Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
Vision Res ; 75: 88-97, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22819833

ABSTRACT

Senior-Løken syndrome (SLS) is an autosomal recessive disease characterized by development of a retinitis pigmentosa (RP)- or Leber congenital amaurosis (LCA)-like retinal dystrophy and a medullary cystic kidney disease, nephronophthisis. Mutations in several genes (called nephrocystins) have been shown to cause SLS. The proteins encoded by these genes are localized in the connecting cilium of photoreceptor cells and in the primary cilium of kidney cells. Nephrocystins are thought to have a role in regulating transport of proteins bound to the outer segment/primary cilium; however, the precise molecular mechanisms are largely undetermined. This review will survey the biochemistry, cell biology and existing animal models for each of the nephrocystins as it relates to photoreceptor biology and pathogenesis of retinal degeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Kidney Diseases, Cystic/genetics , Leber Congenital Amaurosis/genetics , Membrane Proteins/genetics , Optic Atrophies, Hereditary/genetics , Retinitis Pigmentosa/genetics , Ciliopathies , Cytoskeletal Proteins , Humans , Mutation
2.
Proc Natl Acad Sci U S A ; 104(21): 8857-62, 2007 May 22.
Article in English | MEDLINE | ID: mdl-17496142

ABSTRACT

The mouse Pde6d gene encodes a ubiquitous prenyl binding protein, termed PrBP/delta, of largely unknown physiological function. PrBP/delta was originally identified as a putative rod cGMP phosphodiesterase (PDE6) subunit in the retina, where it is relatively abundant. To investigate the consequences of Pde6d deletion in retina, we generated a Pde6d(-/-) mouse by targeted recombination. Although manifesting reduced body weight, the Pde6d(-/-) mouse was viable and fertile and its retina developed normally. Immunocytochemistry showed that farnesylated rhodopsin kinase (GRK1) and prenylated rod PDE6 catalytic subunits partially mislocalized in Pde6d(-/-) rods, whereas rhodopsin was unaffected. In Pde6d(-/-) rod single-cell recordings, sensitivity to single photons was increased and saturating flash responses were prolonged. Pde6d(-/-) scotopic paired-flash electroretinograms indicated a delay in recovery of the dark state, likely due to reduced levels of GRK1 in rod outer segments. In Pde6d(-/-) cone outer segments, GRK1 and cone PDE6alpha' were present at very low levels and the photopic b-wave amplitudes were reduced by 70%. Thus the absence of PrBP/delta in retina impairs transport of prenylated proteins, particularly GRK1 and cone PDE, to rod and cone outer segments, resulting in altered photoreceptor physiology and a phenotype of a slowly progressing rod/cone dystrophy.


Subject(s)
G-Protein-Coupled Receptor Kinase 1/metabolism , Gene Deletion , Phosphoric Diester Hydrolases/deficiency , Phosphoric Diester Hydrolases/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Animals , Catalytic Domain , Cyclic Nucleotide Phosphodiesterases, Type 6 , Electroretinography , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoprene/metabolism , Phosphoric Diester Hydrolases/genetics , Photoreceptor Cells, Vertebrate/chemistry , Protein Transport
3.
Subcell Biochem ; 45: 71-91, 2007.
Article in English | MEDLINE | ID: mdl-18193635

ABSTRACT

Detailed biochemical, structural and physiological studies of the role of Ca2(+)-binding proteins in mammalian retinal neurons have yielded new insights into the function of these proteins in normal and pathological states. In phototransduction, a biochemical process that is responsible for the conversion of light into an electrical impulse, guanylate cyclases (GCs) are regulated by GC-activating proteins (GCAPs). These regulatory proteins respond to changes in cytoplasmic Ca2+ concentrations. Disruption of Ca2+ homeostasis in photoreceptor cells by genetic and environmental factors can result ultimately in degeneration of these cells. Pathogenic mutations in GC1 and GCAP1 cause autosomal recessive Leber congenital amaurosis and autosomal dominant cone dystrophy, respectively. This report provides a recent account of the advances, challenges, and possible future prospects of studying this important step in visual transduction that transcends to other neuronal Ca2+ homeostasis processes.


Subject(s)
Guanylate Cyclase-Activating Proteins/physiology , Guanylate Cyclase/metabolism , Retinal Diseases/physiopathology , Amino Acid Sequence , Amino Acid Substitution , Animals , Calcium Signaling/physiology , EF Hand Motifs , Enzyme Activation , Guanylate Cyclase/genetics , Guanylate Cyclase-Activating Proteins/genetics , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Photoreceptor Cells/physiology , Sequence Alignment , Vision, Ocular/physiology
5.
Biol Chem ; 382(8): 1179-88, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11592399

ABSTRACT

Guanylate cyclase-activating protein 1 (GCAP1) and guanylate cyclase-inhibitory protein (GCIP) are calmodulin-related Ca2+-binding proteins expressed in vertebrate photoreceptor cells. GCAP1 activates photoreceptor guanylate cyclase 1 (GC1) at low free [Ca2+] (<50 nM, in the light), but inhibits it at physiological high [Ca2+] (1 microM, in the dark). GCIP, a Ca2+-binding protein from frog retina, inhibits GC1 at approximately 1 microM [Ca2+], but is unable to stimulate cyclase at low [Ca2+]. In this study, we probed the interaction between GCAP1 and GC1 by producing GCAP1/GCIP chimeras and tested their capability to stimulate GC1. We prepared eight pairs of constructs in which the N-terminal portions of GCIP and GCAP1 were successively replaced by corresponding domains of GCAP1, and GCIP, respectively. The expressed proteins were purified and tested for stimulation of GC1 at 50 nM [Ca2+], and their ability to competitively inhibit GC1 stimulation by a Ca2+-insensitive GCAP1 mutant, GCAP1-tm, at high [Ca2+]. While all GCAP1/GCIP chimeras competitively inhibited GC1 stimulation at high [Ca2+] by GCAP1-tm, several of the GCIP/GCAP1 chimeras had no effect. A chimera consisting of residues 1-20 of GCIP and 21-205 of GCAP1 had no effect on GC1 at low [Ca2+], suggesting that the N-terminal region MGNIMDGKSVEELSSTECHQ, which has no sequence similarity to GCIP, is among the key components necessary for GC1 stimulation. A GCAP1/GCIP chimera consisting of residues 1-43 (including nonfunctional EF1) of GCAP1 and residues 56-206 of GCIP stimulated GC1 at low [Ca2+] and inhibited GC1 at high [Ca2+], suggesting that the essential components required to transform an inhibitory to an activating protein are contained within the N-terminal region of GCAP1 (residues 1-43).


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Eye Proteins/metabolism , Receptors, Cell Surface , Transcription Factors/genetics , Xenopus Proteins , Amino Acid Sequence , Animals , Blotting, Western , Calcium/metabolism , Cattle , Cells, Cultured , Eye Proteins/genetics , Guanylate Cyclase/metabolism , Guanylate Cyclase-Activating Proteins , Insecta/cytology , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Transcription Factors/metabolism
6.
Proc Natl Acad Sci U S A ; 98(17): 9948-53, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11493703

ABSTRACT

The retina's photoreceptor cells adjust their sensitivity to allow photons to be transduced over a wide range of light intensities. One mechanism thought to participate in sensitivity adjustments is Ca(2+) regulation of guanylate cyclase (GC) by guanylate cyclase-activating proteins (GCAPs). We evaluated the contribution of GCAPs to sensitivity regulation in rods by disrupting their expression in transgenic mice. The GC activity from GCAPs-/- retinas showed no Ca(2+) dependence, indicating that Ca(2+) regulation of GCs had indeed been abolished. Flash responses from dark-adapted GCAPs-/- rods were larger and slower than responses from wild-type rods. In addition, the incremental flash sensitivity of GCAPs-/- rods failed to be maintained at wild-type levels in bright steady light. GCAP2 expressed in GCAPs-/- rods restored maximal light-induced GC activity but did not restore normal flash response kinetics. We conclude that GCAPs strongly regulate GC activity in mouse rods, decreasing the flash sensitivity in darkness and increasing the incremental flash sensitivity in bright steady light, thereby extending the rod's operating range.


Subject(s)
Calcium-Binding Proteins/physiology , Cyclic GMP/physiology , Eye Proteins/metabolism , Guanylate Cyclase/metabolism , Photic Stimulation , Retinal Rod Photoreceptor Cells/radiation effects , Second Messenger Systems/physiology , Sensory Thresholds/physiology , Visual Perception/physiology , Adaptation, Physiological , Animals , Calcium Signaling , Calcium-Binding Proteins/genetics , Cattle , Darkness , Enzyme Activation , Female , Gene Expression Regulation , Guanylate Cyclase-Activating Proteins , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Photochemistry , Recombinant Fusion Proteins/physiology , Retinal Rod Photoreceptor Cells/enzymology
7.
J Biol Chem ; 276(46): 43361-73, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11524415

ABSTRACT

Guanylyl cyclase-activating proteins are EF-hand Ca(2+)-binding proteins that belong to the calmodulin superfamily. They are involved in the regulation of photoreceptor membrane-associated guanylyl cyclases that produce cGMP, a second messenger of vertebrate vision. Here, we investigated changes in GCAP1 structure using mutagenesis, chemical modifications, and spectroscopic methods. Two Cys residues of GCAP1 situated in spatially distinct regions of the N-terminal domain (positions 18 and 29) and two Cys residues located within the C-terminal lobe (positions 106 and 125) were employed to detect conformational changes upon Ca(2+) binding. GCAP1 mutants with only a single Cys residue at each of these positions, modified with N,N'-dimethyl-N-(iodoacetyl)-N'-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamine, an environmentally sensitive fluorophore, and with (1-oxy-2,2,5,5-tetramethylpyrroline-3-methyl)methanethiosulfonate, a spin label reagent, were studied using fluorescence and EPR spectroscopy, respectively. Only minor structural changes around Cys(18), Cys(29), Cys(106), and Cys(125) were observed as a function of Ca(2+) concentration. No Ca(2+)-dependent oligomerization of GCAP1 was observed at physiologically relevant Ca(2+) concentrations, in contrast to the observation reported by others for GCAP2. Based on these results and previous studies, we propose a photoreceptor activation model that assumes changes within the flexible central helix upon Ca(2+) dissociation, causing relative reorientation of two structural domains containing a pair of EF-hand motifs and thus switching its partner, guanylyl cyclase, from an inactive (or low activity) to an active conformation.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Electron Spin Resonance Spectroscopy/methods , Amino Acid Motifs , Amino Acid Sequence , Animals , Calcium/chemistry , Calcium/metabolism , Calcium/pharmacology , Cattle , Chromatography, Gel , Cyclic N-Oxides/pharmacology , Cysteine/chemistry , Dose-Response Relationship, Drug , EF Hand Motifs , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Eye/metabolism , Fluorescent Dyes/pharmacology , Guanylate Cyclase/chemistry , Guanylate Cyclase-Activating Proteins , Mesylates/pharmacology , Models, Biological , Models, Chemical , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Oxadiazoles/pharmacology , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Spectrometry, Fluorescence , Spin Labels , Sulfur/chemistry
8.
Prog Retin Eye Res ; 20(4): 469-529, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390257

ABSTRACT

Absorption of light by rhodopsin or cone pigments in photoreceptors triggers photoisomerization of their universal chromophore, 11-cis-retinal, to all-trans-retinal. This photoreaction is the initial step in phototransduction that ultimately leads to the sensation of vision. Currently, a great deal of effort is directed toward elucidating mechanisms that return photoreceptors to the dark-adapted state, and processes that restore rhodopsin and counterbalance the bleaching of rhodopsin. Most notably, enzymatic isomerization of all-trans-retinal to 11-cis-retinal, called the visual cycle (or more properly the retinoid cycle), is required for regeneration of these visual pigments. Regeneration begins in rods and cones when all-trans-retinal is reduced to all-trans-retinol. The process continues in adjacent retinal pigment epithelial cells (RPE), where a complex set of reactions converts all-trans-retinol to 11-cis-retinal. Although remarkable progress has been made over the past decade in understanding the phototransduction cascade, our understanding of the retinoid cycle remains rudimentary. The aim of this review is to summarize recent developments in our current understanding of the retinoid cycle at the molecular level, and to examine the relevance of these reactions to phototransduction.


Subject(s)
Retina/metabolism , Retinoids/metabolism , Vision, Ocular/physiology , Animals , Dark Adaptation/physiology , Humans , Light , Retina/radiation effects , Rhodopsin/physiology , Rhodopsin/radiation effects
9.
Invest Ophthalmol Vis Sci ; 42(3): 826-33, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222546

ABSTRACT

PURPOSE: To study mechanisms leading to photoreceptor degeneration in mouse models for autosomal dominant retinitis pigmentosa (adRP) based on the rhodopsin P23H mutation. METHODS: Mice of a transgenic line expressing a rhodopsin triple mutant, V20G, P23H, and P27L (GHL), were mated with rhodopsin (rho) knockout mice. Littermates of various ages and genotypes (GHL+rho+/+, GHL+rho+/-, and GHL+rho-/-) were examined for outer nuclear layer thickness and outer segment formation (histology), fate of mutant rhodopsin (immunocytochemistry), and photoreceptor function (electroretinogram; ERG). RESULTS: Mice expressing GHL-rhodopsin in the absence of wild-type rhodopsin had severe retinopathy, which was nearly complete by postnatal day (P)30. GHL-rhodopsin formed homodimers nearly exclusively on sodium dodecyl sulfate-polyacrylamide gel electrophoresis gels, whereas wild-type rhodopsin predominantly formed monomers. Expression level of mutant rhodopsin in predegenerate (P10) GHL+rho-/- retinas was low, approximately 10% to 25% of normal levels. No elaboration of disc membrane or outer segment formation was observed at any time point examined. The mutant rhodopsin was found mostly in perinuclear locales (endoplasmic reticulum; ER) as evidenced by colocalization using the antibodies Rho1D4 and calnexin-NT. CONCLUSIONS: GHL-rhodopsin dimerizes, localizes to the ER, and fails to transport and support outer segment formation. Additionally, the mutant protein does not support a scotopic ERG a-wave and accelerates photoreceptor degeneration over that occurring with the rhodopsin knockout alone. These findings indicate a cytotoxic effect of the mutant protein, probably elicited by an unfolded protein response.


Subject(s)
Mutation , Photoreceptor Cells, Vertebrate/ultrastructure , Retinal Degeneration/genetics , Rhodopsin/genetics , Transgenes/genetics , Animals , DNA Primers/chemistry , Electrophoresis, Polyacrylamide Gel , Electroretinography , Female , Fluorescent Antibody Technique, Indirect , Gene Expression , Genotype , Immunoblotting , Male , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Immunoelectron , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Rhodopsin/metabolism
10.
Mol Vis ; 7: 305-13, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11754336

ABSTRACT

PURPOSE: Homozygous inactivation of the mouse gene for GRK1 (G protein-coupled receptor kinase 1, or rhodopsin kinase) causes severe defects in the recovery of cone phototransduction. However, electroretinographic (ERG) analyses of human oguchi patients with defective GRK1 alleles showed normal or slightly abnormal photopic responses. It remains unclear why the loss of GRK1 yields such different phenotypes in the recovery of mouse and human cones. We examined the localization and enzyme activity of GRK7, the human ortholog of the seventh member of the GRK family, in an attempt to understand its potential role in photopic vision. METHODS: Bioinformatic approaches were used to identify the human GRK7 gene. Human and bovine GRK7 cDNAs were isolated by RT-PCR. Recombinant GRK7, expressed in insect cells, was used to phosphorylate activated rhodopsin. Antibodies raised against GRK7 peptides were used to examine the retina specific expression of GRK7 by immunoblotting and its subcellular localization by immunocytochemistry. RESULTS: The human GRK7 gene is located on chromosome 3q21, spans at least 10 Kb and consists of 4 exons. In human, GRK7 is expressed exclusively in the retina and is found in all retinal neurons, and specifically, in cone outer segments. Recombinant human GRK7 catalyzes rhodopsin phosphorylation in a light dependent manner. We provide evidence that GRK1 and GRK7 are co-expressed in human cones. In contrast, mouse GRK7 is expressed in many tissues including retina where photoreceptors apparently do not express GRK7. CONCLUSIONS: The presence of GRK7 in human, but not in mouse, cone outer segments suggests that GRK7 may function to provide the normal photopic vision reported by oguchi patients with a defective GRK1 gene. The absence of GRK7 expression in cone outer segments of mice is consistent with the notion that mouse cones rely solely on GRK1 to shutoff cone visual pigments.


Subject(s)
Eye Proteins , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Retinal Cone Photoreceptor Cells/enzymology , Amino Acid Sequence , Animals , Cattle , Chickens , Chromosomes, Human, Pair 3/genetics , Cloning, Molecular , Fluorescent Antibody Technique, Indirect , G-Protein-Coupled Receptor Kinase 1 , G-Protein-Coupled Receptor Kinases , Genetic Linkage , Humans , Immunoblotting , Immunoenzyme Techniques , Mice , Molecular Sequence Data , Phosphorylation , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Rhodopsin/metabolism , Sequence Homology, Amino Acid , Swine
11.
Biochim Biophys Acta ; 1498(2-3): 233-51, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11108966

ABSTRACT

Examination of the role of Ca(2+)-binding proteins (CaBPs) in mammalian retinal neurons has yielded new insights into the function of these proteins in normal and pathological states. In the last 8 years, studies on guanylate cyclase (GC) regulation by three GC-activating proteins (GCAP1-3) led to several breakthroughs, among them the recent biochemical analysis of GCAP1(Y99) mutants associated with autosomal dominant cone dystrophy. Perturbation of Ca(2+) homeostasis controlled by mutant GCAP1 in photoreceptor cells may result ultimately in degeneration of these cells. Here, detailed analysis of biochemical properties of GCAP1(P50L), which causes a milder form of autosomal dominant cone dystrophy than constitutive active Y99C mutation, showed that the P50L mutation resulted in a decrease of Ca(2+)-binding, without changes in the GC activity profile of the mutant GCAP1. In contrast to this biochemically well-defined regulatory mechanism that involves GCAPs, understanding of other processes in the retina that are regulated by Ca(2+) is at a rudimentary stage. Recently, we have identified five homologous genes encoding CaBPs that are expressed in the mammalian retina. Several members of this subfamily are also present in other tissues. In contrast to GCAPs, the function of this subfamily of calmodulin (CaM)-like CaBPs is poorly understood. CaBPs are closely related to CaM and in biochemical assays CaBPs substitute for CaM in stimulation of CaM-dependent kinase II, and calcineurin, a protein phosphatase. These results suggest that CaM-like CaBPs have evolved into diverse subfamilies that control fundamental processes in cells where they are expressed.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Eye Diseases/genetics , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins/biosynthesis , Calmodulin/biosynthesis , Cattle , Chromosome Mapping , Cloning, Molecular , Enzyme Activation , Escherichia coli/metabolism , Guanylate Cyclase/biosynthesis , Guanylate Cyclase/genetics , Guanylate Cyclase-Activating Proteins , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Photoreceptor Cells, Vertebrate/enzymology , Retina/enzymology , Sequence Alignment
13.
Bioessays ; 22(4): 337-50, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10723031

ABSTRACT

The complex sensation of vision begins with the relatively simple photoisomerization of the visual pigment chromophore 11-cis-retinal to its all-trans configuration. This event initiates a series of biochemical reactions that are collectively referred to as phototransduction, which ultimately lead to a change in the electrochemical signaling of the photoreceptor cell. To operate in a wide range of light intensities, however, the phototransduction pathway must allow for adjustments to background light. These take place through physiological adaptation processes that rely primarily on Ca(2+) ions. While Ca(2+) may modulate some activities directly, it is more often the case that Ca(2+)-binding proteins mediate between transient changes in the concentration of Ca(2+) and the adaptation processes that are associated with phototransduction. Recently, combined genetic, physiological, and biochemical analyses have yielded new insights about the properties and functions of many phototransduction-specific components, including some novel Ca(2+)-binding proteins. Understanding these Ca(2+)-binding proteins will provide a more complete picture of visual transduction, including the mechanisms associated with adaptation, and of related degenerative diseases.


Subject(s)
Calcium-Binding Proteins/physiology , Eye Diseases/etiology , Retina/metabolism , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Guanylate Cyclase-Activating Proteins , Humans , Photoreceptor Cells, Vertebrate/metabolism , Protein Conformation
14.
Gene ; 240(1): 23-34, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10564809

ABSTRACT

An isoform of RGS9 was recently identified as the GTPase activating protein in bovine and mouse rod and cone photoreceptors. To explore the potential role of the RGS9 gene in human retinal disease, we determined its exon/intron arrangement, and investigated its expression in human retina. The results show that the gene, located on 17q24, consists of 19 exons and spans more than 75kb of genomic DNA. The entire gene was found to be contained on a single BAC clone with an insert size of 170kb. The major transcripts of the gene are alternatively spliced into a 9.5kb retina-specific transcript (RGS9-1) and a brain specific 2.5kb transcript (RGS9-2). Exons 1-16 are constitutive and present in both variants. Exon 17 contains the 3' end of the open reading frame and the 3'-UTR of the RGS9-1 variant. In RGS9-2, exon 17 is alternatively spliced and joined to exons 18 and 19 that are not present in the retina variant. Immunolocalization with a monoclonal antibody recognizing the retina and brain variants shows abundant expression in photoreceptors and possibly very low levels in cell types of the inner retina. Owing to the specific expression of RGS9-1 in photoreceptors the RGS9 gene is a candidate gene for RP17, a form of autosomal retinitis pigmentosa, located on the long arm of chromosome 17.


Subject(s)
Alternative Splicing , Genes/genetics , RGS Proteins/genetics , Aged , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , Blotting, Western , Brain/metabolism , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 17/genetics , Corpus Striatum/chemistry , Cricetinae , DNA/analysis , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Exons , Gene Expression Regulation, Developmental , Genetic Variation , Humans , Hybrid Cells , Immunohistochemistry , In Situ Hybridization, Fluorescence , Introns , Mammals/genetics , Molecular Sequence Data , Protein Isoforms/genetics , RGS Proteins/analysis , Retina/chemistry , Retina/metabolism , Sequence Homology, Amino Acid
15.
Mol Vis ; 5: 14, 1999 Jul 28.
Article in English | MEDLINE | ID: mdl-10427104

ABSTRACT

PURPOSE: This study had three objectives: (1) to characterize the structures of the chicken GCAP1 and GCAP2 genes; (2) to determine if GCAP1, GCAP2, and GC1 genes are expressed in chicken pineal gland; (3) if GC1 is expressed in chicken pineal, to determine if the GC1 null mutation carried by the retinal degeneration (rd) chicken is associated with degenerative changes within the pineal glands of these animals. METHODS: GCAP1 and GCAP2 gene structures were determined by analyses of chicken cosmid and cDNA clones. The putative transcription start points for these genes were determined using 5'-RACE. GCAP1, GCAP2 and GC1 transcripts were analyzed using Northern blot and RT-PCR. Routine light microscopy was used to examine pineal morphology. RESULTS: Chicken GCAP1 and GCAP2 genes are arranged in a tail-to-tail array. Each protein is encoded by 4 exons that are interrupted by 3 introns of variable length, the positions of which are identical within each gene. The putative transcription start points for GCAP1 and GCAP2 are 314 and 243 bases upstream of the translation start codons of these genes, respectively. As in retina, GCAP1, GCAP2 and GC1 genes are expressed in the chicken pineal. Although the GC1 null mutation is present in both the retina and pineal of the rd chicken, only the retina appears to undergo degeneration. CONCLUSIONS: The identical arrangement of chicken, human, and mouse GCAP1/2 genes suggests that these genes originated from an ancient gene duplication/inversion event that occurred during evolution prior to vertebrate diversification. The expression of GC1, GCAP1, and GCAP2 in chicken pineal is consistent with the hypothesis that chicken pineal contains a functional phototransduction cascade. The absence of cellular degeneration in the rd pineal gland suggests that GC1 is not critical for pineal cell survival.


Subject(s)
Calcium-Binding Proteins/genetics , Guanylate Cyclase/genetics , Pineal Gland/metabolism , Receptors, Cell Surface , Amino Acid Sequence , Animals , Animals, Newborn , Base Sequence , Blotting, Northern , Calcium-Binding Proteins/biosynthesis , Chickens/genetics , Guanylate Cyclase/biosynthesis , Guanylate Cyclase-Activating Proteins , Humans , Mice , Molecular Sequence Data , Pineal Gland/anatomy & histology , Promoter Regions, Genetic , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Reverse Transcriptase Polymerase Chain Reaction
17.
Brain Res Mol Brain Res ; 70(2): 253-63, 1999 Jul 05.
Article in English | MEDLINE | ID: mdl-10407173

ABSTRACT

We are interested in determining if the visual phototransduction cascade plays a role in light entrainment of photoreceptor circadian oscillators. In this study, we compared mRNA levels of iodopsin and the chicken homolog of Clock (cClock) in the retinas of normal and rd (retinal degeneration) chickens that lack functional rod and cone phototransduction cascades. Iodopsin is a circadian-regulated, photoreceptor-specific gene expressed in chicken retina, and Clock is a transcription factor that has been shown to play a role in the circadian clock mechanism in mouse and Drosophila. The results of our analyses show that cClock and iodopsin transcript levels undergo daily oscillations in retinas of normal animals housed under 12 h light:12 h dark (12L:12D) conditions, and that these oscillations are maintained in the absence of light. Levels of these transcripts in the retinas of rd/rd chickens housed under cyclic light conditions did not change significantly over the course of a 12L:12D cycle; however, there was evidence that the photoreceptor oscillators were entrained in these animals. Comparisons of our normal and rd/rd data suggest that there are at least two light entrainment pathways that impinge on the oscillators found in photoreceptor cells, one of which is effectively disabled by the GC1 null mutation carried by the rd chicken.


Subject(s)
Chickens/genetics , Circadian Rhythm , Gene Expression Regulation , Photoreceptor Cells, Vertebrate/physiology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Pigments/genetics , Rod Opsins , Trans-Activators/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , CLOCK Proteins , DNA Primers , Humans , Light Signal Transduction , Mice , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/metabolism , Restriction Mapping , Sequence Alignment , Sequence Homology, Amino Acid , Trans-Activators/chemistry , Transcription Factors/genetics
18.
J Biol Chem ; 274(28): 19829-37, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10391927

ABSTRACT

Guanylyl cyclase-activating proteins (GCAPs are 23-kDa Ca2+-binding proteins belonging to the calmodulin superfamily. Ca2+-free GCAPs are responsible for activation of photoreceptor guanylyl cyclase during light adaptation. In this study, we characterized GCAP1 mutants in which three endogenous nonessential Trp residues were replaced by Phe residues, eliminating intrinsic fluorescence. Subsequently, hydrophobic amino acids adjacent to each of the three functional Ca2+-binding loops were replaced by reporter Trp residues. Using fluorescence spectroscopy and biochemical assays, we found that binding of Ca2+ to GCAP1 causes a major conformational change especially in the region around the EF3-hand motif. This transition of GCAP1 from an activator to an inhibitor of GC requires an activation energy Ea = 9.3 kcal/mol. When Tyr99 adjacent to the EF3-hand motif was replaced by Cys, a mutation linked to autosomal dominant cone dystrophy in humans, Cys99 is unable to stabilize the inactive GCAP1-Ca2+ complex. Stopped-flow kinetic measurements indicated that GCAP1 rapidly loses its bound Ca2+ (k-1 = 72 s-1 at 37 degrees C) and was estimated to associate with Ca2+ at a rate (k1 > 2 x 10(8) M-1 s-1) close to the diffusion limit. Thus, GCAP1 displays thermodynamic and kinetic properties that are compatible with its involvement early in the phototransduction response.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium/pharmacology , Protein Conformation , Tryptophan/genetics , Animals , Cattle , Guanylate Cyclase/metabolism , Guanylate Cyclase-Activating Proteins , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Rod Cell Outer Segment/enzymology , Spectrometry, Fluorescence , Tryptophan/chemistry
19.
Mol Cell Neurosci ; 13(2): 95-103, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10192768

ABSTRACT

Aoffa-Synuclein, a presynaptic nerve terminal protein, may be an important component of Lewy bodies in Parkinson's disease, dementia with Lewy bodies, and other neurodegenerative diseases. Additionally, recent genetic studies based on linkage analysis and cosegregation of A53T and A30P missense mutations demonstrated that the alpha-synuclein gene may be responsible for the development of at least some cases of familial Parkinson's disease. Despite intense interest in the members of the synuclein family, their function(s) and exact role in the diseases remained unknown. Here we describe a new member of the synuclein family, which we term synoretin, and show that it is expressed in different retinal cells, as well as in the brain, and it may affect the regulation of signal transduction through activation of the Elk1 pathway.


Subject(s)
Eye Proteins/isolation & purification , Lewy Bodies/chemistry , Nerve Tissue Proteins/isolation & purification , Retina/chemistry , Amino Acid Sequence , Animals , Base Sequence , Calcium-Binding Proteins/metabolism , Cattle , Cloning, Molecular , Codon/genetics , Eye Proteins/classification , Eye Proteins/genetics , Eye Proteins/physiology , Gene Expression , Gene Library , Genes , Guanylate Cyclase-Activating Proteins , Humans , Molecular Sequence Data , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Organ Specificity , Parkinson Disease/genetics , Parkinson Disease/metabolism , Rabbits , Recombinant Fusion Proteins/biosynthesis , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Synucleins , Transfection , alpha-Synuclein , gamma-Synuclein
20.
Biochemistry ; 38(9): 2611-6, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10052930

ABSTRACT

Photoreceptor guanylyl cyclase (ROS-GC), converting GTP into cGMP and pyrophosphate, is a key enzyme in the regulation of the visual transduction cascade. ROS-GC requires GC-activating proteins (GCAPs) and low free [Ca] for full activity. We found that when choline or potassium were the major cations present, light caused a 70% inhibition of stimulated ROS-GC in native unstripped membranes. In the presence of sodium ions, however, no inhibition was observed. ROS-GC activity of ROS membranes, stripped of transducin and other components, was not affected by light when reconstituted with GCAP1 only. However, when stripped ROS membranes were reconstituted with both GCAP1 and either transducin (T alpha beta gamma) or the T beta gamma-subunits, the inhibition of ROS-GC by light was restored. The T alpha-subunit alone was ineffective. These results suggest that under saturating light conditions, ROS-GC may be regulated by T beta gamma and cations, providing a possible mechanism of desensitization and light adaptation.


Subject(s)
Guanylate Cyclase/antagonists & inhibitors , Light , Rod Cell Outer Segment/enzymology , Transducin/physiology , Animals , Cations, Monovalent/pharmacology , Cattle , Colorimetry , Enzyme Activation/drug effects , Guanylate Cyclase/metabolism , Hydroxylamine/pharmacology , Models, Biological , Photochemistry , Rhodopsin/metabolism , Sodium/pharmacology , Transducin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...