Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Dis Model Mech ; 14(2)2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33462143

RESUMO

Variants in the LIM homeobox transcription factor 1-beta (LMX1B) gene predispose individuals to elevated intraocular pressure (IOP), a key risk factor for glaucoma. However, the effect of LMX1B mutations varies widely between individuals. To better understand the mechanisms underlying LMX1B-related phenotypes and individual differences, we backcrossed the Lmx1bV265D (also known as Lmx1bIcst ) allele onto the C57BL/6J (B6), 129/Sj (129), C3A/BLiA-Pde6b+ /J (C3H) and DBA/2J-Gpnmb+ (D2-G) mouse strain backgrounds. Strain background had a significant effect on the onset and severity of ocular phenotypes in Lmx1bV265D/+ mutant mice. Mice of the B6 background were the most susceptible to developing abnormal IOP distribution, severe anterior segment developmental anomalies (including malformed eccentric pupils, iridocorneal strands and corneal abnormalities) and glaucomatous nerve damage. By contrast, Lmx1bV265D mice of the 129 background were the most resistant to developing anterior segment abnormalities, had less severe IOP elevation than B6 mutants at young ages and showed no detectable nerve damage. To identify genetic modifiers of susceptibility to Lmx1bV265D -induced glaucoma-associated phenotypes, we performed a mapping cross between mice of the B6 (susceptible) and 129 (resistant) backgrounds. We identified a modifier locus on Chromosome 18, with the 129 allele(s) substantially lessening severity of ocular phenotypes, as confirmed by congenic analysis. By demonstrating a clear effect of genetic background in modulating Lmx1b-induced phenotypes, providing a panel of strains with different phenotypic severities and identifying a modifier locus, this study lays a foundation for better understanding the roles of LMX1B in glaucoma with the goal of developing new treatments.


Assuntos
Segmento Anterior do Olho/fisiopatologia , Anormalidades do Olho/genética , Predisposição Genética para Doença , Glaucoma/genética , Proteínas com Homeodomínio LIM/genética , Fatores de Transcrição/genética , Alelos , Animais , Cruzamentos Genéticos , Modelos Animais de Doenças , Feminino , Genes Homeobox , Patrimônio Genético , Genótipo , Pressão Intraocular , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Nervo Óptico/patologia , Fenótipo , Especificidade da Espécie
2.
PLoS Genet ; 16(4): e1008583, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32236127

RESUMO

The precise control of eye size is essential for normal vision. TMEM98 is a highly conserved and widely expressed gene which appears to be involved in eye size regulation. Mutations in human TMEM98 are found in patients with nanophthalmos (very small eyes) and variants near the gene are associated in population studies with myopia and increased eye size. As complete loss of function mutations in mouse Tmem98 result in perinatal lethality, we produced mice deficient for Tmem98 in the retinal pigment epithelium (RPE), where Tmem98 is highly expressed. These mice have greatly enlarged eyes that are very fragile with very thin retinas, compressed choroid and thin sclera. To gain insight into the mechanism of action we used a proximity labelling approach to discover interacting proteins and identified MYRF as an interacting partner. Mutations of MYRF are also associated with nanophthalmos. The protein is an endoplasmic reticulum-tethered transcription factor which undergoes autoproteolytic cleavage to liberate the N-terminal part which then translocates to the nucleus where it acts as a transcription factor. We find that TMEM98 inhibits the self-cleavage of MYRF, in a novel regulatory mechanism. In RPE lacking TMEM98, MYRF is ectopically activated and abnormally localised to the nuclei. Our findings highlight the importance of the interplay between TMEM98 and MYRF in determining the size of the eye.


Assuntos
Olho/anatomia & histologia , Olho/metabolismo , Proteínas de Membrana/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Animais , Eletrorretinografia , Anormalidades do Olho/genética , Feminino , Deleção de Genes , Mutação com Perda de Função , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Tamanho do Órgão/genética , Ligação Proteica , Transporte Proteico , Epitélio Pigmentado da Retina/anormalidades , Epitélio Pigmentado da Retina/metabolismo , Retinaldeído/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
3.
Sci Rep ; 10(1): 437, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31949211

RESUMO

Fam151b is a mammalian homologue of the C. elegans menorin gene, which is involved in neuronal branching. The International Mouse Phenotyping Consortium (IMPC) aims to knock out every gene in the mouse and comprehensively phenotype the mutant animals. This project identified Fam151b homozygous knock-out mice as having retinal degeneration. We show they have no photoreceptor function from eye opening, as demonstrated by a lack of electroretinograph (ERG) response. Histological analysis shows that during development of the eye the correct number of cells are produced and that the layers of the retina differentiate normally. However, after eye opening at P14, Fam151b mutant eyes exhibit signs of retinal stress and rapidly lose photoreceptor cells. We have mutated the second mammalian menorin homologue, Fam151a, and homozygous mutant mice have no discernible phenotype. Sequence analysis indicates that the FAM151 proteins are members of the PLC-like phosphodiesterase superfamily. However, the substrates and function of the proteins remains unknown.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Proteínas de Membrana/genética , Retina/fisiologia , Homologia de Sequência do Ácido Nucleico , Sequência de Aminoácidos , Animais , Contagem de Células , Técnicas de Inativação de Genes , Humanos , Camundongos , Modelos Moleculares , Mutação , Células Fotorreceptoras de Vertebrados/citologia , Conformação Proteica , Retina/citologia
4.
Invest Ophthalmol Vis Sci ; 60(8): 2875-2887, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31266059

RESUMO

Purpose: We previously found a dominant mutation, Rwhs, causing white spots on the retina accompanied by retinal folds. Here we identify the mutant gene to be Tmem98. In humans, mutations in the orthologous gene cause nanophthalmos. We modeled these mutations in mice and characterized the mutant eye phenotypes of these and Rwhs. Methods: The Rwhs mutation was identified to be a missense mutation in Tmem98 by genetic mapping and sequencing. The human TMEM98 nanophthalmos missense mutations were made in the mouse gene by CRISPR-Cas9. Eyes were examined by indirect ophthalmoscopy and the retinas imaged using a retinal camera. Electroretinography was used to study retinal function. Histology, immunohistochemistry, and electron microscopy techniques were used to study adult eyes. Results: An I135T mutation of Tmem98 causes the dominant Rwhs phenotype and is perinatally lethal when homozygous. Two dominant missense mutations of TMEM98, A193P and H196P, are associated with human nanophthalmos. In the mouse these mutations cause recessive retinal defects similar to the Rwhs phenotype, either alone or in combination with each other, but do not cause nanophthalmos. The retinal folds did not affect retinal function as assessed by electroretinography. Within the folds there was accumulation of disorganized outer segment material as demonstrated by immunohistochemistry and electron microscopy, and macrophages had infiltrated into these regions. Conclusions: Mutations in the mouse orthologue of the human nanophthalmos gene TMEM98 do not result in small eyes. Rather, there is localized disruption of the laminar structure of the photoreceptors.


Assuntos
Proteínas de Membrana/genética , Microftalmia/genética , Mutação de Sentido Incorreto , Células Fotorreceptoras de Vertebrados/patologia , Doenças Retinianas/genética , Animais , Comprimento Axial do Olho/patologia , Sistemas CRISPR-Cas , Eletrorretinografia , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microftalmia/patologia , Microscopia Eletrônica de Transmissão , Oftalmoscopia , Reação em Cadeia da Polimerase , Doenças Retinianas/patologia
5.
Dis Model Mech ; 11(12)2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30478029

RESUMO

Isocitrate dehydrogenase (IDH) is an enzyme required for the production of α-ketoglutarate from isocitrate. IDH3 generates the NADH used in the mitochondria for ATP production, and is a tetramer made up of two α, one ß and one γ subunit. Loss-of-function and missense mutations in both IDH3A and IDH3B have previously been implicated in families exhibiting retinal degeneration. Using mouse models, we investigated the role of IDH3 in retinal disease and mitochondrial function. We identified mice with late-onset retinal degeneration in a screen of ageing mice carrying an ENU-induced mutation, E229K, in Idh3a Mice homozygous for this mutation exhibit signs of retinal stress, indicated by GFAP staining, as early as 3 months, but no other tissues appear to be affected. We produced a knockout of Idh3a and found that homozygous mice do not survive past early embryogenesis. Idh3a-/E229K compound heterozygous mutants exhibit a more severe retinal degeneration compared with Idh3aE229K/E229K homozygous mutants. Analysis of mitochondrial function in mutant cell lines highlighted a reduction in mitochondrial maximal respiration and reserve capacity levels in both Idh3aE229K/E229K and Idh3a-/E229K cells. Loss-of-function Idh3b mutants do not exhibit the same retinal degeneration phenotype, with no signs of retinal stress or reduction in mitochondrial respiration. It has previously been reported that the retina operates with a limited mitochondrial reserve capacity and we suggest that this, in combination with the reduced reserve capacity in mutants, explains the degenerative phenotype observed in Idh3a mutant mice.This article has an associated First Person interview with the first author of the paper.


Assuntos
Isocitrato Desidrogenase/genética , Mitocôndrias/patologia , Mutação/genética , Degeneração Retiniana/genética , Degeneração Retiniana/fisiopatologia , Animais , Fibroblastos/metabolismo , Genótipo , Isocitrato Desidrogenase/metabolismo , Mutação com Perda de Função/genética , Camundongos , Mutação de Sentido Incorreto/genética , Fenótipo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Retina/patologia , Retina/fisiopatologia
6.
Nat Commun ; 7: 12444, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27534441

RESUMO

Determining the genetic bases of age-related disease remains a major challenge requiring a spectrum of approaches from human and clinical genetics to the utilization of model organism studies. Here we report a large-scale genetic screen in mice employing a phenotype-driven discovery platform to identify mutations resulting in age-related disease, both late-onset and progressive. We have utilized N-ethyl-N-nitrosourea mutagenesis to generate pedigrees of mutagenized mice that were subject to recurrent screens for mutant phenotypes as the mice aged. In total, we identify 105 distinct mutant lines from 157 pedigrees analysed, out of which 27 are late-onset phenotypes across a range of physiological systems. Using whole-genome sequencing we uncover the underlying genes for 44 of these mutant phenotypes, including 12 late-onset phenotypes. These genes reveal a number of novel pathways involved with age-related disease. We illustrate our findings by the recovery and characterization of a novel mouse model of age-related hearing loss.


Assuntos
Envelhecimento/genética , Testes Genéticos , Mutagênese/genética , Animais , Cóclea/metabolismo , Modelos Animais de Doenças , Epitélio/ultraestrutura , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Feminino , Audição/genética , Masculino , Camundongos Endogâmicos C57BL , Mutação/genética , Linhagem , Fenótipo
7.
Dis Model Mech ; 8(12): 1555-68, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26542706

RESUMO

Mitogen-activated protein kinase, MAP3K1, plays an important role in a number of cellular processes, including epithelial migration during eye organogenesis. In addition, studies in keratinocytes indicate that MAP3K1 signalling through JNK is important for actin stress fibre formation and cell migration. However, MAP3K1 can also act independently of JNK in the regulation of cell proliferation and apoptosis. We have identified a mouse mutant, goya, which exhibits the eyes-open-at-birth and microphthalmia phenotypes. In addition, these mice also have hearing loss. The goya mice carry a splice site mutation in the Map3k1 gene. We show that goya and kinase-deficient Map3k1 homozygotes initially develop supernumerary cochlear outer hair cells (OHCs) that subsequently degenerate, and a progressive profound hearing loss is observed by 9 weeks of age. Heterozygote mice also develop supernumerary OHCs, but no cellular degeneration or hearing loss is observed. MAP3K1 is expressed in a number of inner-ear cell types, including outer and inner hair cells, stria vascularis and spiral ganglion. Investigation of targets downstream of MAP3K1 identified an increase in p38 phosphorylation (Thr180/Tyr182) in multiple cochlear tissues. We also show that the extra OHCs do not arise from aberrant control of proliferation via p27KIP1. The identification of the goya mutant reveals a signalling molecule involved with hair-cell development and survival. Mammalian hair cells do not have the ability to regenerate after damage, which can lead to irreversible sensorineural hearing loss. Given the observed goya phenotype, and the many diverse cellular processes that MAP3K1 is known to act upon, further investigation of this model might help to elaborate upon the mechanisms underlying sensory hair cell specification, and pathways important for their survival. In addition, MAP3K1 is revealed as a new candidate gene for human sensorineural hearing loss.


Assuntos
Células Ciliadas Auditivas Externas/enzimologia , Células Ciliadas Auditivas Externas/patologia , MAP Quinase Quinase Quinase 1/genética , Mutação Puntual/genética , Animais , Sequência de Bases , Proliferação de Células , Sobrevivência Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Feminino , Células Ciliadas Auditivas Externas/ultraestrutura , Audição , Perda Auditiva/enzimologia , Perda Auditiva/patologia , Perda Auditiva/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dados de Sequência Molecular , Fosforilação , Visão Ocular , beta-Galactosidase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Invest Ophthalmol Vis Sci ; 56(5): 3015-26, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25736793

RESUMO

PURPOSE: As part of a large scale systematic screen to determine the effects of gene knockout mutations in mice, a retinal phenotype was found in mice lacking the Slc9a8 gene, encoding the sodium/hydrogen ion exchange protein NHE8. We aimed to characterize the mutant phenotype and the role of sodium/hydrogen ion exchange in retinal function. METHODS: Detailed histology characterized the pathological consequences of Slc9a8 mutation, and retinal function was assessed by electroretinography (ERG). A conditional allele was used to identify the cells in which NHE8 function is critical for retinal function, and mutant cells analyzed for the effect of the mutation on endosomes. RESULTS: Histology of mutant retinas reveals a separation of photoreceptors from the RPE and infiltration by macrophages. There is a small reduction in photoreceptor length and a mislocalization of visual pigments. The ERG testing reveals a deficit in rod and cone pathway function. The RPE shows abnormal morphology, and mutation of Slc9a8 in only RPE cells recapitulates the mutant phenotype. The NHE8 protein localizes to endosomes, and mutant cells have much smaller recycling endosomes. CONCLUSIONS: The NHE8 protein is required in the RPE to maintain correct regulation of endosomal volume and/or pH which is essential for the cellular integrity and subsequent function of RPE.


Assuntos
Mutação , Doenças Retinianas/genética , Epitélio Pigmentado da Retina/patologia , Trocadores de Sódio-Hidrogênio/genética , Animais , Técnicas de Cultura de Células , Modelos Animais de Doenças , Eletrorretinografia , Imunofluorescência , Técnicas de Inativação de Genes , Inativação Gênica , Pressão Intraocular , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Oftalmoscopia , Plasmídeos , Reação em Cadeia da Polimerase em Tempo Real , Doenças Retinianas/diagnóstico
9.
PLoS Genet ; 10(5): e1004359, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24809698

RESUMO

Mutations in the LIM-homeodomain transcription factor LMX1B cause nail-patella syndrome, an autosomal dominant pleiotrophic human disorder in which nail, patella and elbow dysplasia is associated with other skeletal abnormalities and variably nephropathy and glaucoma. It is thought to be a haploinsufficient disorder. Studies in the mouse have shown that during development Lmx1b controls limb dorsal-ventral patterning and is also required for kidney and eye development, midbrain-hindbrain boundary establishment and the specification of specific neuronal subtypes. Mice completely deficient for Lmx1b die at birth. In contrast to the situation in humans, heterozygous null mice do not have a mutant phenotype. Here we report a novel mouse mutant Icst, an N-ethyl-N-nitrosourea-induced missense substitution, V265D, in the homeodomain of LMX1B that abolishes DNA binding and thereby the ability to transactivate other genes. Although the homozygous phenotypic consequences of Icst and the null allele of Lmx1b are the same, heterozygous Icst elicits a phenotype whilst the null allele does not. Heterozygous Icst causes glaucomatous eye defects and is semi-lethal, probably due to kidney failure. We show that the null phenotype is rescued more effectively by an Lmx1b transgene than is Icst. Co-immunoprecipitation experiments show that both wild-type and Icst LMX1B are found in complexes with LIM domain binding protein 1 (LDB1), resulting in lower levels of functional LMX1B in Icst heterozygotes than null heterozygotes. We conclude that Icst is a dominant-negative allele of Lmx1b. These findings indicate a reassessment of whether nail-patella syndrome is always haploinsufficient. Furthermore, Icst is a rare example of a model of human glaucoma caused by mutation of the same gene in humans and mice.


Assuntos
Genes Dominantes , Genes Letais , Glaucoma/genética , Proteínas com Homeodomínio LIM/genética , Fatores de Transcrição/genética , Alelos , Animais , Padronização Corporal , Dimerização , Heterozigoto , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto
10.
J Neurosci ; 32(22): 7672-84, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649246

RESUMO

Cytoskeleton-associated proteins play key roles not only in regulating cell morphology and migration but also in proliferation. Mutations in the cytoskeleton-associated gene filamin A (FlnA) cause the human disorder periventricular heterotopia (PH). PH is a disorder of neural stem cell development that is characterized by disruption of progenitors along the ventricular epithelium and subsequent formation of ectopic neuronal nodules. FlnA-dependent regulation of cytoskeletal dynamics is thought to direct neural progenitor migration and proliferation. Here we show that embryonic FlnA-null mice exhibited a reduction in brain size and decline in neural progenitor numbers over time. The drop in the progenitor population was not attributable to cell death or changes in premature differentiation, but to prolonged cell cycle duration. Suppression of FlnA led to prolongation of the entire cell cycle length, principally in M phase. FlnA loss impaired degradation of cyclin B1-related proteins, thereby delaying the onset and progression through mitosis. We found that the cdk1 kinase Wee1 bound FlnA, demonstrated increased expression levels after loss of FlnA function, and was associated with increased phosphorylation of cdk1. Phosphorylation of cdk1 inhibited activation of the anaphase promoting complex degradation system, which was responsible for cyclin B1 degradation and progression through mitosis. Collectively, our results demonstrate a molecular mechanism whereby FlnA loss impaired G2 to M phase entry, leading to cell cycle prolongation, compromised neural progenitor proliferation, and reduced brain size.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Córtex Cerebral/fisiologia , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Células-Tronco Neurais/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fatores Etários , Animais , Bromodesoxiuridina/metabolismo , Proteína Quinase CDC2/genética , Ciclo Celular/genética , Diferenciação Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Proteínas Contráteis/deficiência , Ciclina B1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Filaminas , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica/genética , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67 , Camundongos , Camundongos Transgênicos , Microcefalia/genética , Proteínas dos Microfilamentos/deficiência , Heterotopia Nodular Periventricular/genética , Heterotopia Nodular Periventricular/patologia , Fosforilação/genética , Proteínas com Domínio T/metabolismo , Tirosina/metabolismo
11.
BMC Res Notes ; 5: 122, 2012 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-22369496

RESUMO

BACKGROUND: Some abnormalities of mouse corneal epithelial maintenance can be identified by the atypical mosaic patterns they produce in X-chromosome inactivation mosaics and chimeras. Human FLNA/+ females, heterozygous for X-linked, filamin A gene (FLNA) mutations, display a range of disorders and X-inactivation mosaicism is sometimes quantitatively unbalanced. FlnaDilp2/+ mice, heterozygous for an X-linked filamin A (Flna) nonsense mutation have variable eye, skeletal and other abnormalities, but X-inactivation mosaicism has not been investigated. The aim of this study was to determine whether X-inactivation mosaicism in the corneal epithelia of FlnaDilp2/+ mice was affected in any way that might predict abnormal corneal epithelial maintenance. RESULTS: X-chromosome inactivation mosaicism was studied in the corneal epithelium and a control tissue (liver) of FlnaDilp2/+ and wild-type (WT) female X-inactivation mosaics, hemizygous for the X-linked, LacZ reporter H253 transgene, using ß-galactosidase histochemical staining. The corneal epithelia of FlnaDilp2/+ and WT X-inactivation mosaics showed similar radial, striped patterns, implying epithelial cell movement was not disrupted in FlnaDilp2/+ corneas. Corrected stripe numbers declined with age overall (but not significantly for either genotype individually), consistent with previous reports suggesting an age-related reduction in stem cell function. Corrected stripe numbers were not reduced in FlnaDilp2/+ compared with WT X-inactivation mosaics and mosaicism was not significantly more unbalanced in the corneal epithelia or livers of FlnaDilp2/+ than wild-type Flna+/+ X-inactivation mosaics. CONCLUSIONS: Mosaic analysis identified no major effect of the mouse FlnaDilp2 mutation on corneal epithelial maintenance or the balance of X-inactivation mosaicism in the corneal epithelium or liver.


Assuntos
Epitélio Corneano/metabolismo , Proteínas do Olho/genética , Fígado/metabolismo , Mosaicismo , Proteínas do Tecido Nervoso/genética , Inativação do Cromossomo X , Fatores Etários , Animais , Movimento Celular , Epitélio Corneano/citologia , Feminino , Filaminas , Genes Ligados ao Cromossomo X , Genótipo , Heterozigoto , Histocitoquímica , Humanos , Óperon Lac , Fígado/citologia , Camundongos , Camundongos Transgênicos , Mutação , Transgenes , beta-Galactosidase/análise
12.
Hum Mol Genet ; 21(6): 1272-86, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22121117

RESUMO

MKS3, encoding the transmembrane receptor meckelin, is mutated in Meckel-Gruber syndrome (MKS), an autosomal-recessive ciliopathy. Meckelin localizes to the primary cilium, basal body and elsewhere within the cell. Here, we found that the cytoplasmic domain of meckelin directly interacts with the actin-binding protein filamin A, potentially at the apical cell surface associated with the basal body. Mutations in FLNA, the gene for filamin A, cause periventricular heterotopias. We identified a single consanguineous patient with an MKS-like ciliopathy that presented with both MKS and cerebellar heterotopia, caused by an unusual in-frame deletion mutation in the meckelin C-terminus at the region of interaction with filamin A. We modelled this mutation and found it to abrogate the meckelin-filamin A interaction. Furthermore, we found that loss of filamin A by siRNA knockdown, in patient cells, and in tissues from Flna(Dilp2) null mouse embryos results in cellular phenotypes identical to those caused by meckelin loss, namely basal body positioning and ciliogenesis defects. In addition, morpholino knockdown of flna in zebrafish embryos significantly increases the frequency of dysmorphology and severity of ciliopathy developmental defects caused by mks3 knockdown. Our results suggest that meckelin forms a functional complex with filamin A that is disrupted in MKS and causes defects in neuronal migration and Wnt signalling. Furthermore, filamin A has a crucial role in the normal processes of ciliogenesis and basal body positioning. Concurrent with these processes, the meckelin-filamin A signalling axis may be a key regulator in maintaining correct, normal levels of Wnt signalling.


Assuntos
Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/patologia , Proteínas Contráteis/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mutação/genética , Animais , Western Blotting , Transtornos da Motilidade Ciliar/genética , Proteínas Contráteis/antagonistas & inibidores , Proteínas Contráteis/genética , Feminino , Filaminas , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Masculino , Proteínas de Membrana/genética , Camundongos , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/genética , Fenótipo , RNA Interferente Pequeno/genética , Técnicas do Sistema de Duplo-Híbrido , Peixe-Zebra/embriologia
13.
Hum Mol Genet ; 20(2): 223-34, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20943750

RESUMO

Renal-coloboma syndrome, also known as papillorenal syndrome, is an autosomal dominant human disorder in which optic disc coloboma is associated with kidney abnormalities. Mutations in the paired domain transcription factor PAX2 have been found to be the underlying cause of this disease. Disease severity varies between patients, and in some cases, renal hypoplasia has been found in the absence of any retinal defects. Here we report an N-ethyl-N-nitrosourea-induced mouse mutation, Opdc, which is an isoleucinetothreonine missense mutation, I40T, in the first α-helix of the Pax2 paired domain. The mutant protein binds target DNA sequences less strongly than the wild-type protein and acts poorly to transactivate target promoters in culture. The phenotypic consequence of this mutation on the development of the eye and ear is similar to that reported for null alleles of Pax2. However, in homozygotes, cerebellar development is normal on a genetic background in which loss of Pax2 results in failure of cerebellar formation. Moreover, there is a genetic background effect on the heterozygous phenotype such that on some strain backgrounds, kidney development is unaffected. Opdc is the first hypomorphic mutation reported for Pax2 that differs in phenotype from loss-of-function mutations. These results suggest that PAX2 is a strong candidate gene for cases in which human patients have optic disc coloboma not associated with renal dysplasia.


Assuntos
Coloboma/genética , Coloboma/patologia , Mutação de Sentido Incorreto , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo , Fenótipo , Insuficiência Renal/genética , Insuficiência Renal/patologia , Refluxo Vesicoureteral/genética , Refluxo Vesicoureteral/patologia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Animais , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Genótipo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Modelos Animais , Mutação Puntual , Ativação Transcricional/genética
14.
J Cell Sci ; 121(Pt 19): 3140-5, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18765564

RESUMO

The intracellular target of diphtheria toxin is a modified histidine residue, diphthamide, in the translation elongation factor, eEF2 (also known as EFT1). This enigmatic modification occurs in all eukaryotes and is produced in yeast by the action of five gene products, DPH1 to DPH5. Sequence homologues of these genes are present in all sequenced eukaryotic genomes and, in higher eukaryotes, there is functional evidence for DPH1, DPH2, DPH3 and DPH5 acting in diphthamide biosynthesis. We identified a mouse that was mutant for the remaining gene, Dph4. Cells derived from homozygous mutant embryos lacked the diphthamide modification of eEF2 and were resistant to killing by diphtheria toxin. Reporter-tagged DPH4 protein localized to the cytoskeleton, in contrast to the localization of DPH1 and consistent with evidence that DPH4 is not part of a proposed complex containing DPH1, DPH2 and DPH3. Mice that were homozygous for the mutation were retarded in growth and development, and almost always die before birth. Those that survive long enough had preaxial polydactyly, a duplication of digit 1 of the hind foot. This same defect has been seen in embryos that were homozygous for mutation of DPH1, suggesting that lack of diphthamide on eEF2 could result in translational failure of specific proteins, rather than a generalized translation downregulation.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Desenvolvimento Embrionário , Histidina/análogos & derivados , Fator 2 de Elongação de Peptídeos/metabolismo , Animais , Cromossomos de Mamíferos/genética , Cruzamentos Genéticos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Toxina Diftérica/farmacologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Proteínas do Olho/genética , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Testes Genéticos , Genótipo , Histidina/metabolismo , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Camundongos Mutantes , Mutação/genética , Células NIH 3T3 , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Splicing de RNA/efeitos dos fármacos , Proteínas Repressoras/genética , Dedos do Pé/anormalidades
15.
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
16.
Proc Natl Acad Sci U S A ; 103(10): 3704-9, 2006 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-16505357

RESUMO

Chromosome deletions in the mouse have proven invaluable in the dissection of gene function. The brown deletion complex comprises >28 independent genome rearrangements, which have been used to identify several functional loci on chromosome 4 required for normal embryonic and postnatal development. We have constructed a 172-bacterial artificial chromosome contig that spans this 22-megabase (Mb) interval and have produced a contiguous, finished, and manually annotated sequence from these clones. The deletion complex is strikingly gene-poor, containing only 52 protein-coding genes (of which only 39 are supported by human homologues) and has several further notable genomic features, including several segments of >1 Mb, apparently devoid of a coding sequence. We have used sequence polymorphisms to finely map the deletion breakpoints and identify strong candidate genes for the known phenotypes that map to this region, including three lethal loci (l4Rn1, l4Rn2, and l4Rn3) and the fitness mutant brown-associated fitness (baf). We have also characterized misexpression of the basonuclin homologue, Bnc2, associated with the inversion-mediated coat color mutant white-based brown (B(w)). This study provides a molecular insight into the basis of several characterized mouse mutants, which will allow further dissection of this region by targeted or chemical mutagenesis.


Assuntos
Deleção Cromossômica , Glicoproteínas de Membrana/genética , Oxirredutases/genética , Animais , Sequência de Bases , Evolução Biológica , Mapeamento Cromossômico , Cromossomos Artificiais Bacterianos/genética , Feminino , Morte Fetal/genética , Genes Letais , Cor de Cabelo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fenótipo , Polimorfismo de Nucleotídeo Único , Gravidez
17.
Hum Mol Genet ; 14(21): 3161-8, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16159887

RESUMO

Members of the type IV collagen family are essential components of all basement membranes (BMs) and define structural stability as well as tissue-specific functions. The major isoform, alpha1.alpha1.alpha2(IV), contributes to the formation of many BMs and its deficiency causes embryonic lethality in mouse. We have identified an allelic series of three ENU induced dominant mouse mutants with missense mutations in the gene Col4a1 encoding the alpha1(IV) subunit chain. Two severe alleles (Bru and Svc) have mutations affecting the conserved glycine residues in the Gly-Xaa-Yaa collagen repeat. Bru heterozygous mice display defects similar to Axenfeld-Rieger anomaly, including iris defects, corneal opacity, vacuolar cataracts, significant iris/corneal adhesions, buphthalmos and optic nerve cupping, a sign indicative of glaucoma. Kidneys of Bru mice have peripheral glomerulopathy characterized by hypertrophy and hyperplasia of the parietal epithelium of Bowman's capsule. A milder allele (Raw) contains a mutation in the Yaa residue of the collagen repeat and was identified by a silvery appearance of the retinal arterioles. All phenotypes are associated with BM defects that affect the eye, kidney and other tissues. This allelic series shows that mutations affecting the collagen domain cause dominant negative effects on the expression and function of the major collagen IV isoform alpha1(IV), and pathological effects vary with the individual mutations.


Assuntos
Anormalidades Múltiplas/genética , Colágeno Tipo IV/genética , Oftalmopatias/genética , Membrana Basal Glomerular/ultraestrutura , Glomerulonefrite Membranosa/genética , Mutação de Sentido Incorreto/genética , Anormalidades Múltiplas/patologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Colágeno Tipo IV/metabolismo , Eletroforese em Gel de Poliacrilamida , Oftalmopatias/patologia , Genes Dominantes/genética , Glomerulonefrite Membranosa/patologia , Imuno-Histoquímica , Camundongos , Repetições de Microssatélites/genética , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Análise de Sequência de DNA
18.
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
19.
Mamm Genome ; 15(7): 525-36, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15366372

RESUMO

Mice heterozygous for the N-ethyl-N-nitrosourea-induced Waved-5 (Wa5) mutation, isolated in a screen for dominant, visible mutations, exhibit a wavy coat similar to mice homozygous for the recessive Tgfa wa1 or Egfr wa2 alleles. In this study, we show that Wa5 is a new allele of Egfr (Egfr Wa5) containing a missense mutation within the coding region for the highly conserved DFG motif of the tyrosine kinase domain. In vivo analysis of placental development, modification of Apc Min tumorigenesis, and levels of EGF-dependent EGFR phosphorylation demonstrates that Egfr Wa5 functions as an antimorphic allele, recapitulating many abnormalities associated with reduced EGFR activity. Furthermore, Egfr wa5 enhances Egfr Wa2 compound or Tgfa tm1Dcl double mutants exposing additional EGFR-dependent phenotypes. In vitro characterization shows that the antimorphic property of Egfr Wa5 is caused by a kinase-dead receptor acting as a dominant negative.


Assuntos
Alelos , Receptores ErbB/genética , Alquilantes/farmacologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Cricetinae , Análise Mutacional de DNA , Etilnitrosoureia , Genes Dominantes , Teste de Complementação Genética , Genótipo , Heterozigoto , Homozigoto , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Mutagênicos , Mutação , Mutação de Sentido Incorreto , Fenótipo , Fosforilação , Placenta/metabolismo , Placenta/patologia , Mutação Puntual , Estrutura Secundária de Proteína , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Homologia de Sequência de Aminoácidos , Transfecção
20.
Hum Mol Genet ; 13(14): 1433-9, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15150159

RESUMO

Dilp1 is a semi-dominant mouse mutation that causes dilated pupils when heterozygous and is lethal when homozygous. We report here that it is caused by a point mutation that introduces a stop codon close to the start of the coding sequence of the paired-like homeobox transcription factor Phox2b. Mice carrying a targeted allele of Phox2b also have dilated pupils and the two alleles do not complement. Phox2b is necessary for the development of the autonomic nervous system and when absent one of the consequences is that all parasympathetic ganglia fail to form. Constriction of the pupil is a parasympathetic response mediated by the ciliary ganglion and we find that in Phox2b heterozygous mutants it is highly atrophic. The development of other parasympathetic and sympathetic ganglia appears to be largely unaffected indicating that the ciliary ganglion is exquisitely sensitive to a reduction in dose of this transcription factor. PHOX2B has been implicated in human disease. Mutations, principally leading to polyalanine expansions within the protein, have been found in patients with congenital central hypoventilation syndrome (CCHS), the cardinal feature of which is an inability to breathe unassisted when asleep. Additionally, some CCHS patients have ocular abnormalities, including pupillary defects, although they principally have constricted rather than dilated pupils. The apparent phenotypic differences observed between mice carrying a loss-of-function mutation of Phox2b and CCHS patients indicate that PHOX2B mutations found in CCHS patients, all of which can produce proteins with intact DNA-binding domains, are gain-of-function mutations that alter rather than abolish protein function.


Assuntos
Corpo Ciliar/inervação , Gânglios Parassimpáticos/patologia , Proteínas de Homeodomínio/genética , Peptídeos/genética , Distúrbios Pupilares/genética , Apneia do Sono Tipo Central/genética , Fatores de Transcrição/genética , Alelos , Animais , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Mutação , Distúrbios Pupilares/patologia , Síndrome , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA