Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Adv Exp Med Biol ; 723: 391-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22183357

RESUMEN

ENU mutagenesis is an efficient method to identify new animal models of ocular disease. The new alleles described herein will be a useful resource to further examine the role of the affected molecules and the effects of their disruption within the retina.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Ratones Endogámicos C57BL , Degeneración Retiniana/genética , Rodopsina/genética , Alquilantes/toxicidad , Animales , Mapeo Cromosómico/métodos , Análisis Mutacional de ADN/métodos , Etilnitrosourea/toxicidad , Humanos , Ratones , Mutagénesis/fisiología , Degeneración Retiniana/inducido químicamente , Investigación Biomédica Traslacional/métodos
9.
J Biol Chem ; 285(19): 14521-33, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20207741

RESUMEN

Mutations in the Rhodopsin (Rho) gene can lead to autosomal dominant retinitis pigmentosa (RP) in humans. Transgenic mouse models with mutations in Rho have been developed to study the disease. However, it is difficult to know the source of the photoreceptor (PR) degeneration in these transgenic models because overexpression of wild type (WT) Rho alone can lead to PR degeneration. Here, we report two chemically mutagenized mouse models carrying point mutations in Rho (Tvrm1 with an Y102H mutation and Tvrm4 with an I307N mutation). Both mutants express normal levels of rhodopsin that localize to the PR outer segments and do not exhibit PR degeneration when raised in ambient mouse room lighting; however, severe PR degeneration is observed after short exposures to bright light. Both mutations also cause a delay in recovery following bleaching. This defect might be due to a slower rate of chromophore binding by the mutant opsins compared with the WT form, and an increased rate of transducin activation by the unbound mutant opsins, which leads to a constitutive activation of the phototransduction cascade as revealed by in vitro biochemical assays. The mutant-free opsins produced by the respective mutant Rho genes appear to be more toxic to PRs, as Tvrm1 and Tvrm4 mutants lacking the 11-cis chromophore degenerate faster than mice expressing WT opsin that also lack the chromophore. Because of their phenotypic similarity to humans with B1 Rho mutations, these mutants will be important tools in examining mechanisms underlying Rho-induced RP and for testing therapeutic strategies.


Asunto(s)
Luz , Mutación Missense/genética , Células Fotorreceptoras/efectos de la radiación , Rodopsina/genética , Rodopsina/metabolismo , Secuencia de Aminoácidos , Animales , Electrorretinografía , Angiografía con Fluoresceína , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
10.
J Biol Chem ; 285(10): 7697-711, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20048158

RESUMEN

The Neuromutagenesis Facility at the Jackson Laboratory generated a mouse model of retinal vasculopathy, nmf223, which is characterized clinically by vitreal fibroplasia and vessel tortuosity. nmf223 homozygotes also have reduced electroretinogram responses, which are coupled histologically with a thinning of the inner nuclear layer. The nmf223 locus was mapped to chromosome 17, and a missense mutation was identified in Lama1 that leads to the substitution of cysteine for a tyrosine at amino acid 265 of laminin alpha1, a basement membrane protein. Despite normal localization of laminin alpha1 and other components of the inner limiting membrane, a reduced integrity of this structure was suggested by ectopic cells and blood vessels within the vitreous. Immunohistochemical characterization of nmf223 homozygous retinas demonstrated the abnormal migration of retinal astrocytes into the vitreous along with the persistence of hyaloid vasculature. The Y265C mutation significantly reduced laminin N-terminal domain (LN) interactions in a bacterial two-hybrid system. Therefore, this mutation could affect interactions between laminin alpha1 and other laminin chains. To expand upon these findings, a Lama1 null mutant, Lama1(tm1.1Olf), was generated that exhibits a similar but more severe retinal phenotype than that seen in nmf223 homozygotes. The increased severity of the Lama1 null mutant phenotype is probably due to the complete loss of the inner limiting membrane in these mice. This first report of viable Lama1 mouse mutants emphasizes the importance of this gene in retinal development. The data presented herein suggest that hypomorphic mutations in human LAMA1 could lead to retinal disease.


Asunto(s)
Laminina , Mutación Missense , Isoformas de Proteínas , Retina , Enfermedades de la Retina , Vasos Retinianos , Adulto , Secuencia de Aminoácidos , Animales , Astrocitos/citología , Astrocitos/metabolismo , Membrana Basal/citología , Membrana Basal/metabolismo , Electrorretinografía , Femenino , Prueba de Complementación Genética , Humanos , Laminina/genética , Laminina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Fenotipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Retina/anomalías , Retina/anatomía & histología , Retina/fisiología , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Vasos Retinianos/anomalías , Vasos Retinianos/anatomía & histología , Vasos Retinianos/fisiología , Alineación de Secuencia , Transgenes
11.
Hum Mol Genet ; 18(22): 4329-39, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19679561

RESUMEN

The function of the retinitis pigmentosa GTPase regulator interacting protein 1 (RPGRIP1) gene is currently not known. However, mutations within the gene lead to Leber Congenital Amaurosis and autosomal recessive retinitis pigmentosa in human patients. In a previously described knockout mouse model of the long splice variant of Rpgrip1, herein referred to as Rpgrip1(tm1Tili) mice, mislocalization of key outer segment proteins and dysmorphogenesis of outer segment discs preceded subsequent photoreceptor degeneration. In this report, we describe a new mouse model carrying a splice acceptor site mutation in Rpgrip1, herein referred to as Rpgrip1(nmf247) that is phenotypically distinct from Rpgrip1(tm1Tili) mice. Photoreceptor degeneration in homozygous Rpgrip1(nmf247) mice is earlier in onset and more severe when compared with Rpgrip1(tm1Tili) mice. Also, ultrastructural studies reveal that whereas Rpgrip1(nmf247) mutants have a normal structure and number of connecting cilia, unlike Rpgrip1(tm1Tili) mice, they do not elaborate rod outer segments (OS). Therefore, in addition to its role in OS disc morphogenesis, RPGRIP1 is essential for rod OS formation. Our study indicates the absence of multiple Rpgrip1 isoforms in Rpgrip1(nmf247) mice, suggesting different isoforms may play different roles in photoreceptors and underscores the importance of considering splice variants when generating targeted null mutations.


Asunto(s)
Morfogénesis , Proteínas/metabolismo , Retina/crecimiento & desarrollo , Retinitis Pigmentosa/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Mutación , Transporte de Proteínas , Proteínas/química , Proteínas/genética , Retina/metabolismo , Retinitis Pigmentosa/genética , Segmento Externo de la Célula en Bastón/química , Alineación de Secuencia
12.
J Physiol ; 586(18): 4409-24, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18687716

RESUMEN

An electroretinogram (ERG) screen identified a mouse with a normal a-wave but lacking a b-wave, and as such it was designated no b-wave3 (nob3). The nob3 phenotype mapped to chromosome 11 in a region containing the metabotropic glutamate receptor 6 gene (Grm6). Sequence analyses of cDNA identified a splicing error in Grm6, introducing an insertion and an early stop codon into the mRNA of affected mice (designated Grm6(nob3)). Immunohistochemistry of the Grm6(nob3) retina showed that GRM6 was absent. The ERG and visual behaviour abnormalities of Grm6(nob3) mice are similar to Grm6(nob4) animals, and similar deficits were seen in compound heterozygotes (Grm6(nob4/nob3)), indicating that Grm6(nob3) is allelic to Grm6(nob4). Visual responses of Grm6(nob3) retinal ganglion cells (RGCs) to light onset were abnormal. Grm6(nob3) ON RGCs were rarely recorded, but when they were, had ill-defined receptive field (RF) centres and delayed onset latencies. When Grm6(nob3) OFF-centre RGC responses were evoked by full-field stimulation, significantly fewer converted that response to OFF/ON compared to Grm6(nob4) RGCs. Grm6(nob4/nob3) RGC responses verified the conclusion that the two mutants are allelic. We propose that Grm6(nob3) is a new model of human autosomal recessive congenital stationary night blindness. However, an allelic difference between Grm6(nob3) and Grm6(nob4) creates a disparity in inner retinal processing. Because the localization of GRM6 is limited to bipolar cells in the On pathway, the observed difference between RGCs in these mutants is likely to arise from differences in their inputs.


Asunto(s)
Receptores de Glutamato Metabotrópico/genética , Células Ganglionares de la Retina/fisiología , Alelos , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Electrorretinografía , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutación , Estimulación Luminosa , Receptores de Glutamato Metabotrópico/metabolismo , Retina/anatomía & histología , Retina/metabolismo , Análisis de Secuencia de ADN , Campos Visuales
13.
Vis Neurosci ; 25(4): 563-74, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18764959

RESUMEN

A 4 base pair deletion in a splice donor site of the Mfrp (membrane-type frizzled-related protein) gene, herein referred to as Mfrprd6/rd6, is predicted to lead to the skipping of exon 4 and photoreceptor degeneration in retinal degeneration 6 (rd6) mutant mice. Little, however, is known about the function of the protein or how the mutation causes the degenerative retinal phenotype. Here we examine ultrastructural changes in the retina of Mfrprd6/rd6 mice to determine the earliest effects of the mutation. We also extend the reported observations of the expression pattern of the dicistronic Mfrp/C1qtnf5 message and the localization of these and other retinal pigment epithelium (RPE) and retinal proteins during development and assess the ability of RPE cells to phagocytize outer segments (OSs) in mutant and wild-type (WT) mice. At the ultrastructural level, OSs do not develop normally in Mfrprd6/rd6 mutants. They are disorganized and become progressively shorter as mutant mice age. Additionally, there are focal areas in which there is a reduction of apical RPE microvilli. At P25, the rod electroretinogram (ERG) a-wave of Mfrprd6/rd6 mice is reduced in amplitude by ~50% as are ERG components generated by the RPE. Examination of beta-catenin localization and Fos and Tcf-1 expression, intermediates of the canonical Wnt pathway, showed that they were not different between mutant and WT mice, suggesting that MFRP may operate through an alternative pathway. Finally, impaired OS phagocytosis was observed in Mfrprd6/rd6 mice both in standard ambient lighting conditions and with bright light exposure when compared to WT controls.


Asunto(s)
Proteínas del Ojo/fisiología , Proteínas de la Membrana/fisiología , Células Fotorreceptoras/fisiología , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Regulación hacia Abajo , Electrorretinografía , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Eliminación de Gen , Expresión Génica , Luz , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fagocitosis/efectos de la radiación , Células Fotorreceptoras/patología , Epitelio Pigmentado Ocular/patología , Retina/fisiopatología , Retina/ultraestructura , Células Fotorreceptoras Retinianas Bastones/patología , Distribución Tisular
14.
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
15.
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
16.
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
17.
PLoS One ; 9(10): e110299, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25357075

RESUMEN

Mutations in the membrane frizzled-related protein (MFRP/Mfrp) gene, specifically expressed in the retinal pigment epithelium (RPE) and ciliary body, cause nanophthalmia or posterior microphthalmia with retinitis pigmentosa in humans, and photoreceptor degeneration in mice. To better understand MFRP function, microarray analysis was performed on eyes of homozygous Mfrprd6 and C57BL/6J mice at postnatal days (P) 0 and P14, prior to photoreceptor loss. Data analysis revealed no changes at P0 but significant differences in RPE and retina-specific transcripts at P14, suggesting a postnatal influence of the Mfrprd6 allele. A subset of these transcripts was validated by quantitative real-time PCR (qRT-PCR). In Mfrprd6 eyes, a significant 1.5- to 2.0-fold decrease was observed among transcripts of genes linked to retinal degeneration, including those involved in visual cycle (Rpe65, Lrat, Rgr), phototransduction (Pde6a, Guca1b, Rgs9), and photoreceptor disc morphogenesis (Rpgrip1 and Fscn2). Levels of RPE65 were significantly decreased by 2.0-fold. Transcripts of Prss56, a gene associated with angle-closure glaucoma, posterior microphthalmia and myopia, were increased in Mfrprd6 eyes by 17-fold. Validation by qRT-PCR indicated a 3.5-, 14- and 70-fold accumulation of Prss56 transcripts relative to controls at P7, P14 and P21, respectively. This trend was not observed in other RPE or photoreceptor mutant mouse models with similar disease progression, suggesting that Prss56 upregulation is a specific attribute of the disruption of Mfrp. Prss56 and Glul in situ hybridization directly identified Müller glia in the inner nuclear layer as the cell type expressing Prss56. In summary, the Mfrprd6 allele causes significant postnatal changes in transcript and protein levels in the retina and RPE. The link between Mfrp deficiency and Prss56 up-regulation, together with the genetic association of human MFRP or PRSS56 variants and ocular size, raises the possibility that these genes are part of a regulatory network influencing postnatal posterior eye development.


Asunto(s)
Proteínas del Ojo , Regulación Enzimológica de la Expresión Génica , Proteínas de la Membrana , Retina/metabolismo , Serina Proteasas/biosíntesis , Regulación hacia Arriba , Visión Ocular/genética , Animales , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Perfilación de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Microftalmía/genética , Microftalmía/metabolismo , Microftalmía/patología , Retina/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Serina Proteasas/genética
18.
Invest Ophthalmol Vis Sci ; 53(2): 967-74, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22247471

RESUMEN

PURPOSE: Cilia, complex structures found ubiquitously in most vertebrate cells, serve a variety of functions ranging from cell and fluid movement, cell signaling, tissue homeostasis, to sensory perception. Meckelin is a component of ciliary and cell membranes and is encoded by Tmem67 (Mks3). In this study, the retinal morphology and ciliary function in a mouse model for Meckel Syndrome Type 3 (MKS3) throughout the course of photoreceptor development was examined. METHODS: To study the effects of a disruption in the Mks3 gene on the retina, the authors introduced a functional allele of Pde6b into B6C3Fe a/a-bpck/J mice and evaluated their retinas by ophthalmoscopic, histologic, and ultrastructural examination. In addition, immunofluorescence microscopy was used to assess protein trafficking through the connecting cilium and to examine the localization of ciliary and synaptic proteins in Tmem67(bpck) mice and controls. RESULTS: Photoreceptors degenerate early and rapidly in bpck/bpck mutant mice. In addition, phototransduction proteins, such as rhodopsin, arrestin, and transducin, are mislocalized. Ultrastructural examination of photoreceptors reveal morphologically intact connecting cilia but dysmorphic and misoriented outer segment (OS) discs, at the earliest time point examined. CONCLUSIONS: These findings underscore the important role for meckelin in intraciliary transport of phototransduction molecules and their effects on subsequent OS morphogenesis and maintenance.


Asunto(s)
Trastornos de la Motilidad Ciliar/genética , ADN/genética , Encefalocele/genética , Proteínas de la Membrana/genética , Morfogénesis/efectos de los fármacos , Mutación , Enfermedades Renales Poliquísticas/genética , Segmento Externo de la Célula en Bastón/fisiología , Animales , Cilios/genética , Cilios/metabolismo , Cilios/ultraestructura , Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/patología , Modelos Animales de Enfermedad , Encefalocele/metabolismo , Encefalocele/patología , Genotipo , Inmunohistoquímica , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Reacción en Cadena de la Polimerasa , Transporte de Proteínas , Segmento Externo de la Célula en Bastón/ultraestructura
19.
J Ophthalmol ; 2011: 391384, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21052544

RESUMEN

The need for mouse models, with their well-developed genetics and similarity to human physiology and anatomy, is clear and their central role in furthering our understanding of human disease is readily apparent in the literature. Mice carrying mutations that alter developmental pathways or cellular function provide model systems for analyzing defects in comparable human disorders and for testing therapeutic strategies. Mutant mice also provide reproducible, experimental systems for elucidating pathways of normal development and function. Two programs, the Eye Mutant Resource and the Translational Vision Research Models, focused on providing such models to the vision research community are described herein. Over 100 mutant lines from the Eye Mutant Resource and 60 mutant lines from the Translational Vision Research Models have been developed. The ocular diseases of the mutant lines include a wide range of phenotypes, including cataracts, retinal dysplasia and degeneration, and abnormal blood vessel formation. The mutations in disease genes have been mapped and in some cases identified by direct sequencing. Here, we report 3 novel alleles of Crx(tvrm65), Rp1(tvrm64), and Rpe65(tvrm148) as successful examples of the TVRM program, that closely resemble previously reported knockout models.

20.
Invest Ophthalmol Vis Sci ; 52(7): 4703-9, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21436282

RESUMEN

PURPOSE: To determine the basis and to characterize the phenotype of a chemically induced mutation in a mouse model of retinal degeneration. METHODS: Screening by indirect ophthalmoscopy identified a line of N-ethyl-N-nitrosourea (ENU) mutagenized mice demonstrating retinal patches. Longitudinal studies of retinal histologic sections showed photoreceptors in the peripheral retina undergoing slow, progressive degeneration. The mutation was named neuroscience mutagenesis facility 12 (nmf12), and mapping localized the critical region to Chromosome 2. RESULTS: Sequencing of nmf12 DNA revealed a point mutation in the c-mer tyrosine kinase gene, designated Mertk(nmf12). We detected elevated levels of tumor necrosis factor (Tnf, previously Tnfa) in retinas of Mertk(nmf12) homozygotes relative to wild-type controls and investigated whether the increase of TNF, an inflammatory cytokine produced by macrophages/monocytes that signals intracellularly to cause necrosis or apoptosis, could underlie the retinal degeneration observed in Mertk(nmf12) homozygotes. Mertk(nmf12) homozygous mice were mated to mice lacking the entire Tnf gene and partial coding sequences of the Lta (Tnfb) and Ltb (Tnfc) genes.(2) B6.129P2-Ltb/Tnf/Lta(tm1Dvk)/J homozygotes did not exhibit a retinal degeneration phenotype and will, hereafter, be referred to as Tnfabc(-/-) mice. Surprisingly, mice homozygous for both the Mertk(nmf12) and the Ltb/Tnf/Lta(tm1Dvk) allele (Tnfabc(-/-)) demonstrated an increase in the rate of retinal degeneration. CONCLUSIONS: These findings illustrate that a mutation in the Mertk gene leads to a significantly slower progressive retinal degeneration compared with other alleles of Mertk. These results demonstrate that TNF family members play a role in protecting photoreceptors of Mertk(nmf12) homozygotes from cell death.


Asunto(s)
ADN/genética , Etilnitrosourea/toxicidad , Mutación/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Retina/metabolismo , Degeneración Retiniana/genética , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Electrorretinografía , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Oftalmoscopía , Fenotipo , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Retina/efectos de los fármacos , Retina/patología , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Factor de Necrosis Tumoral alfa/biosíntesis , Tirosina Quinasa c-Mer
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA