted retinal next-generation sequencing strategy, which is supportive of novel genetic mechanisms in this population. A novel PDE6B founder variant is likely to account for 16% of recessive inherited retinal dystrophy in Maori. Careful characterization of the clinical presentation permits identification of further Maori patients with a similar phenotype and simplifies the diagnostic algorithm.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , DNA/genética , Mutação , Distrofias Retinianas/genética , Retinose Pigmentar/genética , Adulto , Idoso , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Análise Mutacional de DNA , Feminino , Seguimentos , Testes Genéticos , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Nova Zelândia/epidemiologia , Linhagem , Fenótipo , Polinésia/etnologia , Estudos Prospectivos , Distrofias Retinianas/etnologia , Distrofias Retinianas/metabolismo , Retinose Pigmentar/etnologia , Retinose Pigmentar/metabolismo , Adulto Jovem
3.
Development ; 140(6): 1330-41, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23406904

RESUMO

Dysfunction or death of photoreceptors is the primary cause of vision loss in retinal and macular degenerative diseases. As photoreceptors have an intimate relationship with the retinal pigment epithelium (RPE) for exchange of macromolecules, removal of shed membrane discs and retinoid recycling, an improved understanding of the development of the photoreceptor-RPE complex will allow better design of gene- and cell-based therapies. To explore the epigenetic contribution to retinal development we generated conditional knockout alleles of DNA methyltransferase 1 (Dnmt1) in mice. Conditional Dnmt1 knockdown in early eye development mediated by Rx-Cre did not produce lamination or cell fate defects, except in cones; however, the photoreceptors completely lacked outer segments despite near normal expression of phototransduction and cilia genes. We also identified disruption of RPE morphology and polarization as early as E15.5. Defects in outer segment biogenesis were evident with Dnmt1 exon excision only in RPE, but not when excision was directed exclusively to photoreceptors. We detected a reduction in DNA methylation of LINE1 elements (a measure of global DNA methylation) in developing mutant RPE as compared with neural retina, and of Tuba3a, which exhibited dramatically increased expression in mutant retina. These results demonstrate a unique function of DNMT1-mediated DNA methylation in controlling RPE apicobasal polarity and neural retina differentiation. We also establish a model to study the epigenetic mechanisms and signaling pathways that guide the modulation of photoreceptor outer segment morphogenesis by RPE during retinal development and disease.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , DNA (Citosina-5-)-Metiltransferases/genética , Morfogênese/genética , Segmento Externo das Células Fotorreceptoras da Retina/fisiologia , Epitélio Pigmentado da Retina/fisiologia , Animais , Permeabilidade da Membrana Celular/genética , Polaridade Celular/genética , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA/genética , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Morfogênese/fisiologia , Especificidade de Órgãos/genética , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Epitélio Pigmentado da Retina/embriologia , Epitélio Pigmentado da Retina/crescimento & desenvolvimento , Epitélio Pigmentado da Retina/metabolismo , Transcriptoma
4.
Ophthalmology ; 123(4): 709-22, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26786512

RESUMO

PURPOSE: Corneal dystrophies are a genetically heterogeneous group of disorders. We previously described a family with an autosomal dominant epithelial recurrent erosion dystrophy (ERED). We aimed to identify the underlying genetic cause of ERED in this family and 3 additional ERED families. We sought to characterize the potential function of the candidate genes using the human and zebrafish cornea. DESIGN: Case series study of 4 white families with a similar ERED. An experimental study was performed on human and zebrafish tissue to examine the putative biological function of candidate genes. PARTICIPANTS: Four ERED families, including 28 affected and 17 unaffected individuals. METHODS: HumanLinkage-12 arrays (Illumina, San Diego, CA) were used to genotype 17 family members. Next-generation exome sequencing was performed on an uncle-niece pair. Segregation of potential causative mutations was confirmed using Sanger sequencing. Protein expression was determined using immunohistochemistry in human and zebrafish cornea. Gene expression in zebrafish was assessed using whole-mount in situ hybridization. Morpholino-induced transient gene knockdown was performed in zebrafish embryos. MAIN OUTCOME MEASURES: Linkage microarray, exome analysis, DNA sequence analysis, immunohistochemistry, in situ hybridization, and morpholino-induced genetic knockdown results. RESULTS: Linkage microarray analysis identified a candidate region on chromosome chr10:12,576,562-112,763,135, and exploration of exome sequencing data identified 8 putative pathogenic variants in this linkage region. Two variants segregated in 06NZ-TRB1 with ERED: COL17A1 c.3156C→T and DNAJC9 c.334G→A. The COL17A1 c.3156C→T variant segregated in all 4 ERED families. We showed biologically relevant expression of these proteins in human cornea. Both proteins are expressed in the cornea of zebrafish embryos and adults. Zebrafish lacking Col17a1a and Dnajc9 during development show no gross corneal phenotype. CONCLUSIONS: The COL17A1 c.3156C→T variant is the likely causative mutation in our recurrent corneal erosion families, and its presence in 4 independent families suggests that it is prevalent in ERED. This same COL17A1 c.3156C→T variant recently was identified in a separate pedigree with ERED. Our study expands the phenotypic spectrum of COL17A1 disease from autosomal recessive epidermolysis bullosa to autosomal dominant ERED and identifies COL17A1 as a key protein in maintaining integrity of the corneal epithelium.


Assuntos
Processamento Alternativo/genética , Autoantígenos/genética , Distrofias Hereditárias da Córnea/genética , Epitélio Corneano/patologia , Mutação , Colágenos não Fibrilares/genética , Polimorfismo de Nucleotídeo Único , Adolescente , Adulto , Idoso , Animais , Criança , Distrofias Hereditárias da Córnea/diagnóstico , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/fisiologia , Inativação Gênica , Ligação Genética , Proteínas de Choque Térmico HSP40/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Repetições de Microssatélites , Microscopia Confocal , Pessoa de Meia-Idade , Linhagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra , Colágeno Tipo XVII
5.
BMC Genomics ; 16 Suppl 7: S11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26099324

RESUMO

BACKGROUND: Regulatory regions (e.g. promoters and enhancers) play an essential role in human development and disease. Many computational approaches have been developed to predict the regulatory regions using various genomic features such as sequence motifs and evolutionary conservation. However, these DNA sequence-based approaches do not reflect the tissue-specific nature of the regulatory regions. In this work, we propose to predict regulatory regions using multiple features derived from DNA methylation profile. RESULTS: We discovered several interesting features of the methylated CpG (mCpG) sites within regulatory regions. First, a hypomethylation status of CpGs within regulatory regions, compared to the genomic background methylation level, extended out >1000 bp from the center of the regulatory regions, demonstrating a high degree of correlation between the methylation statuses of neighboring mCpG sites. Second, when a regulatory region was inactive, as determined by histone mark differences between cell lines, methylation level of the mCpG site increased from a hypomethylated state to a hypermethylated state, the level of which was even higher than the genomic background. Third, a distinct set of sequence motifs was overrepresented surrounding mCpG sites within regulatory regions. Using 5 types of features derived from DNA methylation profiles, we were able to predict promoters and enhancers using machine-learning approach (support vector machine). The performances for prediction of promoters and enhancers are quite well, showing an area under the ROC curve (AUC) of 0.992 and 0.817, respectively, which is better than that simply based on methylation level, especially for prediction of enhancers. CONCLUSIONS: Our study suggests that DNA methylation features of mCpG sites can be used to predict regulatory regions.


Assuntos
Metilação de DNA , Elementos Facilitadores Genéticos , Genômica/métodos , Regiões Promotoras Genéticas , Algoritmos , Linhagem Celular , Ilhas de CpG , Humanos , Máquina de Vetores de Suporte
6.
BMC Genomics ; 16: 49, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25652663

RESUMO

BACKGROUND: DNA methylation plays an important role in regulating gene expression during many biological processes. However, the mechanism of DNA-methylation-dependent gene regulation is not fully understood. Here, we explore two possible DNA methylation regulatory mechanisms with opposite modes of gene expression regulation. RESULTS: By comparing the genome-wide methylation and expression patterns in different tissues, we find that majority of tissue-specific differentially methylated regions (T-DMRs) are negatively correlated with expression of their associated genes (negative T-DMRs), consistent with the classical dogma that DNA methylation suppresses gene expression; however, a significant portion of T-DMRs are positively correlated with gene expression (positive T-DMRs). We observe that the positive T-DMRs have similar genomic location as negative T-DMRs, except that the positive T-DMRs are more enriched in the promoter regions. Both positive and negative T-DMRs are enriched in DNase I hypersensitivity sites (DHSs), suggesting that both are likely to be functional. The CpG sites of both positive and negative T-DMRs are also more evolutionarily conserved than the genomic background. Interestingly, the putative target genes of the positive T-DMR are enriched for negative regulators such as transcriptional repressors, suggesting a novel mode of indirect DNA methylation inhibition of expression through transcriptional repressors. Likewise, two distinct sets of DNA sequence motifs exist for positive and negative T-DMRs, suggesting that two distinct sets of transcription factors (TFs) are involved in positive and negative regulation mediated by DNA methylation. CONCLUSIONS: We find both negative and positive association between T-DMRs and gene expression, which implies the existence of two different mechanisms of DNA methylation-dependent gene regulation.


Assuntos
Metilação de DNA/genética , Epigênese Genética , Regulação da Expressão Gênica , Animais , Encéfalo/metabolismo , Ilhas de CpG/genética , Desoxirribonuclease I/genética , Camundongos , Motivos de Nucleotídeos/genética , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Retina/metabolismo
7.
Nucleic Acids Res ; 41(18): 8503-14, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23887936

RESUMO

The exact role of intragenic DNA methylation in regulating tissue-specific gene regulation is unclear. Recently, the DNA-binding protein CTCF has been shown to participate in the regulation of alternative splicing in a DNA methylation-dependent manner. To globally evaluate the relationship between DNA methylation and tissue-specific alternative splicing, we performed genome-wide DNA methylation profiling of mouse retina and brain. In protein-coding genes, tissue-specific differentially methylated regions (T-DMRs) were preferentially located in exons and introns. Gene ontology and evolutionary conservation analysis suggest that these T-DMRs are likely to be biologically relevant. More than 14% of alternatively spliced genes were associated with a T-DMR. T-DMR-associated genes were enriched for developmental genes, suggesting that a specific set of alternatively spliced genes may be regulated through DNA methylation. Novel DNA sequences motifs overrepresented in T-DMRs were identified as being associated with positive and/or negative regulation of alternative splicing in a position-dependent context. The majority of these evolutionarily conserved motifs contain a CpG dinucleotide. Some transcription factors, which recognize these motifs, are known to be involved in splicing. Our results suggest that DNA methylation-dependent alternative splicing is widespread and lay the foundation for further mechanistic studies of the role of DNA methylation in tissue-specific splicing regulation.


Assuntos
Processamento Alternativo , Metilação de DNA , DNA/química , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Encéfalo/metabolismo , Sequência Conservada , Evolução Molecular , Éxons , Íntrons , Camundongos , Camundongos Endogâmicos C57BL , Motivos de Nucleotídeos , Especificidade de Órgãos , Proteínas/genética , Retina/metabolismo
8.
Ophthalmic Genet ; 38(6): 537-543, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28394649

RESUMO

BACKGROUND: To clinically characterize a cohort of patients with ectopia lentis (EL), or Marfanoid features in whom a definite genetic diagnosis of Marfan syndrome (MFS) had been excluded (atypical MFS), and to evaluate the contribution of mutations in ADAMTSL4 (OMIM * 610113), and P3H2 (LEPREL1; OMIM * 610341) to disease in this population. MATERIALS AND METHODS: Subjects underwent comprehensive ophthalmic examination, including keratometry. Mutational analysis of ADAMTSL4 and P3H2 was undertaken using PCR, high resolution melting analysis, and sequencing. The frequency of c.2237G>A; p.(Arg746His) was determined in an unaffected Polynesian cohort. Haplotype analysis used tagged single nucleotide polymorphic markers. RESULTS: Mutational analysis of ADAMTSL4 identified two pathogenic variants in ADAMTSL4 in 11/31 (35%) probands, consistent with the autosomal recessive EL phenotype. A recurrent, rare missense variant in ADAMTSL4, c.2237G>A; p.(Arg746His), was present in 10 probands -(8 homozygotes), predominantly of Polynesian descent, and all shared the same haplotype. p.(Arg746His) affects the Thrombospondin1 (TSP1) domain of the protein and is predicted to be pathogenic. No pathogenic variants in P3H2 were identified. CONCLUSION: A recurrent pathogenic ADAMTSL4 variant is a major cause of early onset autosomal recessive EL in a Cook Island Maori population and associated with a common haplotype, suggesting a founder effect. Children presenting under the age of 5 years, particularly of Cook Island or New Zealand Maori descent, with isolated ectopia lentis, should in the first instance be tested for this single variant.


Assuntos
Proteínas ADAMTS/genética , Ectopia do Cristalino/genética , Efeito Fundador , Mutação , Polimorfismo de Nucleotídeo Único , Criança , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Feminino , Haplótipos , Humanos , Masculino , Nova Zelândia , Linhagem , Reação em Cadeia da Polimerase , Polinésia , Pró-Colágeno-Prolina Dioxigenase/genética , Estudos Prospectivos
9.
Epigenetics ; 10(8): 698-707, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26067391

RESUMO

Age-related macular degeneration (AMD) is a major cause of blindness in the western world. While genetic studies have linked both common and rare variants in genes involved in regulation of the complement system to increased risk of development of AMD, environmental factors, such as smoking and nutrition, can also significantly affect the risk of developing the disease and the rate of disease progression. Since epigenetics has been implicated in mediating, in part, the disease risk associated with some environmental factors, we investigated a possible epigenetic contribution to AMD. We performed genome-wide DNA methylation profiling of blood from AMD patients and controls. No differential methylation site reached genome-wide significance; however, when epigenetic changes in and around known GWAS-defined AMD risk loci were explored, we found small but significant DNA methylation differences in the blood of neovascular AMD patients near age-related maculopathy susceptibility 2 (ARMS2), a top-ranked GWAS locus preferentially associated with neovascular AMD. The methylation level of one of the CpG sites significantly correlated with the genotype of the risk SNP rs10490924, suggesting a possible epigenetic mechanism of risk. Integrating genome-wide DNA methylation analysis of retina samples with and without AMD together with blood samples, we further identified a consistent, replicable change in DNA methylation in the promoter region of protease serine 50 (PRSS50). These methylation changes may identify sites in novel genes that are susceptible to non-genetic factors known to contribute to AMD development and progression.


Assuntos
Metilação de DNA/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Degeneração Macular/genética , Idoso , Idoso de 80 Anos ou mais , Feminino , Genótipo , Humanos , Degeneração Macular/sangue , Degeneração Macular/patologia , Masculino , Polimorfismo de Nucleotídeo Único , Retina/metabolismo , Retina/patologia , Fatores de Risco
10.
Epigenetics Chromatin ; 6(1): 17, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23759032

RESUMO

BACKGROUND: Growing evidence suggests that DNA methylation plays a role in tissue-specific differentiation. Current approaches to methylome analysis using enrichment with the methyl-binding domain protein (MBD) are restricted to large (≥1 µg) DNA samples, limiting the analysis of small tissue samples. Here we present a technique that enables characterization of genome-wide tissue-specific methylation patterns from nanogram quantities of DNA. RESULTS: We have developed a methodology utilizing MBD2b/MBD3L1 enrichment for methylated DNA, kinase pre-treated ligation-mediated PCR amplification (MeKL) and hybridization to the comprehensive high-throughput array for relative methylation (CHARM) customized tiling arrays, which we termed MeKL-chip. Kinase modification in combination with the addition of PEG has increased ligation-mediated PCR amplification over 20-fold, enabling >400-fold amplification of starting DNA. We have shown that MeKL-chip can be applied to as little as 20 ng of DNA, enabling comprehensive analysis of small DNA samples. Applying MeKL-chip to the mouse retina (a limited tissue source) and brain, 2,498 tissue-specific differentially methylated regions (T-DMRs) were characterized. The top five T-DMRs (Rgs20, Hes2, Nfic, Cckbr and Six3os1) were validated by pyrosequencing. CONCLUSIONS: MeKL-chip enables genome-wide methylation analysis of nanogram quantities of DNA with a wide range of observed-to-expected CpG ratios due to the binding properties of the MBD2b/MBD3L1 protein complex. This methodology enabled the first analysis of genome-wide methylation in the mouse retina, characterizing novel T-DMRs.

11.
Cell Rep ; 5(6): 1527-35, 2013 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-24373284

RESUMO

Age-related macular degeneration (AMD) is a leading cause of visual impairment worldwide. Aberrant DNA methylation within the promoter of IL17RC in peripheral blood mononuclear cells has recently been reported in AMD. To validate this association, we examined DNA methylation of the IL17RC promoter in peripheral blood. First, we used Illumina Human Methylation450 Bead Arrays, a widely accepted platform for measuring global DNA methylation. Second, methylation status at multiple sites within the IL17RC promoter was determined by bisulfite pyrosequencing in two cohorts. Third, a methylation-sensitive quantitative PCR-based assay was performed on a subset of samples. In contrast to previous findings, we did not find evidence of differential methylation between AMD cases and age-matched controls. We conclude that hypomethylation within the IL17RC gene promoter in peripheral blood is not suitable for use as a clinical biomarker of AMD. This study highlights the need for considerable replication of epigenetic association studies prior to clinical application.


Assuntos
Metilação de DNA , Leucócitos/metabolismo , Degeneração Macular/genética , Regiões Promotoras Genéticas , Receptores de Interleucina/genética , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Humanos , Degeneração Macular/sangue , Degeneração Macular/metabolismo , Masculino , Pessoa de Meia-Idade , Receptores de Interleucina/sangue
12.
Methods Mol Biol ; 884: 289-304, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22688715

RESUMO

Laser capture microdissection (LCM) is a useful method to isolate specific cells or cell layers of interest from heterogeneous tissues, such as the retina. The collected cells can be used for DNA, RNA, or protein analysis. We have applied LCM technology to isolate cells from the outer nuclear, inner nuclear, and ganglion cell layers of the retina for mRNA and microRNA (miRNA) expression and epigenetic (DNA methylation) analysis. Here, we describe the methods we have employed for sample preparation, LCM-based isolation of retinal layers, RNA/DNA extraction, RNA quality check, microRNA analysis by quantitative PCR, and DNA methylation analysis by bisulfite sequencing.


Assuntos
Metilação de DNA , Perfilação da Expressão Gênica/métodos , Microdissecção e Captura a Laser/métodos , MicroRNAs/genética , RNA Mensageiro/genética , Retina/metabolismo , Animais , DNA Complementar/síntese química , Regulação da Expressão Gênica , Camundongos , Controle de Qualidade , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA
13.
PLoS One ; 7(3): e32602, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22403679

RESUMO

Many studies have demonstrated that epigenetic mechanisms are important in the regulation of gene expression during embryogenesis, gametogenesis, and other forms of tissue-specific gene regulation. We sought to explore the possible role of epigenetics, specifically DNA methylation, in the establishment and maintenance of cell type-restricted gene expression in the retina. To assess the relationship between DNA methylation status and expression level of retinal genes, bisulfite sequence analysis of the 1000 bp region around the transcription start sites (TSS) of representative rod and cone photoreceptor-specific genes and gene expression analysis were performed in the WERI and Y79 human retinoblastoma cell lines. Next, the homologous genes in mouse were bisulfite sequenced in the retina and in non-expressing tissues. Finally, bisulfite sequencing was performed on isolated photoreceptor and non-photoreceptor retinal cells isolated by laser capture microdissection. Differential methylation of rhodopsin (RHO), retinal binding protein 3 (RBP3, IRBP) cone opsin, short-wave-sensitive (OPN1SW), cone opsin, middle-wave-sensitive (OPN1MW), and cone opsin, long-wave-sensitive (OPN1LW) was found in the retinoblastoma cell lines that inversely correlated with gene expression levels. Similarly, we found tissue-specific hypomethylation of the promoter region of Rho and Rbp3 in mouse retina as compared to non-expressing tissues, and also observed hypomethylation of retinal-expressed microRNAs. The Rho and Rbp3 promoter regions were unmethylated in expressing photoreceptor cells and methylated in non-expressing, non-photoreceptor cells from the inner nuclear layer. A third regional hypomethylation pattern of photoreceptor-specific genes was seen in a subpopulation of non-expressing photoreceptors (Rho in cones from the Nrl -/- mouse and Opn1sw in rods). These results demonstrate that a number of photoreceptor-specific genes have cell-specific differential DNA methylation that correlates inversely with their expression level. Furthermore, these cell-specific patterns suggest that DNA methylation may play an important role in modulating photoreceptor gene expression in the developing mammalian retina.


Assuntos
Metilação de DNA , Retina/citologia , Retina/metabolismo , Animais , Linhagem Celular , Clonagem Molecular , Ilhas de CpG/genética , Humanos , Camundongos , Especificidade de Órgãos , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Neurônios Retinianos/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/citologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Análise de Sequência de DNA , Sulfitos/metabolismo
14.
Fertil Steril ; 97(1): 147-53.e7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22112648

RESUMO

OBJECTIVE: To examine an IVF cohort for imprinted and genome-wide DNA methylation abnormalities. DESIGN: Retrospective study. SETTING: Research laboratory. PATIENT(S): DNA samples from a previously described IVF cohort that comprised 66 IVF-conceived prepubertal children (IVF, n = 34; intracytoplasmic sperm injection, n = 32) and 69 matched naturally conceived controls. INTERVENTION(S): DNA methylation was examined at four imprinted gene loci (H19, SNRPN, KCNQ1OT1, and IGF2) and satellite 2 using methylation-sensitive quantitative polymerase chain reaction (MSQ-PCR) followed by bisulfite sequencing at H19, SNRPN, and KCNQ1OT1. Methylated DNA immunoprecipitation (MeDIP) microarray with validation using the Sequenom MassARRAY EpiTYPER(®) platform was also used. MAIN OUTCOME MEASURE(S): Percentage of DNA methylation by MSQ-PCR, differential methylation based on microarray signal intensity, and percentage DNA methylation as determined by Sequenom MassARRAY EpiTYPER were compared. RESULT(S): No differences in percentage of methylation between the IVF and control group were observed at H19, KCNQ1OT1, SNRPN, or IGF2. Absence of aberrant imprinting was confirmed using bisulfite sequencing. Methylation of satellite 2 repeats (a surrogate for global methylation) showed no difference between the IVF and control groups. MeDIP was used to screen for differences in promoter methylation. Subsequent quantification of methylation of eight candidate genes using the Sequenom MassARRAY EpiTYPER system did not reveal any differential methylation. CONCLUSION(S): Low-level imprinting errors are not common in the IVF population.


Assuntos
Metilação de DNA/genética , Fertilização in vitro/estatística & dados numéricos , Doenças Genéticas Inatas/genética , Impressão Genômica/genética , Infertilidade/genética , Resultado da Gravidez/epidemiologia , Criança , Técnicas de Cultura Embrionária/estatística & dados numéricos , Feminino , Doenças Genéticas Inatas/epidemiologia , Predisposição Genética para Doença/epidemiologia , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Humanos , Infertilidade/epidemiologia , Infertilidade/terapia , Masculino , Gravidez , Estudos Retrospectivos , Injeções de Esperma Intracitoplásmicas/estatística & dados numéricos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa