Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
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.
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
5.
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
6.
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
7.
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
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.
Sci Rep ; 12(1): 20866, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36460842

RESUMEN

The house mouse species complex (Mus musculus) is comprised of three primary subspecies. A large number of secondary subspecies have also been suggested on the basis of divergent morphology and molecular variation at limited numbers of markers. While the phylogenetic relationships among the primary M. musculus subspecies are well-defined, relationships among secondary subspecies and between secondary and primary subspecies remain less clear. Here, we integrate de novo genome sequencing of museum-stored specimens of house mice from one secondary subspecies (M. m. bactrianus) and publicly available genome sequences of house mice previously characterized as M. m. helgolandicus, with whole genome sequences from diverse representatives of the three primary house mouse subspecies. We show that mice assigned to the secondary M. m. bactrianus and M. m. helgolandicus subspecies are not genetically differentiated from M. m. castaneus and M. m. domesticus, respectively. Overall, our work suggests that the M. m. bactrianus and M. m. helgolandicus subspecies are not well-justified taxonomic entities, emphasizing the importance of leveraging whole-genome sequence data to inform subspecies designations. Additionally, our investigation provides tailored experimental procedures for generating whole genome sequences from air-dried mouse skins, along with key genomic resources to inform future genomic studies of wild mouse diversity.


Asunto(s)
Genómica , Músculos , Animales , Ratones , Filogenia , Secuenciación Completa del Genoma , Mapeo Cromosómico
11.
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
12.
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
13.
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
14.
Methods Cell Biol ; 105: 87-116, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21951527

RESUMEN

All animals are ecosystems, home to diverse microbial populations. Animal-associated microbes play important roles in the normal development and physiology of their hosts, but can also be agents of infectious disease. Traditionally, mice have been used to study pathogenic and beneficial associations between microbes and vertebrate animals. The zebrafish is emerging as a valuable new model system for host-microbe interaction studies, affording researchers with the opportunity to survey large populations of hosts and to visualize microbe-host associations at a cellular level in living animals. This chapter provides detailed protocols for the analysis of zebrafish-associated microbial communities, the derivation and husbandry of germ-free zebrafish, and the modeling of infectious disease in different stages of zebrafish development via different routes of inoculation. These protocols offer a starting point for researchers to address a multitude of questions about animals' coexistence with microorganisms.


Asunto(s)
Técnicas de Tipificación Bacteriana , Biología Evolutiva/métodos , Vida Libre de Gérmenes , Interacciones Huésped-Patógeno , Hibridación Fluorescente in Situ/métodos , Larva , Microinyecciones/métodos , Pez Cebra , Animales , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/embriología , Infecciones Bacterianas/microbiología , Modelos Animales de Enfermedad , Embrión no Mamífero/microbiología , Embrión no Mamífero/virología , Intestinos/microbiología , Intestinos/virología , Larva/crecimiento & desarrollo , Larva/microbiología , Larva/virología , Ratones , Consorcios Microbianos , Interacciones Microbianas , ARN Bacteriano/análisis , ARN Ribosómico 16S/análisis , Virosis/embriología , Virosis/virología , Virus/crecimiento & desarrollo , Pez Cebra/embriología , Pez Cebra/microbiología , Pez Cebra/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA