Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 18: 582-594, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32775493

ABSTRACT

No treatment is available for nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1)-associated retinal degeneration, an inherited disease that leads to severe vision loss early in life. Although the causative gene, NMNAT1, plays an essential role in nuclear nicotinamide adenine dinucleotide (NAD)+ metabolism in tissues throughout the body, NMNAT1-associated disease is isolated to the retina. Since this condition is recessive, supplementing the retina with a normal copy of NMNAT1 should protect vulnerable cells from disease progression. We tested this hypothesis in a mouse model that harbors the p.Val9Met mutation in Nmnat1 and consequently develops a retinal degenerative phenotype that recapitulates key features of the human disease. Gene augmentation therapy, delivered by subretinal injection of adeno-associated virus (AAV) carrying a normal human copy of NMNAT1, rescued retinal structure and function. Due to the early-onset profile of the phenotype, a rapidly activating self-complementary AAV was required to initiate transgene expression during the narrow therapeutic window. These data represent the first proof of concept for a therapy to treat patients with NMNAT1-associated disease.

2.
Adv Exp Med Biol ; 1185: 91-96, 2019.
Article in English | MEDLINE | ID: mdl-31884594

ABSTRACT

Mutations in USH2A gene account for most cases of Usher syndrome type II (USH2), characterized by a combination of congenital hearing loss and progressive vision loss. In particular, approximately 30% of USH2A patients harbor a single base pair deletion, c.2299delG, in exon 13 that creates a frameshift and premature stop codon, leading to a nonfunctional USH2A protein. The USH2A protein, also known as usherin, is an extremely large transmembrane protein (5202 aa), which limits the use of conventional AAV-mediated gene therapy; thus development of alternative approaches is required for the treatment of USH2A patients. As usherin contains multiple repetitive domains, we hypothesize that removal of one or more of those domains encoded by mutant exon(s) in the USH2A gene may reconstitute the reading frame and restore the production of a shortened yet adequately functional protein. In this study, we deleted the exon 12 of mouse Ush2a gene (corresponding to exon 13 of human USH2A) using CRISPR/Cas9-based exon-skipping approach and revealed that a shortened form of Ush2a that lacks exon 12 (Ush2a-∆Ex12) is produced and localized correctly in the cochlea. When the Ush2a-∆Ex12 allele is expressed on an Ush2a null background, the Ush2a-∆Ex12 protein can successfully restore the impaired hair cell structure and the auditory function in the Ush2a-/- mice. These results demonstrate that CRISPR/Cas9-based exon-skipping strategy holds a great therapeutic potential for the treatment of USH2A patients.


Subject(s)
Extracellular Matrix Proteins/genetics , Usher Syndromes/therapy , Animals , CRISPR-Cas Systems , Exons , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Usher Syndromes/genetics
3.
Curr Eye Res ; 41(11): 1498-1506, 2016 11.
Article in English | MEDLINE | ID: mdl-27158920

ABSTRACT

PURPOSE: Anterior ischemic optic neuropathy (AION) is the most common cause of non-glaucomatous optic nerve head (ONH) injury among older adults. AION results from a sudden ischemic insult to the proximal portion of the optic nerve, typically leading to visual impairment. Here, we present an experimental model of photodynamically induced ONH injury that can be used to study neuroprotective modalities. METHODS: Intraperitoneal injection of mesoporphyrin IX was followed by photodynamic treatment of the ONH in one eye of Brown-Norway rats; the fellow eye received the reverse sequence as a sham control. Fluorescein angiography (FA), spectral domain optical coherence tomography (SD-OCT), and visual evoked potential (VEP) recordings were performed at different time points following laser treatment. Immunohistochemistry was used to monitor apoptotic cell death (TUNEL) and macrophage infiltration (CD68). Cytokine levels were evaluated using enzyme-linked immunosorbent assay (ELISA). RESULTS: FA showed early hyperfluorescence and late leakage of the ONH, while SD-OCT revealed optic nerve edema. No leakage or other abnormalities were detected in control eyes. VEPs were significantly reduced in amplitude and showed prolonged responses compared to sham eyes. The number of apoptotic retinal ganglion cells was elevated one day after laser treatment (13.77 ± 4.49, p < 0.01) and peaked on day 7 (57.22 ± 11.34, p < 0.01). ONH macrophage infiltration also peaked on day 7 (101.8 ± 9.8, p < 0.05). ELISAs performed showed upregulation of macrophage chemoattractant protein-1 and macrophage inflammatory protein-2 on days 3 and 1, respectively. CONCLUSIONS: Photodynamic treatment of the ONH after administration of mesoporphyrin IX leads to macroscopic, histologic, and physiologic evidence of ONH injury. Given the long half-life of mesoporphyrin IX and the ease of intraperitoneal injections, this new model of photodynamically induced ONH injury may be a useful tool for studying optic nerve injury and possible neuroprotective treatments.


Subject(s)
Optic Disk/pathology , Optic Neuropathy, Ischemic/pathology , Photochemotherapy/adverse effects , Retinal Ganglion Cells/pathology , Animals , Apoptosis , Disease Models, Animal , Evoked Potentials, Visual , Fluorescein Angiography , Fundus Oculi , Male , Optic Neuropathy, Ischemic/etiology , Optic Neuropathy, Ischemic/physiopathology , Rats , Rats, Inbred BN , Tomography, Optical Coherence/methods
4.
Neuron ; 86(1): 247-63, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25801704

ABSTRACT

Organismal development requires the precise coordination of genetic programs to regulate cell fate and function. MEF2 transcription factors (TFs) play essential roles in this process but how these broadly expressed factors contribute to the generation of specific cell types during development is poorly understood. Here we show that despite being expressed in virtually all mammalian tissues, in the retina MEF2D binds to retina-specific enhancers and controls photoreceptor cell development. MEF2D achieves specificity by cooperating with a retina-specific factor CRX, which recruits MEF2D away from canonical MEF2 binding sites and redirects it to retina-specific enhancers that lack the consensus MEF2-binding sequence. Once bound to retina-specific enhancers, MEF2D and CRX co-activate the expression of photoreceptor-specific genes that are critical for retinal function. These findings demonstrate that broadly expressed TFs acquire specific functions through competitive recruitment to enhancers by tissue-specific TFs and through selective activation of these enhancers to regulate tissue-specific genes.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Photoreceptor Cells/physiology , Retina/cytology , Trans-Activators/metabolism , Adaptation, Ocular/genetics , Age Factors , Animals , Animals, Newborn , Chromatin Immunoprecipitation , Electroretinography , Embryo, Mammalian , Eye Proteins/metabolism , Genome , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Retina/growth & development
5.
Nat Neurosci ; 17(9): 1190-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25086610

ABSTRACT

Age-related decreases in neural function result in part from alterations in synapses. To identify molecular defects that lead to such changes, we focused on the outer retina, in which synapses are markedly altered in old rodents and humans. We found that the serine/threonine kinase LKB1 and one of its substrates, AMPK, regulate this process. In old mice, synaptic remodeling was accompanied by specific decreases in the levels of total LKB1 and active (phosphorylated) AMPK. In the absence of either kinase, young adult mice developed retinal defects similar to those that occurred in old wild-type animals. LKB1 and AMPK function in rod photoreceptors where their loss leads to aberrant axonal retraction, the extension of postsynaptic dendrites and the formation of ectopic synapses. Conversely, increasing AMPK activity genetically or pharmacologically attenuates and may reverse age-related synaptic alterations. Together, these results identify molecular determinants of age-related synaptic remodeling and suggest strategies for attenuating these changes.


Subject(s)
AMP-Activated Protein Kinases/genetics , Aging/physiology , Protein Serine-Threonine Kinases/genetics , Rod Cell Outer Segment/pathology , Rod Cell Outer Segment/physiology , AMP-Activated Protein Kinases/metabolism , Aging/pathology , Amacrine Cells/pathology , Amacrine Cells/physiology , Animals , Electroretinography , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/physiology , Protein Serine-Threonine Kinases/metabolism , Retinal Bipolar Cells/pathology , Retinal Bipolar Cells/physiology , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Substrate Specificity , Synapses/pathology , Synapses/physiology
6.
Invest Ophthalmol Vis Sci ; 55(2): 856-64, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24425853

ABSTRACT

PURPOSE: Exfoliation syndrome (ES) is commonly associated with glaucoma, premature cataracts, and other ocular and systemic pathologies. LOXL1 gene variants are significantly associated with ES; however, the role of the protein in ES development remains unclear. The purpose of this study was to characterize the ocular phenotype in Loxl1(-/-) (null) mice. METHODS: Loxl1 null mice and strain-matched controls (C57BL) were evaluated by clinical and histologic analyses. RESULTS: Anterior segment histology showed a pronounced vesiculation of the anterior lens in the null mice. The lesions were subcapsular and in direct apposition with the posterior iris surface. Fluorescein angiography showed increased diffusion of fluorescein into the anterior chamber of the null mice compared with age-matched controls (P = 0.003, two-tailed, unequal variance t-test), suggesting compromise of the blood-aqueous barrier. Intraocular pressure measurements were within the normal range (16.5 ± 2.0 mm Hg) in null mice up to 1 year of age. Immunohistochemistry showed decreased elastin in the iris and ciliary body in the null mouse compared with controls. CONCLUSIONS: Elimination of LOXL1 in mice impairs the blood-aqueous humor barrier in the ocular anterior segment and causes lens abnormalities consistent with cataract formation, but does not result in deposition of macromolecular material or glaucoma. These results show that mice lacking LOXL1 have some ES features but that complete disease manifestation requires other factors that could be genetic and/or environmental.


Subject(s)
Amino Acid Oxidoreductases/genetics , Blood-Aqueous Barrier/pathology , Cataract/pathology , Exfoliation Syndrome/pathology , Gene Expression Regulation, Enzymologic/physiology , Lens, Crystalline/ultrastructure , Animals , Anterior Chamber/metabolism , Blood-Aqueous Barrier/enzymology , Cataract/enzymology , Ciliary Body/metabolism , Elastin/metabolism , Exfoliation Syndrome/enzymology , Fluorescein/metabolism , Fluorescein Angiography , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes/metabolism , Immunoblotting , Intraocular Pressure , Iris/metabolism , Lens, Crystalline/enzymology , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Phenotype , Polymerase Chain Reaction
7.
Am J Pathol ; 180(5): 1917-27, 2012 May.
Article in English | MEDLINE | ID: mdl-22429967

ABSTRACT

The increasing popularity of the Cre/loxP recombination system has led to the generation of numerous transgenic mouse lines in which Cre recombinase is expressed under the control of organ- or cell-specific promoters. Alterations in retinal pigment epithelium (RPE), a multifunctional cell monolayer that separates the retinal photoreceptors from the choroid, are prevalent in the pathogenesis of a number of ocular disorders, including age-related macular degeneration. To date, six transgenic mouse lines have been developed that target Cre to the RPE under the control of various gene promoters. However, multiple lines of evidence indicate that high levels of Cre expression can be toxic to mammalian cells. In this study, we report that in the Trp1-Cre mouse, a commonly used transgenic Cre strain for RPE gene function studies, Cre recombinase expression alone leads to RPE dysfunction and concomitant disorganization of RPE layer morphology, large areas of RPE atrophy, retinal photoreceptor dysfunction, and microglial cell activation in the affected areas. The phenotype described herein is similar to previously published reports of conditional gene knockouts that used the Trp1-Cre mouse, suggesting that Cre toxicity alone could account for some of the reported phenotypes and highlighting the importance of the inclusion of Cre-expressing mice as controls in conditional gene targeting studies.


Subject(s)
Integrases/physiology , Retinal Pigment Epithelium/enzymology , Animals , Atrophy/enzymology , Atrophy/pathology , Disease Models, Animal , Electroretinography/methods , Gene Expression Regulation , Integrases/genetics , Integrases/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Transgenic , Microglia/pathology , Microglia/physiology , Microscopy, Electron , Oxidoreductases/genetics , Oxidoreductases/physiology , Phenotype , Photoreceptor Cells, Vertebrate/physiology , Recombinant Fusion Proteins/genetics , Retinal Dystrophies/enzymology , Retinal Dystrophies/pathology , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/physiopathology , Retinal Pigment Epithelium/ultrastructure
8.
PLoS Genet ; 6(5): e1000955, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20502675

ABSTRACT

Mutations in whirlin cause either Usher syndrome type II (USH2), a deafness-blindness disorder, or nonsyndromic deafness. The molecular basis for the variable disease expression is unknown. We show here that only the whirlin long isoform, distinct from a short isoform by virtue of having two N-terminal PDZ domains, is expressed in the retina. Both long and short isoforms are expressed in the inner ear. The N-terminal PDZ domains of the long whirlin isoform mediates the formation of a multi-protein complex that includes usherin and VLGR1, both of which are also implicated in USH2. We localized this USH2 protein complex to the periciliary membrane complex (PMC) in mouse photoreceptors that appears analogous to the frog periciliary ridge complex. The latter is proposed to play a role in photoreceptor protein trafficking through the connecting cilium. Mice carrying a targeted disruption near the N-terminus of whirlin manifest retinal and inner ear defects, reproducing the clinical features of human USH2 disease. This is in contrast to mice with mutations affecting the C-terminal portion of whirlin in which the phenotype is restricted to the inner ear. In mice lacking any one of the USH2 proteins, the normal localization of all USH2 proteins is disrupted, and there is evidence of protein destabilization. Taken together, our findings provide new insights into the pathogenic mechanism of Usher syndrome. First, the three USH2 proteins exist as an obligatory functional complex in vivo, and loss of one USH2 protein is functionally close to loss of all three. Second, defects in the three USH2 proteins share a common pathogenic process, i.e., disruption of the PMC. Third, whirlin mutations that ablate the N-terminal PDZ domains lead to Usher syndrome, but non-syndromic hearing loss will result if they are spared.


Subject(s)
Extracellular Matrix Proteins/physiology , Hearing Loss/genetics , Membrane Proteins/physiology , Protein Isoforms/physiology , Vision Disorders/genetics , Animals , Extracellular Matrix Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Protein Isoforms/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
Hum Gene Ther ; 21(8): 993-1004, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20384479

ABSTRACT

RPGR-interacting protein-1 (RPGRIP1) is localized in the photoreceptor-connecting cilium, where it anchors the RPGR (retinitis pigmentosa GTPase regulator) protein, and its function is essential for photoreceptor maintenance. Genetic defect in RPGRIP1 is a known cause of Leber congenital amaurosis (LCA), a severe, early-onset form of retinal degeneration. We evaluated the efficacy of replacement gene therapy in a murine model of LCA carrying a targeted disruption of RPGRIP1. The replacement construct, packaged in an adeno-associated virus serotype 8 (AAV8) vector, used a rhodopsin kinase gene promoter to drive RPGRIP1 expression. Both promoter and transgene were of human origin. After subretinal delivery of the replacement gene in the mutant mice, human RPGRIP1 was expressed specifically in photoreceptors, localized correctly in the connecting cilia, and restored the normal localization of RPGR. Electroretinogram and histological examinations showed better preservation of rod and cone photoreceptor function and improved photoreceptor survival in the treated eyes. This study demonstrates the efficacy of human gene replacement therapy and validates a gene therapy design for future clinical trials in patients afflicted with this condition. Our results also have therapeutic implications for other forms of retinal degenerations attributable to a ciliary defect.


Subject(s)
Genetic Therapy , Leber Congenital Amaurosis/therapy , Photoreceptor Connecting Cilium/metabolism , Proteins/genetics , Animals , Cytoskeletal Proteins , Disease Models, Animal , Electroretinography , G-Protein-Coupled Receptor Kinase 1/metabolism , Genetic Vectors , Humans , Leber Congenital Amaurosis/genetics , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Proteins/therapeutic use , Retinal Degeneration/genetics , Retinal Degeneration/therapy
10.
PLoS One ; 4(12): e8438, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20041177

ABSTRACT

BACKGROUND: In one group of gene mutations that cause photoreceptor degeneration in human patients, guanylyl cyclase is overactive in the dark. The ensuing excess opening of cGMP-gated cation channels causes intracellular calcium to rise to toxic levels. The Y99C mutation in guanylate cyclase-activating protein 1 (GCAP1) has been shown to act this way. We determined whether prolonged light exposure, which lowers cGMP levels through activation of phototransduction, might protect photoreceptors in a line of transgenic mice carrying the GCAP1-Y99C. METHODOLOGY/PRINCIPAL FINDINGS: We reared cohorts of GCAP1-Y99C transgenic mice under standard cyclic, constant dark and constant light conditions. Mouse eyes were analyzed by histology and by immunofluorescence for GFAP upregulation, a non-specific marker for photoreceptor degeneration. Full-field electroretinograms (ERGs) were recorded to assess retinal function. Consistent with our hypothesis, constant darkness accelerated disease, while continuous lighting arrested photoreceptor degeneration. CONCLUSIONS/SIGNIFICANCE: In contrast to most forms of retinal degeneration, which are exacerbated by increased exposure to ambient light, a subset with mutations that cause overly active guanylyl cyclase and high intracellular calcium benefitted from prolonged light exposure. These findings may have therapeutic implications for patients with these types of genetic defects.


Subject(s)
Calcium/metabolism , Darkness , Photoreceptor Cells, Vertebrate/pathology , Animals , Disease Models, Animal , Guanylate Cyclase-Activating Proteins/metabolism , Mice , Mice, Mutant Strains , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/radiation effects , Retina/metabolism , Retina/physiopathology , Retina/radiation effects
11.
Hum Mol Genet ; 18(12): 2099-114, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19299492

ABSTRACT

Defects in the photoreceptor-specific gene encoding aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) are clinically heterogeneous and present as Leber Congenital Amaurosis, the severest form of early-onset retinal dystrophy and milder forms of retinal dystrophies such as juvenile retinitis pigmentosa and dominant cone-rod dystrophy. [Perrault, I., Rozet, J.M., Gerber, S., Ghazi, I., Leowski, C., Ducroq, D., Souied, E., Dufier, J.L., Munnich, A. and Kaplan, J. (1999) Leber congenital amaurosis. Mol. Genet. Metab., 68, 200-208.] Although not yet fully elucidated, AIPL1 is likely to function as a specialized chaperone for rod phosphodiesterase (PDE). We evaluate whether AAV-mediated gene replacement therapy is able to improve photoreceptor function and survival in retinal degeneration associated with AIPL1 defects. We used two mouse models of AIPL1 deficiency simulating three different rates of photoreceptor degeneration. The Aipl1 hypomorphic (h/h) mouse has reduced Aipl1 levels and a relatively slow degeneration. Under light acceleration, the rate of degeneration in the Aipl1 h/h mouse is increased by 2-3-fold. The Aipl1-/- mouse has no functional Aipl1 and has a very rapid retinal degeneration. To treat the different rates of degeneration, two pseudotypes of recombinant adeno-associated virus (AAV) exhibiting different transduction kinetics are used for gene transfer. We demonstrate restoration of cellular function and preservation of photoreceptor cells and retinal function in Aipl1 h/h mice following gene replacement therapy using an AAV2/2 vector and in the light accelerated Aipl1 h/h model and Aipl1-/- mice using an AAV2/8 vector. We have thus established the potential of gene replacement therapy in varying rates of degeneration that reflect the clinical spectrum of disease. This is the first gene replacement study to report long-term rescue of a photoreceptor-specific defect and to demonstrate effective rescue of a rapid photoreceptor degeneration.


Subject(s)
Carrier Proteins/genetics , Genetic Therapy , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/genetics , Humans , Mice , Mice, Transgenic , Optic Atrophy, Hereditary, Leber/physiopathology , Photoreceptor Cells, Vertebrate/metabolism , Retinitis Pigmentosa/physiopathology
12.
Cell ; 130(4): 730-741, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17719549

ABSTRACT

Circadian clocks are widely distributed in mammalian tissues, but little is known about the physiological functions of clocks outside the suprachiasmatic nucleus of the brain. The retina has an intrinsic circadian clock, but its importance for vision is unknown. Here we show that mice lacking Bmal1, a gene required for clock function, had abnormal retinal transcriptional responses to light and defective inner retinal electrical responses to light, but normal photoreceptor responses to light and retinas that appeared structurally normal by light and electron microscopy. We generated mice with a retina-specific genetic deletion of Bmal1, and they had defects of retinal visual physiology essentially identical to those of mice lacking Bmal1 in all tissues and lacked a circadian rhythm of inner retinal electrical responses to light. Our findings indicate that the intrinsic circadian clock of the retina regulates retinal visual processing in vivo.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Retina/physiology , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Electroretinography , Gene Deletion , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Retina/cytology , Retina/ultrastructure
13.
Invest Ophthalmol Vis Sci ; 48(9): 3954-61, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17724172

ABSTRACT

PURPOSE: Gene therapy for retinal degeneration requires well-defined promoters that drive expression in rod and cone photoreceptors. This study was undertaken to develop short, active derivatives of the human rhodopsin kinase (RK) gene promoter for targeting transgene expression in rods and cones. RK, also known as G protein-coupled receptor kinase 1 (GRK1), is a component of the light adaptation pathway expressed in rods and cones. METHODS: Human RK (hRK) promoter and its concatemers or derivatives extending into the conserved 5' untranslated region (5'-UTR) were assayed for promoter activity in WERI retinoblastoma or Crx/Sp1-supplemented HEK-293 cells. The derivative displaying the highest activity was linked to a GFP reporter and packaged in a pseudotyped adenoassociated viral vector (AAV2/5). The AAV vector was tested in vivo by subretinal injections in wild-type mice, in the all-cone Nrl(-/-) mice, and in the cone-rich diurnal Nile grass rat (Arvicanthis niloticus). Control eyes received a similar AAV2/5 vector carrying a mouse rod opsin (mOps) promoter-controlled GFP reporter. RESULTS: The hRK promoter with the full 5' untranslated sequence (-112 to +180) was the most active in cell culture. Delivered by the AAV2/5 vector, RK promoter drove GFP expression specifically in photoreceptors. In rods, hRK promoter-mediated expression was as efficient as, but appeared more uniform than, mOps promoter-mediated expression. In cones, the hRK promoter drove expression, whereas the mOps promoter did not. CONCLUSIONS: The hRK promoter is active and specific for rod and cone photoreceptors. Because of its small size and proven activity in cones, it is a promoter of choice for somatic gene transfer and gene therapy targeting rods and cones.


Subject(s)
Dependovirus/genetics , G-Protein-Coupled Receptor Kinase 1/genetics , Gene Expression , Green Fluorescent Proteins/genetics , Photoreceptor Cells, Vertebrate/metabolism , Promoter Regions, Genetic/genetics , Animals , Gene Targeting , Genetic Vectors , Kidney/embryology , Luminescent Agents , Mice , Plasmids , Rats , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Rod Opsins/genetics , Transfection , Transgenes , Tumor Cells, Cultured
14.
J Neurochem ; 101(5): 1294-302, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17442056

ABSTRACT

Retinal abnormalities are well documented in patients with ganglioside storage diseases. The total content and distribution of retinal glycosphingolipids was studied for the first time in control mice and in Sandhoff disease (SD) and GM1 gangliosidosis mice. Light and electron microscopy of the SD and the GM1 retinas revealed storage in ganglion cells. Similar to previous findings in rat retina, GD3 was the major ganglioside in mouse retina, while GM2 and GM1 were minor species. Total ganglioside content was 44% and 40% higher in the SD and the GM1 retinas, respectively, than in the control retinas. Furthermore, GM2 and GM1 content were 11-fold and 51-fold higher in the SD and the GM1 retinas than in the control retinas, respectively. High concentrations of asialo-GM2 and asialo-GM1 were found in the SD and the GM1 retinas, respectively, but were undetectable in the control retinas. The GSL abnormalities in the SD and the GM1 retinas reflect significant reductions in beta-hexosaminidase and beta-galactosidase enzyme activities, respectively. Although electroretinograms appeared normal in the SD and the GM1 mice, visual evoked potentials were subnormal in both mutants, indicating visual impairments. Our findings present a model system for assessing retinal pathobiology and therapies for the gangliosidoses.


Subject(s)
Gangliosidosis, GM1/genetics , Retina/metabolism , Retina/pathology , Sandhoff Disease/pathology , Sandhoff Disease/physiopathology , Age Factors , Animals , Chromatography, High Pressure Liquid/methods , Disease Models, Animal , Electroretinography , Evoked Potentials, Visual/genetics , Female , Gangliosidoses/metabolism , Gene Expression Regulation, Enzymologic/genetics , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Retina/physiopathology , Retina/ultrastructure , Sandhoff Disease/enzymology , Sandhoff Disease/genetics , beta-Galactosidase/genetics , beta-N-Acetylhexosaminidases/deficiency , beta-N-Acetylhexosaminidases/metabolism
15.
Hum Mol Genet ; 16(9): 1017-29, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17341488

ABSTRACT

Membrane palmitoylated protein 4 (Mpp4) is a member of the membrane-associated guanylate kinase family. We show that Mpp4 localizes specifically to the plasma membrane of photoreceptor synaptic terminals. Plasma membrane Ca(2+) ATPases (PMCAs), the Ca(2+) extrusion pumps, interact with an Mpp4-dependent presynaptic membrane protein complex that includes Veli3 and PSD95. In mice lacking Mpp4, PMCAs were lost from rod photoreceptor presynaptic membranes. Synaptic ribbons were enlarged, a phenomenon known to correlate with higher Ca(2+). SERCA2 (sarcoplasmic-endoplasmic reticulum Ca(2+) ATPase, type 2), which pumps cytosolic Ca(2+) into intracellular Ca(2+) stores and localizes next to the ribbons, was increased. The distribution of IP(3)RII (InsP(3) receptor, type 2), which releases Ca(2+) from the stores, was shifted away from the synaptic terminals. Synaptic transmission to second-order neurons was maintained but was reduced in amplitude. These data suggest that loss of Mpp4 disrupts a Ca(2+) extrusion mechanism at the presynaptic membranes, with ensuing adaptive responses by the photoreceptor to restore Ca(2+) homeostasis. We propose that Mpp4 organizes a presynaptic protein complex that includes PMCAs and has a role in modulating Ca(2+) homeostasis and synaptic transmission in rod photoreceptors.


Subject(s)
Calcium/metabolism , Membrane Proteins/physiology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Presynaptic Terminals/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Disks Large Homolog 4 Protein , Fluorescent Antibody Technique , Gene Deletion , Guanylate Kinases , Homeostasis , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Models, Biological , Multiprotein Complexes/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Presynaptic Terminals/ultrastructure , Protein Binding , Retinal Rod Photoreceptor Cells/ultrastructure , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Two-Hybrid System Techniques
16.
Proc Natl Acad Sci U S A ; 104(11): 4413-8, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360538

ABSTRACT

Usher syndrome type IIA (USH2A), characterized by progressive photoreceptor degeneration and congenital moderate hearing loss, is the most common subtype of Usher syndrome. In this article, we show that the USH2A protein, also known as usherin, is an exceptionally large ( approximately 600-kDa) matrix protein expressed specifically in retinal photoreceptors and developing cochlear hair cells. In mammalian photoreceptors, usherin is localized to a spatially restricted membrane microdomain at the apical inner segment recess that wraps around the connecting cilia, corresponding to the periciliary ridge complex described for amphibian photoreceptors. In sensory hair cells of the cochlea, it is associated transiently with the hair bundles during postnatal development. Targeted disruption of the Ush2a gene in mice leads to progressive photoreceptor degeneration and a moderate but nonprogressive hearing impairment, mimicking the visual and hearing deficits in USH2A patients. These data suggest that usherin is required for the long-term maintenance of retinal photoreceptors and for the development of cochlear hair cells. We propose a model in which usherin in photoreceptors is tethered via its C terminus to the plasma membrane and its large extracellular domain projecting into the periciliary matrix, where they may interact with the connecting cilium to fulfill important structural or signaling roles.


Subject(s)
Cochlea/growth & development , Extracellular Matrix Proteins/physiology , Gene Expression Regulation, Developmental , Hair Cells, Auditory/growth & development , Retina/growth & development , Retinitis Pigmentosa/pathology , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Molecular Sequence Data , Retinitis Pigmentosa/metabolism
17.
Am J Ophthalmol ; 142(3): 515-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16935610

ABSTRACT

PURPOSE: We investigated whether opsin mislocalization occurs in photoreceptors in a female carrier of X-linked retinitis pigmentosa with a Gly436Asp mutation in the retinitis pigmentosa GTPase regulator gene (RPGR). DESIGN: Histologic findings in autopsy eyes from a carrier were compared with those from a normal female. METHODS: Frozen retinal sections from the periphery of one eye of the carrier and the normal were stained with antibodies against either human red or green opsins, blue cone opsin, or rhodopsin and labeled with fluorochrome conjugated secondary antibodies. Cell nuclei were counterstained with Hoechst dye. Fellow eyes were evaluated with light microscopy. RESULTS: Fluorescent labeling showed mislocalized cone and rod opsins in photoreceptor cells only in the carrier. The carrier also showed some loss of photoreceptor nuclei. CONCLUSIONS: A defect in trafficking of opsins to outer segments exists in a carrier with the RPGR Gly436Asp mutation.


Subject(s)
Eye Proteins/genetics , Genetic Diseases, X-Linked/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Point Mutation , Retinitis Pigmentosa/metabolism , Rod Opsins/metabolism , Aged , Female , Fluorescent Antibody Technique, Indirect , Genetic Diseases, X-Linked/genetics , Guanine Nucleotide Exchange Factors/genetics , Heterozygote , Humans , Retinitis Pigmentosa/genetics
18.
Invest Ophthalmol Vis Sci ; 47(5): 2185-94, 2006 May.
Article in English | MEDLINE | ID: mdl-16639031

ABSTRACT

PURPOSE: To investigate the impact of aryl hydrocarbon receptor-interacting protein-like (AIPL)-1 on photoreception in rods. METHODS: Photoresponses of mouse rods expressing lowered amounts of AIPL1 were studied by single-cell and electroretinogram (ERG) recordings. Phototransduction protein levels and enzymatic activities were determined in biochemical assays. Ca2+ dynamics were probed with a fluorescent dye. Comparisons were made to rods expressing mutant Y99C guanylate cyclase activating protein (GCAP)-1, to understand which effects arose from elevated dark levels of cGMP and Ca2+. RESULTS: Except for PDE, transduction protein levels were normal in low-AIPL1 retinas, as were guanylate cyclase (GC), rhodopsin kinase (RK), and normalized phosphodiesterase (PDE) activities. Y99C and low-AIPL1 rods were more sensitive to flashes than normal, but flash responses of low-AIPL1 rods showed an abnormal delay, reduced rate of increase, and longer recovery not present in Y99C rod responses. In addition, low-AIPL1 rods but not Y99C rods failed to reach the normal light-induced minimum in Ca2+ concentration. CONCLUSIONS: Reduced AIPL1 delayed the photoresponse, decreased its amplification constant, slowed a rate-limiting step in its recovery, and limited the light-induced decrease in Ca2+. Not all changes were attributable to decreased PDE or to elevated cGMP and Ca2+ in darkness. Therefore, AIPL1 directly or indirectly affects more than one component of phototransduction.


Subject(s)
Carrier Proteins/physiology , Retinal Rod Photoreceptor Cells/physiology , Vision, Ocular/physiology , Adaptor Proteins, Signal Transducing , Animals , Calcium/metabolism , Chromatography, Gas , Cyclic GMP/metabolism , Electroretinography , Fatty Acids/metabolism , G-Protein-Coupled Receptor Kinase 1/metabolism , Guanylate Cyclase/metabolism , Guanylate Cyclase-Activating Proteins/metabolism , Mice , Mice, Transgenic , Phosphoric Diester Hydrolases/metabolism , Photic Stimulation , Retinal Rod Photoreceptor Cells/radiation effects , Transducin/metabolism
19.
Cancer Cell ; 9(1): 13-22, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16413468

ABSTRACT

Cyclin D1 is a multifunctional protein that activates CDK4 and CDK6, titrates Cip/Kip CDK inhibitors to increase CDK2 activity, and modulates the function of certain transcription factors. To specifically test the importance of cyclin D1-associated kinase activity, we generated "knockin" mice expressing mutant cyclin D1 deficient in activating CDK4/6. The development of several cyclin D1-dependent compartments, including mammary glands, proceeds relatively normally in these animals, demonstrating that cyclin D1-associated kinase activity is largely dispensable for development of these tissues. Strikingly, knockin mice were resistant to breast cancers initiated by ErbB-2. These results demonstrate a differential requirement for cyclin D1-CDK4/6 kinase activity in development versus tumorigenesis and strongly support cyclin D1-dependent kinase activity as a specific therapeutic target in breast cancer.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/metabolism , Animals , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Enzyme Activation , Female , Genes, erbB-2 , Mammary Glands, Animal/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Mutant Strains , Mutation , Protein Binding , Retina/enzymology , Retina/growth & development
20.
Am J Pathol ; 168(2): 519-28, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16436666

ABSTRACT

Pelvic floor disorders, a group of conditions affecting adult women, include pelvic organ prolapse and urinary incontinence. Vaginal childbirth and aging are risk factors, and weakening of the pelvic support structures is a major aspect of the pathology. However, the underlying molecular mechanism remains unknown. Female reproductive organs are rich in elastic fibers that turn over slowly in most adult tissues but undergo massive remodeling in the reproductive organs through pregnancy and birth. Here we show that a failure to maintain elastic fiber homeostasis in mice causes pelvic floor disorders. Lysyl oxidase-like-1 (LOXL1), a protein essential for the postnatal deposition of elastic fibers, was highly expressed and regulated in the reproductive tract of the mouse, and its expression was diminished during aging. LOXL1 deficiency caused an inability of reproductive tissues to replenish elastic fibers after parturition, leading to pelvic organ prolapse, weakening of the vaginal wall, paraurethral pathology, and lower urinary tract dysfunction. These data demonstrate the importance of elastic fibers for maintaining structural and functional integrity of the female pelvic floor. Our findings raise the possibility that a failure of elastic fiber homeostasis, either due to genetic predisposition or advancing age, could underlie the etiology of pelvic floor dysfunction in women.


Subject(s)
Amino Acid Oxidoreductases/physiology , Elastic Tissue/metabolism , Homeostasis/physiology , Pelvic Floor/physiopathology , Urinary Incontinence, Stress/etiology , Uterine Prolapse/etiology , Amino Acid Oxidoreductases/genetics , Animals , Female , Mice , Mice, Knockout , Urinary Incontinence, Stress/physiopathology , Uterine Prolapse/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...