Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 136(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37732459

RESUMEN

A characteristic of normal aging and age-related diseases is the remodeling of the cellular organization of a tissue through polyploid cell growth. Polyploidy arises from an increase in nuclear ploidy or the number of nuclei per cell. However, it is not known whether age-induced polyploidy is an adaption to stressors or a precursor to degeneration. Here, we find that abdominal epithelium of the adult fruit fly becomes polyploid with age through generation of multinucleated cells by cell fusion. Inhibition of fusion does not improve the lifespan of the fly, but does enhance its biomechanical fitness, a measure of the healthspan of the animal. Remarkably, Drosophila can maintain their epithelial tension and abdominal movements with age when cell fusion is inhibited. Epithelial cell fusion also appears to be dependent on a mechanical cue, as knockdown of Rho kinase, E-cadherin or α-catenin is sufficient to induce multinucleation in young animals. Interestingly, mutations in α-catenin in mice result in retina pigment epithelial multinucleation associated with macular disease. Therefore, we have discovered that polyploid cells arise by cell fusion and contribute to the decline in the biomechanical fitness of the animal with age.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Ratones , Drosophila/genética , alfa Catenina , Fusión Celular , Proteínas de Drosophila/genética , Poliploidía
2.
PLoS Genet ; 18(6): e1009798, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35675330

RESUMEN

Mutations in the apicobasal polarity gene CRB1 lead to diverse retinal diseases, such as Leber congenital amaurosis, cone-rod dystrophy, retinitis pigmentosa (with and without Coats-like vasculopathy), foveal retinoschisis, macular dystrophy, and pigmented paravenous chorioretinal atrophy. Limited correlation between disease phenotypes and CRB1 alleles, and evidence that patients sharing the same alleles often present with different disease features, suggest that genetic modifiers contribute to clinical variation. Similarly, the retinal phenotype of mice bearing the Crb1 retinal degeneration 8 (rd8) allele varies with genetic background. Here, we initiated a sensitized chemical mutagenesis screen in B6.Cg-Crb1rd8/Pjn, a strain with a mild clinical presentation, to identify genetic modifiers that cause a more severe disease phenotype. Two models from this screen, Tvrm266 and Tvrm323, exhibited increased retinal dysplasia. Genetic mapping with high-throughput exome and candidate-gene sequencing identified causative mutations in Arhgef12 and Prkci, respectively. Epistasis analysis of both strains indicated that the increased dysplastic phenotype required homozygosity of the Crb1rd8 allele. Retinal dysplastic lesions in Tvrm266 mice were smaller and caused less photoreceptor degeneration than those in Tvrm323 mice, which developed an early, large diffuse lesion phenotype. At one month of age, Müller glia and microglia mislocalization at dysplastic lesions in both modifier strains was similar to that in B6.Cg-Crb1rd8/Pjn mice but photoreceptor cell mislocalization was more extensive. External limiting membrane disruption was comparable in Tvrm266 and B6.Cg-Crb1rd8/Pjn mice but milder in Tvrm323 mice. Immunohistological analysis of mice at postnatal day 0 indicated a normal distribution of mitotic cells in Tvrm266 and Tvrm323 mice, suggesting normal early development. Aberrant electroretinography responses were observed in both models but functional decline was significant only in Tvrm323 mice. These results identify Arhgef12 and Prkci as modifier genes that differentially shape Crb1-associated retinal disease, which may be relevant to understanding clinical variability and underlying disease mechanisms in humans.


Asunto(s)
Proteínas del Tejido Nervioso , Displasia Retiniana , Factores de Intercambio de Guanina Nucleótido Rho , Animales , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Retina/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Displasia Retiniana/genética , Displasia Retiniana/metabolismo , Displasia Retiniana/patología , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo
3.
Adv Exp Med Biol ; 1415: 27-36, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37440010

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness in the global aging population. Familial aggregation and genome-wide association (GWA) studies have identified gene variants associated with AMD, implying a strong genetic contribution to AMD development. Two loci, on human Chr 1q31 and 10q26, respectively, represent the most influential of all genetic factors. While the role of CFH at Chr 1q31 is well established, uncertainty remains about the genes ARMS2 and HTRA1, at the Chr 10q26 locus. Since both genes are in strong linkage disequilibrium, assigning individual gene effects is difficult. In this chapter, we review current literature about ARMS2 and HTRA1 and their relevance to AMD risk. Future studies will be necessary to unravel the mechanisms by which they contribute to AMD.


Asunto(s)
Degeneración Macular , Proteínas , Humanos , Anciano , Proteínas/genética , Serina Endopeptidasas/genética , Estudio de Asociación del Genoma Completo , Serina Peptidasa A1 que Requiere Temperaturas Altas/genética , Degeneración Macular/genética , Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple , Factor H de Complemento/genética , Genotipo
4.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-35163536

RESUMEN

Adipor1tm1Dgen and Mfrprd6 mutant mice share similar eye disease characteristics. Previously, studies established a functional relationship of ADIPOR1 and MFRP proteins in maintaining retinal lipidome homeostasis and visual function. However, the independent and/or interactive contribution of both genes to similar disease phenotypes, including fundus spots, decreased axial length, and photoreceptor degeneration has yet to be examined. We performed a gene-interaction study where homozygous Adipor1tm1Dgen and Mfrprd6 mice were bred together and the resulting doubly heterozygous F1 offspring were intercrossed to produce 210 F2 progeny. Four-month-old mice from all nine genotypic combinations obtained in the F2 generation were assessed for white spots by fundus photo documentation, for axial length by caliper measurements, and for photoreceptor degeneration by histology. Two-way factorial ANOVA was performed to study individual as well as gene interaction effects on each phenotype. Here, we report the first observation of reduced axial length in Adipor1tmlDgen homozygotes. We show that while Adipor1 and Mfrp interact to affect spotting and degeneration, they act independently to control axial length, highlighting the complex functional association between these two genes. Further examination of the molecular basis of this interaction may help in uncovering mechanisms by which these genes perturb ocular homeostasis.


Asunto(s)
Proteínas del Ojo/genética , Proteínas de la Membrana/genética , Mutación , Receptores de Adiponectina/genética , Degeneración Retiniana/patología , Animales , Cruzamiento , Modelos Animales de Enfermedad , Epistasis Genética , Proteínas del Ojo/metabolismo , Homocigoto , Proteínas de la Membrana/metabolismo , Ratones , Oftalmoscopía , Fenotipo , Receptores de Adiponectina/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo
5.
Int J Mol Sci ; 23(18)2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-36142331

RESUMEN

Transcriptomic analysis of the mammalian retinal pigment epithelium (RPE) aims to identify cellular networks that influence ocular development, maintenance, function, and disease. However, available evidence points to RPE cell heterogeneity within native tissue, which adds complexity to global transcriptomic analysis. Here, to assess cell heterogeneity, we performed single-cell RNA sequencing of RPE cells from two young adult male C57BL/6J mice. Following quality control to ensure robust transcript identification limited to cell singlets, we detected 13,858 transcripts among 2667 and 2846 RPE cells. Dimensional reduction by principal component analysis and uniform manifold approximation and projection revealed six distinct cell populations. All clusters expressed transcripts typical of RPE cells; the smallest (C1, containing 1-2% of total cells) exhibited the hallmarks of stem and/or progenitor (SP) cells. Placing C1-6 along a pseudotime axis suggested a relative decrease in melanogenesis and SP gene expression and a corresponding increase in visual cycle gene expression upon RPE maturation. K-means clustering of all detected transcripts identified additional expression patterns that may advance the understanding of RPE SP cell maintenance and the evolution of cellular metabolic networks during development. This work provides new insights into the transcriptome of the mouse RPE and a baseline for identifying experimentally induced transcriptional changes in future studies of this tissue.


Asunto(s)
Perfilación de la Expresión Génica , Epitelio Pigmentado de la Retina , Animales , Perfilación de la Expresión Génica/métodos , Masculino , Mamíferos , Ratones , Ratones Endogámicos C57BL , Epitelio Pigmentado de la Retina/metabolismo , Análisis de Secuencia de ARN , Transcriptoma
6.
Int J Mol Sci ; 23(4)2022 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-35216333

RESUMEN

Fluid and solute transporters of the retinal pigment epithelium (RPE) are core components of the outer blood-retinal barrier. Characterizing these transporters and their role in retinal homeostasis may provide insights into ocular function and disease. Here, we describe RPE defects in tvrm77 mice, which exhibit hypopigmented patches in the central retina. Mapping and nucleotide sequencing of tvrm77 mice revealed a disrupted 5' splice donor sequence in Slc4a5, a sodium bicarbonate cotransporter gene. Slc4a5 expression was reduced 19.7-fold in tvrm77 RPE relative to controls, and alternative splice variants were detected. SLC4A5 was localized to the Golgi apparatus of cultured human RPE cells and in apical and basal membranes. Fundus imaging, optical coherence tomography, microscopy, and electroretinography (ERG) of tvrm77 mice revealed retinal detachment, hypopigmented patches corresponding to neovascular lesions, and retinal folds. Detachment worsened and outer nuclear layer thickness decreased with age. ERG a- and b-wave response amplitudes were initially normal but declined in older mice. The direct current ERG fast oscillation and light peak were reduced in amplitude at all ages, whereas other RPE-associated responses were unaffected. These results link a new Slc4a5 mutation to subretinal fluid accumulation and altered light-evoked RPE electrophysiological responses, suggesting that SLC4A5 functions at the outer blood-retinal barrier.


Asunto(s)
Mutación/genética , Empalme del ARN/genética , Retina/patología , Desprendimiento de Retina/genética , Epitelio Pigmentado de la Retina/patología , Simportadores de Sodio-Bicarbonato/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Desprendimiento de Retina/patología , Tomografía de Coherencia Óptica/métodos
7.
Int J Mol Sci ; 23(19)2022 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-36233305

RESUMEN

Congenital disorders of glycosylation (CDG) are a heterogenous group of primarily autosomal recessive mendelian diseases caused by disruptions in the synthesis of lipid-linked oligosaccharides and their transfer to proteins. CDGs usually affect multiple organ systems and vary in presentation, even within families. There is currently no cure, and treatment is aimed at ameliorating symptoms and improving quality of life. Here, we describe a chemically induced mouse mutant, tvrm76, with early-onset photoreceptor degeneration. The recessive mutation was mapped to Chromosome 9 and associated with a missense mutation in the Dpagt1 gene encoding UDP-N-acetyl-D-glucosamine:dolichyl-phosphate N-acetyl-D-glucosaminephosphotransferase (EC 2.7.8.15). The mutation is predicted to cause a substitution of aspartic acid with glycine at residue 166 of DPAGT1. This represents the first viable animal model of a Dpagt1 mutation and a novel phenotype for a CDG. The increased expression of Ddit3, and elevated levels of HSPA5 (BiP) suggest the presence of early-onset endoplasmic reticulum (ER) stress. These changes were associated with the induction of photoreceptor apoptosis in tvrm76 retinas. Mutations in human DPAGT1 cause myasthenic syndrome-13 and severe forms of a congenital disorder of glycosylation Type Ij. In contrast, Dpagt1tvrm76 homozygous mice present with congenital photoreceptor degeneration without overt muscle or muscular junction involvement. Our results suggest the possibility of DPAGT1 mutations in human patients that present primarily with retinitis pigmentosa, with little or no muscle disease. Variants in DPAGT1 should be considered when evaluating cases of non-syndromic retinal degeneration.


Asunto(s)
Trastornos Congénitos de Glicosilación , Enfermedades de la Retina , Acetilglucosamina , Animales , Ácido Aspártico/genética , Trastornos Congénitos de Glicosilación/genética , Glicina/genética , Humanos , Ratones , Debilidad Muscular , Mutación , Mutación Missense , Fosfatos , Calidad de Vida , Uridina Difosfato
8.
Hum Mol Genet ; 27(19): 3340-3352, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29947801

RESUMEN

Photoreceptor dysplasia, characterized by formation of folds and (pseudo-)rosettes in the outer retina, is associated with loss of functional nuclear receptor subfamily 2 group E member 3 (NR2E3) and neural retina leucine-zipper (NRL) in both humans and mice. A sensitized chemical mutagenesis study to identify genetic modifiers that suppress photoreceptor dysplasia in Nr2e3rd7mutant mice identified line Tvrm222, which exhibits a normal fundus appearance in the presence of the rd7 mutation. The Tvrm222 modifier of Nr2e3rd7/rd7 was localized to Chromosome 6 and identified as a missense mutation in the FERM domain containing 4B (Frmd4b) gene. The variant is predicted to cause the substitution of a serine residue 938 with proline (S938P). The Frmd4bTvrm222 allele was also found to suppress outer nuclear layer (ONL) rosettes in Nrl-/- mice. Fragmentation of the external limiting membrane (ELM), normally observed in rd7 and Nrl-/-mouse retinas, was absent in the presence of the Frmd4bTvrm222 allele. FRMD4B, a binding partner of cytohesin 3, is proposed to participate in cell junction remodeling. Its biological function in photoreceptor dysplasia has not been previously examined. In vitro experiments showed that the FRMD4B938P variant fails to be efficiently recruited to the cell surface upon insulin stimulation. In addition, we found a reduction in protein kinase B phosphorylation and increased levels of cell junction proteins, Catenin beta 1 and tight junction protein 1, associated with the cell membrane in Tvrm222 retinas. Taken together, this study reveals a critical role of FRMD4B in maintaining ELM integrity and in rescuing morphological abnormalities of the ONL in photoreceptor dysplasia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Enfermedades Hereditarias del Ojo/genética , Proteínas del Ojo/genética , Receptores Nucleares Huérfanos/genética , Degeneración Retiniana/genética , Trastornos de la Visión/genética , Animales , Enfermedades Hereditarias del Ojo/metabolismo , Enfermedades Hereditarias del Ojo/patología , Fondo de Ojo , Humanos , Ratones , Mutación Missense , Dominios Proteicos/genética , Retina/crecimiento & desarrollo , Retina/patología , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Segmento Externo de las Células Fotorreceptoras Retinianas , Trastornos de la Visión/metabolismo , Trastornos de la Visión/patología
9.
Adv Exp Med Biol ; 1074: 545-551, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721986

RESUMEN

The formation of solid tissues is not a simple aggregation of individual cells but rather an ordered assembly of cells connected by junctions. These junctions provide a diffusion barrier as well as mechanical support and a conduit for signalling changes in the environment to the cells. Cell junctions are functionally categorized as occluding (e.g. tight junctions, TJs), anchoring (e.g. adherens junctions, AJs) and communicating junctions (e.g. gap junctions). Each type of the cell junction is formed by protein complexes with extracellular domains and/or intracellular domains, which bind partners that provide scaffolding and signalling components. Cell junctions are ubiquitously expressed in multiple tissues and organs, including the retina. In the retina, their biological impact is not limited to regulating tissue growth and development. Disruption of the complexes mediates both congenital and postnatal pathogenesis. In this review, we will focus on cell junctions, specifically AJs and TJs in the external limiting membrane, in order to articulate their influence on pathophysiology of the retina.


Asunto(s)
Uniones Adherentes/fisiología , Retina/ultraestructura , Enfermedades de la Retina/fisiopatología , Uniones Estrechas/fisiología , Uniones Adherentes/ultraestructura , Comunicación Celular , Proteínas del Ojo/genética , Proteínas del Ojo/fisiología , Uniones Comunicantes/fisiología , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Retina/fisiología , Retina/fisiopatología , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/patología , Enfermedades de la Retina/terapia , Tomografía de Coherencia Óptica
10.
Proc Natl Acad Sci U S A ; 112(42): 12962-7, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26438849

RESUMEN

Sphingolipids typically have an 18-carbon (C18) sphingoid long chain base (LCB) backbone. Although sphingolipids with LCBs of other chain lengths have been identified, the functional significance of these low-abundance sphingolipids is unknown. The LCB chain length is determined by serine palmitoyltransferase (SPT) isoenzymes, which are trimeric proteins composed of two large subunits (SPTLC1 and SPTLC2 or SPTLC3) and a small subunit (SPTssa or SPTssb). Here we report the identification of an Sptssb mutation, Stellar (Stl), which increased the SPT affinity toward the C18 fatty acyl-CoA substrate by twofold and significantly elevated 20-carbon (C20) LCB production in the mutant mouse brain and eye, resulting in surprising neurodegenerative effects including aberrant membrane structures, accumulation of ubiquitinated proteins on membranes, and axon degeneration. Our work demonstrates that SPT small subunits play a major role in controlling SPT activity and substrate affinity, and in specifying sphingolipid LCB chain length in vivo. Moreover, our studies also suggest that excessive C20 LCBs or C20 LCB-containing sphingolipids impair protein homeostasis and neural functions.


Asunto(s)
Carbono/química , Mutación , Enfermedades Neurodegenerativas/enzimología , Serina C-Palmitoiltransferasa/química , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Datos de Secuencia Molecular , Enfermedades Neurodegenerativas/genética , Homología de Secuencia de Aminoácido , Serina C-Palmitoiltransferasa/genética , Ubiquitinación
11.
Hum Mol Genet ; 24(24): 6958-74, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26405179

RESUMEN

Human gene mutations have revealed that a significant number of ADAMTS (a disintegrin-like and metalloproteinase (reprolysin type) with thrombospondin type 1 motifs) proteins are necessary for normal ocular development and eye function. Mutations in human ADAMTSL4, encoding an ADAMTS-like protein which has been implicated in fibrillin microfibril biogenesis, cause ectopia lentis (EL) and EL et pupillae. Here, we report the first ADAMTSL4 mouse model, tvrm267, bearing a nonsense mutation in Adamtsl4. Homozygous Adamtsl4(tvrm267) mice recapitulate the EL phenotype observed in humans, and our analysis strongly suggests that ADAMTSL4 is required for stable anchorage of zonule fibers to the lens capsule. Unexpectedly, homozygous Adamtsl4(tvrm267) mice exhibit focal retinal pigment epithelium (RPE) defects primarily in the inferior eye. RPE dedifferentiation was indicated by reduced pigmentation, altered cellular morphology and a reduction in RPE-specific transcripts. Finally, as with a subset of patients with ADAMTSL4 mutations, increased axial length, relative to age-matched controls, was observed and was associated with the severity of the RPE phenotype. In summary, the Adamtsl4(tvrm267) model provides a valuable tool to further elucidate the molecular basis of zonule formation, the pathophysiology of EL and ADAMTSL4 function in the maintenance of the RPE.


Asunto(s)
Proteínas ADAM/genética , Desplazamiento del Cristalino/genética , Procolágeno N-Endopeptidasa/genética , Trastornos de la Pupila/genética , Epitelio Pigmentado de la Retina/citología , Proteínas ADAM/fisiología , Proteína ADAMTS4 , Animales , Longitud Axial del Ojo , Diferenciación Celular , Codón sin Sentido , Colágeno/genética , Modelos Animales de Enfermedad , Desplazamiento del Cristalino/patología , Colágenos Asociados a Fibrillas , Regulación de la Expresión Génica , Homocigoto , Humanos , Cristalino/citología , Cristalino/patología , Ratones , Ratones Mutantes , Procolágeno N-Endopeptidasa/fisiología , Pupila , Trastornos de la Pupila/patología , Epitelio Pigmentado de la Retina/patología
12.
Am J Pathol ; 186(7): 1925-1938, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27207593

RESUMEN

The nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) enzyme is essential for regenerating the nuclear pool of NAD(+) in all nucleated cells in the body, and mounting evidence also suggests that it has a separate role in neuroprotection. Recently, mutations in the NMNAT1 gene were associated with Leber congenital amaurosis, a severe retinal degenerative disease that causes blindness during infancy. Availability of a reliable mammalian model of NMNAT1-Leber congenital amaurosis would assist in determining the mechanisms through which disruptions in NMNAT1 lead to retinal cell degeneration and would provide a resource for testing treatment options. To this end, we identified two separate N-ethyl-N-nitrosourea-generated mouse lines that harbor either a p.V9M or a p.D243G mutation. Both mouse models recapitulate key aspects of the human disease and confirm the pathogenicity of mutant NMNAT1. Homozygous Nmnat1 mutant mice develop a rapidly progressing chorioretinal disease that begins with photoreceptor degeneration and includes attenuation of the retinal vasculature, optic atrophy, and retinal pigment epithelium loss. Retinal function deteriorates in both mouse lines, and, in the more rapidly progressing homozygous Nmnat1(V9M) mutant mice, the electroretinogram becomes undetectable and the pupillary light response weakens. These mouse models offer an opportunity for investigating the cellular mechanisms underlying disease pathogenesis, evaluating potential therapies for NMNAT1-Leber congenital amaurosis, and conducting in situ studies on NMNAT1 function and NAD(+) metabolism.


Asunto(s)
Modelos Animales de Enfermedad , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/fisiopatología , Nicotinamida-Nucleótido Adenililtransferasa/genética , Animales , Genotipo , Humanos , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa
13.
Mol Vis ; 23: 140-148, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28356706

RESUMEN

PURPOSE: Familial exudative vitreoretinopathy (FEVR) is caused by mutations in the genes encoding low-density lipoprotein receptor-related protein (LRP5) or its interacting partners, namely frizzled class receptor 4 (FZD4) and norrin cystine knot growth factor (NDP). Mouse models for Lrp5, Fzd4, and Ndp have proven to be important for understanding the retinal pathophysiology underlying FEVR and systemic abnormalities related to defective Wnt signaling. Here, we report a new mouse mutant, tvrm111B, which was identified by electroretinogram (ERG) screening of mice generated in the Jackson Laboratory Translational Vision Research Models (TVRM) mutagenesis program. METHODS: ERGs were used to examine outer retinal physiology. The retinal vasculature was examined by in vivo retinal imaging, as well as by histology and immunohistochemistry. The tvrm111B locus was identified by genetic mapping of mice generated in a cross to DBA/2J, and subsequent sequencing analysis. Gene expression was examined by real-time PCR of retinal RNA. Bone mineral density (BMD) was examined by peripheral dual-energy X-ray absorptiometry. RESULTS: The tvrm111B allele is inherited as an autosomal recessive trait. Genetic mapping of the decreased ERG b-wave phenotype of tvrm111B mice localized the mutation to a region on chromosome 19 that included Lrp5. Sequencing of Lrp5 identified the insertion of a cytosine (c.4724_4725insC), which is predicted to cause a frameshift that disrupts the last three of five conserved PPPSPxS motifs in the cytoplasmic domain of LRP5, culminating in a premature termination. In addition to a reduced ERG b-wave, Lrp5tvrm111B homozygotes have low BMD and abnormal features of the retinal vasculature that have been reported previously in Lrp5 mutant mice, including persistent hyaloid vessels, leakage on fluorescein angiography, and an absence of the deep retinal capillary bed. CONCLUSIONS: The phenotype of the Lrp5tvrm111B mutant includes abnormalities of the retinal vasculature and of BMD. This model may be a useful resource to further our understanding of the biological role of LRP5 and to evaluate experimental therapies for FEVR or other conditions associated with LRP5 dysfunction.


Asunto(s)
Densidad Ósea , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Mutagénesis/genética , Mutación/genética , Vasos Retinianos/anomalías , Vasos Retinianos/fisiopatología , Animales , Electrorretinografía , Regulación de la Expresión Génica , Homocigoto , Masculino , Ratones Endogámicos C57BL , Tamaño de los Órganos/genética , Fenotipo , Vasos Retinianos/diagnóstico por imagen , Vasos Retinianos/patología , Vía de Señalización Wnt/genética
14.
Adv Exp Med Biol ; 854: 745-50, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26427484

RESUMEN

Regulation of vesicle trafficking to lysosomes and lysosome-related organelles (LROs) as well as regulation of the size of these organelles are critical to maintain their functions. Disruption of the lysosomal trafficking regulator (LYST) results in Chediak-Higashi syndrome (CHS), a rare autosomal recessive disorder characterized by oculocutaneous albinism, prolonged bleeding, severe immunodeficiency, recurrent bacterial infection, neurologic dysfunction and hemophagocytic lympohistiocytosis (HLH). The classic diagnostic feature of the syndrome is enlarged LROs in all cell types, including lysosomes, melanosomes, cytolytic granules and platelet dense bodies. The most striking CHS ocular pathology observed is an enlargement of melanosomes in the retinal pigment epithelium (RPE), which leads to aberrant distribution of eye pigmentation, and results in photophobia and decreased visual acuity. Understanding the molecular function of LYST and identification of its interacting partners may provide therapeutic targets for CHS and other diseases associated with the regulation of LRO size and/or vesicle trafficking, such as asthma, urticaria and Leishmania amazonensis infections.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Lisosomas/metabolismo , Melanosomas/metabolismo , Orgánulos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Síndrome de Chediak-Higashi/metabolismo , Síndrome de Chediak-Higashi/fisiopatología , Humanos , Fotofobia/metabolismo , Fotofobia/fisiopatología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/fisiopatología , Agudeza Visual
15.
Adv Exp Med Biol ; 854: 177-83, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26427409

RESUMEN

Mouse models provide important resources for many areas of vision research, pertaining to retinal development, retinal function and retinal disease. The Translational Vision Research Models (TVRM) program uses chemical mutagenesis to generate new mouse models for vision research. In this chapter, we report the identification of mouse models for Grm1, Grk1 and Lrit3. Each of these is characterized by a primary defect in the electroretinogram. All are available without restriction to the research community.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Mutación , Retina/metabolismo , Enfermedades de la Retina/genética , Alelos , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Oftalmopatías/diagnóstico , Oftalmopatías/genética , Oftalmopatías/fisiopatología , Quinasa 1 del Receptor Acoplado a Proteína-G/genética , Pruebas Genéticas/métodos , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Mutagénesis , Receptores de Glutamato Metabotrópico/genética , Retina/patología , Retina/fisiopatología , Enfermedades de la Retina/diagnóstico , Investigación Biomédica Traslacional/métodos , Visión Ocular/genética , Visión Ocular/fisiología
16.
Hum Mol Genet ; 22(3): 558-67, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23108158

RESUMEN

Approximately 36 000 cases of simplex and familial retinitis pigmentosa (RP) worldwide are caused by a loss in phosphodiesterase (PDE6) function. In the preclinical Pde6α(nmf363) mouse model of this disease, defects in the α-subunit of PDE6 result in a progressive loss of photoreceptors and neuronal function. We hypothesized that increasing PDE6α levels using an AAV2/8 gene therapy vector could improve photoreceptor survival and retinal function. We utilized a vector with the cell-type-specific rhodopsin (RHO) promoter: AAV2/8(Y733F)-Rho-Pde6α, to transduce Pde6α(nmf363) retinas and monitored its effects over a 6-month period (a quarter of the mouse lifespan). We found that a single injection enhanced survival of photoreceptors and improved retinal function. At 6 months of age, the treated eyes retained photoreceptor cell bodies, while there were no detectable photoreceptors remaining in the untreated eyes. More importantly, the treated eyes demonstrated functional visual responses even after the untreated eyes had lost all vision. Despite focal rescue of the retinal structure adjacent to the injection site, global functional rescue of the entire retina was observed. These results suggest that RP due to PDE6α deficiency in humans, in addition to PDE6ß deficiency, is also likely to be treatable by gene therapy.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Terapia Genética/métodos , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Electrorretinografía , Vectores Genéticos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Retina/anatomía & histología , Retina/fisiopatología , Retinitis Pigmentosa/fisiopatología , Rodopsina/genética , Rodopsina/metabolismo , Transducción Genética
17.
Am J Hum Genet ; 90(2): 331-9, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22325362

RESUMEN

Complete congenital stationary night blindness (cCSNB) is a clinically and genetically heterogeneous group of retinal disorders characterized by nonprogressive impairment of night vision, absence of the electroretinogram (ERG) b-wave, and variable degrees of involvement of other visual functions. We report here that mutations in GPR179, encoding an orphan G protein receptor, underlie a form of autosomal-recessive cCSNB. The Gpr179(nob5/nob5) mouse model was initially discovered by the absence of the ERG b-wave, a component that reflects depolarizing bipolar cell (DBC) function. We performed genetic mapping, followed by next-generation sequencing of the critical region and detected a large transposon-like DNA insertion in Gpr179. The involvement of GPR179 in DBC function was confirmed in zebrafish and humans. Functional knockdown of gpr179 in zebrafish led to a marked reduction in the amplitude of the ERG b-wave. Candidate gene analysis of GPR179 in DNA extracted from patients with cCSNB identified GPR179-inactivating mutations in two patients. We developed an antibody against mouse GPR179, which robustly labeled DBC dendritic terminals in wild-type mice. This labeling colocalized with the expression of GRM6 and was absent in Gpr179(nob5/nob5) mutant mice. Our results demonstrate that GPR179 plays a critical role in DBC signal transduction and expands our understanding of the mechanisms that mediate normal rod vision.


Asunto(s)
Mutación , Miopía/genética , Miopía/fisiopatología , Ceguera Nocturna/genética , Ceguera Nocturna/fisiopatología , Receptores Acoplados a Proteínas G/genética , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/fisiología , Animales , Mapeo Cromosómico/métodos , Adaptación a la Oscuridad/genética , Electrorretinografía/métodos , Enfermedades Hereditarias del Ojo , Técnicas de Silenciamiento del Gen/métodos , Enfermedades Genéticas Ligadas al Cromosoma X , Heterocigoto , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Miopía/metabolismo , Ceguera Nocturna/metabolismo , Linaje , Receptores de Glutamato Metabotrópico/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/fisiología , Transducción de Señal , Pez Cebra
18.
Am J Pathol ; 184(10): 2721-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25088982

RESUMEN

Mutations in the RP1 gene can cause retinitis pigmentosa. We identified a spontaneous L66P mutation caused by two adjacent point mutations in the Rp1 gene in a colony of C57BL/6J mice. Mice homozygous for the L66P mutation exhibited slow, progressive photoreceptor degeneration throughout their lifespan. Optical coherence tomography imaging found abnormal photoreceptor reflectivity at 1 month of age. Histology found shortening and disorganization of the photoreceptor inner and outer segments and progressive thinning of the outer nuclear layer. Electroretinogram a- and b-wave amplitudes were decreased with age. Western blot analysis found that the quantity and size of the mutated retinitis pigmentosa 1 (RP1) protein were normal. However, immunohistochemistry found that the mutant Rp1 protein partially mislocalized to the transition zone of the shortened axonemes. This mutation disrupted colocalization with cytoplasmic microtubules in vitro. In conclusion, the L66P mutation in the first doublecortin domain of the Rp1 gene impairs Rp1 protein localization and function, leading to abnormalities in photoreceptor outer segment structure and progressive photoreceptor degeneration. This is the first missense mutation in Rp1 shown to cause retinal degeneration. It provides a unique, slowly progressive photoreceptor degeneration model that mirrors the slow degeneration kinetics in most patients with retinitis pigmentosa.


Asunto(s)
Axonema/metabolismo , Proteínas del Ojo/genética , Proteínas Asociadas a Microtúbulos/genética , Degeneración Retiniana/genética , Animales , Células COS , Chlorocebus aethiops , Electrorretinografía , Proteínas del Ojo/metabolismo , Femenino , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación Missense , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/metabolismo
19.
Exp Eye Res ; 118: 30-5, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24200520

RESUMEN

The identification of genes that modify pathological ocular phenotypes in mouse models may improve our understanding of disease mechanisms and lead to new treatment strategies. Here, we identify modifier loci affecting photoreceptor cell loss in homozygous Mfrp(rd6) mice, which exhibit a slowly progressive photoreceptor degeneration. A cohort of 63 F2 homozygous Mfrp(rd6) mice from a (B6.C3Ga-Mfrp(rd6)/J × CAST/EiJ) F1 intercross exhibited a variable number of cell bodies in the retinal outer nuclear layer at 20 weeks of age. Mice were genotyped with a panel of single nucleotide polymorphism markers, and genotypes were correlated with phenotype by quantitative trait locus (QTL) analysis to map modifier loci. A genome-wide scan revealed a statistically significant, protective candidate locus on CAST/EiJ Chromosome 1 and suggestive modifier loci on Chromosomes 6 and 11. Multiple regression analysis of a three-QTL model indicated that the modifier loci on Chromosomes 1 and 6 together account for 26% of the observed phenotypic variation, while the modifier locus on Chromosome 11 explains only an additional 4%. Our findings indicate that the severity of the Mfrp(rd6) retinal degenerative phenotype in mice depends on the strain genetic background and that a significant modifier locus on CAST/EiJ Chromosome 1 protects against Mfrp(rd6)-associated photoreceptor loss.


Asunto(s)
ADN/genética , Proteínas del Ojo/genética , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Retina/metabolismo , Degeneración Retiniana/genética , Animales , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Genotipo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Reacción en Cadena de la Polimerasa , Retina/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología
20.
Hum Mol Genet ; 20(3): 482-96, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21078623

RESUMEN

Nephronophthisis (NPHP) is an autosomal recessive kidney disease that is often associated with vision and/or brain defects. To date, 11 genes are known to cause NPHP. The gene products, while structurally unrelated, all localize to cilia or centrosomes. Although mouse models of NPHP are available for 9 of the 11 genes, none has been described for nephronophthisis 4 (Nphp4). Here we report a novel, chemically induced mutant, nmf192, that bears a nonsense mutation in exon 4 of Nphp4. Homozygous mutant Nphp4(nmf192/nmf192) mice do not exhibit renal defects, phenotypes observed in human patients bearing mutations in NPHP4, but they do develop severe photoreceptor degeneration and extinguished rod and cone ERG responses by 9 weeks of age. Photoreceptor outer segments (OS) fail to develop properly, and some OS markers mislocalize to the inner segments and outer nuclear layer in the Nphp4(nmf192/nmf192) mutant retina. Despite NPHP4 localization to the transition zone in the connecting cilia (CC), the CC appear to be normal in structure and ciliary transport function is partially retained. Likewise, synaptic ribbons develop normally but then rapidly degenerate by P14. Finally, Nphp4(nmf192/nmf192) male mutants are sterile and show reduced sperm motility and epididymal sperm counts. Although Nphp4(nmf192/nmf192) mice fail to recapitulate the kidney phenotype of NPHP, they will provide a valuable tool to further elucidate how NPHP4 functions in the retina and male reproductive organs.


Asunto(s)
Células Fotorreceptoras de Vertebrados/fisiología , Células Fotorreceptoras de Vertebrados/ultraestructura , Proteínas/genética , Proteínas/fisiología , Degeneración Retiniana/genética , Maduración del Esperma/genética , Sinapsis/fisiología , Sinapsis/ultraestructura , Animales , Western Blotting , Mapeo Cromosómico , Cilios/metabolismo , Codón sin Sentido , Electrorretinografía , Infertilidad Masculina/genética , Enfermedades Renales Quísticas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Mutación , Compuestos de Nitrosourea/farmacología , Fenotipo , Retina/anomalías , Análisis de Secuencia de ADN , Motilidad Espermática
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA