Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cells ; 12(1)2022 12 30.
Article in English | MEDLINE | ID: mdl-36611941

ABSTRACT

Arf-like protein 2 (ARL2) is a ubiquitously expressed small GTPase with multiple functions. In a cell culture, ARL2 participates with tubulin cofactor D (TBCD) in the neogenesis of tubulin αß-heterodimers, the building blocks of microtubules. To evaluate this function in the retina, we conditionally deleted ARL2 in mouse retina at two distinct stages, either during the embryonic development (retArl2-/-) or after ciliogenesis specifically in rods (rodArl2-/-). retArl2-/- retina sections displayed distorted nuclear layers and a disrupted microtubule cytoskeleton (MTC) as early as postnatal day 6 (P6). Rod and cone outer segments (OS) did not form. By contrast, the rod ARL2 knockouts were stable at postnatal day 35 and revealed normal ERG responses. Cytoplasmic dynein is reduced in retArl2-/- inner segments (IS), suggesting that dynein may be unstable in the absence of a normal MTC. We investigated the microtubular stability in the absence of either ARL2 (retARL2-/-) or DYNC1H1 (retDync1h1-/-), the dynein heavy chain, and found that both the retArl2-/- and retDync1h1-/- retinas exhibited reduced microtubules and nuclear layer distortion. The results suggest that ARL2 and dynein depend on each other to generate a functional MTC during the early photoreceptor development.


Subject(s)
Dyneins , Tubulin , Mice , Animals , Tubulin/metabolism , Microtubules/metabolism , Photoreceptor Cells/metabolism , Retina/metabolism
2.
Invest Ophthalmol Vis Sci ; 62(14): 23, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34807236

ABSTRACT

Purpose: Cytoplasmic dynein-1 (henceforth dynein) moves cargo in conjunction with dynactin toward the minus end of microtubules. The dynein heavy chain, DYNC1H1, comprises the backbone of dynein, a retrograde motor. Deletion of Dync1h1 abrogates dynein function. The purpose of this communication is to demonstrate effects of photoreceptor dynein inactivation during late postnatal development and in adult retina. Methods: We mated Dync1h1F/F mice with iCre75 and Prom1-CreERT2 mice to generate conditional rod and tamoxifen-induced knockout in rods and cones, respectively. We documented retina degeneration with confocal microscopy at postnatal day (P) 10 to P30 for the iCre75 line and 1 to 4 weeks post tamoxifen induction (wPTI) for the Prom1-CreERT2 line. We performed scotopic and photopic electroretinography (ERG) at P16 to P30 in the iCre75 line and at 1-week increments in the Prom1-CreERT2 line. Results were evaluated statistically using Student's t-test, two-factor ANOVA, and Welch's ANOVA. Results: Cre-induced homologous recombination of Dync1h1F/F mice truncated DYNC1H1 after exon 23. rodDync1h1-/- photoreceptors degenerated after P14, reducing outer nuclear layer (ONL) thickness and combined inner segment/outer segment (IS/OS) length significantly by P18. Scotopic ERG a-wave amplitudes decreased by P16 and were extinguished at P30. Cones were stable under rod-knockout conditions until P21 but inactive at P30. In tamDync1h1-/- photoreceptors, the IS/OS began shortening by 3wPTI and were nearly eliminated by 4wPTI. The ONL shrank significantly over this interval, indicating rapid photoreceptor degeneration following the loss of dynein. Conclusions: Our results demonstrate dynein is essential for the secretory pathway, formation of outer segments, and photoreceptor maintenance.


Subject(s)
Cytoplasmic Dyneins/genetics , Gene Deletion , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/genetics , Alcohol Oxidoreductases/metabolism , Animals , Animals, Newborn , Co-Repressor Proteins/metabolism , Color Vision/physiology , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Electroretinography , Estrogen Antagonists/toxicity , Eye Proteins/metabolism , Female , Genotyping Techniques , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Night Vision/physiology , Photic Stimulation , Tamoxifen/toxicity
3.
PLoS One ; 16(3): e0248354, 2021.
Article in English | MEDLINE | ID: mdl-33705456

ABSTRACT

Cytoplasmic dynein (dynein 1), a major retrograde motor of eukaryotic cells, is a 1.4 MDa protein complex consisting of a pair of heavy chains (DYNC1H1) and a set of heterodimeric noncatalytic accessory components termed intermediate, light intermediate and light chains. DYNC1H1 (4644 amino acids) is the dynein backbone encoded by a gene consisting of 77 exons. We generated a floxed Dync1h1 allele that excises exons 24 and 25 and truncates DYNC1H1 during Six3Cre-induced homologous recombination. Truncation results in loss of the motor and microtubule-binding domain. Dync1h1F/F;Six3Cre photoreceptors degenerated rapidly within two postnatal weeks. In the postnatal day 6 (P6) Dync1h1F/F;Six3Cre central retina, outer and inner nuclear layers were severely disorganized and lacked a recognizable outer plexiform layer (OPL). Although the gene was effectively silenced by P6, DYNC1H1 remnants persisted and aggregated together with rhodopsin, PDE6 and centrin-2-positive centrosomes in the outer nuclear layer. As photoreceptor degeneration is delayed in the Dync1h1F/F;Six3Cre retina periphery, retinal lamination and outer segment elongation are in part preserved. DYNC1H1 strongly persisted in the inner plexiform layer (IPL) beyond P16 suggesting lack of clearance of the DYNC1H1 polypeptide. This persistence of DYNC1H1 allows horizontal, rod bipolar, amacrine and ganglion cells to survive past P12. The results show that cytoplasmic dynein is essential for retina lamination, nuclear positioning, vesicular trafficking of photoreceptor membrane proteins and inner/outer segment elaboration.


Subject(s)
Amacrine Cells/metabolism , Cell Membrane/metabolism , Cytoplasmic Dyneins/deficiency , Retinal Ganglion Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Amacrine Cells/pathology , Animals , Animals, Newborn , Cell Membrane/genetics , Cell Membrane/pathology , Cytoplasmic Dyneins/metabolism , Gene Deletion , Mice , Mice, Knockout , Retinal Ganglion Cells/pathology , Retinal Rod Photoreceptor Cells/pathology
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.
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
6.
Sci Rep ; 8(1): 4807, 2018 Mar 14.
Article in English | MEDLINE | ID: mdl-29540812

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

7.
Sci Rep ; 7(1): 6690, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28751656

ABSTRACT

Cones are responsible for daylight, central, high acuity and color vision. Three proteins found in human cones, i.e. long-wavelength (L)-, middle-wavelength (M)-, and short-wavelength sensitive (S)-opsins, are responsible for red, green and blue color recognition, respectively. Human blue cone monochromacy (BCM) is characterized by functional loss of both L- and M-cone opsins due to mutations in the OPN1LW/OPN1MW gene cluster on the X chromosome. BCM patients, who rely on their vision from only S-cones and rods, suffer severely reduced visual acuity and impaired color vision. Recent studies show that there is sufficient cone structure remaining in the central fovea of BCM patients to consider AAV-mediated gene augmentation therapy. In contrast, mouse retina has only two opsins, S-opsin and M-opsin, but no L-opsin. We generated an M-opsin knockout mouse (Opn1mw -/-) expressing only S-opsin as a model for human BCM. We show that recombinant M-opsin delivered by AAV5 vectors rescues M-cone function in Opn1mw -/- mice. We also show that AAV delivered M-opsin localizes in the dorsal cone outer segments, and co-localizes with S-opsin in the ventral retina. Our study demonstrates that cones without M-opsin remain viable and respond to gene augmentation therapy, thereby providing proof-of-concept for cone function restoration in BCM patients.


Subject(s)
Color Vision Defects/genetics , Color Vision Defects/therapy , Genetic Therapy , Animals , Dependovirus/metabolism , Disease Models, Animal , Electroretinography , Humans , Mice , Mice, Knockout , Opsins/genetics , Opsins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology
8.
PLoS One ; 11(8): e0161236, 2016.
Article in English | MEDLINE | ID: mdl-27529348

ABSTRACT

Rab11a and Rab8a are ubiquitous small GTPases shown as required for rhodopsin transport in Xenopus laevis and zebrafish photoreceptors by dominant negative (dn) disruption of function. Here, we generated retina-specific Rab11a (retRab11a) and Rab8a (retRab8a) single and double knockout mice to explore the consequences in mouse photoreceptors. Rhodopsin and other outer segment (OS) membrane proteins targeted correctly to OS and electroretinogram (ERG) responses in all three mutant mouse lines were indistinguishable from wild-type (WT). Further, AAV (adeno-associated virus)-mediated expression of dnRab11b in retRab11a-/- retina, or expression of dnRab8b in retRab8a-/- retina did not cause OS protein mislocalization. Finally, a retRab8a-/- retina injected at one month of age with AAVs expressing dnRab11a, dnRab11b, dnRab8b, and dnRab10 (four dn viruses on Rab8a-/- background) and harvested three months later exhibited normal OS protein localization. In contrast to results obtained with dnRab GTPases in Xenopus and zebrafish, mouse Rab11a and Rab8a are dispensable for proper rhodopsin and outer segment membrane protein targeting. Absence of phenotype after expression of four dn Rab GTPases in a Rab8a-/- retina suggests that Rab8b and Rab11b paralogs maybe dispensable as well. Our data thus demonstrate significant interspecies variation in photoreceptor membrane protein and rhodopsin trafficking.


Subject(s)
Photoreceptor Cells/metabolism , Rhodopsin/metabolism , rab GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Protein Transport , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/deficiency , rab GTP-Binding Proteins/genetics
9.
Biochem Biophys Res Commun ; 473(4): 1211-1217, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27079236

ABSTRACT

PURPOSE: UNC119 proteins are involved in G protein trafficking in mouse retinal photoreceptors and Caenorhabditis elegans olfactory neurons. An Unc119 null allele is associated with cone-rod dystrophy in mouse, but the mechanism leading to disease is not understood. We studied the role of Unc119 paralogs and Arl3l2 in zebrafish vision and retinal organization resulting from unc119c and arl3l2 knockdown. METHODS: Zebrafish unc119c was amplified by PCR from retina and pineal gland cDNA. Its expression pattern in the eye and pineal gland was determined by whole-mount in-situ hybridization. unc119c and arl3l2 were knocked down using morpholino-modified oligonucleotides (MO). Their visual function was assessed with a quantitative optomotor assay on 6 days post-fertilization larvae. Retinal morphology was analyzed using immunohistochemistry with anti-cone arrestin (zpr-1) and anti-cone transducin-α (GNAT2) antibodies. RESULTS: The zebrafish genome contains four genes encoding unc119 paralogs located on different chromosomes. The exon/intron arrangements of these genes are identical. Three Unc119 paralogs are expressed in the zebrafish retina, termed Unc119a-c. Based on sequence similarity, Unc119a and Unc119b are orthologs of mammalian UNC119a and UNC119b, respectively. A third, Unc119c, is unique and not present in mammals. Whole mount in-situ hybridization revealed that unc119a and unc119b RNA are ubiquitously expressed in the CNS, and unc119c is specifically expressed in photoreceptive tissues (pineal gland and retina). A Unc119 interactant, Arl3l2 also localizes to the pineal gland and the retina. As measured by the optomotor response, unc119c and arl3l2 knockdown resulted in significantly lower vision compared to wild-type zebrafish larvae and control morpholino (MO). Immunohistological analysis with anti-cone transducin and anti-cone arrestin (zpr-1) indicates that knockdown of unc119c leads to photoreceptor degeneration mostly affecting cones. CONCLUSIONS: Our results suggest that Unc119c is the only Unc119 paralog that is highly specific to the retina in zebrafish. Unc119c and Arl3l2 proteins are important for the function of cones.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Eye Diseases, Hereditary/complications , Eye Diseases, Hereditary/physiopathology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Dystrophies/complications , Retinal Dystrophies/physiopathology , Vision Disorders/etiology , Vision Disorders/physiopathology , Animals , Eye Diseases, Hereditary/pathology , Gene Knockdown Techniques , Retinal Cone Photoreceptor Cells/pathology , Retinal Dystrophies/pathology , Vision Disorders/pathology , Zebrafish
10.
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
11.
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
12.
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
13.
Vision Res ; 75: 26-32, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23000199

ABSTRACT

The mechanism by which myristoylated proteins are targeted to specific subcellular membrane compartments is poorly understood. Two novel acyl-binding proteins, UNC119A and UNC119B, have been shown recently to function as chaperones/co-factors in the transport of myristoylated G protein α-subunits and src-type tyrosine kinases. UNC119 polypeptides feature an immunoglobulin-like ß-sandwich fold that forms a hydrophobic pocket capable of binding lauroyl (C12) and myristoyl (C14) side chains. UNC119A in rod photoreceptors facilitates the transfer of transducin α subunits (Tα) from inner segment to outer segment membranes by forming an intermediate diffusible UNC119-Tα complex. Similar complexes are formed in other sensory neurons, as the G proteins ODR-3 and GPA-13 in Caenorhabditis elegans unc-119 mutants traffic inappropriately. UNC119B knockdown in IMCD3 cells prevents trafficking ofmyristoylated nephrocystin-3 (NPHP3), a protein associated with nephronophthisis, to cilia. Further, UNC119A was shown to transport myristoylated src-type tyrosine kinases to cell membranes and to affect T-cell receptor (TCR) and interleukin-5 receptor (IL-5R) activities. These interactions establish UNC119 polypeptides as novel lipid-binding chaperones with specificity for a diverse subset of myristoylated proteins.


Subject(s)
Acyl Carrier Protein/physiology , Acyl Carrier Protein/chemistry , Animals , Caenorhabditis elegans/physiology , Cilia/physiology , GTP-Binding Protein alpha Subunits/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Chaperones/physiology , Protein Structure, Tertiary , Protein Transport/physiology , Retinal Rod Photoreceptor Cells/physiology , src-Family Kinases/metabolism
14.
Nat Neurosci ; 14(7): 874-80, 2011 Jun 05.
Article in English | MEDLINE | ID: mdl-21642972

ABSTRACT

UNC119 is widely expressed among vertebrates and other phyla. We found that UNC119 recognized the acylated N terminus of the rod photoreceptor transducin α (Tα) subunit and Caenorhabditis elegans G proteins ODR-3 and GPA-13. The crystal structure of human UNC119 at 1.95-Å resolution revealed an immunoglobulin-like ß-sandwich fold. Pulldowns and isothermal titration calorimetry revealed a tight interaction between UNC119 and acylated Gα peptides. The structure of co-crystals of UNC119 with an acylated Tα N-terminal peptide at 2.0 Å revealed that the lipid chain is buried deeply into UNC119's hydrophobic cavity. UNC119 bound Tα-GTP, inhibiting its GTPase activity, thereby providing a stable UNC119-Tα-GTP complex capable of diffusing from the inner segment back to the outer segment after light-induced translocation. UNC119 deletion in both mouse and C. elegans led to G protein mislocalization. Thus, UNC119 is a Gα subunit cofactor essential for G protein trafficking in sensory cilia.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Gene Expression Regulation/physiology , Sensory Receptor Cells/metabolism , Transducin/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cattle , Dark Adaptation/genetics , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits/deficiency , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go , Gene Expression Regulation/genetics , Glycine/genetics , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Knockout , Models, Chemical , Models, Molecular , Mutation/genetics , Protein Binding/genetics , Protein Structure, Quaternary/genetics , Protein Transport/genetics , Signal Transduction/genetics , Time Factors , Transducin/deficiency , Transducin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...