Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.302
Filter
1.
Elife ; 122024 May 13.
Article in English | MEDLINE | ID: mdl-38739438

ABSTRACT

The retina consumes massive amounts of energy, yet its metabolism and substrate exploitation remain poorly understood. Here, we used a murine explant model to manipulate retinal energy metabolism under entirely controlled conditions and utilised 1H-NMR spectroscopy-based metabolomics, in situ enzyme detection, and cell viability readouts to uncover the pathways of retinal energy production. Our experimental manipulations resulted in varying degrees of photoreceptor degeneration, while the inner retina and retinal pigment epithelium were essentially unaffected. This selective vulnerability of photoreceptors suggested very specific adaptations in their energy metabolism. Rod photoreceptors were found to rely strongly on oxidative phosphorylation, but only mildly on glycolysis. Conversely, cone photoreceptors were dependent on glycolysis but insensitive to electron transport chain decoupling. Importantly, photoreceptors appeared to uncouple glycolytic and Krebs-cycle metabolism via three different pathways: (1) the mini-Krebs-cycle, fuelled by glutamine and branched chain amino acids, generating N-acetylaspartate; (2) the alanine-generating Cahill-cycle; (3) the lactate-releasing Cori-cycle. Moreover, the metabolomics data indicated a shuttling of taurine and hypotaurine between the retinal pigment epithelium and photoreceptors, likely resulting in an additional net transfer of reducing power to photoreceptors. These findings expand our understanding of retinal physiology and pathology and shed new light on neuronal energy homeostasis and the pathogenesis of neurodegenerative diseases.


Subject(s)
Citric Acid Cycle , Glycolysis , Oxidative Phosphorylation , Retina , Animals , Mice , Retina/metabolism , Energy Metabolism , Metabolomics , Retinal Pigment Epithelium/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Mice, Inbred C57BL , Retinal Cone Photoreceptor Cells/metabolism
2.
PLoS One ; 19(5): e0300584, 2024.
Article in English | MEDLINE | ID: mdl-38709779

ABSTRACT

Though rod and cone photoreceptors use similar phototransduction mechanisms, previous model calculations have indicated that the most important differences in their light responses are likely to be differences in amplification of the G-protein cascade, different decay rates of phosphodiesterase (PDE) and pigment phosphorylation, and different rates of turnover of cGMP in darkness. To test this hypothesis, we constructed TrUx;GapOx rods by crossing mice with decreased transduction gain from decreased transducin expression, with mice displaying an increased rate of PDE decay from increased expression of GTPase-activating proteins (GAPs). These two manipulations brought the sensitivity of TrUx;GapOx rods to within a factor of 2 of WT cone sensitivity, after correcting for outer-segment dimensions. These alterations did not, however, change photoreceptor adaptation: rods continued to show increment saturation though at a higher background intensity. These experiments confirm model calculations that rod responses can mimic some (though not all) of the features of cone responses after only a few changes in the properties of transduction proteins.


Subject(s)
Retinal Cone Photoreceptor Cells , Retinal Rod Photoreceptor Cells , Transducin , Animals , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Mice , Transducin/metabolism , Transducin/genetics , Retina/metabolism , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/genetics
3.
Sci Rep ; 14(1): 10498, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38714794

ABSTRACT

Prominin 1 (PROM1) is a pentaspan transmembrane glycoprotein localized on the nascent photoreceptor discs. Mutations in PROM1 are linked to various retinal diseases. In this study, we assessed the role of PROM1 in photoreceptor biology and physiology using the PROM1 knockout murine model (rd19). Our study found that PROM1 is essential for vision and photoreceptor development. We found an early reduction in photoreceptor response beginning at post-natal day 12 (P12) before eye opening in the absence of PROM1 with no apparent loss in photoreceptor cells. However, at this stage, we observed an increased glial cell activation, indicative of cell damage. Contrary to our expectations, dark rearing did not mitigate photoreceptor degeneration or vision loss in PROM1 knockout mice. In addition to physiological defects seen in PROM1 knockout mice, ultrastructural analysis revealed malformed outer segments characterized by whorl-like continuous membranes instead of stacked disks. In parallel to the reduced rod response at P12, proteomics revealed a significant reduction in the levels of protocadherin, a known interactor of PROM1, and rod photoreceptor outer segment proteins, including rhodopsin. Overall, our results underscore the indispensable role of PROM1 in photoreceptor development and maintenance of healthy vision.


Subject(s)
AC133 Antigen , Mice, Knockout , Animals , Mice , AC133 Antigen/metabolism , AC133 Antigen/genetics , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Rhodopsin/metabolism , Rhodopsin/genetics , Photoreceptor Cells, Vertebrate/metabolism
4.
Proc Natl Acad Sci U S A ; 121(21): e2404763121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38743626

ABSTRACT

Congenital stationary night blindness (CSNB) is an inherited retinal disease that causes a profound loss of rod sensitivity without severe retinal degeneration. One well-studied rhodopsin point mutant, G90D-Rho, is thought to cause CSNB because of its constitutive activity in darkness causing rod desensitization. However, the nature of this constitutive activity and its precise molecular source have not been resolved for almost 30 y. In this study, we made a knock-in (KI) mouse line with a very low expression of G90D-Rho (equal in amount to ~0.1% of normal rhodopsin, WT-Rho, in WT rods), with the remaining WT-Rho replaced by REY-Rho, a mutant with a very low efficiency of activating transducin due to a charge reversal of the highly conserved ERY motif to REY. We observed two kinds of constitutive noise: one being spontaneous isomerization (R*) of G90D-Rho at a molecular rate (R* s-1) 175-fold higher than WT-Rho and the other being G90D-Rho-generated dark continuous noise comprising low-amplitude unitary events occurring at a very high molecular rate equivalent in effect to ~40,000-fold of R* s-1 from WT-Rho. Neither noise type originated from G90D-Opsin because exogenous 11-cis-retinal had no effect. Extrapolating the above observations at low (0.1%) expression of G90D-Rho to normal disease exhibited by a KI mouse model with RhoG90D/WTand RhoG90D/G90D genotypes predicts the disease condition very well quantitatively. Overall, the continuous noise from G90D-Rho therefore predominates, constituting the major equivalent background light causing rod desensitization in CSNB.


Subject(s)
Eye Diseases, Hereditary , Genetic Diseases, X-Linked , Myopia , Night Blindness , Rhodopsin , Animals , Night Blindness/genetics , Night Blindness/metabolism , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/metabolism , Mice , Rhodopsin/genetics , Rhodopsin/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Myopia/genetics , Myopia/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Darkness , Transducin/genetics , Transducin/metabolism , Gene Knock-In Techniques , Disease Models, Animal
5.
J Med Chem ; 67(10): 8396-8405, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38688030

ABSTRACT

Retinitis pigmentosa (RP) is a form of retinal degeneration affecting a young population with an unmet medical need. Photoreceptor degeneration has been associated with increased guanosine 3',5'-cyclic monophosphate (cGMP), which reaches toxic levels for photoreceptors. Therefore, inhibitory cGMP analogues attract interest for RP treatments. Here we present the synthesis of dithio-CN03, a phosphorodithioate analogue of cGMP, prepared using the H-phosphonothioate route. Two crystal modifications were identified as a trihydrate and a tetrahydrofuran monosolvates. Dithio-CN03 featured a lower aqueous solubility than its RP-phosphorothioate counterpart CN03, a drug candidate, and this characteristic might be favorable for sustained-release formulations aimed at retinal delivery. Dithio-CN03 was tested in vitro for its neuroprotective effects in photoreceptor models of RP. The comparison of dithio-CN03 to CN03 and its diastereomer SP-CN03, and to their phosphate derivative oxo-CN03 identifies dithio-CN03 as the compound with the highest efficacy in neuroprotection and thus as a promising new candidate for the treatment of RP.


Subject(s)
Cyclic GMP , Neuroprotective Agents , Retinal Rod Photoreceptor Cells , Animals , Cyclic GMP/metabolism , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Mice , Humans , Retinal Degeneration/drug therapy , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Structure-Activity Relationship
6.
FASEB J ; 38(8): e23606, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648465

ABSTRACT

Rhodopsin mislocalization encompasses various blind conditions. Rhodopsin mislocalization is the primary factor leading to rod photoreceptor dysfunction and degeneration in autosomal dominant retinitis pigmentosa (adRP) caused by class I mutations. In this study, we report a new knock-in mouse model that harbors a class I Q344X mutation in the endogenous rhodopsin gene, which causes rod photoreceptor degeneration in an autosomal dominant pattern. In RhoQ344X/+ mice, mRNA transcripts from the wild-type (Rho) and RhoQ344X mutant rhodopsin alleles are expressed at equal levels. However, the amount of RHOQ344X mutant protein is 2.7 times lower than that of wild-type rhodopsin, a finding consistent with the rapid degradation of the mutant protein. Immunofluorescence microscopy indicates that RHOQ344X is mislocalized to the inner segment and outer nuclear layers of rod photoreceptors in both RhoQ344X/+ and RhoQ344X/Q344X mice, confirming the essential role of the C-terminal VxPx motif in promoting OS delivery of rhodopsin. The mislocalization of RHOQ344X is associated with the concurrent mislocalization of wild-type rhodopsin in RhoQ344X/+ mice. To understand the global changes in proteostasis, we conducted quantitative proteomics analysis and found attenuated expression of rod-specific OS membrane proteins accompanying reduced expression of ciliopathy causative gene products, including constituents of BBSome and axonemal dynein subunit. Those studies unveil a novel negative feedback regulation involving ciliopathy-associated proteins. In this process, a defect in the trafficking signal leads to a reduced quantity of the trafficking apparatus, culminating in a widespread reduction in the transport of ciliary proteins.


Subject(s)
Disease Models, Animal , Gene Knock-In Techniques , Retinal Rod Photoreceptor Cells , Retinitis Pigmentosa , Rhodopsin , Animals , Rhodopsin/metabolism , Rhodopsin/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Mice , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Cilia/metabolism , Cilia/pathology
7.
Dev Biol ; 511: 39-52, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38548147

ABSTRACT

The fovea is a small region within the central retina that is responsible for our high acuity daylight vision. Chickens also have a high acuity area (HAA), and are one of the few species that enables studies of the mechanisms of HAA development, due to accessible embryonic tissue and methods to readily perturb gene expression. To enable such studies, we characterized the development of the chick HAA using single molecule fluorescent in situ hybridization (smFISH), along with more classical methods. We found that Fgf8 provides a molecular marker for the HAA throughout development and into adult stages, allowing studies of the cellular composition of this area over time. The radial dimension of the ganglion cell layer (GCL) was seen to be the greatest at the HAA throughout development, beginning during the period of neurogenesis, suggesting that genesis, rather than cell death, creates a higher level of retinal ganglion cells (RGCs) in this area. In contrast, the HAA acquired its characteristic high density of cone photoreceptors post-hatching, which is well after the period of neurogenesis. We also confirmed that rod photoreceptors are not present in the HAA. Analyses of cell death in the developing photoreceptor layer, where rods would reside, did not show apoptotic cells, suggesting that lack of genesis, rather than death, created the "rod-free zone" (RFZ). Quantification of each cone photoreceptor subtype showed an ordered mosaic of most cone subtypes. The changes in cellular densities and cell subtypes between the developing and mature HAA provide some answers to the overarching strategy used by the retina to create this area and provide a framework for future studies of the mechanisms underlying its formation.


Subject(s)
Retina , Retinal Ganglion Cells , Animals , Chick Embryo , Retinal Ganglion Cells/cytology , Retina/embryology , Retinal Cone Photoreceptor Cells/metabolism , Chickens , Neurogenesis/physiology , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factor 8/genetics , In Situ Hybridization, Fluorescence , Fovea Centralis/embryology , Visual Acuity , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/cytology , Gene Expression Regulation, Developmental
8.
Mol Ther ; 32(5): 1445-1460, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38504520

ABSTRACT

Age-related macular degeneration (AMD) is the most common cause of untreatable blindness in the developed world. Recently, CDHR1 has been identified as the cause of a subset of AMD that has the appearance of the "dry" form, or geographic atrophy. Biallelic variants in CDHR1-a specialized protocadherin highly expressed in cone and rod photoreceptors-result in blindness from shortened photoreceptor outer segments and progressive photoreceptor cell death. Here we demonstrate long-term morphological, ultrastructural, functional, and behavioral rescue following CDHR1 gene therapy in a relevant murine model, sustained to 23-months after injection. This represents the first demonstration of rescue of a monogenic cadherinopathy in vivo. Moreover, the durability of CDHR1 gene therapy seems to be near complete-with morphological findings of the rescued retina not obviously different from wildtype throughout the lifespan of the mouse model. A follow-on clinical trial in patients with CDHR1-associated retinal degeneration is warranted. Hypomorphic CDHR1 variants may mimic advanced dry AMD. Accurate clinical classification is now critical, as their pathogenesis and treatment are distinct.


Subject(s)
Cadherin Related Proteins , Cadherins , Disease Models, Animal , Genetic Therapy , Nerve Tissue Proteins , Retinal Cone Photoreceptor Cells , Retinal Degeneration , Retinal Rod Photoreceptor Cells , Animals , Mice , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Cadherins/genetics , Cadherins/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Retinal Degeneration/etiology , Humans , Genetic Therapy/methods , Macular Degeneration/therapy , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/etiology , Macular Degeneration/metabolism
9.
Cell Rep Med ; 5(4): 101459, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38518771

ABSTRACT

Retinitis pigmentosa (RP) is one of the most common forms of hereditary neurodegeneration. It is caused by one or more of at least 3,100 mutations in over 80 genes that are primarily expressed in rod photoreceptors. In RP, the primary rod-death phase is followed by cone death, regardless of the underlying gene mutation that drove the initial rod degeneration. Dampening the oxidation of glycolytic end products in rod mitochondria enhances cone survival in divergent etiological disease models independent of the underlying rod-specific gene mutations. Therapeutic editing of the prolyl hydroxylase domain-containing protein gene (PHD2, also known as Egln1) in rod photoreceptors led to the sustained survival of both diseased rods and cones in both preclinical autosomal-recessive and dominant RP models. Adeno-associated virus-mediated CRISPR-based therapeutic reprogramming of the aerobic glycolysis node may serve as a gene-agnostic treatment for patients with various forms of RP.


Subject(s)
Retinal Rod Photoreceptor Cells , Retinitis Pigmentosa , Animals , Humans , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/therapy , Retinal Cone Photoreceptor Cells/metabolism , Disease Models, Animal
10.
Hum Mol Genet ; 33(9): 802-817, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38297980

ABSTRACT

Mutations in Cytosolic Carboxypeptidase-like Protein 5 (CCP5) are associated with vision loss in humans. To decipher the mechanisms behind CCP5-associated blindness, we generated a novel mouse model lacking CCP5. In this model, we found that increased tubulin glutamylation led to progressive cone-rod dystrophy, with cones showing a more pronounced and earlier functional loss than rod photoreceptors. The observed functional reduction was not due to cell death, levels, or the mislocalization of major phototransduction proteins. Instead, the increased tubulin glutamylation caused shortened photoreceptor axonemes and the formation of numerous abnormal membranous whorls that disrupted the integrity of photoreceptor outer segments (OS). Ultimately, excessive tubulin glutamylation led to the progressive loss of photoreceptors, affecting cones more severely than rods. Our results highlight the importance of maintaining tubulin glutamylation for normal photoreceptor function. Furthermore, we demonstrate that murine cone photoreceptors are more sensitive to disrupted tubulin glutamylation levels than rods, suggesting an essential role for axoneme in the structural integrity of the cone outer segment. This study provides valuable insights into the mechanisms of photoreceptor diseases linked to excessive tubulin glutamylation.


Subject(s)
Cone-Rod Dystrophies , Tubulin , Humans , Mice , Animals , Tubulin/genetics , Tubulin/metabolism , Cone-Rod Dystrophies/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Mutation
11.
Am J Pathol ; 194(5): 796-809, 2024 May.
Article in English | MEDLINE | ID: mdl-38395146

ABSTRACT

α-Synuclein (α-Syn) is a key determinator of Parkinson disease (PD) pathology, but synapse and microcircuit pathologies in the retina underlying visual dysfunction are poorly understood. Herein, histochemical and ultrastructural analyses and ophthalmologic measurements in old transgenic M83 PD model (mice aged 16 to 18 months) indicated that abnormal α-Syn aggregation in the outer plexiform layer (OPL) was associated with degeneration in the C-terminal binding protein 2 (CtBP2)+ ribbon synapses of photoreceptor terminals and protein kinase C alpha (PKCα)+ rod bipolar cell terminals, whereas α-Syn aggregates in the inner retina correlated with the reduction and degeneration of tyrosine hydroxylase- and parvalbumin-positive amacrine cells. Phosphorylated Ser129 α-synuclein expression was strikingly restricted in the OPL, with the most severe degenerations in the entire retina, including mitochondrial degeneration and loss of ribbon synapses in 16- to 18-month-old mice. These synapse- and microcircuit-specific deficits of the rod pathway at the CtBP2+ rod terminals and PKCα+ rod bipolar and amacrine cells were associated with attenuated a- and b-wave amplitudes and oscillatory potentials on the electroretinogram. They were also associated with the impairment of visual functions, including reduced contrast sensitivity and impairment of the middle range of spatial frequencies. Collectively, these findings demonstrate that α-Syn aggregates cause the synapse- and microcircuit-specific deficits of the rod pathway and the most severe damage to the OPL, providing the retinal synaptic and microcircuit basis for visual dysfunctions in PD.


Subject(s)
Protein Kinase C-alpha , alpha-Synuclein , Animals , Mice , alpha-Synuclein/metabolism , Amacrine Cells/metabolism , Protein Kinase C-alpha/metabolism , Retina/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/ultrastructure , Synapses/metabolism , Transcription Factors/metabolism
12.
JCI Insight ; 9(4)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38227383

ABSTRACT

AMP-activated protein kinase (AMPK) plays a crucial role in maintaining ATP homeostasis in photoreceptor neurons. AMPK is a heterotrimeric protein consisting of α, ß, and γ subunits. The independent functions of the 2 isoforms of the catalytic α subunit, PRKAA1 and PRKAA2, are uncharacterized in specialized neurons, such as photoreceptors. Here, we demonstrate in mice that rod photoreceptors lacking PRKAA2, but not PRKAA1, showed altered levels of cGMP, GTP, and ATP, suggesting isoform-specific regulation of photoreceptor metabolism. Furthermore, PRKAA2-deficient mice displayed visual functional deficits on electroretinography and photoreceptor outer segment structural abnormalities on transmission electron microscopy consistent with neuronal dysfunction, but not neurodegeneration. Phosphoproteomics identified inosine monophosphate dehydrogenase (IMPDH) as a molecular driver of PRKAA2-specific photoreceptor dysfunction, and inhibition of IMPDH improved visual function in Prkaa2 rod photoreceptor-knockout mice. These findings highlight a therapeutically targetable PRKAA2 isoform-specific function of AMPK in regulating photoreceptor metabolism and function through a potentially previously uncharacterized mechanism affecting IMPDH activity.


Subject(s)
AMP-Activated Protein Kinases , Retinal Rod Photoreceptor Cells , Animals , Mice , AMP-Activated Protein Kinases/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Protein Isoforms/metabolism , Electroretinography , Mice, Knockout , Adenosine Triphosphate/metabolism
13.
J Biol Chem ; 300(2): 105608, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38159849

ABSTRACT

Phototransduction in retinal rods occurs when the G protein-coupled photoreceptor rhodopsin triggers the activation of phosphodiesterase 6 (PDE6) by GTP-bound alpha subunits of the G protein transducin (GαT). Recently, we presented a cryo-EM structure for a complex between two GTP-bound recombinant GαT subunits and native PDE6, that included a bivalent antibody bound to the C-terminal ends of GαT and the inhibitor vardenafil occupying the active sites on the PDEα and PDEß subunits. We proposed GαT-activated PDE6 by inducing a striking reorientation of the PDEγ subunits away from the catalytic sites. However, questions remained including whether in the absence of the antibody GαT binds to PDE6 in a similar manner as observed when the antibody is present, does GαT activate PDE6 by enabling the substrate cGMP to access the catalytic sites, and how does the lipid membrane enhance PDE6 activation? Here, we demonstrate that 2:1 GαT-PDE6 complexes form with either recombinant or retinal GαT in the absence of the GαT antibody. We show that GαT binding is not necessary for cGMP nor competitive inhibitors to access the active sites; instead, occupancy of the substrate binding sites enables GαT to bind and reposition the PDE6γ subunits to promote catalytic activity. Moreover, we demonstrate by reconstituting GαT-stimulated PDE6 activity in lipid bilayer nanodiscs that the membrane-induced enhancement results from an increase in the apparent binding affinity of GαT for PDE6. These findings provide new insights into how the retinal G protein stimulates rapid catalytic turnover by PDE6 required for dim light vision.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Models, Molecular , Transducin , Cyclic Nucleotide Phosphodiesterases, Type 6/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Guanosine Triphosphate/metabolism , Retinal Rod Photoreceptor Cells/enzymology , Retinal Rod Photoreceptor Cells/metabolism , Transducin/chemistry , Transducin/genetics , Transducin/metabolism , Animals , Cattle , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Protein Structure, Quaternary , Protein Binding/drug effects , Catalytic Domain , 1-Methyl-3-isobutylxanthine/pharmacology , Lipid Bilayers/metabolism , Enzyme Activation
14.
Cell Mol Life Sci ; 80(12): 362, 2023 Nov 18.
Article in English | MEDLINE | ID: mdl-37979052

ABSTRACT

A hallmark of inherited retinal degenerative diseases such as retinitis pigmentosa (RP) is progressive structural and functional remodeling of the remaining retinal cells as photoreceptors degenerate. Extensive remodeling of the retina stands as a barrier for the successful implementation of strategies to restore vision. To understand the molecular basis of remodeling, we performed analyses of single-cell transcriptome data from adult zebrafish retina of wild type AB strain (WT) and a P23H mutant rhodopsin transgenic model of RP with continuous degeneration and regeneration. Retinas from both female and male fish were pooled to generate each library, combining data from both sexes. We provide a benchmark atlas of retinal cell type transcriptomes in zebrafish and insight into how each retinal cell type is affected in the P23H model. Oxidative stress is found throughout the retina, with increases in reliance on oxidative metabolism and glycolysis in the affected rods as well as cones, bipolar cells, and retinal ganglion cells. There is also transcriptional evidence for widespread synaptic remodeling and enhancement of glutamatergic transmission in the inner retina. Notably, changes in circadian rhythm regulation are detected in cones, bipolar cells, and retinal pigmented epithelium. We also identify the transcriptomic signatures of retinal progenitor cells and newly formed rods essential for the regenerative process. This comprehensive transcriptomic analysis provides a molecular road map to understand how the retina remodels in the context of chronic retinal degeneration with ongoing regeneration.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Animals , Male , Female , Zebrafish/genetics , Retina/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Disease Models, Animal
15.
J Biol Chem ; 299(12): 105412, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37918805

ABSTRACT

A major unsolved question in vertebrate photoreceptor biology is the mechanism of rhodopsin transport to the outer segment. In rhodopsin-like class A G protein-coupled receptors, hydrophobic interactions between C-terminal α-helix 8 (H8), and transmembrane α-helix-1 (TM1) have been shown to be important for transport to the plasma membrane, however whether this interaction is important for rhodopsin transport to ciliary rod outer segments is not known. We examined the crystal structures of vertebrate rhodopsins and class A G protein-coupled receptors and found a conserved network of predicted hydrophobic interactions. In Xenopus rhodopsin (xRho), this interaction corresponds to F313, L317, and L321 in H8 and M57, V61, and L68 in TM1. To evaluate the role of H8-TM1 hydrophobic interactions in rhodopsin transport, we expressed xRho-EGFP where hydrophobic residues were mutated in Xenopus rods and evaluated the efficiency of outer segment enrichment. We found that substituting L317 and M57 with hydrophilic residues had the strongest impact on xRho mislocalization. Substituting hydrophilic amino acids at positions L68, F313, and L321 also had a significant impact. Replacing L317 with M resulted in significant mislocalization, indicating that the hydrophobic interaction between residues 317 and 57 is exquisitely sensitive. The corresponding experiment in bovine rhodopsin expressed in HEK293 cells had a similar effect, showing that the H8-TM1 hydrophobic network is essential for rhodopsin transport in mammalian species. Thus, for the first time, we show that a hydrophobic interaction between H8 and TM1 is critical for efficient rhodopsin transport to the vertebrate photoreceptor ciliary outer segment.


Subject(s)
Retinal Rod Photoreceptor Cells , Rhodopsin , Animals , Cattle , Humans , HEK293 Cells , Hydrophobic and Hydrophilic Interactions , Receptors, G-Protein-Coupled/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Rhodopsin/genetics , Rhodopsin/chemistry , Rod Cell Outer Segment/metabolism , Vertebrates
16.
Cell Death Dis ; 14(10): 701, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37880237

ABSTRACT

Neurodegenerative diseases are accompanied by dynamic changes in gene expression, including the upregulation of hallmark stress-responsive genes. While the transcriptional pathways that impart adaptive and maladaptive gene expression signatures have been the focus of intense study, the role of higher order nuclear organization in this process is less clear. Here, we examine the role of the nuclear lamina in genome organization during the degeneration of rod photoreceptors. Two proteins had previously been shown to be necessary and sufficient to tether heterochromatin at the nuclear envelope. The lamin B receptor (Lbr) is expressed during development, but downregulates upon rod differentiation. A second tether is the intermediate filament lamin A (LA), which is not normally expressed in murine rods. Here, we show that in the rd1 model of retinitis pigmentosa, LA ectopically upregulates in rod photoreceptors at the onset of degeneration. LA upregulation correlated with increased heterochromatin tethering at the nuclear periphery in rd1 rods, suggesting that LA reorganizes the nucleus. To determine how heterochromatin tethering affects the genome, we used in vivo electroporation to misexpress LA or Lbr in mature rods in the absence of degeneration, resulting in the restoration of conventional nuclear architecture. Using scRNA-seq, we show that reorganizing the nucleus via LA/Lbr misexpression has relatively minor effects on rod gene expression. Next, using ATAC-seq, we show that LA and Lbr both lead to marked increases in genome accessibility. Novel ATAC-seq peaks tended to be associated with stress-responsive genes. Together, our data reveal that heterochromatin tethers have a global effect on genome accessibility, and suggest that heterochromatin tethering primes the photoreceptor genome to respond to stress.


Subject(s)
Heterochromatin , Retinal Degeneration , Mice , Animals , Heterochromatin/genetics , Heterochromatin/metabolism , Lamin Type A/metabolism , Up-Regulation/genetics , Cell Nucleus/metabolism , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/metabolism
17.
Proc Natl Acad Sci U S A ; 120(42): e2308204120, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37812728

ABSTRACT

Migration is essential for the laminar stratification and connectivity of neurons in the central nervous system. In the retina, photoreceptors (PRs) migrate to positions according to birthdate, with early-born cells localizing to the basal-most side of the outer nuclear layer. It was proposed that apical progenitor mitoses physically drive these basal translocations non-cell autonomously, but direct evidence is lacking, and whether other mechanisms participate is unknown. Here, combining loss- or gain-of-function assays to manipulate cell cycle regulators (Sonic hedgehog, Cdkn1a/p21) with an in vivo lentiviral labelling strategy, we demonstrate that progenitor division is one of two forces driving basal translocation of rod soma. Indeed, replacing Shh activity rescues abnormal rod translocation in retinal explants. Unexpectedly, we show that rod differentiation also promotes rod soma translocation. While outer segment function or formation is dispensable, Crx and SNARE-dependent synaptic function are essential. Thus, both non-cell and cell autonomous mechanisms underpin PR soma sublaminar positioning in the mammalian retina.


Subject(s)
Neurosecretion , Retinal Rod Photoreceptor Cells , Animals , Retinal Rod Photoreceptor Cells/metabolism , Hedgehog Proteins/metabolism , Retina/metabolism , Cell Differentiation , Mammals
18.
Sci Rep ; 13(1): 12899, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37558693

ABSTRACT

Enhancers function with a basal promoter to control the transcription of target genes. Enhancer regulatory activity is often studied using reporter-based transgene assays. However, unmatched results have been reported when selected enhancers are silenced in situ. In this study, using genomic deletion analysis in mice, we investigated the roles of two previously identified enhancers and the promoter of the Rho gene that codes for the visual pigment rhodopsin. The Rho gene is robustly expressed by rod photoreceptors of the retina, and essential for the subcellular structure and visual function of rod photoreceptors. Mutations in RHO cause severe vision loss in humans. We found that each Rho regulatory region can independently mediate local epigenomic changes, but only the promoter is absolutely required for establishing active Rho chromatin configuration and transcription and maintaining the cell integrity and function of rod photoreceptors. To our surprise, two Rho enhancers that enable strong promoter activation in reporter assays are largely dispensable for Rho expression in vivo. Only small and age-dependent impact is detectable when both enhancers are deleted. Our results demonstrate context-dependent roles of enhancers and highlight the importance of studying functions of cis-regulatory regions in the native genomic context.


Subject(s)
Retinal Rod Photoreceptor Cells , Rhodopsin , Humans , Animals , Mice , Rhodopsin/genetics , Rhodopsin/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retina/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Promoter Regions, Genetic
19.
Stem Cells Dev ; 32(21-22): 681-692, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37470211

ABSTRACT

Retinitis pigmentosa (RP) is a prevalent inherited retinal degenerative disease resulting from photoreceptor and pigment epithelial apoptosis. The Rhodopsin (RHO) is the most commonly associated pathogenic gene in RP. However, RHO mutations (c.512C>T P171L) have been infrequently reported, and the RP pathogenesis caused by these mutations remains unclear. The objective of this study was to investigate the impact of RHO (c.512C>T P171L) mutation on retinal cell differentiation and elucidate the underlying mechanisms of RP. An effective retinal organoid induction scheme for inhibiting the Wnt signaling pathway was selected for further experiments, and the established cell line chHES-406 was demonstrated to be heterozygous for RHO c.512C>T, with a normal karyotype and pluripotency potential. Furthermore, the development of chHES-406 organoids may be delayed, and apoptosis detection and co-localization revealed that chHES-406 organoids had more apoptotic cells than chHES-90 in the outer nuclear layer (ONL), mutant RHO protein was mislocalized in the endoplasmic reticulum (ER), and stress-related and apoptotic gene expression increased. Overall, our study elucidated a possible mechanism by which ER stress caused by RHO P171L protein mislocalization may lead to ONL cell apoptosis.


Subject(s)
Retinitis Pigmentosa , Rhodopsin , Humans , Rhodopsin/genetics , Rhodopsin/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Endoplasmic Reticulum Stress/genetics , Apoptosis/genetics , Mutation/genetics
20.
Tissue Cell ; 83: 102150, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37423033

ABSTRACT

Rod photoreceptors in the adult teleost retina are produced by rod precursors located in the outer nuclear layer (ONL). Annual fishes of the genus Austrolebias exhibit extensive adult retinal cell proliferation and neurogenesis, as well as surprising adaptive strategies to their extreme and changing environment, including adult retinal plasticity. Thus, here we identify and characterize rod precursors in the ONL of the Austrolebias charrua retina. For this aim we used classical histological techniques, transmission electron microscopy, detection of cell proliferation, and immunohistochemistry. Through these complementary approaches, we describe a cell population clearly distinguishable from photoreceptors in the ONL of the adult retina of A. charrua, which we propose corresponds to the rod precursor population. These cells exhibited particular morphological and ultrastructural characteristics, uptake of cell proliferation markers (BrdU+) and expression of stem cell markers (Sox2+). Determining the existence of the population of rod precursors is crucial to understand the sequence of events related to retinal plasticity and regeneration.


Subject(s)
Retina , Retinal Rod Photoreceptor Cells , Animals , Retinal Rod Photoreceptor Cells/metabolism , Fishes , Cell Proliferation
SELECTION OF CITATIONS
SEARCH DETAIL
...