Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Exp Eye Res ; 101: 60-71, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22687918

RESUMEN

Oxidative stress in the retinal pigment epithelium (RPE) is hypothesized to be a major contributor to the development of age-related macular degeneration (AMD). Mitochondrial manganese superoxide dismutase (MnSOD) is a critical antioxidant protein that scavenges the highly reactive superoxide radical. We speculated that specific reduction of MnSOD in the RPE will increase the level of reactive oxygen species in the retina/RPE/choroid complex leading to pathogenesis similar to geographic atrophy. To test this hypothesis, an Sod2-specific hammerhead ribozyme (Rz), delivered by AAV2/1 and driven by the human VMD2 promoter was injected subretinally into C57BL/6J mice. Dark-adapted full field electroretinogram (ERG) detected a decrease in the response to light. We investigated the age-dependent phenotypic and morphological changes of the outer retina using digital fundus imaging and SD-OCT measurement of ONL thickness. Fundus microscopy revealed pigmentary abnormalities in the retina and these corresponded to sub-retinal and sub-RPE deposits seen in SD-OCT B-scans. Light and electron microscopy documented the localization of apical deposits and thickening of the RPE. In RPE flat-mounts we observed abnormally displaced nuclei and regions of apparent fibrosis in the central retina of the oldest mice. This region was surrounded by enlarged and irregular RPE cells that have been observed in eyes donated by AMD patients and in other mouse models of AMD.


Asunto(s)
Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica/fisiología , Atrofia Geográfica/patología , Mitocondrias/enzimología , Estrés Oxidativo , Epitelio Pigmentado de la Retina/ultraestructura , Superóxido Dismutasa/genética , Animales , Dependovirus/genética , Electrorretinografía , Angiografía con Fluoresceína , Silenciador del Gen/fisiología , Vectores Genéticos , Atrofia Geográfica/enzimología , Atrofia Geográfica/genética , Humanos , Ratones , Ratones Endogámicos C57BL , ARN Catalítico/genética , Epitelio Pigmentado de la Retina/enzimología , Tomografía de Coherencia Óptica
11.
Front Mol Neurosci ; 15: 1080136, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36698779

RESUMEN

During routine screening of mouse strains and stocks by the Eye Mutant Resource at The Jackson Laboratory for genetic mouse models of human ocular disorders, we identified cpfl9, a mouse model with cone photoreceptor function loss. The mice exhibited an early-onset phenotype that was easily recognized by the absence of a cone-mediated b-wave electroretinography response and by a reduction in rod-mediated photoresponses at four weeks of age. By genetic mapping and high-throughput sequencing of a whole exome capture library of cpfl9, a homozygous 25 bp deletion within exon 11 of the Gucy2e gene was identified, which is predicted to result in a frame shift leading to premature termination. The corresponding protein in human, retinal guanylate cyclase 1 (GUCY2D), plays an important role in rod and cone photoreceptor cell function. Loss-of-function mutations in human GUCY2D cause LCA1, one of the most common forms of Leber congenital amaurosis, which results in blindness at birth or in early childhood. The early loss of cone and reduced rod photoreceptor cell function in the cpfl9 mutant is accompanied by a later, progressive loss of cone and rod photoreceptor cells, which may be relevant to understanding disease pathology in a subset of LCA1 patients and in individuals with cone-rod dystrophy caused by recessive GUCY2D variants. cpfl9 mice will be useful for studying the role of Gucy2e in the retina.

12.
Exp Eye Res ; 93(6): 862-72, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21989111

RESUMEN

Regenerative medicine holds the promise of restoring cells and tissues that are destroyed in human disease, including degenerative eye disorders. However, development of this approach in the eye has been limited by a lack of animal models that show robust regeneration of ocular tissue. Here, we test whether MRL/MpJ mice, which exhibit enhanced wound healing, can efficiently regenerate the retinal pigment epithelium (RPE) after an injury that mimics the loss of this tissue in age-related macular degeneration. The RPE of MRL/MpJ and control AKR/J mice was injured by retro-orbital injection of sodium iodate at 20 mg/kg body weight, which titration studies indicated was optimal for highlighting strain differences in the response to injury. Five days after sodium iodate injection at this dose, electroretinography of both strains revealed equivalent retinal responses that were significantly reduced compared to untreated mice. At one and two months post-injection, retinal responses were restored in MRL/MpJ but not AKR/J mice. Bright field and fluorescence microscopy of eyecup cryosections indicated an initial central loss of RPE cells and RPE65 immunostaining in MRL/MpJ and AKR/J mice, with preservation of peripheral RPE. Phalloidin staining of posterior eye whole mounts confirmed this pattern of RPE loss, and revealed a transition region characterized by RPE cell shedding and restructuring in both strains, suggesting a similar initial response to injury. At one month post-injection, central RPE cells, RPE65 immunostaining and phalloidin staining were restored in MRL/MpJ but not AKR/J mice. BrdU incorporation was observed throughout the RPE of MRL/MpJ but not AKR/J mice after one month of administration following sodium iodate treatment, consistent with RPE proliferation. These findings provide evidence for a dramatic regeneration of the RPE after injury in MRL/MpJ mice that supports full recovery of retinal function, which has not been observed previously in mammalian eyes. This model should prove useful for understanding molecular mechanisms that underlie regeneration, and for identifying factors that promote RPE regeneration in age-related macular degeneration and related diseases.


Asunto(s)
Proliferación Celular , Degeneración Macular/patología , Regeneración , Epitelio Pigmentado de la Retina/patología , Animales , Proteínas Portadoras/metabolismo , Forma de la Célula , Modelos Animales de Enfermedad , Electrorretinografía , Potenciales Evocados Visuales , Proteínas del Ojo/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Yodatos , Degeneración Macular/inducido químicamente , Degeneración Macular/genética , Degeneración Macular/metabolismo , Degeneración Macular/fisiopatología , Ratones , Ratones Endogámicos AKR , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Estimulación Luminosa , Recuperación de la Función , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/fisiopatología , Factores de Tiempo , cis-trans-Isomerasas
13.
Cells ; 9(4)2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32290105

RESUMEN

Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated. The progression of PR cell loss with postnatal age was documented in mutant alleles of genes grouped by biological function. As anticipated, a wide range in the onset and rate of cell loss was observed among the reported models. The analysis underscored relationships between RD genes and ciliary function, transcription-coupled DNA damage repair, and cellular chloride homeostasis. Comparing the mouse gene list to human RD genes identified in the RetNet database revealed that mouse models are available for 40% of the known human diseases, suggesting opportunities for future research. This work may provide insight into the molecular players and pathways through which PR degenerative disease occurs and may be useful for planning translational studies.


Asunto(s)
Modelos Animales de Enfermedad , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Animales , Humanos , Ratones , Degeneración Retiniana/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
14.
Trends Biochem Sci ; 29(12): 648-55, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15544951

RESUMEN

Integral membrane proteins (IMPs) are essential components of the plasma and organellar membranes of the eukaryotic cell. Non-native IMPs, which can arise as a result of mutations, errors during biosynthesis or cellular stress, can disrupt these membranes and potentially lead to cell death. To protect against this outcome, the cell possesses quality control (QC) systems that detect and dispose of non-native IMPs from cellular membranes. Recent studies suggest that recognition of non-native IMPs by the QC machinery is correlated with the thermodynamic stability of these proteins. Consistent with this, small molecules known as chemical and pharmacological chaperones have been identified that stabilize non-native IMPs and enable them to evade QC. These findings have far-reaching implications for treating human diseases caused by defective IMPs.


Asunto(s)
Membrana Celular/fisiología , Proteínas de la Membrana/fisiología , Modelos Biológicos , Chaperonas Moleculares , Pliegue de Proteína , Control de Calidad
15.
Annu Rev Vis Sci ; 5: 99-122, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31226014

RESUMEN

Inflammation of the blood vessels that serve the central nervous system has been increasingly identified as an early and possibly initiating event among neurodegenerative conditions such as Alzheimer's disease and related dementias. However, the causal relevance of vascular inflammation to major retinal degenerative diseases is unresolved. Here, we describe how genetics, aging-associated changes, and environmental factors contribute to vascular inflammation in age-related macular degeneration, diabetic retinopathy, and glaucoma. We highlight the importance of mouse models in studying the underlying mechanisms and possible treatments for these diseases. We conclude that data support vascular inflammation playing a central if not primary role in retinal degenerative diseases, and this association should be a focus of future research.


Asunto(s)
Retinopatía Diabética/fisiopatología , Glaucoma/fisiopatología , Degeneración Macular/fisiopatología , Vasculitis Retiniana/fisiopatología , Envejecimiento/fisiología , Animales , Modelos Animales de Enfermedad , Interacción Gen-Ambiente , Humanos , Inflamación/fisiopatología , Factores de Riesgo
16.
Curr Protoc Mouse Biol ; 7(3): 176-190, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28884793

RESUMEN

Comparing 3D structural information obtained by optical coherence tomography (OCT) requires accurate alignment of images acquired from individual subjects. Despite the widespread use of OCT to image the anterior and posterior mouse eye, few approaches to align the resulting image data have been described, in part due to a lack of well-characterized landmarks that are suitable for alignment. Here, we provide an OCT acquisition and analysis protocol that incorporates the use of the long posterior ciliary arteries as landmarks. In mammals, these two large choroidal vessels lie in a plane approximately parallel to the horizon. Our OCT imaging approach resolves these vessels in the mouse eye and suggests that their location is reproducible. The protocol may be useful for preparing 3D OCT data to compare experimental cohorts of mice and for standardizing results from independent research laboratories. © 2017 by John Wiley & Sons, Inc.


Asunto(s)
Coroides/irrigación sanguínea , Arterias Ciliares/diagnóstico por imagen , Imagenología Tridimensional , Animales , Ratones , Tomografía de Coherencia Óptica
17.
PLoS One ; 12(8): e0183837, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28859131

RESUMEN

Mouse models provide a valuable tool for exploring pathogenic mechanisms underlying inherited human disease. Here, we describe seven mouse models identified through the Translational Vision Research Models (TVRM) program, each carrying a new allele of a gene previously linked to retinal developmental and/or degenerative disease. The mutations include four alleles of three genes linked to human nonsyndromic ocular diseases (Aipl1tvrm119, Aipl1tvrm127, Rpgrip1tvrm111, RhoTvrm334) and three alleles of genes associated with human syndromic diseases that exhibit ocular phentoypes (Alms1tvrm102, Clcn2nmf289, Fkrptvrm53). Phenotypic characterization of each model is provided in the context of existing literature, in some cases refining our current understanding of specific disease attributes. These murine models, on fixed genetic backgrounds, are available for distribution upon request and may be useful for understanding the function of the gene in the retina, the pathological mechanisms induced by its disruption, and for testing experimental approaches to treat the corresponding human ocular diseases.


Asunto(s)
Oftalmopatías/genética , Degeneración Retiniana/genética , Investigación Biomédica Traslacional , Visión Ocular/genética , Alelos , Animales , Canales de Cloruro CLC-2 , Proteínas de Ciclo Celular , Canales de Cloruro/genética , Proteínas del Citoesqueleto , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Oftalmopatías/patología , Humanos , Ratones , Mutación , Pentosiltransferasa , Proteínas/genética , Retina/patología , Degeneración Retiniana/patología , Transferasas
18.
Protein Sci ; 15(7): 1679-90, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16815918

RESUMEN

The mechanisms by which G-protein-coupled receptors (GPCRs) activate G-proteins are not well understood due to the lack of atomic structures of GPCRs in an active form or in GPCR/G-protein complexes. For study of GPCR/G-protein interactions, we have generated a series of chimeras by replacing the third cytoplasmic loop of a scaffold protein bacteriorhodopsin (bR) with various lengths of cytoplasmic loop 3 of bovine rhodopsin (Rh), and one such chimera containing loop 3 of the human beta2-adrenergic receptor. The chimeras expressed in the archaeon Halobacterium salinarum formed purple membrane lattices thus facilitating robust protein purification. Retinal was correctly incorporated into the chimeras, as determined by spectrophotometry. A 2D crystal (lattice) was evidenced by circular dichroism analysis, and proper organization of homotrimers formed by the bR/Rh loop 3 chimera Rh3C was clearly illustrated by atomic force microscopy. Most interestingly, Rh3C (and Rh3G to a lesser extent) was functional in activation of GTPgamma35S/GDP exchange of the transducin alpha subunit (Galphat) at a level 3.5-fold higher than the basal exchange. This activation was inhibited by GDP and by a high-affinity peptide analog of the Galphat C terminus, indicating specificity in the exchange reaction. Furthermore, a specific physical interaction between the chimera Rh3C loop 3 and the Galphat C terminus was demonstrated by cocentrifugation of transducin with Rh3C. This Galphat-activating bR/Rh chimera is highly likely to be a useful tool for studying GPCR/G-protein interactions.


Asunto(s)
Bacteriorodopsinas/química , Rodopsina/química , Transducina/metabolismo , Animales , Bacteriorodopsinas/genética , Bovinos , Citoplasma , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/genética , Receptores Acoplados a Proteínas G/química , Proteínas Recombinantes de Fusión/química , Retinaldehído/química , Rodopsina/genética
19.
Methods Mol Biol ; 1438: 395-415, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27150100

RESUMEN

Noninvasive live imaging has been used extensively for ocular phenotyping in mouse vision research. Bright-field imaging and optical coherence tomography (OCT) are two methods that are particularly useful for assessing the posterior mouse eye (fundus), including the retina, retinal pigment epithelium, and choroid, and are widely applied due to the commercial availability of sophisticated instruments and software. Here, we provide a guide to using these approaches with an emphasis on post-acquisition image processing using Fiji, a bundled version of the Java-based public domain software ImageJ. A bright-field fundus imaging protocol is described for acquisition of multi-frame videos, followed by image registration to reduce motion artifacts, averaging to reduce noise, shading correction to compensate for uneven illumination, filtering to improve image detail, and rotation to adjust orientation. An OCT imaging protocol is described for acquiring replicate volume scans, with subsequent registration and averaging to yield three-dimensional datasets that show reduced motion artifacts and enhanced detail. The Fiji algorithms used in these protocols are designed for batch processing and are freely available. The image acquisition and processing approaches described here may facilitate quantitative phenotyping of the mouse eye in drug discovery, mutagenesis screening, and the functional cataloging of mouse genes by individual laboratories and large-scale projects, such as the Knockout Mouse Phenotyping Project and International Mouse Phenotyping Consortium.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/instrumentación , Retina/diagnóstico por imagen , Tomografía de Coherencia Óptica/métodos , Algoritmos , Animales , Fondo de Ojo , Ratones , Ratones Noqueados , Modelos Animales , Programas Informáticos , Tomografía de Coherencia Óptica/instrumentación
20.
Nat Genet ; 48(2): 144-51, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26691986

RESUMEN

Butterfly-shaped pigment dystrophy is an eye disease characterized by lesions in the macula that can resemble the wings of a butterfly. Here we report the identification of heterozygous missense mutations in the CTNNA1 gene (encoding α-catenin 1) in three families with butterfly-shaped pigment dystrophy. In addition, we identified a Ctnna1 missense mutation in a chemically induced mouse mutant, tvrm5. Parallel clinical phenotypes were observed in the retinal pigment epithelium (RPE) of individuals with butterfly-shaped pigment dystrophy and in tvrm5 mice, including pigmentary abnormalities, focal thickening and elevated lesions, and decreased light-activated responses. Morphological studies in tvrm5 mice demonstrated increased cell shedding and the presence of large multinucleated RPE cells, suggesting defects in intercellular adhesion and cytokinesis. This study identifies CTNNA1 gene variants as a cause of macular dystrophy, indicates that CTNNA1 is involved in maintaining RPE integrity and suggests that other components that participate in intercellular adhesion may be implicated in macular disease.


Asunto(s)
Mutación Missense , Distrofias Retinianas/genética , Epitelio Pigmentado de la Retina/patología , alfa Catenina/genética , Animales , Femenino , Humanos , Luz , Masculino , Ratones , Ratones Mutantes , Linaje , Distrofias Retinianas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA