Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 17(24): 3929-41, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18805803

ABSTRACT

We observed that a naturally occurring mouse strain developed age-related retinal degeneration (arrd2). These mice had normal fundi, electroretinograms (ERGs) and retinal histology at 6 months of age; vessel attenuation, RPE atrophy and pigmentary abnormalities at 14 months, which progressed to complete loss of photoreceptors and extinguished ERG by 22 months. Genetic analysis revealed that the retinal degeneration in arrd2 segregates in an autosomal recessive manner and the disease gene localizes to mouse chromosome 10. A positional candidate cloning approach detected a nonsense mutation in the mouse double minute-1 gene (Mdm1), which results in the truncation of the putative protein from 718 amino acids to 398. We have identified a novel transcript of the Mdm1 gene, which is the predominant transcript in the retina. The Mdm1 transcript is localized to the nuclear layers of neural retina. Expression of Mdm1 in the retina increases steadily from post-natal day 30 to 1 year, and a high level of Mdm1 are subsequently maintained. The Mdm1 transcript was found to be significantly depleted in the retina of arrd2 mice and the transcript was observed to degrade by nonsense-mediated decay. These results indicate that the depletion of the Mdm1 transcript may underlie the mechanism leading to late-onset progressive retinal degeneration in arrd2 mice. Analysis of a cohort of patients with age-related macular degeneration (AMD) wherein the susceptibility locus maps to chromosome 12q, a region bearing the human ortholog to MDM1, did not reveal association between human MDM1 and AMD.


Subject(s)
Aging/genetics , Codon, Nonsense/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Animals , Electroretinography , Female , Gene Deletion , Humans , Male , Mice , Mice, Inbred C57BL , Sequence Analysis, DNA
2.
Pigment Cell Melanoma Res ; 21(5): 565-78, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18715234

ABSTRACT

Spontaneous mutations altering mouse coat colors have been a classic resource for discovery of numerous molecular pathways. Although often overlooked, the mouse iris is also densely pigmented and easily observed, thus representing a similarly powerful opportunity for studying pigment cell biology. Here, we present an analysis of iris phenotypes among 16 mouse strains with mutations influencing melanosomes. Many of these strains exhibit biologically and medically relevant phenotypes, including pigment dispersion, a common feature of several human ocular diseases. Pigment dispersion was identified in several strains with mutant alleles known to influence melanosomes, including beige, light, and vitiligo. Pigment dispersion was also detected in the recently arising spontaneous coat color variant, nm2798. We have identified the nm2798 mutation as a missense mutation in the Dct gene, an identical re-occurrence of the slaty light mutation. These results suggest that dysregulated events of melanosomes can be potent contributors to the pigment dispersion phenotype. Combined, these findings illustrate the utility of studying iris phenotypes as a means of discovering new pathways, and re-linking old ones, to processes of pigmented cells in health and disease.


Subject(s)
Iris , Phenotype , Pigmentation/genetics , Pigments, Biological/metabolism , Animals , Base Sequence , Hair , Humans , Iris/anatomy & histology , Iris/physiology , Melanosomes/physiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mutation , Oxidoreductases/genetics , Oxidoreductases/metabolism , Sequence Analysis, DNA
3.
J Physiol ; 586(18): 4409-24, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18687716

ABSTRACT

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.


Subject(s)
Receptors, Metabotropic Glutamate/genetics , Retinal Ganglion Cells/physiology , Alleles , Amino Acid Sequence , Animals , Chromosome Mapping , Electroretinography , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Photic Stimulation , Receptors, Metabotropic Glutamate/metabolism , Retina/anatomy & histology , Retina/metabolism , Sequence Analysis, DNA , Visual Fields
4.
Invest Ophthalmol Vis Sci ; 49(10): 4278-83, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18586879

ABSTRACT

PURPOSE: To test AAV-mediated gene therapy in the rd10 mouse, a natural model of recessive RP caused by mutation of the beta-subunit of rod photoreceptor cGMP phosphodiesterase. METHODS: One eye of a cohort of rd10 mice kept in a dark environment was subretinally injected at postnatal day (P) 14 with 1 microL AAV5-smCBA-PDEbeta. The contralateral eye was not injected. The animals were then maintained for 2 weeks in the dark before they were moved to a normal 12-hour light/12-hour dark cycling light environment for visually guided behavioral training. Three weeks after injection, treated rd10 mice were examined by scotopic and photopic electroretinography and then killed for biochemical and morphologic examination. RESULTS: Substantial scotopic ERG signals were maintained in treated rd10 eyes, whereas untreated eyes in the same animals showed minimal signals. Treated eyes showed photopic ERG b-wave amplitudes similar to those of the normal eyes; in untreated partner eyes, only half the normal amplitudes remained. Strong PDEbeta expression was observed in photoreceptor outer segments only in treated eyes. Light microscopy showed a substantial preservation of the outer nuclear layer in most parts of the treated retina only. Electron microscopy showed good outer segment preservation only in treated eyes. A visually guided water maze behavioral test under dim light showed significantly improved performance in one eye-treated rd10 mice compared with untreated mice. CONCLUSIONS: These data demonstrate that P14 administration of AAV5-smCBA-PDEbeta can prevent retinal degeneration in rd10 mice, as reflected by significant structural, biochemical, electrophysiological, and behavioral preservation/restoration. These results serve as a baseline for studying long-term retinal rescue in rd10 mice.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Mutation , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/prevention & control , Animals , Animals, Congenic , Animals, Newborn , Blotting, Western , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Electroretinography , Female , Fluorescent Antibody Technique, Indirect , Genes, Recessive , Mice , Mice, Inbred C57BL , Pregnancy , Psychomotor Performance/physiology , Retina/metabolism , Retina/pathology , Retina/physiopathology , Retinitis Pigmentosa/physiopathology
5.
Invest Ophthalmol Vis Sci ; 49(1): 304-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18172107

ABSTRACT

PURPOSE: To identify the causative gene mutation for a new dominant cataract in mice and to investigate the molecular basis for how the mutated gene leads to a dense nuclear cataract. METHODS: Genomewide linkage analysis and DNA sequencing were used to determine the gene mutation. Histology, immunohistochemistry, and Western blotting were used to characterize lens phenotypes. Ion concentrations were measured by an inductively coupled plasma-optical emission spectrometer (ICP-OES). RESULTS: A point mutation (A to C) of the gammaB-crystallin gene, which results in the gammaB-S11R mutant protein, was identified in this cataractous mouse line. Homozygous mutant mice developed dense nuclear cataracts associated with disrupted inner lens fiber cells. Immunohistochemistry data revealed gamma-crystallin aggregates at the cell boundaries of inner mature fibers that lose actin filaments. Western blotting showed an increased degradation of crystallin proteins correlated with the nuclear cataract. ICP-OES confirmed a substantial elevation of calcium concentration in mutant lenses. CONCLUSIONS: This dominant cataract was caused by the gammaB-S11R mutation. Mutant gammaB-S11R proteins triggered the gamma-crystallin aggregation that probably disrupted membrane-cytoskeleton structures of inner fiber cells, causing increased calcium influxes. Subsequent activation of calcium-dependent protein degradation and degeneration of inner mature fiber cells led to the dense nuclear cataract.


Subject(s)
Cataract/genetics , Disease Models, Animal , Lens Nucleus, Crystalline/pathology , Point Mutation , gamma-Crystallins/genetics , Animals , Blotting, Western , Calcium/metabolism , Cataract/metabolism , Chromosome Mapping , Crystallins/metabolism , Genes, Dominant , Lens Nucleus, Crystalline/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Confocal , Phenotype , Sequence Analysis, DNA , Spectrometry, X-Ray Emission
6.
Genetics ; 177(1): 307-11, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17890364

ABSTRACT

Mice heterozygous for missense mutations of the Notch ligand Jagged1 (Jag1) exhibit head-shaking behavior indicative of an inner ear vestibular defect. In contrast, mice heterozygous for a targeted deletion of the Jag1 gene (Jag1del1) do not demonstrate obvious head-shaking behavior. To determine whether the differences in inner ear phenotypes were due to the types of Jag1 mutations or to differences in genetic background, we crossed Jag1del1 heterozygous mice onto the same genetic background as the missense mutants. This analysis revealed that variation of the Jag1 mutant inner ear phenotype is caused by genetic background differences and is not due to the type of Jag1 mutation. Genome scans of N2 backcross mice identified a significant modifier locus on chromosome 7, as well as a suggestive locus on chromosome 14. We also analyzed modifiers of an eye defect in Jag1del1 heterozygous mice from this same cross.


Subject(s)
Calcium-Binding Proteins/physiology , Cochlear Diseases/etiology , Ear, Inner/physiology , Eye Diseases/etiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Animals , Behavior, Animal , Calcium-Binding Proteins/genetics , Cochlear Diseases/pathology , Eye Diseases/pathology , Female , Heterozygote , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Phenotype , Serrate-Jagged Proteins
7.
Nat Med ; 13(6): 685-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17515894

ABSTRACT

Loss of cone function in the central retina is a pivotal event in the development of severe vision impairment for many prevalent blinding diseases. Complete achromatopsia is a genetic defect resulting in cone vision loss in 1 in 30,000 individuals. Using adeno-associated virus (AAV) gene therapy, we show that it is possible to target cones and rescue both the cone-mediated electroretinogram response and visual acuity in the Gnat2 ( cpfl3 ) mouse model of achromatopsia.


Subject(s)
Color Vision Defects/therapy , Disease Models, Animal , Genetic Therapy , Retinal Cone Photoreceptor Cells/physiology , Animals , Color Vision Defects/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Mice , Mice, Transgenic
8.
Am J Hum Genet ; 79(6): 1059-70, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17186464

ABSTRACT

The rd3 mouse is one of the oldest identified models of early-onset retinal degeneration. Using the positional candidate approach, we have identified a C-->T substitution in a novel gene, Rd3, that encodes an evolutionarily conserved protein of 195 amino acids. The rd3 mutation results in a predicted stop codon after residue 106. This change is observed in four rd3 lines derived from the original collected mice but not in the nine wild-type mouse strains that were examined. Rd3 is preferentially expressed in the retina and exhibits increasing expression through early postnatal development. In transiently transfected COS-1 cells, the RD3-fusion protein shows subnuclear localization adjacent to promyelocytic leukemia-gene-product bodies. The truncated mutant RD3 protein is detectable in COS-1 cells but appears to get degraded rapidly. To explore potential association of the human RD3 gene at chromosome 1q32 with retinopathies, we performed a mutation screen of 881 probands from North America, India, and Europe. In addition to several alterations of uncertain significance, we identified a homozygous alteration in the invariant G nucleotide of the RD3 exon 2 donor splice site in two siblings with Leber congenital amaurosis. This mutation is predicted to result in premature truncation of the RD3 protein, segregates with the disease, and is not detected in 121 ethnically matched control individuals. We suggest that the retinopathy-associated RD3 protein is part of subnuclear protein complexes involved in diverse processes, such as transcription and splicing.


Subject(s)
Cell Nucleus Structures/metabolism , Nuclear Proteins/genetics , Retinal Degeneration/genetics , Amino Acid Sequence , Animals , COS Cells/metabolism , Case-Control Studies , Chlorocebus aethiops , Disease Models, Animal , Europe , Gene Expression Regulation, Developmental , Humans , India , Mice , Mice, Mutant Strains , Molecular Sequence Data , Mutation , North America , Nuclear Proteins/metabolism , Retinal Diseases/genetics , Sequence Homology, Amino Acid
9.
Genomics ; 88(1): 44-51, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16595169

ABSTRACT

Lens opacity 11 (lop11) is an autosomal recessive mouse cataract mutation that arose spontaneously in the RIIIS/J strain. At 3 weeks of age mice exhibit total cataracts with vacuoles. The lop11 locus was mapped to mouse chromosome 8. Analysis of the mouse genome for the lop11 critical region identified Hsf4 as a candidate gene. Molecular evaluation of Hsf4 revealed an early transposable element (ETn) in intron 9 inserted 61 bp upstream of the intron/exon junction. The same mutation was also identified in a previously mapped cataract mutant, ldis1. The ETn insertion altered splicing and expression of the Hsf4 gene, resulting in the truncated Hsf4 protein. In humans, mutations in HSF4 have been associated with both autosomal dominant and recessive cataracts. The lop11 mouse is an excellent resource for evaluating the role of Hsf4 in transparency of the lens.


Subject(s)
Cataract/genetics , DNA Transposable Elements/genetics , DNA-Binding Proteins/genetics , Eye Diseases, Hereditary/genetics , Eye Proteins/genetics , Introns , Transcription Factors/genetics , Animals , Base Sequence , Cataract/physiopathology , DNA-Binding Proteins/physiology , Disease Models, Animal , Eye Diseases, Hereditary/physiopathology , Eye Proteins/physiology , Genes, Recessive , Heat Shock Transcription Factors , Mice , Mice, Inbred Strains , Molecular Sequence Data , Transcription Factors/physiology
10.
Invest Ophthalmol Vis Sci ; 47(3): 1201-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16505059

ABSTRACT

PURPOSE: Viable motheaten mutant mice (abbreviated allele symbol me(v)) are deficient in Src-homology 2-domain phosphatase (SHP)-1, a critical negative regulator of signal transduction in hematopoietic cells. These mice exhibit immune dysfunction, hyperproliferation of myeloid cells, and regenerative anemia. This study focused on the role of SHP-1 in retinal homeostasis. METHODS: Ophthalmoscopy, histology, transmission electron microscopy (TEM), electroretinography (ERG), immunohistochemistry, Western blot, bone marrow transplantation, and genetic crosses were performed for phenotypic characterization and functional studies of retinal degeneration (RD) in me(v)/me(v) mice. RESULTS: Fundus examinations of me(v)/me(v) mice revealed numerous, small white spots. Histologic examination demonstrated photoreceptor loss beginning at 3 weeks of age, and TEM revealed disorganization and reduction in the number of outer segments, as well as the presence of phagocytic cells in the subretinal space. Rod- and cone-mediated ERGs were abnormal. SHP-1 protein was expressed in mouse and human retinal lysates and was localized to the outer nuclear layer of the retina in me(v)/me(v) and control mice. Autoantibodies are not necessary for RD, as B-cell-deficient me(v)/me(v) Igh-6(tm1Cgn) mice had no attenuation of photoreceptor cell loss compared with age-matched me(v)/me(v) mice. Histologic examination of lungs and retinas from normal recipients of me(v)/me(v) marrow revealed the classic acidophilic macrophage pneumonia of me(v)/me(v) mice, but no retinal degeneration. CONCLUSIONS: me(v)/me(v) mice exhibit normal retinal development with the onset of RD at 3 weeks of age and a rapidly progressive loss of photoreceptors. These findings support the hypothesis that SHP-1 plays a critical role in retinal homeostasis.


Subject(s)
Intracellular Signaling Peptides and Proteins/deficiency , Protein Tyrosine Phosphatases/deficiency , Retinal Degeneration/enzymology , Animals , Apoptosis , Blotting, Western , Bone Marrow Transplantation , Crosses, Genetic , Electroretinography , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Ophthalmoscopy , Photoreceptor Cells, Vertebrate/enzymology , Photoreceptor Cells, Vertebrate/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Retinal Degeneration/physiopathology
11.
Mol Vis ; 11: 152-62, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15765048

ABSTRACT

PURPOSE: To report the phenotype and characterization of a new, naturally occurring mouse model of hereditary retinal degeneration (rd12). METHODS: The retinal phenotype of rd12 mice were studied using serial indirect ophthalmoscopy, fundus photography, electroretinography (ERG), genetic analysis including linkage studies and gene identification, immunohistochemistry, and biochemical analysis. RESULTS: Mice homozygous for the rd12 mutation showed small punctate white spots on fundus examination at 5 months of age. The retina in the rd12 homozygote had a normal appearance at the light microscopic level until 6 weeks of age when occasional voids appeared in the outer segments (OS) of the photoreceptor (PR) cells. The outer nuclear layer (ONL) appeared normal until 3 months of age though more obvious voids were detected in the OS. By 7 months of age, 6 to 8 layers of ONL remained in the mutant retina, and the OS were obviously shorter. The first sign of retinal degeneration was detected at the electron microscopic level around 3 weeks of age when occasional small lipid-like droplets were detected in the retinal pigment epithelium (RPE). By 3 months of age, much larger, lipid-like droplets accumulated in RPE cells accompanied by some OS degeneration. While the histology indicated a relatively slow retinal degeneration in the rd12 homozygous mutant mice, the rod ERG response was profoundly diminished even at 3 weeks of age. Genetic analysis showed that rd12 was an autosomal recessive mutation and mapped to mouse chromosome 3 closely linked to D3Mit19, a location known to be near the mouse Rpe65 gene. Sequence analysis showed that the mouse retinal degeneration is caused by a nonsense mutation in exon 3 of the Rpe65 gene, and the gene symbol for the rd12 mutation has been updated to Rpe65rd12 to reflect this. No RPE65 expression, 11-cis retinal, or rhodopsin could be detected in retinas from rd12 homozygotes, while retinyl esters were found to accumulate in the retinal pigment epithelium (RPE). CONCLUSIONS: Mutations in the retinal pigment epithelium gene encoding RPE65 cause an early onset autosomal recessive form of human retinitis pigmentosa, known as Leber congenital amaurosis (LCA), which results in blindness or severely impaired vision in children. A naturally arising mouse Rpe65 mutation provides a good model for studying the pathology of human RPE65 mutations and the effects of retinyl ester accumulation.


Subject(s)
Blindness/congenital , Codon, Nonsense , Disease Models, Animal , Eye Proteins/genetics , Retinal Degeneration/genetics , Animals , Blindness/metabolism , Blindness/pathology , Carrier Proteins , Electroretinography , Exons/genetics , Eye Proteins/metabolism , Female , Fluorescent Antibody Technique, Indirect , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Ophthalmoscopy , Phenotype , Photoreceptor Cells, Vertebrate/pathology , Pigment Epithelium of Eye/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinaldehyde/metabolism , Rhodopsin/metabolism , cis-trans-Isomerases
12.
Hum Mol Genet ; 12(23): 3075-86, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14519688

ABSTRACT

We mapped two new recessive mutations causing circling behavior and deafness to the same region on chromosome 7 and showed they are allelic by complementation analysis. One was named 'deaf circler' (allele symbol dfcr) and the other 'deaf circler 2 Jackson' (allele symbol dfcr-2J). Both were shown to be mutations of the Ush1c gene, the mouse ortholog of the gene responsible for human Usher syndrome type IC and for the non-syndromic deafness disorder DFNB18. The Ush1c gene contains 28 exons, 20 that are constitutive and eight that are alternatively spliced. The dfcr mutation is a 12.8 kb intragenic deletion that eliminates three constitutive and five alternatively spliced exons. The dfcr-2J mutation is a 1 bp deletion in an alternatively spliced exon that creates a transcriptional frame shift, changing 38 amino acid codons before introducing a premature stop codon. Both mutations cause congenital deafness and severe balance deficits due to inner ear dysfunction. The stereocilia of cochlear hair cells are disorganized and splayed in mutant mice, with subsequent degeneration of the hair cells and spiral ganglion cells. Harmonin, the protein encoded by Ush1c, has been shown to bind, by means of its PDZ-domains, with the products of other Usher syndrome genes, including Myo7a, Cdh23 and Sans. The complexes formed by these protein interactions are thought to be essential for maintaining the integrity of hair cell stereocilia. The Ush1c mutant mice described here provide a means to directly investigate these interactions in vivo and to evaluate gene structure-function relationships that affect inner ear and eye phenotypes.


Subject(s)
Carrier Proteins/genetics , Deafness/genetics , Mutation , Animals , Base Sequence , Cell Cycle Proteins , Chromosome Mapping , Cochlea/abnormalities , Cochlea/ultrastructure , Cytoskeletal Proteins , Eye/ultrastructure , Genes, Recessive , Genetic Complementation Test , Hair Cells, Auditory, Inner/ultrastructure , Mice , Phenotype , Protein Biosynthesis , Transcription, Genetic
13.
J Bone Miner Res ; 18(9): 1612-21, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12968670

ABSTRACT

UNLABELLED: A missense mutation in the mouse Col2a1 gene has been discovered, resulting in a mouse phenotype with similarities to human spondyloepiphyseal dysplasia (SED) congenita. In addition, SED patients have been identified with a similar molecular mutation in human COL2A1. This mouse model offers a useful tool for molecular and biological studies of bone development and pathology. INTRODUCTION: A new mouse autosomal recessive mutation has been discovered and named spondyloepiphyseal dysplasia congenita (gene symbol sedc). MATERIALS AND METHODS: Homozygous sedc mice can be identified at birth by their small size and shortened trunk. Adults have shortened noses, dysplastic vertebrae, femora, and tibias, plus retinoschisis and hearing loss. The mutation was mapped to Chr15, and Col2a1 was identified as a candidate gene. RESULTS: Sequence analyses revealed that the affected gene is Col2a1, which has a missense mutation at exon 48 causing an amino acid change of arginine to cysteine at position 1417. Two human patients with spondyloepiphyseal dysplasia (SED) congenita have been reported with the same amino acid substitution at position 789 in the human COL2A1 gene. CONCLUSIONS: Thus, sedc/sedc mice provide a valuable model of human SED congenita with molecular and phenotypic homology. Further biochemical analyses, molecular modeling, and cell culture studies using sedc/sedc mice could provide insight into mechanisms of skeletal development dependent on Col2a1 and its role in fibril formation and cartilage template organization.


Subject(s)
Collagen Type II/genetics , Hearing Loss/genetics , Mutation, Missense , Osteochondrodysplasias/genetics , Retinoschisis/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Collagen Type II/physiology , DNA, Complementary/genetics , Disease Models, Animal , Female , Genes, Recessive , Growth Plate/abnormalities , Humans , Male , Mice , Mice, Inbred AKR , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Osteochondrodysplasias/congenital , Phenotype
14.
Retina ; 23(4): 518-22, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12972764

ABSTRACT

PURPOSE: To characterize the phenotype and report a reliable genetic model of retinal angiogenesis and subretinal neovascularization in the mouse. METHODS: The mouse phenotype was characterized using ophthalmoscopy, fundus photography, fluorescein angiography, electroretinography, histology, gene sequencing, and linkage analysis. RESULTS: Scattered pink-gray retinal lesions were found on ophthalmoscopy and were confirmed to be subretinal neovascularization on fluorescein angiography. On histologic examination, outer plexiform retinal neovascularization with growth into the subretinal space was found as early as postnatal Day 15. On genetic analysis, homozygosity of the Vldlr mutation always segregated with the retinal angiogenesis, whereas normal and heterozygous mice had no neovascularization. The histologic studies 15 to 18 days consistently showed new outer plexiform neovascular vessels drawn to the subretinal space by 20 days, and by 30 to 50 days, subretinal hemorrhages and choroidal anastomoses were common. Mice by 8 months had increased vascularity of the iris and ciliary body. CONCLUSIONS: The Vldlr mutation in the mouse provides a good model for retinal angiogenesis and subretinal neovascularization. Finding a strong association between retinal angiogenesis and a very low density lipid receptor mutation is new, and study of lipid receptor physiology may broaden the understanding of retinal angiogenesis.


Subject(s)
Arteriovenous Fistula/genetics , Choroid/blood supply , Disease Models, Animal , Mutation , Receptors, LDL/genetics , Retinal Neovascularization/genetics , Retinal Vessels/abnormalities , Animals , Arteriovenous Fistula/pathology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Electroretinography , Female , Fluorescein Angiography , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Photography , Retinal Neovascularization/pathology
15.
Hum Mol Genet ; 12(17): 2179-89, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12915475

ABSTRACT

Mutations within the CRB1 gene have been shown to cause human retinal diseases including retinitis pigmentosa and Leber congenital amaurosis. We have recently identified a mouse model, retinal degeneration 8 (rd8) with a single base deletion in the Crb1 gene. This mutation is predicted to cause a frame shift and premature stop codon which truncates the transmembrane and cytoplasmic domain of CRB1. Like in Drosophila crumbs (crb) mutants, staining for adherens junction proteins known to localize to the external limiting membrane, the equivalent of the zonula adherens in the mammalian retina, is discontinuous and fragmented. Shortened photoreceptor inner and outer segments are observed as early as 2 weeks after birth, suggesting a developmental defect in these structures rather than a degenerative process. Photoreceptor degeneration is observed only within regions of retinal spotting, which is seen predominantly in the inferior nasal quadrant of the eye, and is caused by retinal folds and pseudorosettes. Photoreceptor dysplasia and degeneration in Crb1 mutants strongly vary with genetic background, suggesting that the variability in phenotypes of human patients that carry mutations in CRB1 may be due to interactions with background modifiers in addition to allelic variations. The Crb1rd8 mouse model will facilitate the analysis of Crb1 function in the neural retina and the identification of interacting factors as candidate retinal disease genes.


Subject(s)
Basement Membrane/metabolism , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/physiology , Photoreceptor Cells, Vertebrate/cytology , Retina/embryology , Alternative Splicing , Animals , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Chromosome Mapping , Frameshift Mutation , Mice , Mice, Inbred C57BL , Morphogenesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Peptide Fragments/immunology , Photoreceptor Cells, Vertebrate/metabolism , Protein Isoforms , Retina/growth & development , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Schizosaccharomyces pombe Proteins/immunology , Schizosaccharomyces pombe Proteins/metabolism
16.
Hum Mol Genet ; 11(16): 1879-86, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12140190

ABSTRACT

The autosomal recessive mouse mutation retinal degeneration 6 (rd6) causes small, white retinal spots and progressive photoreceptor degeneration similar to that observed in human flecked retinal diseases. Using a positional cloning approach, we determined that rd6 mice carry a splice donor mutation in the mouse homolog of the human membrane-type frizzled-related protein (Mfrp) gene that results in the skipping of exon 4. We found that mRNA of Mfrp is predominantly expressed in the eye, and at a lower level in the brain. To determine where in the eye Mfrp is expressed, in situ hybridization was done and showed that Mfrp is expressed specifically in the retinal pigment epithelium (RPE) and ciliary epithelium of the eye. The deduced amino acid sequence of MFRP contains a region with similarities to the cysteine-rich domain (CRD) of frizzled, a gene originally found in Drosophila that controls tissue polarity. The CRD is essential for Wnt binding and signaling. Wnt signaling has been shown to be involved in the control of gene expression, cell adhesion, planar polarity, proliferation and apoptosis. We also observed the localization of Wnt family proteins in the apical membrane of the RPE. Our results provide genetic evidence for an involvement of the Mfrp gene expressed by RPE in the degeneration of photoreceptors.


Subject(s)
Eye Proteins , Membrane Proteins/genetics , Mutation , Retinal Degeneration/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA/genetics , Disease Models, Animal , Gene Expression , Humans , In Situ Hybridization , Mice , Mice, Mutant Strains , Molecular Sequence Data , Sequence Homology, Amino Acid
17.
Behav Brain Res ; 132(2): 145-58, 2002 May 14.
Article in English | MEDLINE | ID: mdl-11997145

ABSTRACT

A new spontaneous mouse mutation named fierce (frc) is deleted for the nuclear receptor Nr2e1 gene (also known as Tlx, mouse homolog of Drosophila tailless). The fierce mutation is genetically and phenotypically similar to Nr2e1 targeted mutations previously studied on segregating genetic backgrounds. However, we have characterized the fierce brain, eye, and behavioural phenotypes on three defined genetic backgrounds (C57BL/6J, 129P3/JEms, and B6129F1). The data revealed many novel and background-dependent phenotypic characteristics. Whereas abnormalities in brain development, hypoplasia of cerebrum and olfactory lobes, were consistent on all three backgrounds, our novel finding of enlarged ventricles in 100% and overt hydrocephalus in up to 30% of fierce mice were unique to the C57BL/6J background. Developmental eye abnormalities were also background-dependent with B6129F1-frc mice having less severe thinning of optic layers and less affected electroretinogram responses. Impaired regression of hyaloid vessels was observed in all backgrounds. Furthermore, retinal vessels were deficient in size and number in 129P3/JEms-frc and B6129F1-frc mice but almost entirely absent in C57BL/6J-frc mice. We present the first standardized behavioural tests conducted on Nr2e1 mutant mice and show that C57BL/6J-frc and B6129F1-frc mice have deficits in sensorimotor assays and are hyperaggressive in both sexes and backgrounds. However, C57BL/6J-frc mice were significantly more aggressive than B6129F1-frc mice. Overall, this extensive characterization of the fierce mutation is essential to its application for the study of behavioural, and brain and eye developmental disorders. In addition, the background-dependent differences revealed will enable the identification of important genetic modifiers.


Subject(s)
Aggression , Eye Abnormalities/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Behavior, Animal/physiology , Blotting, Northern , Blotting, Southern , Brain/growth & development , Brain/pathology , Corticosterone/blood , Electroretinography , Eye Abnormalities/pathology , Female , Gene Deletion , Hearing/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Phenotype , Retina/abnormalities , Retina/pathology , Sexual Behavior, Animal/physiology , Smell/genetics , Testosterone/blood
18.
Hum Mol Genet ; 11(5): 507-13, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11875045

ABSTRACT

Mutations of connexin alpha 8 (GJA8 or Cx50) and connexin alpha 3 (GJA3 or Cx46) in humans have been reported to cause cataracts with semi-dominant inheritance patterns. Targeted null mutations in Gja8 and Gja3 in mice cause cataracts with recessive inheritance. The molecular bases for these differences in inheritance patterns and the mechanism for cataractogenesis in these mutants are poorly understood. We recently mapped an autosomal semi-dominant cataract [lens opacity 10 (Lop10)] mutation to mouse chromosome 3 and identified a missense mutation (G-->C) in the Gja8 gene, which causes glycine at codon 22 to be replaced with arginine (G22R). Moreover, we demonstrated that the alpha 8 G22R isoform is a loss-of-function mutant for alpha 8, as well as a dominant mutation for reducing the phosphorylated forms of alpha 3 connexin in vivo. To test the hypothesis that the alteration of endogenous alpha 3 connexin in Lop10 mice led to a unique lens phenotype, we generated double mutant offspring between Lop10 and the Gja3(tm1) (alpha 3(-/-)) mice. The double homozygous mutant mice (Lop10/Lop10 alpha 3(-/-)) showed relatively normal lens cortical fibers compared to the Lop10 mice. A functional impairment of endogenous alpha 3 connexin is therefore partly responsible for cellular phenotypes in the Lop10 mice. This study has provided some novel molecular insights into mouse and human cataractogenesis caused by alpha 8 and alpha 3 mutations. These mouse models will be useful for investigating the mechanistic relationship between gap junction impairment and cataract formation.


Subject(s)
Cataract/genetics , Connexins/genetics , Eye Proteins/genetics , Point Mutation/genetics , Amino Acid Substitution , Animals , Arginine/metabolism , Base Sequence , Cataract/pathology , Chromosome Mapping , Codon , Crosses, Genetic , Female , Genes, Dominant , Genetic Linkage , Genetic Markers , Genotype , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Lens, Crystalline/ultrastructure , Male , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Microphthalmos/genetics , Microphthalmos/pathology , Pedigree , Phenotype , Sequence Analysis, DNA
19.
Nat Genet ; 30(1): 81-5, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11743578

ABSTRACT

Pigmentary glaucoma is a significant cause of human blindness. Abnormally liberated iris pigment and cell debris enter the ocular drainage structures, leading to increased intraocular pressure (IOP) and glaucoma. DBA/2J (D2) mice develop a form of pigmentary glaucoma involving iris pigment dispersion (IPD) and iris stromal atrophy (ISA). Using high-resolution mapping techniques, sequencing and functional genetic tests, we show that IPD and ISA result from mutations in related genes encoding melanosomal proteins. IPD is caused by a premature stop codon mutation in the Gpnmb (GpnmbR150X) gene, as proved by the occurrence of IPD only in D2 mice that are homozygous with respect to GpnmbR150X; otherwise, similar D2 mice that are not homozygous for GpnmbR150X do not develop IPD. ISA is caused by the recessive Tyrp1b mutant allele and rescued by the transgenic introduction of wildtype Tyrp1. We hypothesize that IPD and ISA alter melanosomes, allowing toxic intermediates of pigment production to leak from melanosomes, causing iris disease and subsequent pigmentary glaucoma. This is supported by the rescue of IPD and ISA in D2 eyes with substantially decreased pigment production. These data indicate that pigment production and mutant melanosomal protein genes may contribute to human pigmentary glaucoma. The fact that hypopigmentation profoundly alleviates the D2 disease indicates that therapeutic strategies designed to decrease pigment production may be beneficial in human pigmentary glaucoma.


Subject(s)
Eye Proteins/genetics , Glaucoma, Open-Angle/genetics , Melanosomes/metabolism , Membrane Glycoproteins/genetics , Oxidoreductases , Pigments, Biological/metabolism , Proteins/genetics , Animals , Atrophy , Chromosome Mapping , Chromosomes, Artificial, Bacterial , Codon, Nonsense , Codon, Terminator , Crosses, Genetic , Epistasis, Genetic , Genetic Predisposition to Disease , Haplotypes , Humans , Iris/chemistry , Iris/pathology , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Recombination, Genetic , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL
...