Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Invest Ophthalmol Vis Sci ; 60(5): 1442-1453, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30947334

ABSTRACT

Purpose: The purpose of this study was to test the extent of light damage in different models of night blindness and apply these paradigms in testing the therapeutic efficacy of combination therapy by drugs acting on the Gi, Gs, and Gq protein-coupled receptors. Methods: Acute bright light exposure was used to test susceptibility to light damage in mice lacking the following crucial phototransduction proteins: rod transducin (GNAT1), cone transducin (GNAT2), visual arrestin 1 (ARR1), and rhodopsin kinase 1 (GRK1). Mice were intraperitoneally injected with either vehicle or drug combination consisting of metoprolol (ß1-receptor antagonist), bromocriptine (dopamine family-2 receptor agonist) and tamsulosin (α1-receptor antagonist) before bright light exposure. Light damage was primarily assessed with optical coherence tomography and inspection of cone population in retinal whole mounts. Retinal inflammation was assessed in a subset of experiments using autofluorescence imaging by scanning laser ophthalmoscopy and by postmortem inspection of microglia and astrocyte activity. Results: The Gnat1-/- mice showed slightly increased susceptibility to rod light damage, whereas the Gnat2-/- mice were very resistant. The Arr1-/- and Grk1-/- mice were sensitive for both rod and cone light damage and showed robust retinal inflammation 7 days after bright light exposure. Pretreatment with metoprolol + bromocriptine + tamsulosin rescued the retina in all genetic backgrounds, starting at doses of 0.2 mg/kg metoprolol, 0.02 mg/kg bromocriptine, and 0.01 mg/kg tamsulosin in the Gnat1-/- mice. The therapeutic drug doses increased in parallel with light-damage severity. Conclusions: Our results suggest that congenital stationary night blindness and Oguchi disease patients can be at an elevated risk of the toxic effects of bright light. Furthermore, systems pharmacology drug regimens that stimulate Gi signaling and attenuate Gs and Gq signaling present a promising disease-modifying therapy for photoreceptor degenerative diseases.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/therapeutic use , Adrenergic beta-1 Receptor Antagonists/therapeutic use , Bromocriptine/pharmacology , Light/adverse effects , Metoprolol/pharmacology , Night Blindness/drug therapy , Tamsulosin/pharmacology , Animals , Arrestins/deficiency , Disease Models, Animal , G-Protein-Coupled Receptor Kinase 1/deficiency , Mice , Transducin/deficiency , United States , United States Food and Drug Administration
2.
Mol Vis ; 21: 1281-94, 2015.
Article in English | MEDLINE | ID: mdl-26664249

ABSTRACT

PURPOSE: Well-established laboratory mouse lines are important in creating genetically engineered knockout mouse models; however, these routinely used inbred strains are prone to spontaneous and deleterious mutations. One of these strains, the commonly used C57BL/6N (B6N), was discovered to carry a point mutation in the Crumbs homolog 1 (Crb1(rd8) ) gene, which codes for a developmental protein involved in tight junction formation at the outer limiting membrane (OLM). This mutation disrupts photoreceptor polarity and leads to retinal degeneration. It was hypothesized that the G-protein receptor kinase 1 knockouts (Grk1(-/-) ), which were based on the B6N strain, would exhibit abnormal morphological phenotypes in their offspring not related to GRK1's major phosphorylation function. The hypothesis was tested by examining Grk1(-/-) with or without the Crb1(rd8) mutation. METHODS: The mice strains tested were C57BL/6J (B6J), B6N, and Grk1(-/-) on either a B6J (Grk1(-/-) (;B6J)) or B6N background (Grk1(-/-) (;B6N)) and were verified with PCR genotype analysis for Grk1(-/-) and Crb (rd8) . The mice were bred and raised in complete darkness until 1 or 3 months of age and then exposed to 1,000 lux light for 24 h, followed by processing for immunohistochemistry (IHC) analysis on the retinal structure to investigate the morphological effects of light exposure. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) was performed to detect photoreceptor apoptosis. RESULTS: The microanatomy of the retinal sections revealed disorganization of the outer nuclear layer (ONL) in the B6N and Grk1(-/-) (;B6N) mice and a significant decrease in the thickness of the ONL in the 3-month-old Grk1(-/-) (;B6N) mice. The adherens-junction-associated protein, Zona occludens-1 (ZO-1), formed a continuous line at the OLM in the 1- and 3-month-old control B6J and Grk1(-/-) (;B6J) mice. In contrast, the B6N and Grk1(-/-) (;B6N) retinas showed discontinuous and fragmented staining for ZO-1 at the OLM at both ages. After the mice were exposed to light, TUNEL analysis showed a significant increase in photoreceptor cell death in the Grk1(-/-) (;B6J) and Grk1(-/-) (;B6N) retinas versus either the B6J or B6N retinas at 1 and 3 months of age and a small significant difference between the Grk1(-/-) (;B6J) and Grk1(-/-) (;B6N) retinas at 1 month. In addition, glial fibrillary acidic protein (GFAP) expression was enhanced in the Grk1(-/-) (;B6J) and Grk1(-/-) (;B6N) retinas at 1 and 3 months. Occasional sprouting processes of rod bipolar cells were detected in the B6N and Grk1(-/-) (;B6N) retinas, but sprouting was not detected in the B6J or Grk1(-/-) (;B6J) retinas at either age. CONCLUSIONS: The B6N strain background exhibited abnormal phenotypes in the Grk1(-/-) (;B6N) retina. This study demonstrates that the B6N background can influence the phenotype of a genetic mouse knockout and introduces potential visual functional consequences of the Crb1 mutation.


Subject(s)
G-Protein-Coupled Receptor Kinase 1/genetics , Nerve Tissue Proteins/genetics , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/genetics , Age Factors , Animals , Apoptosis , Darkness , Female , G-Protein-Coupled Receptor Kinase 1/deficiency , Gene Expression Regulation , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nerve Tissue Proteins/metabolism , Photic Stimulation , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Signal Transduction , Species Specificity , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
3.
J Neurosci ; 30(20): 6815-24, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20484624

ABSTRACT

The remarkable ability of our vision to function under ever-changing conditions of ambient illumination is mediated by multiple molecular mechanisms regulating the light sensitivity of rods and cones. One such mechanism involves massive translocation of signaling proteins, including the G-protein transducin, into and out of the light-sensitive photoreceptor outer segment compartment. Transducin translocation extends the operating range of rods, but in cones transducin never translocates, which is puzzling because cones typically function in much brighter light than rods. Using genetically manipulated mice in which the rates of transducin activation and inactivation were altered, we demonstrate that, like in rods, transducin translocation in cones can be triggered when transducin activation exceeds a critical level, essentially saturating the photoresponse. However, this level is never achieved in wild-type cones: their superior ability to tightly control the rates of transducin activation and inactivation, responsible for avoiding saturation by light, also accounts for the prevention of transducin translocation at any light intensity.


Subject(s)
Light Signal Transduction/physiology , Retinal Cone Photoreceptor Cells/metabolism , Transducin/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Aging/genetics , Aging/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Electroretinography/methods , Eye Proteins , G-Protein-Coupled Receptor Kinase 1/deficiency , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Gene Expression Regulation/genetics , Light , Light Signal Transduction/genetics , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Protein Transport/genetics , Protein Transport/physiology , RGS Proteins/deficiency , Retinal Rod Photoreceptor Cells/metabolism
4.
J Neurosci ; 30(7): 2496-503, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20164334

ABSTRACT

Rpe65(-/-) mice are unable to produce 11-cis-retinal, the chromophore of visual pigments. Consequently, the pigment is present as the apoprotein opsin with a minute level of pigment containing 9-cis-retinal as chromophore. Notably, a 10-20% fraction of this opsin is mono-phosphorylated independently of light conditions. To determine the role of rhodopsin kinase (GRK1) in phosphorylating this opsin and to test whether eliminating this phosphorylation would accelerate photoreceptor degeneration, we generated the Rpe65(-/-)Grk1(-/-) mouse. The retinae of Rpe65(-/-)Grk1(-/-) mice had negligible opsin phosphorylation, extensive degeneration with decreased opsin levels, and diminished light-evoked rod responses relative to Rpe65(-/-) mice. These data show that opsin phosphorylation in the Rpe65(-/-) mouse is due to the action of GRK1 and is neuroprotective. However, despite the higher activity of unphosphorylated opsin, the severe loss of opsin in the rapidly degenerating Rpe65(-/-)Grk1(-/-) mice resulted in lower overall opsin activity and in higher rod sensitivity compared with Rpe65(-/-) mice. In Rpe65(-/-)Grk1(-/-)Gnat1(-/-) mice where transduction activation was blocked, degeneration was only partially prevented. Therefore, increased opsin activity in the absence of phosphorylation was not the only mechanism for the accelerated retinal degeneration. Finally, the deletion of GRK1 triggered retinal degeneration in Grk1(-/-) mice after 1 month, even in the absence of apo-opsin. This degeneration was independent of light conditions and occurred even in the absence of transducin in Grk1(-/-)Gnat1(-/-) mice. Taken together, our results demonstrate a light-independent mechanism for retinal degeneration in the absence of GRK1, suggesting a second, not previously recognized role for that kinase.


Subject(s)
G-Protein-Coupled Receptor Kinase 1/deficiency , Retinal Degeneration/genetics , Transducin/metabolism , Vision, Ocular/genetics , Adaptation, Ocular/genetics , Animals , Biophysics/methods , Carrier Proteins/genetics , Eye Proteins/genetics , GTP-Binding Protein alpha Subunits/deficiency , Mice , Mice, Knockout , Opsins/metabolism , Phosphorylation/genetics , Photic Stimulation/methods , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/physiology , cis-trans-Isomerases
SELECTION OF CITATIONS
SEARCH DETAIL
...