Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
PLoS Genet ; 18(9): e1010154, 2022 09.
Article in English | MEDLINE | ID: mdl-36074756

ABSTRACT

Centrosomal protein of 164 kDa (CEP164) is located at distal appendages of primary cilia and is necessary for basal body (BB) docking to the apical membrane. To investigate the function of photoreceptor CEP164 before and after BB docking, we deleted CEP164 during retina embryonic development (Six3Cre), in postnatal rod photoreceptors (iCre75) and in mature retina using tamoxifen induction (Prom1-ETCre). BBs dock to the cell cortex during postnatal day 6 (P6) to extend a connecting cilium (CC) and an axoneme. P6 retina-specific knockouts (retCep164-/-) are unable to dock BBs, thereby preventing formation of CC or outer segments (OSs). In rod-specific knockouts (rodCep164-/-), Cre expression starts after P7 and CC/OS form. P16 rodCep164-/- rods have nearly normal OS lengths, and maintain OS attachment through P21 despite loss of CEP164. Intraflagellar transport components (IFT88, IFT57 and IFT140) were reduced at P16 rodCep164-/- BBs and CC tips and nearly absent at P21, indicating impaired intraflagellar transport. Nascent OS discs, labeled with a fluorescent dye on P14 and P18 and harvested on P19, showed continued rodCep164-/- disc morphogenesis but absence of P14 discs mid-distally, indicating OS instability. Tamoxifen induction with PROM1ETCre;Cep164F/F (tamCep164-/-) adult mice affected maintenance of both rod and cone OSs. The results suggest that CEP164 is key towards recruitment and stabilization of IFT-B particles at the BB/CC. IFT impairment may be the main driver of ciliary malfunction observed with hypomorphic CEP164 mutations.


Subject(s)
Basal Bodies , Fluorescent Dyes , Animals , Basal Bodies/metabolism , Cilia/metabolism , Fluorescent Dyes/metabolism , Mice , Protein Transport/genetics , Retinal Cone Photoreceptor Cells , Tamoxifen
2.
Exp Eye Res ; 229: 109429, 2023 04.
Article in English | MEDLINE | ID: mdl-36863431

ABSTRACT

The macular carotenoids lutein and zeaxanthin are taken up from the bloodstream into the human retina through a selective process, for which the HDL cholesterol receptor scavenger receptor BI (SR-BI) in the cells of retinal pigment epithelium (RPE) is thought to be a key mediator. However, the mechanism of SR-BI-mediated selective uptake of macular carotenoids is still not fully understood. Here, we investigate possible mechanisms using biological assays and cultured HEK293 cells, a cell line without endogenous SR-BI expression. Binding affinities between SR-BI and various carotenoids were measured by surface plasmon resonance (SPR) spectroscopy, which shows that SR-BI cannot bind lutein or zeaxanthin specifically. Overexpression of SR-BI in HEK293 cells results in more lutein and zeaxanthin taken up than ß-carotene, and this effect can be eliminated by an SR-BI mutant (C384Y) whose cholesterol uptake tunnel is blocked. Next, we determined the effects of HDL and hepatic lipase (LIPC), SR-BI's partners in HDL cholesterol transport, on SR-BI-mediated carotenoid uptake. HDL addition dramatically reduced lutein, zeaxanthin, and ß-carotene in HEK293 cells expressing SR-BI, but the cellular lutein and zeaxanthin are higher than ß-carotene. LIPC addition increases the uptake of all three carotenoids in HDL-treated cells, and promotes the transport of lutein and zeaxanthin better than ß-carotene. Our results suggest that SR-BI and its HDL cholesterol partner HDL and LIPC may be involved in the selective uptake of macular carotenoids.


Subject(s)
Carotenoids , Lutein , Humans , beta Carotene , Carotenoids/metabolism , CD36 Antigens , Cholesterol , Cholesterol, HDL/metabolism , HEK293 Cells , Lutein/pharmacology , Receptors, Scavenger/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Zeaxanthins
3.
Proc Natl Acad Sci U S A ; 117(22): 12352-12358, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32409609

ABSTRACT

Lutein and zeaxanthin are xanthophyll carotenoids that are highly concentrated in the human macula, where they protect the eye from oxidative damage and improve visual performance. Distinguishing lutein from zeaxanthin in images of the human retina in vivo or in donor eye tissues has been challenging because no available technology has been able to reliably differentiate between these two carotenoids, which differ only in the position of one C = C bond. Here, we report the differential distributions of lutein and zeaxanthin in human donor retinas mapped with confocal resonance Raman microscopy. Zeaxanthin is highly concentrated in the fovea, extending from the inner to the outer limiting membranes, with especially high concentrations in the outer plexiform layer, while lutein is much more diffuse at relatively lower concentration. Our results imply that zeaxanthin may play a more important role than lutein in human macular health and disease.


Subject(s)
Lutein/analysis , Retina/chemistry , Zeaxanthins/analysis , Humans , Microscopy, Confocal/methods , Xanthophylls/analysis
4.
J Biol Chem ; 296: 100529, 2021.
Article in English | MEDLINE | ID: mdl-33711342

ABSTRACT

INPP5E, also known as pharbin, is a ubiquitously expressed phosphatidylinositol polyphosphate 5-phosphatase that is typically located in the primary cilia and modulates the phosphoinositide composition of membranes. Mutations to or loss of INPP5E is associated with ciliary dysfunction. INPP5E missense mutations of the phosphatase catalytic domain cause Joubert syndrome in humans-a syndromic ciliopathy affecting multiple tissues including the brain, liver, kidney, and retina. In contrast to other primary cilia, photoreceptor INPP5E is prominently expressed in the inner segment and connecting cilium and absent in the outer segment, which is a modified primary cilium dedicated to phototransduction. To investigate how loss of INPP5e causes retina degeneration, we generated mice with a retina-specific KO (Inpp5eF/F;Six3Cre, abbreviated as retInpp5e-/-). These mice exhibit a rapidly progressing rod-cone degeneration resembling Leber congenital amaurosis that is nearly completed by postnatal day 21 (P21) in the central retina. Mutant cone outer segments contain vesicles instead of discs as early as P8. Although P10 mutant outer segments contain structural and phototransduction proteins, axonemal structure and disc membranes fail to form. Connecting cilia of retInpp5e-/- rods display accumulation of intraflagellar transport particles A and B at their distal ends, suggesting disrupted intraflagellar transport. Although INPP5E ablation may not prevent delivery of outer segment-specific proteins by means of the photoreceptor secretory pathway, its absence prevents the assembly of axonemal and disc components. Herein, we suggest a model for INPP5E-Leber congenital amaurosis, proposing how deletion of INPP5E may interrupt axoneme extension and disc membrane elaboration.


Subject(s)
Axoneme/pathology , Morphogenesis , Phosphoric Monoester Hydrolases/physiology , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/pathology , Animals , Axoneme/metabolism , Eye Proteins/physiology , Mice , Mice, Knockout , Protein Transport , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/etiology , Retinal Rod Photoreceptor Cells/metabolism
5.
Arch Biochem Biophys ; 716: 109111, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34942193

ABSTRACT

Supplementation with antioxidant carotenoids is a therapeutic strategy to protect against age-related macular degeneration (AMD); however, the transport mechanism of carotenoids from the liver to the retina is still not fully understood. Here, we investigate if HDL serves as the primary transporter for the macular carotenoids. ApoA-I, the key apolipoprotein of HDL, was genetically deleted from BCO2 knockout (Bco2-/-) mice, a macular pigment mouse model capable of accumulating carotenoids in the retina. We then conducted a feeding experiment with a mixed carotenoid chow (lutein:zeaxanthin:ß-carotene = 1:1:1) for one month. HPLC data demonstrated that the total carotenoids were increased in the livers but decreased in the serum, retinal pigment epithelium (RPE)/choroids, and retinas of ApoA-I-/-/Bco2-/- mice compared to Bco2-/- mice. In detail, ApoA-I deficiency caused a significant increase of ß-carotene but not lutein and zeaxanthin in the liver, decreased all three carotenoids in the serum, blocked the majority of zeaxanthin and ß-carotene transport to the RPE/choroid, and dramatically reduced ß-carotene and zeaxanthin but not lutein in the retina. Furthermore, surface plasmon resonance spectroscopy (SPR) data showed that the binding affinity between ApoA-I and ß-carotene â‰« zeaxanthin > lutein. Our results show that carotenoids are transported from the liver to the eye mainly by HDL, and ApoA-I may be involved in the selective delivery of macular carotenoids to the RPE.


Subject(s)
Apolipoprotein A-I/genetics , Carotenoids/metabolism , Dioxygenases/genetics , Lipoproteins, HDL2/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Carotenoids/blood , Disease Models, Animal , Humans , Liver , Lutein/metabolism , Macular Degeneration/metabolism , Mice , Mice, Knockout , Retina , Zeaxanthins/metabolism , beta Carotene/metabolism
6.
J Biol Chem ; 294(11): 3957-3973, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30647131

ABSTRACT

Centrins (CETN1-4) are ubiquitous and conserved EF-hand-family Ca2+-binding proteins associated with the centrosome, basal body, and transition zone. Deletion of CETN1 or CETN2 in mice causes male infertility or dysosmia, respectively, without affecting photoreceptor function. However, it remains unclear to what extent centrins are redundant with each other in photoreceptors. Here, to explore centrin redundancy, we generated Cetn3GT/GT single-knockout and Cetn2-/-;Cetn3GT/GT double-knockout mice. Whereas the Cetn3 deletion alone did not affect photoreceptor function, simultaneous ablation of Cetn2 and Cetn3 resulted in attenuated scotopic and photopic electroretinography (ERG) responses in mice at 3 months of age, with nearly complete retina degeneration at 1 year. Removal of CETN2 and CETN3 activity from the lumen of the connecting cilium (CC) destabilized the photoreceptor axoneme and reduced the CC length as early as postnatal day 22 (P22). In Cetn2-/-;Cetn3GT/GT double-knockout mice, spermatogenesis-associated 7 (SPATA7), a key organizer of the photoreceptor-specific distal CC, was depleted gradually, and CETN1 was condensed to the mid-segment of the CC. Ultrastructural analysis revealed that in this double knockout, the axoneme of the CC expanded radially at the distal end, with vertically misaligned outer segment discs and membrane whorls. These observations suggest that CETN2 and CETN3 cooperate in stabilizing the CC/axoneme structure.


Subject(s)
Axoneme/metabolism , Calcium-Binding Proteins/metabolism , Cilia/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Animals , Calcium-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Biol Chem ; 401(5): 573-584, 2020 04 28.
Article in English | MEDLINE | ID: mdl-31811799

ABSTRACT

Photoreceptors are polarized neurons, with specific subcellular compartmentalization and unique requirements for protein expression and trafficking. Each photoreceptor contains an outer segment (OS) where vision begins, an inner segment (IS) where protein synthesis occurs and a synaptic terminal for signal transmission to second-order neurons. The OS is a large, modified primary cilium attached to the IS by a slender connecting cilium (CC), the equivalent of the transition zone (TZ). Daily renewal of ~10% of the OS requires massive protein biosynthesis in the IS with reliable transport and targeting pathways. Transport of lipidated ('sticky') proteins depends on solubilization factors, phosphodiesterase δ (PDEδ) and uncoordinated protein-119 (UNC119), and the cargo dispensation factor (CDF), Arf-like protein 3-guanosine triphosphate (ARL3-GTP). As PDE6 and transducin still reside prominently in the OS of PDEδ and UNC119 germline knockout mice, respectively, we propose the existence of an alternate trafficking pathway, whereby lipidated proteins migrate in rhodopsin-containing vesicles of the secretory pathway.


Subject(s)
Lipid Metabolism , Photoreceptor Cells/metabolism , Animals , Diffusion , Humans , Protein Transport
8.
J Biol Chem ; 293(45): 17546-17558, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30228185

ABSTRACT

RAB28, a member of the RAS oncogene family, is a ubiquitous, farnesylated, small GTPase of unknown function present in photoreceptors and the retinal pigmented epithelium (RPE). Nonsense mutations of the human RAB28 gene cause recessive cone-rod dystrophy 18 (CRD18), characterized by macular hyperpigmentation, progressive loss of visual acuity, RPE atrophy, and severely attenuated cone and rod electroretinography (ERG) responses. In an attempt to elucidate the disease-causing mechanism, we generated Rab28-/- mice by deleting exon 3 and truncating RAB28 after exon 2. We found that Rab28-/- mice recapitulate features of the human dystrophy (i.e. they exhibited reduced cone and rod ERG responses and progressive retina degeneration). Cones of Rab28-/- mice extended their outer segments (OSs) to the RPE apical processes and formed enlarged, balloon-like distal tips before undergoing degeneration. The visual pigment content of WT and Rab28-/- cones was comparable before the onset of degeneration. Cone phagosomes were almost absent in Rab28-/- mice, whereas rod phagosomes displayed normal levels. A protein-protein interaction screen identified several RAB28-interacting proteins, including the prenyl-binding protein phosphodiesterase 6 δ-subunit (PDE6D) and voltage-gated potassium channel subfamily J member 13 (KCNJ13) present in the RPE apical processes. Of note, the loss of PDE6D prevented delivery of RAB28 to OSs. Taken together, these findings reveal that RAB28 is required for shedding and phagocytosis of cone OS discs.


Subject(s)
Phagocytosis , Retinal Cone Photoreceptor Cells/enzymology , Retinal Pigment Epithelium/enzymology , rab GTP-Binding Proteins/metabolism , Animals , Cone-Rod Dystrophies/enzymology , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/pathology , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Mice , Mice, Knockout , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Pigment Epithelium/pathology , Retinal Rod Photoreceptor Cells/enzymology , Retinal Rod Photoreceptor Cells/pathology , rab GTP-Binding Proteins/genetics
9.
J Biol Chem ; 292(52): 21442-21456, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29089384

ABSTRACT

Arf-like protein 13b (ARL13b) is a small GTPase that functions as a guanosine nucleotide exchange factor (GEF) for ARL3-GDP. ARL13b is located exclusively in photoreceptor outer segments (OS) presumably anchored to discs by palmitoylation, whereas ARL3 is an inner segment cytoplasmic protein. Hypomorphic mutations affecting the ARL13b G-domain inactivate GEF activity and lead to Joubert syndrome (JS) in humans. However, the molecular mechanisms in ARL13b mutation-induced Joubert syndrome, particularly the function of primary cilia, are still incompletely understood. Because Arl13b germline knockouts in mouse are lethal, we generated retina-specific deletions of ARL13b in which ARL3-GTP formation is impaired. In mouse retArl13b-/- central retina at postnatal day 6 (P6) and older, outer segments were absent, thereby preventing trafficking of outer segment proteins to their destination. Ultrastructure of postnatal day 10 (P10) central retArl13b-/- photoreceptors revealed docking of basal bodies to cell membranes, but mature transition zones and disc structures were absent. Deletion of ARL13b in adult mice via tamoxifen-induced Cre/loxP recombination indicated that axonemes gradually shorten and outer segments progressively degenerate. IFT88, essential for anterograde intraflagellar transport (IFT), was significantly reduced at tamArl13b-/- basal bodies, suggesting impairment of intraflagellar transport. AAV2/8 vector-mediated ARL13b expression in the retArl13b-/- retina rescued ciliogenesis.


Subject(s)
ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/physiology , Photoreceptor Cells/ultrastructure , ADP-Ribosylation Factors/genetics , Abnormalities, Multiple , Animals , Axoneme/metabolism , Basal Bodies/metabolism , Cell Membrane/metabolism , Cerebellum/abnormalities , Cilia/metabolism , Crystallography, X-Ray/methods , Eye Abnormalities , Guanine Nucleotide Exchange Factors/metabolism , Kidney Diseases, Cystic , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Knockout , Photoreceptor Cells/metabolism , Protein Transport , Retina/abnormalities , Retina/metabolism , Retina/physiology
10.
Mol Vis ; 24: 834-846, 2018.
Article in English | MEDLINE | ID: mdl-30713422

ABSTRACT

Purpose: Recessive mutations in the human IQCB1/NPHP5 gene are associated with Senior-Løken syndrome (SLS), a ciliopathy presenting with nephronophthisis and Leber congenital amaurosis (LCA). Nphp5-knockout mice develop LCA without nephronophthisis. Mutant rods rapidly degenerate while mutant cones survive for months. The purpose of this study was to reinitiate cone ciliogenesis in a Nphp5 -/-; Nrl -/- mouse with viral expression of full-length NPHP5 and rescue function. Methods: Nphp5 -/- mice were mated with Nrl -/- mice to generate Nphp5-/-; Nrl-/- double-knockouts. Nphp5-/-; Nrl-/- mice and Nphp5+/-; Nrl-/- controls were phenotyped with confocal microscopy from postnatal day 10 (P10) until 6 months of age. Nphp5-/-; Nrl-/- mice and Nphp5+/-; Nrl-/- controls were injected at P15 with self-complementary adenoassociated virus 8 (Y733F) (AAV8(Y733F)) expressing GRK1-FL-cNPHP5. Expression of mutant NPHP5 was verified with confocal microscopy and electroretinography (ERG). Results: In the Nphp5 -/- and cone-only Nphp5 -/-; Nrl -/- mice, cone outer segments did not form, but mutant cones continued to express cone pigments in the inner segments without obvious signs of cone cell death. The mutant cone outer nuclear layer (ONL) and the inner segments were stable for more than 6 months in the cone-only Nphp5 -/-; Nrl -/- retinas. Viral expression of NPHP5 initiated after eye opening showed that connecting cilia and RP1-positive axonemes were formed. Furthermore, cone pigments and other cone outer segment proteins (cone transducin and cone PDE6) were present in the nascent mutant cone outer segments, and rescued mutant cones exhibited a significant photopic b-wave (30% of Nphp5 +/-; Nrl -/- controls). Conclusions: Nphp5-/-; Nrl-/- cones persistently express cone pigments in the inner segments without obvious degeneration, providing an extended duration interval for viral gene expression. Viral expression of full-length NPHP5 initiates ciliogenesis between P15 and P60, and mutant cones are, in part, functional, encouraging future retina gene replacement therapy.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Calmodulin-Binding Proteins/genetics , Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Retinal Cone Photoreceptor Cells/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Amino Acid Sequence , Animals , Axoneme/metabolism , Axoneme/ultrastructure , Basic-Leucine Zipper Transcription Factors/deficiency , Calmodulin-Binding Proteins/deficiency , Cilia/metabolism , Cilia/ultrastructure , Crosses, Genetic , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Disease Models, Animal , Eye Proteins/metabolism , Female , G-Protein-Coupled Receptor Kinase 1/genetics , G-Protein-Coupled Receptor Kinase 1/metabolism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Gene Expression Regulation , Genetic Therapy/methods , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phenotype , Retinal Cone Photoreceptor Cells/pathology , Sequence Alignment , Sequence Homology, Amino Acid , Transducin/genetics , Transducin/metabolism
11.
Arch Biochem Biophys ; 649: 22-28, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29742455

ABSTRACT

Carotenoid supplementation can improve human visual performance, but there is still no validated rodent model to test their effects on visual function in laboratory animals. We recently showed that mice deficient in ß-carotene oxygenase 2 (BCO2) and/or ß-carotene oxygenase 1 (BCO1) enzymes can accumulate carotenoids in their retinas, allowing us to investigate the effects of carotenoids on the visual performance of mice. Using OptoMotry, a device to measure visual function in rodents, we examined the effect of zeaxanthin, lutein, and ß-carotene on visual performance of various BCO knockout mice. We then transgenically expressed the human zeaxanthin-binding protein GSTP1 (hGSTP1) in the rods of bco2-/- mice to examine if delivering more zeaxanthin to retina will improve their visual function further. The visual performance of bco2-/- mice fed with zeaxanthin or lutein was significantly improved relative to control mice fed with placebo beadlets. ß-Carotene had no significant effect in bco2-/- mice but modestly improved cone visual function of bco1-/- mice. Expression of hGSTP1 in the rods of bco2-/-mice resulted in a 40% increase of retinal zeaxanthin and further improvement of visual performance. This work demonstrates that these "macular pigment mice" may serve as animal models to study carotenoid function in the retina.


Subject(s)
Carotenoids/pharmacology , Functional Food , Retina/drug effects , Vision, Ocular/drug effects , Animals , Female , Functional Food/analysis , Glutathione S-Transferase pi/genetics , Humans , Lutein/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Retina/physiology , Zeaxanthins/pharmacology , beta Carotene/pharmacology , beta-Carotene 15,15'-Monooxygenase/genetics
12.
Adv Exp Med Biol ; 1074: 317-325, 2018.
Article in English | MEDLINE | ID: mdl-29721959

ABSTRACT

UNC119 and PDEδ are lipid-binding proteins and are thought to form diffusible complexes with transducin-α and prenylated OS proteins, respectively, to mediate their trafficking to photoreceptor outer segments. Here, we investigate mechanisms of trafficking which are controlled by Arf-like protein 3 (Arl3), a small GTPase. The activity of ARL3 is regulated by a GEF (ARL13b) and a GAP (RP2). In a mouse germline knockout of RP2, ARL3-GTP is abundant as its intrinsic GTPase activity is extremely low. High levels of ARL3-GTP impair binding and trafficking of cargo to the outer segment. Germline knockout of ARL3 is embryonically lethal generating a syndromic ciliopathy-like phenotype. Retina- and rod-specific knockout of ARL3 allow to determine the precise mechanisms leading to photoreceptor degeneration. The knockouts reveal binary functions of ARL3-GTP as a key molecule in late-stage photoreceptor ciliogenesis and cargo displacement factor.


Subject(s)
ADP-Ribosylation Factors/physiology , Protein Transport/physiology , ADP-Ribosylation Factors/deficiency , ADP-Ribosylation Factors/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/metabolism , Cone-Rod Dystrophies/pathology , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , GTP-Binding Proteins , Genes, Lethal , Guanosine Triphosphate/metabolism , Lipoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Organ Specificity , Protein Prenylation , Pyrophosphatases/deficiency , Pyrophosphatases/physiology , Rod Cell Outer Segment/metabolism
13.
J Biol Chem ; 291(13): 7142-55, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26814127

ABSTRACT

Arf-like protein 3 (ARL3) is a ubiquitous small GTPase expressed in ciliated cells of plants and animals. Germline deletion ofArl3in mice causes multiorgan ciliopathy reminiscent of Bardet-Biedl or Joubert syndromes. As photoreceptors are elegantly compartmentalized and have cilia, we probed the function of ARL3 (ADP-ribosylation factor (Arf)-like 3 protein) by generating rod photoreceptor-specific (prefix(rod)) and retina-specific (prefix(ret))Arl3deletions. In predegenerate(rod)Arl3(-/-)mice, lipidated phototransduction proteins showed trafficking deficiencies, consistent with the role of ARL3 as a cargo displacement factor for lipid-binding proteins. By contrast,(ret)Arl3(-/-)rods and cones expressing Cre recombinase during embryonic development formed neither connecting cilia nor outer segments and degenerated rapidly. Absence of cilia infers participation of ARL3 in ciliogenesis and axoneme formation. Ciliogenesis was rescued, and degeneration was reversed in part by subretinal injection of adeno-associated virus particles expressing ARL3-EGFP. The conditional knock-out phenotypes permitted identification of two ARL3 functions, both in the GTP-bound form as follows: one as a regulator of intraflagellar transport participating in photoreceptor ciliogenesis and the other as a cargo displacement factor transporting lipidated protein to the outer segment. Surprisingly, a farnesylated inositol polyphosphate phosphatase only trafficked from the endoplasmic reticulum to the Golgi, thereby excluding it from a role in photoreceptor cilia physiology.


Subject(s)
ADP-Ribosylation Factors/genetics , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , ADP-Ribosylation Factors/deficiency , Age Factors , Animals , Cilia/metabolism , Cilia/pathology , Dependovirus/genetics , Electroretinography , Embryo, Mammalian , Eye Proteins/genetics , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Integrases/genetics , Integrases/metabolism , Light Signal Transduction , Mice , Mice, Knockout , Organogenesis/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology
14.
FASEB J ; 30(10): 3400-3412, 2016 10.
Article in English | MEDLINE | ID: mdl-27328943

ABSTRACT

Null mutations in the human IQCB1/NPHP5 (nephrocystin-5) gene that encodes NPHP5 are the most frequent cause of Senior-Løken syndrome, a ciliopathy that is characterized by Leber congenital amaurosis and nephronophthisis. We generated germline Nphp5-knockout mice by placing a ß-Geo gene trap in intron 4, thereby truncating NPHP5 at Leu87 and removing all known functional domains. At eye opening, Nphp5-/- mice exhibited absence of scotopic and photopic electroretinogram responses, a phenotype that resembles Leber congenital amaurosis. Outer segment transmembrane protein accumulation in Nphp5-/- endoplasmic reticulum was evident as early as postnatal day (P)6. EGFP-CETN2, a centrosome and transition zone marker, identified basal bodies in Nphp5-/- photoreceptors, but without fully developed transition zones. Ultrastructure of P6 and 10 Nphp5-/- photoreceptors revealed aberrant transition zones of reduced diameter. Nphp5-/- photoreceptor degeneration was complete at 1 mo of age but was delayed significantly in Nphp5-/-;Nrl-/- (cone only) retina. Nphp5-/- mouse embryonic fibroblast developed normal cilia, and Nphp5-/- kidney histology at 1 yr of age showed no significant pathology. Results establish that nephrocystin-5 is essential for photoreceptor outer segment formation but is dispensable for kidney and mouse embryonic fibroblast ciliary formation.-Ronquillo, C. C., Hanke-Gogokhia, C., Revelo, M. P., Frederick, J. M., Jiang, L., Baehr, W. Ciliopathy-associated IQCB1/NPHP5 protein is required for mouse photoreceptor outer segment formation.


Subject(s)
Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Mutation/genetics , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Animals , Cilia/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Guanylate Cyclase/genetics , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/metabolism , Mice, Knockout , Optic Atrophies, Hereditary/genetics , Optic Atrophies, Hereditary/metabolism , Retinal Degeneration/metabolism
15.
Proc Natl Acad Sci U S A ; 111(28): 10173-8, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982131

ABSTRACT

The macula of the primate retina uniquely concentrates high amounts of the xanthophyll carotenoids lutein, zeaxanthin, and meso-zeaxanthin, but the underlying biochemical mechanisms for this spatial- and species-specific localization have not been fully elucidated. For example, despite abundant retinal levels in mice and primates of a binding protein for zeaxanthin and meso-zeaxanthin, the pi isoform of glutathione S-transferase (GSTP1), only human and monkey retinas naturally contain detectable levels of these carotenoids. We therefore investigated whether or not differences in expression, localization, and activity between mouse and primate carotenoid metabolic enzymes could account for this species-specific difference in retinal accumulation. We focused on ß,ß-carotene-9',10'-dioxygenase (BCO2, also known as BCDO2), the only known mammalian xanthophyll cleavage enzyme. RT-PCR, Western blot analysis, and immunohistochemistry (IHC) confirmed that BCO2 is expressed in both mouse and primate retinas. Cotransfection of expression plasmids of human or mouse BCO2 into Escherichia coli strains engineered to produce zeaxanthin demonstrated that only mouse BCO2 is an active zeaxanthin cleavage enzyme. Surface plasmon resonance (SPR) binding studies showed that the binding affinities between human BCO2 and lutein, zeaxanthin, and meso-zeaxanthin are 10- to 40-fold weaker than those for mouse BCO2, implying that ineffective capture of carotenoids by human BCO2 prevents cleavage of xanthophyll carotenoids. Moreover, BCO2 knockout mice, unlike WT mice, accumulate zeaxanthin in their retinas. Our results provide a novel explanation for how primates uniquely concentrate xanthophyll carotenoids at high levels in retinal tissue.


Subject(s)
Dioxygenases/metabolism , Eye Proteins/metabolism , Lutein/metabolism , Retina/enzymology , Xanthophylls/metabolism , Animals , Dioxygenases/genetics , Eye Proteins/genetics , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/metabolism , Humans , Lutein/genetics , Mice , Mice, Knockout , Retina/cytology , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Xanthophylls/genetics , Zeaxanthins
16.
J Biol Chem ; 290(20): 12765-78, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25825494

ABSTRACT

Anterograde intraflagellar transport (IFT) employing kinesin-2 molecular motors has been implicated in trafficking of photoreceptor outer segment proteins. We generated embryonic retina-specific (prefix "emb") and adult tamoxifen-induced (prefix "tam") deletions of KIF3a and IFT88 in adult mice to study photoreceptor ciliogenesis and protein trafficking. In (emb)Kif3a(-/-) and in (emb)Ift88(-/-) mice, basal bodies failed to extend transition zones (connecting cilia) with outer segments, and visual pigments mistrafficked. In contrast, (tam)Kif3a(-/-) and (tam)Ift88(-/-) photoreceptor axonemes disintegrated slowly post-induction, starting distally, but rhodopsin and cone pigments trafficked normally for more than 2 weeks, a time interval during which the outer segment is completely renewed. The results demonstrate that visual pigments transport to the retinal outer segment despite removal of KIF3 and IFT88, and KIF3-mediated anterograde IFT is responsible for photoreceptor transition zone and axoneme formation.


Subject(s)
Axoneme/metabolism , Kinesins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Rhodopsin/metabolism , Animals , Axoneme/genetics , Basal Bodies/metabolism , Kinesins/genetics , Mice , Mice, Knockout , Protein Transport/physiology , Retinal Cone Photoreceptor Cells/cytology , Rhodopsin/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
FASEB J ; 29(12): 4866-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26229057

ABSTRACT

In Caenorhabditis elegans, homodimeric [kinesin family (KIF) 17, osmotic avoidance abnormal-3 (OSM-3)] and heterotrimeric (KIF3) kinesin-2 motors are required to establish sensory cilia by intraflagellar transport (IFT) where KIF3 and KIF17 cooperate to build the axoneme core and KIF17 builds the distal segments. However, the function of KIF17 in vertebrates is unresolved. We expressed full-length and motorless KIF17 constructs in mouse rod photoreceptors using adeno-associated virus in Xenopus laevis rod photoreceptors using a transgene and in ciliated IMCD3 cells. We found that tagged KIF17 localized along the rod outer segment axoneme when expressed in mouse and X. laevis photoreceptors, whereas KIF3A was restricted to the proximal axoneme. Motorless KIF3A and KIF17 mutants caused photoreceptor degeneration, likely through dominant negative effects on IFT. KIF17 mutant lacking the motor domain translocated to nuclei after exposure of a C-terminal nuclear localization signal. Germ-line deletion of Kif17 in mouse did not affect photoreceptor function. A rod-specific Kif3/Kif17 double knockout mouse demonstrated that KIF17 and KIF3 do not act synergistically and did not prevent rhodopsin trafficking to rod outer segments. In summary, the nematode model of KIF3/KIF17 cooperation apparently does not apply to mouse photoreceptors in which the photosensory cilium is built exclusively by KIF3.


Subject(s)
Kinesins/physiology , Photoreceptor Cells, Vertebrate/physiology , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Kinesins/chemistry , Kinesins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , NIH 3T3 Cells , Protein Transport , Rhodopsin/metabolism , Xenopus laevis
18.
FASEB J ; 29(3): 932-42, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25422369

ABSTRACT

The retinitis pigmentosa 2 polypeptide (RP2) functions as a GTPase-activating protein (GAP) for ARL3 (Arf-like protein 3), a small GTPase. ARL3 is an effector of phosphodiesterase 6 Δ (PDE6D), a prenyl-binding protein and chaperone of prenylated protein in photoreceptors. Mutations in the human RP2 gene cause X-linked retinitis pigmentosa (XLRP) and cone-rod dystrophy (XL-CORD). To study mechanisms causing XLRP, we generated an RP2 knockout mouse. The Rp2h(-/-) mice exhibited a slowly progressing rod-cone dystrophy simulating the human disease. Rp2h(-/-) scotopic a-wave and photopic b-wave amplitudes declined at 1 mo of age and continued to decline over the next 6 mo. Prenylated PDE6 subunits and G-protein coupled receptor kinase 1 (GRK1) were unable to traffic effectively to the Rp2h(-/-) outer segments. Mechanistically, absence of RP2 GAP activity increases ARL3-GTP levels, forcing PDE6D to assume a predominantly "closed" conformation that impedes binding of lipids. Lack of interaction disrupts trafficking of PDE6 and GRK1 to their destination, the photoreceptor outer segments. We propose that hyperactivity of ARL3-GTP in RP2 knockout mice and human patients with RP2 null alleles leads to XLRP resembling recessive rod-cone dystrophy.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Eye Proteins/physiology , G-Protein-Coupled Receptor Kinase 1/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Protein Prenylation , Retinitis Pigmentosa/metabolism , Animals , Antibody Formation , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Cilia/metabolism , Electroretinography , Female , GTP-Binding Proteins , Guanosine Triphosphate/metabolism , Humans , Immunoblotting , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells, Vertebrate/metabolism , Protein Transport , Rabbits , Retinitis Pigmentosa/pathology
19.
Adv Exp Med Biol ; 854: 655-61, 2016.
Article in English | MEDLINE | ID: mdl-26427472

ABSTRACT

Arf-like proteins (ARLs) are ubiquitously expressed small G proteins of the RAS superfamily. In photoreceptors, ARL2 and ARL3 participate in the trafficking of lipidated membrane-associated proteins and colocalize in the inner segment with UNC119A and PDEδ. UNC119A and PDEδ are acyl- and prenyl-binding proteins, respectively, involved in trafficking of acylated (transducin-α subunit, nephrocystin NPHP3) and prenylated proteins (GRK1, PDE6). Germline Arl3 knockout mice do not survive beyond postnatal day 21 and display ciliary defects in multiple organs (kidney, liver and pancreas) as well as retinal degeneration. Conditional knockouts will be necessary to delineate mechanisms of protein transport in retina disease.


Subject(s)
ADP-Ribosylation Factors/metabolism , Eye Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , ADP-Ribosylation Factors/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Eye Proteins/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Mice, Knockout , Molecular Sequence Data , Protein Binding , Protein Transport , Sequence Homology, Amino Acid
20.
J Neurosci ; 34(18): 6377-88, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24790208

ABSTRACT

Centrins are ancient calmodulin-related Ca(2+)-binding proteins associated with basal bodies. In lower eukaryotes, Centrin2 (CETN2) is required for basal body replication and positioning, although its function in mammals is undefined. We generated a germline CETN2 knock-out (KO) mouse presenting with syndromic ciliopathy including dysosmia and hydrocephalus. Absence of CETN2 leads to olfactory cilia loss, impaired ciliary trafficking of olfactory signaling proteins, adenylate cyclase III (ACIII), and cyclic nucleotide-gated (CNG) channel, as well as disrupted basal body apical migration in postnatal olfactory sensory neurons (OSNs). In mutant OSNs, cilia base-anchoring of intraflagellar transport components IFT88, the kinesin-II subunit KIF3A, and cytoplasmic dynein 2 appeared compromised. Although the densities of mutant ependymal and respiratory cilia were largely normal, the planar polarity of mutant ependymal cilia was disrupted, resulting in uncoordinated flow of CSF. Transgenic expression of GFP-CETN2 rescued the Cetn2-deficiency phenotype. These results indicate that mammalian basal body replication and ciliogenesis occur independently of CETN2; however, mouse CETN2 regulates protein trafficking of olfactory cilia and participates in specifying planar polarity of ependymal cilia.


Subject(s)
Calcium-Binding Proteins/physiology , Cilia/metabolism , Cilia/pathology , Epithelium/pathology , Gene Expression Regulation, Developmental/genetics , Olfactory Bulb/pathology , Protein Transport/genetics , Animals , Animals, Newborn , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cell Polarity/genetics , Cilia/ultrastructure , Disease Models, Animal , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Hydrocephalus/complications , Hydrocephalus/genetics , Hydrocephalus/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Odorants , Olfaction Disorders/complications , Olfaction Disorders/genetics , Olfaction Disorders/pathology , Pentanols/pharmacology , Protein Transport/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL