Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Invest Ophthalmol Vis Sci ; 54(5): 3569-78, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23633653

RESUMO

PURPOSE: A mouse mutant identified during a recessive N-ethyl-N-nitrosourea (ENU) mutagenesis screen exhibited ocular hemorrhaging resulting in a blood-filled orbit, and hence was named "redeye." We aimed to identify the causal mutation in redeye, and evaluate it as a model for diabetic retinopathy (DR). METHODS: The causative gene mutation in redeye was identified by haplotype mapping followed by exome sequencing. Glucose tolerance tests, detailed histologic and immunofluorescence analyses, and vascular permeability assays were performed to determine the affect of redeye on glucose metabolism, pericyte recruitment, and the development of the retinal vasculature and blood-retinal barrier (BRB). RESULTS: A mutation was identified in the Pdgfrb gene at position +2 of intron 6. We show that this change causes partial loss of normal splicing resulting in a frameshift and premature termination, and, therefore, a substantial reduction in normal Pdgfrb transcript. The animals exhibit defective pericyte recruitment restricted to the central nervous system (CNS) causing basement membrane and vascular patterning defects, impaired vascular permeability, and aberrant BRB development, resulting in vascular leakage and retinal ganglion cell apoptosis. Despite exhibiting classic features of diabetic retinopathy, redeye glucose tolerance is normal. CONCLUSIONS: The Pdgfrb(redeye/redeye) mice exhibit all of the features of nonproliferative DR, including retinal neurodegeneration. In addition, the perinatal onset of the CNS-specific vascular phenotype negates the need to age animals or manage diabetic complications in other organs. Therefore, they are a more useful model for diseases involving pericyte deficiencies, such as DR, than those currently being used.


Assuntos
Barreira Hematorretiniana/patologia , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Animais , Sequência de Bases , Membrana Basal/patologia , Barreira Hematorretiniana/metabolismo , Códon sem Sentido/genética , Retinopatia Diabética/metabolismo , Modelos Animais de Doenças , Éxons/genética , Feminino , Mutação da Fase de Leitura/genética , Teste de Tolerância a Glucose , Haplótipos , Íntrons/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Mutantes , Dados de Sequência Molecular , Mutagênese , Pericitos/patologia , Sítios de Splice de RNA/genética
2.
PLoS One ; 8(1): e54173, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23326594

RESUMO

The transcription factor Pax6 is a developmental regulator with a crucial role in development of the eye, brain, and olfactory system. Pax6 is also required for correct development of the endocrine pancreas and specification of hormone producing endocrine cell types. Glucagon-producing cells are almost completely lost in Pax6-null embryos, and insulin-expressing beta and somatostatin-expressing delta cells are reduced. While the developmental role of Pax6 is well-established, investigation of a further role for Pax6 in the maintenance of adult pancreatic function is normally precluded due to neonatal lethality of Pax6-null mice. Here a tamoxifen-inducible ubiquitous Cre transgene was used to inactivate Pax6 at 6 months of age in a conditional mouse model to assess the effect of losing Pax6 function in adulthood. The effect on glucose homeostasis and the expression of key islet cell markers was measured. Homozygous Pax6 deletion mice, but not controls, presented with all the symptoms of classical diabetes leading to severe weight loss requiring termination of the experiment five weeks after first tamoxifen administration. Immunohistochemical analysis of the pancreata revealed almost complete loss of Pax6 and much reduced expression of insulin, glucagon, and somatostatin. Several other markers of islet cell function were also affected. Notably, strong upregulation in the number of ghrelin-expressing endocrine cells was observed. These findings demonstrate that Pax6 is essential for adult maintenance of glucose homeostasis and function of the endocrine pancreas.


Assuntos
Diabetes Mellitus , Proteínas do Olho/metabolismo , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Ilhotas Pancreáticas/crescimento & desenvolvimento , Fatores de Transcrição Box Pareados/metabolismo , Pâncreas/crescimento & desenvolvimento , Proteínas Repressoras/metabolismo , Animais , Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatologia , Proteínas do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Grelina/genética , Grelina/metabolismo , Glucose/genética , Proteínas de Homeodomínio/genética , Ilhotas Pancreáticas/metabolismo , Camundongos , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Pâncreas/citologia , Pâncreas/metabolismo , Proteínas Repressoras/genética , Regulação para Cima , Redução de Peso
3.
Invest Ophthalmol Vis Sci ; 53(7): 3951-8, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22577079

RESUMO

PURPOSE: Mutations in the retinitis pigmentosa (RP) GTPase regulator (RPGR) gene account for more than 70% of X-linked RP cases. This study aims to characterize the proximal promoter region of the human RPGR gene. METHODS: The 5'-flanking region (5 kb) of human RPGR was cloned and sequenced. A potential transcription start site and transcription factor binding motifs were identified by bioinformatic analysis. Constructs containing the putative human RPGR promoter region upstream of a luciferase reporter gene were generated and analyzed by transient transfection and luciferase assays. Transgenic mouse lines carrying a 3-kb human RPGR promoter sequence fused to lacZ were generated and RPGR proximal promoter activity was analyzed by X-gal staining. RESULTS: Bioinformatic analyses of the human RPGR 5'-flanking region uncovered potential transcription factor binding sites and a CpG island. Transient transfection assays with RPGR promoter/luciferase reporter constructs revealed a 980-bp fragment (-952 to +28) that produced higher levels of luciferase activity. Mutagenesis identified a putative Sp1 binding site that was critical for regulating transcriptional activity. We generated transgenic mice in which a lacZ reporter gene was controlled by the 3-kb upstream region of RPGR. ß-galactosidase expression was predominantly found in mouse retina, brain, and kidney. In the retina, the photoreceptor cell layer showed the strongest ß-galactosidase staining. CONCLUSIONS: Our study defined the human RPGR proximal promoter region in which a 3-kb fragment contained sufficient regulatory elements to control RPGR expression in mouse retina and other tissues. Characterization of the RPGR promoter will facilitate understanding of the functional role of RPGR in the retina and gene therapy of X-linked RP.


Assuntos
Proteínas do Olho/genética , Regulação da Expressão Gênica , Mutação , Retina/metabolismo , Retinose Pigmentar/genética , Animais , Linhagem Celular , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Proteínas do Olho/biossíntese , Feminino , Fatores de Troca do Nucleotídeo Guanina , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Retina/patologia , Retinose Pigmentar/metabolismo , Retinose Pigmentar/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional
4.
Hum Mol Genet ; 15(16): 2457-67, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16825286

RESUMO

The X-linked gene filamin A (Flna) encodes a widely expressed actin-binding protein that crosslinks actin into orthogonal networks and interacts with a variety of other proteins including membrane proteins, integrins, transmembrane receptor complexes and second messengers, thus forming an important intracellular signalling scaffold. Heterozygous loss of function of human FLNA causes periventricular nodular heterotopia in females and is generally lethal (cause unknown) in hemizygous males. Missense FLNA mutations underlie a spectrum of disorders affecting both sexes that feature skeletal dysplasia accompanied by a variety of other abnormalities. Dilp2 is an X-linked male-lethal mouse mutation that was induced by N-ethyl-N-nitrosourea. We report here that Dilp2 is caused by a T-to-A transversion that converts a tyrosine codon to a stop codon in the Flna gene (Y2388X), leading to absence of the Flna protein and male lethality because of incomplete septation of the outflow tract of the heart, which produces common arterial trunk. A proportion of both male and female mutant mice have other cardiac defects including ventricular septal defect. In addition, mutant males have midline fusion defects manifesting as sternum and palate abnormalities. Carrier females exhibit milder sternum and palate defects and misshapen pupils. These results define crucial roles for Flna in development, demonstrate that X-linked male lethal mutations can be recovered from ENU mutagenesis screens and suggest possible explanations for lethality of human males hemizygous for null alleles of FLNA.


Assuntos
Osso e Ossos/anormalidades , Proteínas Contráteis/genética , Proteínas Contráteis/fisiologia , Cardiopatias Congênitas/genética , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/fisiologia , Osteogênese/genética , Animais , Perda do Embrião/etiologia , Perda do Embrião/genética , Feminino , Filaminas , Expressão Gênica , Genes Letais , Genes Ligados ao Cromossomo X/fisiologia , Cardiopatias Congênitas/ultraestrutura , Heterozigoto , Perda de Heterozigosidade/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Proteínas Mutantes/fisiologia , Palato/anormalidades , Fenótipo , Mutação Puntual/fisiologia , Gravidez , Distúrbios Pupilares/genética , Caracteres Sexuais
5.
Mol Cell Endocrinol ; 253(1-2): 14-21, 2006 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-16698177

RESUMO

In order to purify and characterize nestin-positive cells in the developing pancreas a transgenic mouse was generated, in which the enhanced green fluorescent protein (EGFP) was driven by the nestin second intronic enhancer and upstream promoter. In keeping with previous studies on the distribution of nestin, EGFP was expressed in the developing embryo in neurones in the brain, eye, spinal cord, tail bud and glial cells in the small intestine. In the pancreas there was no detectable EGFP at embryonic day 11.5 (E11.5). EGFP expression appeared at E12.5 and increased in intensity through E14.5, E18.5 and post-natal day 1. Flow cytometry was used to quantify and purify the EGFP positive population in the E15.5 pancreas. The purified (96%) EGFP-expressing cells, which represent 20% of the total cell population, were shown by RT/PCR to express exocrine cell markers (amylase and P48) and endocrine cell markers (insulin 1, insulin 2, and Ngn3). They also expressed, at a lower level, PDX-1, Isl-1, and the islet hormones pancreatic polypeptide, glucagon and somatostatin as well as GLUT2, the stem cell marker ABCG2 and PECAM, a marker of endothelial cells. It was further shown by immunocytochemistry of the E15.5 pancreas that EGFP colocalised in separate subpopulations of cells that expressed nestin, insulin and amylase. These results support the conclusion that nestin expressing cells can give rise to both endocrine and exocrine cells. The ability to purify these putative progenitor cells may provide further insights into their properties and function.


Assuntos
Proteínas de Fluorescência Verde/genética , Proteínas de Filamentos Intermediários/genética , Ilhotas Pancreáticas/embriologia , Proteínas do Tecido Nervoso/genética , Pâncreas Exócrino/enzimologia , Amilases/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/enzimologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Nestina , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo
6.
Invest Ophthalmol Vis Sci ; 46(9): 3443-50, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16123450

RESUMO

PURPOSE: To identify the underlying molecular defects causing retinal degeneration in seven N-ethyl-N-nitrosourea (ENU) induced mutant alleles of the Pde6b gene and to analyze the timescale of retinal degeneration in these new models of retinitis pigmentosa. METHODS: Conformation sensitive capillary electrophoresis and DNA sequencing were used to identify the mutations in the Pde6b gene. Visual acuity testing was performed with a visual-tracking drum at ages ranging from postnatal day 25 to week 10. Retinal examination was performed with an indirect ophthalmoscope. Animals were killed and eyes were prepared for histologic analysis. RESULTS: Point mutations in the seven new alleles of Pde6b were identified: Three generated premature stop codons, two were missense mutations, and two were splice mutations. The three stop codon mutants and one of the splice mutants had phenotypes indistinguishable from the Pde6b(rd1) mouse in rapidity of onset of retinal degeneration, suggesting that they are null alleles. However, the remaining alleles showed slower onset of retinal degeneration, as determined by visual acuity testing, fundus examination, and histology, indicating that they are hypomorphic alleles. CONCLUSIONS: These data demonstrate a correlation between genotype and phenotype. Four of the mutants with severe genetic lesions have rapid onset of retinal degeneration, as determined by fundus examination. These mice were indistinguishable from Pde6b(rd1) mice, which are effectively blind by 3 weeks of age. In contrast, the milder genetic lesions show a slower progression of the disease and provide the community with models that more closely mimic human retinitis pigmentosa.


Assuntos
Mutação , Diester Fosfórico Hidrolases/genética , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/genética , Alquilantes/toxicidade , Alelos , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6 , Análise Mutacional de DNA , Modelos Animais de Doenças , Progressão da Doença , Eletroforese Capilar , Etilnitrosoureia/toxicidade , Feminino , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Fenótipo , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/enzimologia , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/enzimologia , Degeneração Retiniana/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Acuidade Visual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA