Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38558979

ABSTRACT

Progressive Rod-Cone Degeneration (PRCD) is an integral membrane protein found in photoreceptor outer segment (OS) disc membranes and its function remains unknown. Mutations in Prcd are implicated in Retinitis pigmentosa (RP) in humans and multiple dog breeds. PRCD-deficient models exhibit decreased levels of cholesterol in the plasma. However, potential changes in the retinal cholesterol remain unexplored. In addition, impaired phagocytosis observed in these animal models points to potential deficits in the retinal pigment epithelium (RPE). Here, using a Prcd -/- murine model we investigated the alterations in the retinal cholesterol levels and impairments in the structural and functional integrity of the RPE. Lipidomic and immunohistochemical analyses show a 5-fold increase in the levels of cholesteryl esters (C.Es) and accumulation of neutral lipids in the PRCD-deficient retina, respectively, indicating alterations in total retinal cholesterol. Longitudinal fundus and spectral domain optical coherence tomography (SD-OCT) examinations showed focal lesions and RPE hyperreflectivity. Strikingly, the RPE of Prcd -/- mice exhibited age-related pathological features such as neutral lipid deposits, lipofuscin accumulation, Bruch's membrane (BrM) thickening and drusenoid focal deposits, mirroring an Age-related Macular Degeneration (AMD)-like phenotype. We propose that the extensive lipofuscin accumulation likely impairs lysosomal function, leading to the defective phagocytosis observed in Prcd -/- mice. Our findings support the dysregulation of retinal cholesterol homeostasis in the absence of PRCD. Further, we demonstrate that progressive photoreceptor degeneration in Prcd -/- mice is accompanied by progressive structural and functional deficits in the RPE, which likely exacerbates vision loss over time.

2.
Adv Exp Med Biol ; 1415: 389-394, 2023.
Article in English | MEDLINE | ID: mdl-37440062

ABSTRACT

The outer segments of photoreceptors are specialized sensory cilia crucial for light detection. Any disruption that alters outer segment morphology can impair photoreceptor function and therefore vision. Progressive rod-cone degeneration (PRCD) is an integral membrane protein exclusively present in the photoreceptor OS with an unknown function. Multiple mutations in PRCD are linked with retinitis pigmentosa. The most common PRCD mutation observed in both human and multiple dog breeds, PRCD-C2Y, lacks the lipid modification "palmitoylation," which is crucial for protein stability and trafficking to the OS. Previous studies including ours show impaired disc morphogenesis and rhodopsin distributions in the absence of PRCD, but the precise role of PRCD in maintaining OS structure and function remains unclear. In this chapter, we discuss the potential role of PRCD in the maintenance of photoreceptor OS structural and functional integrity.


Subject(s)
Cilia , Eye Proteins , Animals , Dogs , Humans , Eye Proteins/genetics , Cilia/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Homeostasis , Lipids
3.
Bio Protoc ; 13(8): e4654, 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37113337

ABSTRACT

Palmitoylation is a unique and reversible posttranslational lipid modification (PTM) that plays a critical role in many cellular events, including protein stability, activity, membrane association, and protein-protein interactions. The dynamic nature of palmitoylation dictates the efficient sorting of various retinal proteins to specific subcellular compartments. However, the underlying mechanism through which palmitoylation supports efficient protein trafficking in the retina remains unclear. Recent studies show that palmitoylation can also function as a signaling PTM, underlying epigenetic regulation and homeostasis in the retina. Efficient isolation of retinal palmitoyl proteome will pave the way to a better understanding of the role(s) for palmitoylation in visual function. The standard methods for detecting palmitoylated proteins employ 3H- or 14C-radiolabeled palmitic acid and have many limitations, including poor sensitivity. Relatively recent studies use thiopropyl Sepharose 6B resin, which offers efficient detection of palmitoylated proteome but is now discontinued from the market. Here, we describe a modified acyl resin-assisted capture (Acyl-RAC) method using agarose S3 high-capacity resin to purify palmitoylated proteins from the retina and other tissues, which is greatly compatible with downstream processing by LC-MS/MS. Unlike other palmitoylation assays, the present protocol is easy to perform and cost-effective. Graphical abstract.

4.
Int J Mol Sci ; 23(18)2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36142714

ABSTRACT

Progressive rod-cone degeneration (PRCD) is a photoreceptor outer segment (OS) disc-specific protein essential for maintaining OS structures while contributing to rhodopsin packaging densities and distribution in disc membranes. Previously, we showed PRCD undergoing palmitoylation at the sole cysteine (Cys2), where a mutation linked with retinitis pigmentosa (RP) in humans and dogs demonstrates the importance of palmitoylation for protein stability and trafficking to the OS. We demonstrate a mutation, in the polybasic region (PBR) of PRCD (Arg17Cys) linked with RP where an additional lipidation is observed through acyl-RAC. Immunolocalization of transiently expressed R17C in hRPE1 cells depicts similar characteristics to wild-type PRCD; however, a double mutant lacking endogenous palmitoylation at Cys2Tyr with Arg17Cys is comparable to the C2Y protein as both aggregate, mislocalized to the subcellular compartments within the cytoplasm. Subretinal injection of PRCD mutant constructs followed by electroporation in murine retina exhibit mislocalization in the inner segment. Despite being additionally lipidated and demonstrating strong membrane association, the mutation in the PBR affects protein stability and localization to the OS. Acylation within the PBR alone neither compensates for protein stability nor trafficking, revealing defects in the PBR likely lead to dysregulation of PRCD protein associated with blinding diseases.


Subject(s)
Retinitis Pigmentosa , Rhodopsin , Animals , Cysteine/genetics , Dogs , Eye Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mutation , Protein Stability , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism
5.
Elife ; 102021 12 08.
Article in English | MEDLINE | ID: mdl-34878972

ABSTRACT

Despite mounting evidence that the mammalian retina is exceptionally reliant on proper NAD+ homeostasis for health and function, the specific roles of subcellular NAD+ pools in retinal development, maintenance, and disease remain obscure. Here, we show that deletion of the nuclear-localized NAD+ synthase nicotinamide mononucleotide adenylyltransferase-1 (NMNAT1) in the developing murine retina causes early and severe degeneration of photoreceptors and select inner retinal neurons via multiple distinct cell death pathways. This severe phenotype is associated with disruptions to retinal central carbon metabolism, purine nucleotide synthesis, and amino acid pathways. Furthermore, transcriptomic and immunostaining approaches reveal dysregulation of a collection of photoreceptor and synapse-specific genes in NMNAT1 knockout retinas prior to detectable morphological or metabolic alterations. Collectively, our study reveals previously unrecognized complexity in NMNAT1-associated retinal degeneration and suggests a yet-undescribed role for NMNAT1 in gene regulation during photoreceptor terminal differentiation.


Subject(s)
Gene Deletion , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Photoreceptor Cells, Vertebrate/enzymology , Retinal Degeneration/enzymology , Retinal Neurons/enzymology , Animals , Female , Male , Mice , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Neurons/pathology
6.
Sci Rep ; 10(1): 17885, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087780

ABSTRACT

Progressive rod-cone degeneration (PRCD) is a small protein localized to photoreceptor outer segment (OS) disc membranes. Several mutations in PRCD are linked to retinitis pigmentosa (RP) in canines and humans, and while recent studies have established that PRCD is required for high fidelity disc morphogenesis, its precise role in this process remains a mystery. To better understand the part which PRCD plays in disease progression as well as its contribution to photoreceptor OS disc morphogenesis, we generated a Prcd-KO animal model using CRISPR/Cas9. Loss of PRCD from the retina results in reduced visual function accompanied by slow rod photoreceptor degeneration. We observed a significant decrease in rhodopsin levels in Prcd-KO retina prior to photoreceptor degeneration. Furthermore, ultrastructural analysis demonstrates that rod photoreceptors lacking PRCD display disoriented and dysmorphic OS disc membranes. Strikingly, atomic force microscopy reveals that many disc membranes in Prcd-KO rod photoreceptor neurons are irregular, containing fewer rhodopsin molecules and decreased rhodopsin packing density compared to wild-type discs. This study strongly suggests an important role for PRCD in regulation of rhodopsin incorporation and packaging density into disc membranes, a process which, when dysregulated, likely gives rise to the visual defects observed in patients with PRCD-associated RP.


Subject(s)
Eye Proteins/metabolism , Membrane Proteins/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/metabolism , Rhodopsin/metabolism , Animals , Disease Models, Animal , Eye Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Microscopy, Atomic Force , Retina/metabolism , Retinitis Pigmentosa/genetics
7.
Proc Natl Acad Sci U S A ; 116(9): 3530-3535, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808746

ABSTRACT

Glucose metabolism in vertebrate retinas is dominated by aerobic glycolysis (the "Warburg Effect"), which allows only a small fraction of glucose-derived pyruvate to enter mitochondria. Here, we report evidence that the small fraction of pyruvate in photoreceptors that does get oxidized by their mitochondria is required for visual function, photoreceptor structure and viability, normal neuron-glial interaction, and homeostasis of retinal metabolism. The mitochondrial pyruvate carrier (MPC) links glycolysis and mitochondrial metabolism. Retina-specific deletion of MPC1 results in progressive retinal degeneration and decline of visual function in both rod and cone photoreceptors. Using targeted-metabolomics and 13C tracers, we found that MPC1 is required for cytosolic reducing power maintenance, glutamine/glutamate metabolism, and flexibility in fuel utilization.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Retina/metabolism , Vision, Ocular/genetics , Animals , Glucose/metabolism , Glycolysis/genetics , Humans , Mice , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters , Pyruvic Acid/metabolism , Retina/pathology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology
8.
J Neurosci ; 39(8): 1347-1364, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30573647

ABSTRACT

Mutations in the Joubert syndrome-associated small GTPase ARL13B are linked to photoreceptor impairment and vision loss. To determine the role of ARL13B in the development, function, and maintenance of ciliated photoreceptors, we generated a pan-retina knock-out (Six3-Cre) and a rod photoreceptor-specific inducible conditional knock-out (Pde6g-CreERT2) of ARL13B using murine models. Embryonic deletion of ARL13B led to defects in retinal development with reduced cell proliferation. In the absence of ARL13B, photoreceptors failed to develop outer segment (OS) membranous discs and axonemes, resulting in loss of function and rapid degeneration. Additionally, the majority of photoreceptor basal bodies did not dock properly at the apical edge of the inner segments. The removal of ARL13B in adult rod photoreceptor cells after maturation of OS resulted in loss of photoresponse and vesiculation in the OS. Before changes in photoresponse, removal of ARL13B led to mislocalization of rhodopsin, prenylated phosphodiesterase-6 (PDE6), and intraflagellar transport protein-88 (IFT88). Our findings show that ARL13B is required at multiple stages of retinogenesis, including early postnatal proliferation of retinal progenitor cells, development of photoreceptor cilia, and morphogenesis of photoreceptor OS discs regardless of sex. Last, our results establish a need for ARL13B in photoreceptor maintenance and protein trafficking.SIGNIFICANCE STATEMENT The normal development of photoreceptor cilia is essential to create functional, organized outer segments with stacked membrane discs that house the phototransduction proteins necessary for sight. Our study identifies a complex role for ARL13B, a small GTPase linked to Joubert syndrome and visual impairment, at various stages of photoreceptor development. Loss of ARL13B led to defects in retinal proliferation, altered placement of basal bodies crucial for components of the cilium (transition zone) to emanate, and absence of photoreceptor-stacked discs. These defects led to extinguished visual response and dysregulated protein trafficking. Our findings show the complex role ARL13B plays in photoreceptor development, viability, and function. Our study accounts for the severe retinal impairment observed in ARL13B-linked Joubert syndrome patients.


Subject(s)
ADP-Ribosylation Factors/physiology , Retina/metabolism , Rod Cell Outer Segment/metabolism , ADP-Ribosylation Factors/deficiency , ADP-Ribosylation Factors/genetics , Aging/metabolism , Animals , Axoneme/metabolism , Axoneme/ultrastructure , Cilia/metabolism , Cilia/ultrastructure , Eye Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Organelle Biogenesis , Protein Transport/physiology , Retina/abnormalities , Retina/embryology , Retina/growth & development , Rod Cell Outer Segment/radiation effects , Sensory Rhodopsins/metabolism
9.
Front Mol Neurosci ; 11: 233, 2018.
Article in English | MEDLINE | ID: mdl-30038560

ABSTRACT

Rod and cone phosphodiesterase 6 (PDE6) are key effector enzymes of the vertebrate phototransduction pathway. Rod PDE6 consists of two catalytic subunits PDE6α and PDE6ß and two identical inhibitory PDE6γ subunits, while cone PDE6 is composed of two identical PDE6α' catalytic subunits and two identical cone-specific PDE6γ' inhibitory subunits. Despite their prominent function in regulating cGMP levels and therefore rod and cone light response properties, it is not known how each subunit contributes to the functional differences between rods and cones. In this study, we generated an rd10/cpfl1 mouse model lacking rod PDE6ß and cone PDE6α' subunits. Both rod and cone photoreceptor cells are degenerated with age and all PDE6 subunits degrade in rd10/cpfl1 mice. We expressed cone PDE6α' in both rods and cones of rd10/cpfl1 mice by adeno-associated virus (AAV)-mediated delivery driven by the ubiquitous, constitutive small chicken ß-actin promoter. We show that expression of PDE6α' rescues rod function in rd10/cpfl1 mice, and the restoration of rod light sensitivity is attained through restoration of endogenous rod PDE6γ and formation of a functional PDE6α'γ complex. However, improved photopic cone responses were achieved only after supplementation of both cone PDE6α' and PDE6γ' subunits but not by PDE6α' treatment alone. We observed a two fold increase of PDE6α' levels in the eyes injected with both PDE6α' plus PDE6γ' relative to eyes receiving PDE6α' alone. Despite the presence of both PDE6γ' and PDE6γ, the majority of PDE6α' formed functional complexes with PDE6γ', suggesting that PDE6α' has a higher association affinity for PDE6γ' than for PDE6γ. These results suggest that the presence of PDE6γ' augments cone PDE6 assembly and enhances its stability. Our finding has important implication for gene therapy of PDE6α'-associated achromatopsia.

10.
Front Mol Neurosci ; 11: 16, 2018.
Article in English | MEDLINE | ID: mdl-29410614

ABSTRACT

Primary cilia are microtubule-based organelles, which protrude from the plasma membrane and receive a wide range of extracellular signals. Various cilia use G protein-coupled receptors (GPCRs) for the detection of these signals. For instance, vertebrate rod photoreceptors use their cilia (also called outer segments) as antennae detecting photons by GPCR rhodopsin. Rhodopsin recognizes incoming light and activates its G protein, transducin, which is composed of three subunits α, ß, and γ. Similar to all G protein γ subunits, the transducin Gγ1 subunit undergoes C-terminal prenylation resulting in the addition of an isoprenoid farnesyl; however, the significance of this posttranslational modification is unclear. To study the role of the farnesyl group, we genetically introduced a mutant Gγ1 that lacked the prenylation site into the retinal photoreceptors of mice. The biochemical and physiological analyses of these mice revealed that mutant Gγ1 dimerizes with the endogenous transducin Gß1 subunit and that the resulting Gßγ dimers display reduced hydrophobicity. Although mutant Gßγ dimers could form a heterotrimeric G protein, they could not mediate phototransduction. This deficiency was due to a strong exclusion of non-farnesylated Gßγ complexes from the cilia (rod outer segments). Our results provide the first evidence that farnesylation is required for trafficking of G-protein ßγ subunits to the cilium of rod photoreceptors.

11.
J Biol Chem ; 291(44): 23036-23046, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27613864

ABSTRACT

Progressive rod-cone degeneration (PRCD) is a photoreceptor outer segment (OS) disc-specific protein with unknown function that is associated with retinitis pigmentosa (RP). The most common mutation in PRCD linked with severe RP phenotype is substitution of the only cysteine to tyrosine (C2Y). In this study, we find that PRCD is post-translationally modified by a palmitoyl lipid group at the cysteine residue linked with RP. Disrupting PRCD palmitoylation either chemically or by genetically eliminating the modified cysteine dramatically affects the stability of PRCD. Furthermore, in vivo electroporation of PRCD C2Y mutant in the mouse retina demonstrates that the palmitoylation of PRCD is important for its proper localization in the photoreceptor OS. Mutant PRCD C2Y was found in the inner segment in contrast to normal localization of WT PRCD in the OS. Our results also suggest that zDHHC3, a palmitoyl acyltransferase (PAT), catalyzes the palmitoylation of PRCD in the Golgi compartment. In conclusion, we find that the palmitoylation of PRCD is crucial for its trafficking to the photoreceptor OS and mislocalization of this protein likely leads to RP-related phenotypes.


Subject(s)
Eye Proteins/chemistry , Eye Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Retinitis Pigmentosa/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Amino Acid Motifs , Animals , Eye Proteins/genetics , Humans , Lipoylation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Protein Stability , Protein Transport , Retinitis Pigmentosa/enzymology , Retinitis Pigmentosa/genetics
12.
J Neurosci ; 36(18): 5107-14, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27147662

ABSTRACT

UNLABELLED: Retinal neurons use multiple strategies to fine-tune visual signal transduction, including post-translational modifications of proteins, such as addition of an isoprenyl lipid to a carboxyl-terminal cysteine in proteins that terminate with a "CAAX motif." We previously showed that RAS converting enzyme 1 (RCE1)-mediated processing of isoprenylated proteins is required for photoreceptor maintenance and function. However, it is not yet known whether the requirement for the RCE1-mediated protein processing is related to the absence of the endoproteolytic processing step, the absence of the subsequent methylation step by isoprenylcysteine methyltransferase (ICMT), or both. To approach this issue and to understand the significance of protein methylation, we generated mice lacking Icmt expression in the retina. In the absence of Icmt expression, rod and cone light-mediated responses diminished progressively. Lack of ICMT-mediated methylation led to defective association of isoprenylated transducin and cone phosphodiesterase 6 (PDE6α') with photoreceptor membranes and resulted in decreased levels of transducin, PDE6α', and cone G-protein coupled receptor kinase-1 (GRK1). In contrast to our earlier findings with retina-specific Rce1 knock-out mice, rod PDE6 in Icmt-deficient mice trafficked normally to the photoreceptor outer segment, suggesting that the failure to remove the -AAX is responsible for blocking the movement of PDE6 to the outer segment. Our findings demonstrate that carboxyl methylation of isoprenylated proteins is crucial for maintenance of photoreceptor function. SIGNIFICANCE STATEMENT: In this report, we show that an absence of isoprenylcysteine methyltransferase-mediated protein methylation leads to progressive loss of vision. Photoreceptors also degenerate, although at a slower pace than the rate of visual loss. The reduction in photoresponses is due to defective association of crucial players in phototransduction cascade. Unlike the situation with RCE1 deficiency, where both methylation and removal of -AAX were affected, the transport of isoprenylated proteins in isoprenylcysteine methyltransferase-deficient retinas was not dependent on methylation. This finding implies that the retention of the -AAX in PDE6 catalytic subunits in Rce1(-/-) mice is responsible for impeding their transport to the rod photoreceptor outer segment. In conclusion, lack of methylation of isoprenylcysteines leads to age-dependent photoreceptor dysfunction.


Subject(s)
Photoreceptor Cells, Vertebrate , Protein Methyltransferases/deficiency , Protein Methyltransferases/genetics , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Electroretinography , Light Signal Transduction , Mice , Mice, Knockout , Neurons/physiology , Protein Processing, Post-Translational/genetics , Retina/cytology , Retina/metabolism , Retina/physiopathology , Retinal Photoreceptor Cell Outer Segment , Vision Disorders/genetics , Vision Disorders/physiopathology
13.
Methods Mol Biol ; 1421: 269-86, 2016.
Article in English | MEDLINE | ID: mdl-26965271

ABSTRACT

In vivo alternative splicing is controlled in a tissue and cell type specific manner. Often individual cellular components of complex tissues will express different splicing programs. Thus, when studying splicing in multicellular organisms it is critical to determine the exon inclusion levels in individual cells positioned in the context of their native tissue or organ. Here we describe how a fluorescent splicing reporter in combination with in vivo electroporation can be used to visualize alternative splicing in individual cells within mature tissues. In a test case we show how the splicing of a photoreceptor specific exon can be visualized within the mouse retina. The retina was chosen as an example of a complex tissue that is fragile and whose cells cannot be studied in culture. With minor modifications to the injection and electroporation procedure, the protocol we outline can be applied to other tissues and organs.


Subject(s)
Alternative Splicing , Electroporation/methods , Microscopy, Fluorescence/methods , RNA Precursors/genetics , Retina/metabolism , Animals , Dissection/methods , Exons , Genes, Reporter , Mice , Polymerase Chain Reaction/methods , RNA Precursors/isolation & purification , Retina/cytology
14.
Mol Cell Biol ; 35(10): 1860-70, 2015 May.
Article in English | MEDLINE | ID: mdl-25776555

ABSTRACT

Bardet-Biedl syndrome (BBS) is a genetic disorder affecting multiple systems and organs in the body. Several mutations in genes associated with BBS affect only photoreceptor cells and cause nonsyndromic retinitis pigmentosa (RP), raising the issue of why certain mutations manifest as a systemic disorder whereas other changes in the same gene affect only a specific cell type. Here, we show that cell-type-specific alternative splicing is responsible for confining the phenotype of the A-to-G substitution in the 3' splice site of BBS8 exon 2A (IVS1-2A>G mutation) in the BBS8 gene to photoreceptor cells. The IVS1-2A>G mutation leads to missplicing of BBS8 exon 2A, producing a frameshift in the BBS8 reading frame and thus eliminating the protein specifically in photoreceptor cells. Cell types other than photoreceptors skip exon 2A from the mature BBS8 transcript, which renders them immune to the mutation. We also show that the splicing of Bbs8 exon 2A in photoreceptors is directed exclusively by redundant splicing enhancers located in the adjacent introns. These intronic sequences are sufficient for photoreceptor-cell-specific splicing of heterologous exons, including an exon with a randomized sequence.


Subject(s)
Alternative Splicing , Cornea/metabolism , Microtubule-Associated Proteins/genetics , Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Animals , Animals, Newborn/anatomy & histology , Animals, Newborn/genetics , Cytoskeletal Proteins , Enhancer Elements, Genetic , Exons , Frameshift Mutation , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Organ Specificity , Retinitis Pigmentosa/pathology
15.
Invest Ophthalmol Vis Sci ; 55(5): 3081-92, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24736053

ABSTRACT

PURPOSE: Mutations in the photoreceptor cell-specific gene encoding aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) lead to Leber congenital amaurosis (LCA4), retinitis pigmentosa, and cone-rod dystrophy. Gene therapy appears to be promising in the treatment for AIPL1-mediated vision loss in humans. Prior to initiating these treatments, however, it is crucial to understand how the retinal neurons remodel themselves in response to photoreceptor cell degeneration. In this study, using an animal model for AIPL1-LCA, Aipl1(-/-) mice, we investigate the changes in postreceptoral retinal neurons during the course of photoreceptor cell loss. METHODS: Morphology of the Aipl1(-/-) retina from postnatal day 8 to 150 was compared to that of age-matched, wild-type C57Bl6/J retina (WT) by immunocytochemistry using cell-specific markers. RESULTS: Expression of postsynaptic proteins in bipolar cells is reduced prior to photoreceptor cell degeneration at postnatal day 8. Bipolar and horizontal cells retract their dendrites. Cell bodies and axons of bipolar and horizontal cells are disorganized during the course of degeneration. Müller cell processes become hypertrophic and form a dense fibrotic layer outside the inner nuclear layer. CONCLUSIONS: An early defect in photoreceptor cells in the AIPL1-LCA mouse model affects the expression of postsynaptic markers, suggesting abnormal development of bipolar synapses. Once degeneration of photoreceptor cells is initiated, remodeling of retinal neurons in the Aipl1(-/-) animal is rapid.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Retinal Diseases/metabolism , Retinal Neurons/metabolism , Alcohol Oxidoreductases , Animals , Biomarkers/metabolism , Co-Repressor Proteins , DNA-Binding Proteins/metabolism , Disease Models, Animal , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Phosphoproteins/metabolism , Retinal Diseases/genetics , Retinal Diseases/pathology , Retinal Neurons/pathology , Synaptic Membranes/metabolism
16.
Adv Exp Med Biol ; 801: 43-8, 2014.
Article in English | MEDLINE | ID: mdl-24664679

ABSTRACT

Mutations in Aryl hydrocarbon receptor interacting protein like-1 (AIPL1) are linked to Leber congenital amaurosis (LCA), a severe blinding disease that occurs in early childhood. The severity of disease is due to requirement for AIPL1 in both rod and cone photoreceptor cell survival and function. Aipl1 is expressed very early during retinal development in both rods and cones. In adult primates, robust expression of Aipl1 is found in rods but not in cones. Mouse models revealed the importance of AIPL1 in stability and function of heteromeric phosphodiesterase 6 (PDE6), an enzyme needed for visual response. However, the need for AIPL1 in cone cell survival and function is not clearly understood. In this chapter, using results obtained from multiple lines of animal models, we discuss the role for AIPL1 in photoreceptors.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carrier Proteins/physiology , Eye Proteins/physiology , Leber Congenital Amaurosis/physiopathology , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/physiology , Animals , Carrier Proteins/genetics , Cell Survival/physiology , Disease Models, Animal , Eye Proteins/genetics , Humans , Leber Congenital Amaurosis/pathology , Mice
17.
Hum Mol Genet ; 23(4): 1002-12, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24108108

ABSTRACT

Defects in the photoreceptor-specific gene encoding aryl hydrocarbon receptor interacting protein like-1 (AIPL1) are linked to blinding diseases, including Leber congenital amaurosis (LCA) and cone dystrophy. While it is apparent that AIPL1 is needed for rod and cone function, the role of AIPL1 in cones is not clear. In this study, using an all-cone animal model lacking Aipl1, we show a light-independent degeneration of M- and S-opsin containing cones that proceeds in a ventral-to-dorsal gradient. Aipl1 is needed for stability, assembly and membrane association of cone PDE6, an enzyme crucial for photoreceptor function and survival. Furthermore, RetGC1, a protein linked to LCA that is needed for cGMP synthesis, was dramatically reduced in cones lacking Aipl1. A defect in RetGC1 is supported by our finding that cones lacking Aipl1 exhibited reduced levels of cGMP. These findings are in contrast to the role of Aipl1 in rods, where destabilization of rod PDE6 results in an increase in cGMP levels, which drives rapid rod degeneration. Our results illustrate mechanistic differences behind the death of rods and cones in retinal degenerative disease caused by deficiencies in AIPL1.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Blindness/enzymology , Cyclic GMP/metabolism , Leber Congenital Amaurosis/enzymology , Retinal Cone Photoreceptor Cells/enzymology , Adaptor Proteins, Signal Transducing/deficiency , Animals , Catalytic Domain , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Enzyme Stability , Guanylate Cyclase/metabolism , Humans , Mice , Mice, Knockout , Protein Transport , Receptors, Cell Surface/metabolism , Retina/pathology
18.
Proc Natl Acad Sci U S A ; 110(30): 12468-73, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23836670

ABSTRACT

In rod photoreceptors, several phototransduction components display light-dependent translocation between cellular compartments. Notably, the G protein transducin translocates from rod outer segments to inner segments/spherules in bright light, but the functional consequences of translocation remain unclear. We generated transgenic mice where light-induced transducin translocation is impaired. These mice exhibited slow photoreceptor degeneration, which was prevented if they were dark-reared. Physiological recordings showed that control and transgenic rods and rod bipolar cells displayed similar sensitivity in darkness. After bright light exposure, control rods were more strongly desensitized than transgenic rods. However, in rod bipolar cells, this effect was reversed; transgenic rod bipolar cells were more strongly desensitized than control. This sensitivity reversal indicates that transducin translocation in rods enhances signaling to rod bipolar cells. The enhancement could not be explained by modulation of inner segment conductances or the voltage sensitivity of the synaptic Ca(2+) current, suggesting interactions of transducin with the synaptic machinery.


Subject(s)
Retinal Rod Photoreceptor Cells/physiology , Synaptic Transmission/physiology , Transducin/metabolism , Animals , Darkness , Light , Mice , Mice, Transgenic , Protein Transport , Retinal Degeneration/prevention & control , Retinal Rod Photoreceptor Cells/cytology , Transducin/physiology
19.
J Neurosci ; 33(29): 11745-53, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23864662

ABSTRACT

Phosphodiesterase-6 (PDE6) is the key effector enzyme of the vertebrate phototransduction pathway in rods and cones. Rod PDE6 catalytic core is composed of two distinct subunits, PDE6α and PDE6ß, whereas two identical PDE6α' subunits form the cone PDE6 catalytic core. It is not known whether this difference in PDE6 catalytic subunit identity contributes to the functional differences between rods and cones. To address this question, we expressed cone PDE6α' in the photoreceptor cells of the retinal degeneration 10 (rd10) mouse that carries a mutation in rod PDEß subunit. We show that adeno-associated virus-mediated subretinal delivery of PDE6α' rescues rod electroretinogram responses and preserves retinal structure, indicating that cone PDE6α' can couple effectively to the rod phototransduction pathway. We also show that restoration of light sensitivity in rd10 rods is attributable to assembly of PDE6α' with rod PDE6γ. Single-cell recordings revealed that, surprisingly, rods expressing cone PDE6α' are twofold more sensitive to light than wild-type rods, most likely because of the slower shutoff of their light responses. Unlike in wild-type rods, the response kinetics in PDE6α'-treated rd10 rods accelerated with increasing flash intensity, indicating a possible direct feedback modulation of cone PDE6α' activity. Together, these results demonstrate that cone PDE6α' can functionally substitute for rod PDEαß in vivo, conferring treated rods with distinct physiological properties.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Catalytic Domain , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Gene Transfer Techniques , Mice , Mice, Knockout , Patch-Clamp Techniques , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism
20.
J Biol Chem ; 286(38): 33252-9, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21799013

ABSTRACT

Rod and cone photoreceptor neurons utilize discrete PDE6 enzymes that are crucial for phototransduction. Rod PDE6 is composed of heterodimeric catalytic subunits (αß), while the catalytic core of cone PDE6 (α') is a homodimer. It is not known if variations between PDE6 subunits preclude rod PDE6 catalytic subunits from coupling to the cone phototransduction pathway. To study this issue, we generated a cone-dominated mouse model lacking cone PDE6 (Nrl(-/-) cpfl1). In this animal model, using several independent experimental approaches, we demonstrated the expression of rod PDE6 (αß) and the absence of cone PDE6 (α') catalytic subunits. The rod PDE6 enzyme expressed in cone cells is active and contributes to the hydrolysis of cGMP in response to light. In addition, rod PDE6 expressed in cone cells couples to the light signaling pathway to produce S-cone responses. However, S-cone responses and light-dependent cGMP hydrolysis were eliminated when the ß-subunit of rod PDE6 was removed (Nrl(-/-) cpfl1 rd). We conclude that either rod or cone PDE6 can effectively couple to the cone phototransduction pathway to mediate visual signaling. Interestingly, we also found that functional PDE6 is required for trafficking of M-opsin to cone outer segments.


Subject(s)
Catalytic Domain , Retinal Cone Photoreceptor Cells/enzymology , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/enzymology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Electroretinography , Gene Expression Regulation, Enzymologic , Light , Mice , Models, Animal , Opsins/metabolism , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...