Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
FASEB J ; 38(11): e23720, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38837708

ABSTRACT

Recessive Stargardt disease (STGD1) is an inherited juvenile maculopathy caused by mutations in the ABCA4 gene, for which there is no suitable treatment. Loss of functional ABCA4 in the retinal pigment epithelium (RPE) alone, without contribution from photoreceptor cells, was shown to induce STGD1 pathology. Here, we identified cathepsin D (CatD), the primary RPE lysosomal protease, as a key molecular player contributing to endo-lysosomal dysfunction in STGD1 using a newly developed "disease-in-a-dish" RPE model from confirmed STGD1 patients. Induced pluripotent stem cell (iPSC)-derived RPE originating from three STGD1 patients exhibited elevated lysosomal pH, as previously reported in Abca4-/- mice. CatD protein maturation and activity were impaired in RPE from STGD1 patients and Abca4-/- mice. Consequently, STGD1 RPE cells have reduced photoreceptor outer segment degradation and abnormal accumulation of α-synuclein, the natural substrate of CatD. Furthermore, dysfunctional ABCA4 in STGD1 RPE cells results in intracellular accumulation of autofluorescent material and phosphatidylethanolamine (PE). The altered distribution of PE associated with the internal membranes of STGD1 RPE cells presumably compromises LC3-associated phagocytosis, contributing to delayed endo-lysosomal degradation activity. Drug-mediated re-acidification of lysosomes in the RPE of STGD1 restores CatD functional activity and reduces the accumulation of immature CatD protein loads. This preclinical study validates the contribution of CatD deficiencies to STGD1 pathology and provides evidence for an efficacious therapeutic approach targeting RPE cells. Our findings support a cell-autonomous RPE-driven pathology, informing future research aimed at targeting RPE cells to treat ABCA4-mediated retinopathies.


Subject(s)
ATP-Binding Cassette Transporters , Cathepsin D , Lysosomes , Retinal Pigment Epithelium , Stargardt Disease , Cathepsin D/metabolism , Cathepsin D/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Stargardt Disease/metabolism , Stargardt Disease/pathology , Stargardt Disease/genetics , Animals , Humans , Mice , Lysosomes/metabolism , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/genetics , Induced Pluripotent Stem Cells/metabolism , Mice, Knockout , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics
2.
Pharm Res ; 41(4): 807-817, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38443629

ABSTRACT

OBJECTIVE: Current gene therapy of inherited retinal diseases is achieved mainly by subretinal injection, which is invasive with severe adverse effects. Intravitreal injection is a minimally invasive alternative for gene therapy of inherited retinal diseases. This work explores the efficacy of intravitreal delivery of PEGylated ECO (a multifunctional pH-sensitive amphiphilic amino lipid) plasmid DNA (pGRK1-ABCA4-S/MAR) nanoparticles (PEG-ELNP) for gene therapy of Stargardt disease. METHODS: Pigmented Abca4-/- knockout mice received 1 µL of PEG-ELNP solution (200 ng/uL, pDNA concentration) by intravitreal injections at an interval of 1.5 months. The expression of ABCA4 in the retina was determined by RT-PCR and immunohistochemistry at 6 months after the second injection. A2E levels in the treated eyes and untreated controls were determined by HPLC. The safety of treatment was monitored by scanning laser ophthalmoscopy and electroretinogram (ERG). RESULTS: PEG-ELNP resulted in significant ABCA4 expression at both mRNA level and protein level at]6 months after 2 intravitreal injections, and a 40% A2E accumulation reduction compared with non-treated controls. The PEG-ELNP also demonstrated excellent safety as shown by scanning laser ophthalmoscopy, and the eye function evaluation from electroretinogram. CONCLUSIONS: Intravitreal delivery of the PEG-ELNP of pGRK1-ABCA4-S/MAR is a promising approach for gene therapy of Stargardt Disease, which can also be a delivery platform for gene therapy of other inherited retinal diseases.


Subject(s)
Nanoparticles , Retina , Mice , Animals , Stargardt Disease/genetics , Stargardt Disease/metabolism , Stargardt Disease/therapy , Retina/metabolism , Genetic Therapy/methods , Plasmids/genetics , DNA/metabolism , Mice, Knockout , Polyethylene Glycols/metabolism , Intravitreal Injections , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism
3.
Proc Natl Acad Sci U S A ; 120(50): e2314698120, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38064509

ABSTRACT

Mutations in many visual cycle enzymes in photoreceptors and retinal pigment epithelium (RPE) cells can lead to the chronic accumulation of toxic retinoid byproducts, which poison photoreceptors and the underlying RPE if left unchecked. Without a functional ATP-binding cassette, sub-family A, member 4 (ABCA4), there is an elevation of all-trans-retinal and prolonged buildup of all-trans-retinal adducts, resulting in a retinal degenerative disease known as Stargardt-1 disease. Even in this monogenic disorder, there is significant heterogeneity in the time to onset of symptoms among patients. Using a combination of molecular techniques, we studied Abca4 knockout (simulating human noncoding disease variants) and Abca4 knock-in mice (simulating human misfolded, catalytically inactive protein variants), which serve as models for Stargardt-1 disease. We compared the two strains to ascertain whether they exhibit differential responses to agents that affect cytokine signaling and/or ceramide metabolism, as alterations in either of these pathways can exacerbate retinal degenerative phenotypes. We found different degrees of responsiveness to maraviroc, a known immunomodulatory CCR5 antagonist, and to the ceramide-lowering agent AdipoRon, an agonist of the ADIPOR1 and ADIPOR2 receptors. The two strains also display different degrees of transcriptional deviation from matched WT controls. Our phenotypic comparison of the two distinct Abca4 mutant-mouse models sheds light on potential therapeutic avenues previously unexplored in the treatment of Stargardt disease and provides a surrogate assay for assessing the effectiveness for genome editing.


Subject(s)
Macular Degeneration , Retinal Degeneration , Humans , Mice , Animals , Stargardt Disease/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/genetics , Macular Degeneration/metabolism , Retinaldehyde/metabolism , Retina/metabolism , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Disease Models, Animal , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism
4.
Int J Mol Sci ; 24(8)2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37108442

ABSTRACT

The retina-specific ATP-binding cassette transporter protein ABCA4 is responsible for properly continuing the visual cycle by removing toxic retinoid byproducts of phototransduction. Functional impairment caused by ABCA4 sequence variations is the leading cause of autosomal recessive inherited retinal disorders, including Stargardt disease, retinitis pigmentosa, and cone-rod dystrophy. To date, more than 3000 ABCA4 genetic variants have been identified, approximately 40 percent of which have not been able to be classified for pathogenicity assessments. This study examined 30 missense ABCA4 variants using AlphaFold2 protein modeling and computational structure analysis for pathogenicity prediction. All variants classified as pathogenic (n = 10) were found to have deleterious structural consequences. Eight of the ten benign variants were structurally neutral, while the remaining two resulted in mild structural changes. This study's results provided multiple lines of computational pathogenicity evidence for eight ABCA4 variants of uncertain clinical significance. Overall, in silico analyses of ABCA4 can provide a valuable tool for understanding the molecular mechanisms of retinal degeneration and their pathogenic impact.


Subject(s)
Cone-Rod Dystrophies , Retinal Degeneration , Retinitis Pigmentosa , Humans , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retina/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Stargardt Disease/genetics , Stargardt Disease/metabolism , Cone-Rod Dystrophies/metabolism , Mutation , Pedigree , ATP-Binding Cassette Transporters/metabolism
5.
J Biol Chem ; 299(5): 104686, 2023 05.
Article in English | MEDLINE | ID: mdl-37031820

ABSTRACT

Dry age-related macular degeneration (AMD) and recessive Stargardt's disease (STGD1) lead to irreversible blindness in humans. The accumulation of all-trans-retinal (atRAL) induced by chaos in visual cycle is closely associated with retinal atrophy in dry AMD and STGD1 but its critical downstream signaling molecules remain ambiguous. Here, we reported that activation of eukaryotic translation initiation factor 2α (eIF2α) by atRAL promoted retinal degeneration and photoreceptor loss through activating c-Jun N-terminal kinase (JNK) signaling-dependent apoptosis and gasdermin E (GSDME)-mediated pyroptosis. We determined that eIF2α activation by atRAL in photoreceptor cells resulted from endoplasmic reticulum homeostasis disruption caused at least in part by reactive oxygen species production, and it activated JNK signaling independent of and dependent on activating transcription factor 4 and the activating transcription factor 4/transcription factor C/EBP homologous protein (CHOP) axis. CHOP overexpression induced apoptosis of atRAL-loaded photoreceptor cells through activating JNK signaling rather than inhibiting the expression of antiapoptotic gene Bcl2. JNK activation by eIF2α facilitated photoreceptor cell apoptosis caused by atRAL via caspase-3 activation and DNA damage. Additionally, we demonstrated that eIF2α was activated in neural retina of light-exposed Abca4-/-Rdh8-/- mice, a model that shows severe defects in atRAL clearance and displays primary features of human dry AMD and STGD1. Of note, inhibition of eIF2α activation by salubrinal effectively ameliorated retinal degeneration and photoreceptor apoptosis in Abca4-/-Rdh8-/- mice upon light exposure. The results of this study suggest that eIF2α is an important target to develop drug therapies for the treatment of dry AMD and STGD1.


Subject(s)
Eukaryotic Initiation Factor-2 , Retinal Degeneration , Retinaldehyde , Stargardt Disease , Animals , Humans , Mice , Activating Transcription Factor 4/metabolism , Apoptosis , ATP-Binding Cassette Transporters/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Retinaldehyde/metabolism , Stargardt Disease/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism
6.
J Biol Chem ; 299(5): 104614, 2023 05.
Article in English | MEDLINE | ID: mdl-36931393

ABSTRACT

N-retinylidene-phosphatidylethanolamine (N-Ret-PE), the Schiff-base conjugate formed through the reversible reaction of retinal (Vitamin A-aldehyde) and phosphatidylethanolamine, plays a crucial role in the visual cycle and visual pigment photoregeneration. However, N-Ret-PE can react with another molecule of retinal to form toxic di-retinoids if not removed from photoreceptors through its transport across photoreceptor membranes by the ATP-binding-cassette transporter ABCA4. Loss-of-function mutations in ABCA4 are known to cause Stargardt disease (STGD1), an inherited retinal degenerative disease associated with the accumulation of fluorescent di-retinoids and severe loss in vision. A larger assessment of retinal-phospholipid Schiff-base conjugates in photoreceptors is needed, along with further investigation of ABCA4 residues important for N-Ret-PE binding. In this study we show that N-Ret-PE formation is dependent on pH and phospholipid content. When retinal is added to liposomes or photoreceptor membranes, 40 to 60% is converted to N-Ret-PE at physiological pH. Phosphatidylserine and taurine also react with retinal to form N-retinylidene-phosphatidylserine and N-retinylidene-taurine, respectively, but at significantly lower levels. N-retinylidene-phosphatidylserine is not a substrate for ABCA4 and reacts poorly with retinal to form di-retinoids. Additionally, amino acid residues within the binding pocket of ABCA4 that contribute to its interaction with N-Ret-PE were identified and characterized using site-directed mutagenesis together with functional and binding assays. Substitution of arginine residues and hydrophobic residues with alanine or residues implicated in STGD1 significantly reduced or eliminated substrate-activated ATPase activity and substrate binding. Collectively, this study provides important insight into conditions which affect retinal-phospholipid Schiff-base formation and mechanisms underlying the pathogenesis of STGD1.


Subject(s)
Phospholipids , Stargardt Disease , Humans , ATP-Binding Cassette Transporters/metabolism , Phosphatidylserines , Retinoids/metabolism , Stargardt Disease/metabolism
7.
J Biol Chem ; 298(2): 101553, 2022 02.
Article in English | MEDLINE | ID: mdl-34973334

ABSTRACT

The breakdown of all-trans-retinal (atRAL) clearance is closely associated with photoreceptor cell death in dry age-related macular degeneration (AMD) and autosomal recessive Stargardt's disease (STGD1), but its mechanisms remain elusive. Here, we demonstrate that activation of gasdermin E (GSDME) but not gasdermin D promotes atRAL-induced photoreceptor damage by activating pyroptosis and aggravating apoptosis through a mitochondria-mediated caspase-3-dependent signaling pathway. Activation of c-Jun N-terminal kinase was identified as one of the major causes of mitochondrial membrane rupture in atRAL-loaded photoreceptor cells, resulting in the release of cytochrome c from mitochondria to the cytosol, where it stimulated caspase-3 activation required for cleavage of GSDME. Aggregation of the N-terminal fragment of GSDME in the mitochondria revealed that GSDME was likely to penetrate mitochondrial membranes in photoreceptor cells after atRAL exposure. ABC (subfamily A, member 4) and all-trans-retinol dehydrogenase 8 are two key proteins responsible for clearing atRAL in the retina. Abca4-/-Rdh8-/- mice exhibit serious defects in atRAL clearance upon light exposure and serve as an acute model for dry AMD and STGD1. We found that N-terminal fragment of GSDME was distinctly localized in the photoreceptor outer nuclear layer of light-exposed Abca4-/-Rdh8-/- mice. Of note, degeneration and caspase-3 activation in photoreceptors were significantly alleviated in Abca4-/-Rdh8-/-Gsdme-/- mice after exposure to light. The results of this study indicate that GSDME is a common causative factor of photoreceptor pyroptosis and apoptosis arising from atRAL overload, suggesting that repressing GSDME may represent a potential treatment of photoreceptor atrophy in dry AMD and STGD1.


Subject(s)
Photoreceptor Cells , Pore Forming Cytotoxic Proteins , Retina , Retinaldehyde , Stargardt Disease , ATP-Binding Cassette Transporters/metabolism , Animals , Caspase 3/metabolism , Mice , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Pore Forming Cytotoxic Proteins/metabolism , Retina/metabolism , Retina/pathology , Retinaldehyde/metabolism , Stargardt Disease/metabolism , Stargardt Disease/pathology
8.
Nat Commun ; 12(1): 5902, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34625547

ABSTRACT

ABCA4 is an ATP-binding cassette (ABC) transporter that flips N-retinylidene-phosphatidylethanolamine (N-Ret-PE) from the lumen to the cytoplasmic leaflet of photoreceptor membranes. Loss-of-function mutations cause Stargardt disease (STGD1), a macular dystrophy associated with severe vision loss. To define the mechanisms underlying substrate binding and STGD1, we determine the cryo-EM structure of ABCA4 in its substrate-free and bound states. The two structures are similar and delineate an elongated protein with the two transmembrane domains (TMD) forming an outward facing conformation, extended and twisted exocytoplasmic domains (ECD), and closely opposed nucleotide binding domains. N-Ret-PE is wedged between the two TMDs and a loop from ECD1 within the lumen leaflet consistent with a lateral access mechanism and is stabilized through hydrophobic and ionic interactions with residues from the TMDs and ECDs. Our studies provide a framework for further elucidating the molecular mechanism associated with lipid transport and disease and developing promising disease interventions.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Cryoelectron Microscopy/methods , Stargardt Disease/metabolism , Binding Sites , Biological Transport , HEK293 Cells , Humans , Macular Degeneration/genetics , Mutation , Phosphatidylethanolamines , Protein Domains , Retinoids , Stargardt Disease/genetics
9.
PLoS One ; 16(6): e0253716, 2021.
Article in English | MEDLINE | ID: mdl-34170959

ABSTRACT

PURPOSE: Preclinical research provides evidence for the complement system as a potential common pathway in Stargardt disease (STGD1) and age-related macular degeneration (AMD) leading to retinal pigment epithelium (RPE) loss. However, systemic complement activation has not yet been assessed in STGD1 patients. We conducted a cross-sectional case-control study to assess systemic complement activation in STGD1 patients and its association with disease severity. METHODS: Systemic concentrations of complement component C3 and its degradation product C3d were compared between 80 STGD1 patients and 80 controls that were frequency matched for age and sex. The C3d/C3 ratio was used as parameter of systemic complement activation. Within the STGD1 cohort, we additionally examined the association between the C3d/C3 ratio, demographic and behavioural factors (age, sex, smoking and BMI), and measures of disease severity (age at onset, visual acuity, and area of atrophy). RESULTS: The C3d/C3 ratio did not significantly differ between patients (mean C3d/C3 ratio 3.5±1.4) and controls (mean C3d/C3 ratio 3.6±1.0), mean difference -0.156 (p = 0.804, independent samples t-test). The overall effect size was 8% (95% confidence interval, 3-15%). Elevated C3d/C3 ratios (>8.1) were found in three patients who all had a concomitant inflammatory condition at the time of blood draw. Within the patient cohort, C3 levels were associated with sex (mean difference -134, p = 0.001, independent samples t-test) and BMI (correlation coefficient 0.463, p<0.001, Spearman's Correlation). CONCLUSIONS: Systemic complement levels were not elevated in STGD1 patients compared to age and sex matched controls and was not associated with STGD1 severity. Considering the continued absent proof of a systemic contribution of the complement system to RPE loss in STGD1 patients, we hypothesize that complement activation in STGD1 is more likely a local process. In light of upcoming complement-targeted therapies, further studies are needed that measure complement levels in the eye of STGD1 patients.


Subject(s)
Complement Activation , Complement C3d/metabolism , Retinal Pigment Epithelium/metabolism , Sex Characteristics , Stargardt Disease/blood , Stargardt Disease/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Humans , Male , Middle Aged
10.
Acta Ophthalmol ; 99(8): e1396-e1404, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33973369

ABSTRACT

PURPOSE: The aim of our study was to evaluate retinal microvascular changes recorded with optical coherence tomography angiography (OCTA) and the metabolic function measured with retinal oximetry (RO) in patients with retinitis pigmentosa (RP) and Stargardt disease (STGD). METHODS: In this prospective, noninterventional study, OCTA and RO were performed on 107 eyes (56 subjects): 53 eyes diagnosed with RP without the presence of macular oedema (no-ME-RP), 26 eyes with STGD, and 28 control eyes. Main outcome measures were the mean superficial (FAZ-S; mm2 ) and deep foveal avascular zone (FAZ-D; mm2 ) measured with OCTA as well as the mean arterial (A-SO2 ; %), venular (V-SO2 ; %) oxygen saturation, their difference (A-V SO2 ; %) and the corresponding mean diameters of the peripapillary retinal arterioles (D-A; µm) and venules (D-V; µm) determined with RO. RESULTS: Stargardt disease (STGD) patients differed from controls and no-ME-RP by an enlarged FAZ-S and reduced A-SO2 and V-SO2 (p ≤ 0.013). No-ME-RP eyes presented with attenuated vessels (p < 0.001) and increased A-SO2 and V-SO2 (p ≤ 0.012) compared to controls and STGD. The FAZ-D showed significant interactions with A-SO2 (p = 0.003) in no-ME-RP while the FAZ-S correlated with visual acuity in no-ME-RP (p = 0.007) and STGD (p = 0.034). CONCLUSION: Retinitis pigmentosa (RP) and Stargardt disease (STGD) patients suffer from microvascular and metabolic alterations, however, showing a different pattern. A combined microvascular-metabolic model may therefore allow to more precisely characterize RP and STGD as well as presumably other inherited retinal diseases.


Subject(s)
Microvessels/metabolism , Oxygen Saturation/physiology , Retinal Vessels/metabolism , Retinitis Pigmentosa/metabolism , Stargardt Disease/metabolism , Visual Acuity , Adult , Aged , Aged, 80 and over , Electroretinography , Fluorescein Angiography/methods , Fundus Oculi , Humans , Microvessels/physiopathology , Middle Aged , Oximetry/methods , Prospective Studies , Retinal Vessels/diagnostic imaging , Retinal Vessels/physiopathology , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/physiopathology , Stargardt Disease/diagnosis , Stargardt Disease/physiopathology , Tomography, Optical Coherence/methods , Young Adult
11.
J Biol Chem ; 296: 100259, 2021.
Article in English | MEDLINE | ID: mdl-33837742

ABSTRACT

The ability of iron to transfer electrons enables the contribution of this metal to a variety of cellular activities even as the redox properties of iron are also responsible for the generation of hydroxyl radicals (•OH), the most destructive of the reactive oxygen species. We previously showed that iron can promote the oxidation of bisretinoid by generating highly reactive hydroxyl radical (•OH). Now we report that preservation of iron regulation in the retina is not sufficient to prevent iron-induced bisretinoid oxidative degradation when blood iron levels are elevated in liver-specific hepcidin knockout mice. We obtained evidence for the perpetuation of Fenton reactions in the presence of the bisretinoid A2E and visible light. On the other hand, iron chelation by deferiprone was not associated with changes in postbleaching recovery of 11-cis-retinal or dark-adapted ERG b-wave amplitudes indicating that the activity of Rpe65, a rate-determining visual cycle protein that carries an iron-binding domain, is not affected. Notably, iron levels were elevated in the neural retina and retinal pigment epithelial (RPE) cells of Abca4-/- mice. Consistent with higher iron content, ferritin-L immunostaining was elevated in RPE of a patient diagnosed with ABCA4-associated disease and in RPE and photoreceptor cells of Abca4-/- mice. In neural retina of the mutant mice, reduced Tfrc mRNA was also an indicator of retinal iron overload. Thus iron chelation may defend retina when bisretinoid toxicity is implicated in disease processes.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Genes, Recessive , Retinaldehyde/metabolism , Retinoids/metabolism , Stargardt Disease/metabolism , cis-trans-Isomerases/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Mice , Mice, Knockout , Oxidation-Reduction , Retinaldehyde/genetics , Retinoids/genetics , Stargardt Disease/genetics , Stargardt Disease/pathology , cis-trans-Isomerases/genetics
12.
Sci Rep ; 11(1): 1466, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33446864

ABSTRACT

Spatially-resolved retinal function can be measured by psychophysical testing like fundus-controlled perimetry (FCP or 'microperimetry'). It may serve as a performance outcome measure in emerging interventional clinical trials for macular diseases as requested by regulatory agencies. As FCP constitute laborious examinations, we have evaluated a machine-learning-based approach to predict spatially-resolved retinal function ('inferred sensitivity') based on microstructural imaging (obtained by spectral domain optical coherence tomography) and patient data in recessive Stargardt disease. Using nested cross-validation, prediction accuracies of (mean absolute error, MAE [95% CI]) 4.74 dB [4.48-4.99] were achieved. After additional inclusion of limited FCP data, the latter reached 3.89 dB [3.67-4.10] comparable to the test-retest MAE estimate of 3.51 dB [3.11-3.91]. Analysis of the permutation importance revealed, that the IS&OS and RPE thickness were the most important features for the prediction of retinal sensitivity. 'Inferred sensitivity', herein, enables to accurately estimate differential effects of retinal microstructure on spatially-resolved function in Stargardt disease, and might be used as quasi-functional surrogate marker for a refined and time-efficient investigation of possible functionally relevant treatment effects or disease progression.


Subject(s)
Retina/physiopathology , Stargardt Disease/physiopathology , Visual Field Tests/methods , Adult , Female , Fundus Oculi , Humans , Image Processing, Computer-Assisted/methods , Machine Learning , Macular Degeneration/physiopathology , Male , Middle Aged , Retinal Diseases/physiopathology , Stargardt Disease/metabolism , Tomography, Optical Coherence/methods , Visual Acuity , Visual Fields
13.
J Biol Chem ; 296: 100187, 2021.
Article in English | MEDLINE | ID: mdl-33334878

ABSTRACT

The death of photoreceptor cells in dry age-related macular degeneration (AMD) and autosomal recessive Stargardt disease (STGD1) is closely associated with disruption in all-trans-retinal (atRAL) clearance in neural retina. In this study, we reveal that the overload of atRAL leads to photoreceptor degeneration through activating ferroptosis, a nonapoptotic form of cell death. Ferroptosis of photoreceptor cells induced by atRAL resulted from increased ferrous ion (Fe2+), elevated ACSL4 expression, system Xc- inhibition, and mitochondrial destruction. Fe2+ overload, tripeptide glutathione (GSH) depletion, and damaged mitochondria in photoreceptor cells exposed to atRAL provoked reactive oxygen species (ROS) production, which, together with ACSL4 activation, promoted lipid peroxidation and thereby evoked ferroptotic cell death. Moreover, exposure of photoreceptor cells to atRAL activated COX2, a well-accepted biomarker for ferroptosis onset. In addition to GSH supplement, inhibiting either Fe2+ by deferoxamine mesylate salt (DFO) or lipid peroxidation with ferrostatin-1 (Fer-1) protected photoreceptor cells from ferroptosis caused by atRAL. Abca4-/-Rdh8-/- mice exhibiting defects in atRAL clearance is an animal model for dry AMD and STGD1. We observed that ferroptosis was indeed present in neural retina of Abca4-/-Rdh8-/- mice after light exposure. More importantly, photoreceptor atrophy and ferroptosis in light-exposed Abca4-/-Rdh8-/- mice were effectively alleviated by intraperitoneally injected Fer-1, a selective inhibitor of ferroptosis. Our study suggests that ferroptosis is one of the important pathways of photoreceptor cell death in retinopathies arising from excess atRAL accumulation and should be pursued as a novel target for protection against dry AMD and STGD1.


Subject(s)
Ferroptosis , Lipid Peroxidation , Macular Degeneration/pathology , Photoreceptor Cells, Vertebrate/pathology , Retinaldehyde/analogs & derivatives , Animals , Macular Degeneration/metabolism , Mice , Mice, Inbred C57BL , Oxidative Stress , Photoreceptor Cells, Vertebrate/metabolism , Reactive Oxygen Species/metabolism , Retinaldehyde/metabolism , Stargardt Disease/metabolism , Stargardt Disease/pathology
14.
Exp Eye Res ; 202: 108389, 2021 01.
Article in English | MEDLINE | ID: mdl-33301772

ABSTRACT

ABCA4 gene associated retinal dystrophies (ABCA4-RD) are a group of inherited eye diseases caused by ABCA4 gene mutations, including Stargardt disease, cone-rod dystrophy and retinitis pigmentosa. With the development of next-generation sequencing (NGS), numerous clinical and genetic studies on ABCA4-RD have been performed, and the genotype and phenotype spectra have been elucidated. However, most of the studies focused on the Caucasian population and limited studies of large Chinese ABCA4-RD cohorts were reported. In this study, we summarized the phenotypic and genotypic characteristics of 129 Chinese patients with ABCA4-RD. We found a mutation spectrum of Chinese patients which is considerably different from that of the Caucasian population and identified 35 novel ABCA4 mutations. We also reported some rare and special cases, such as, pedigrees with patients in two generations, patients diagnosed with cone-rod dystrophy or retinitis pigmentosa, patients with subretinal fibrosis and patients with preserved foveal structure. At the same time, we focused on the correlation between the genotypes and phenotypes. By the comprehensive analysis of multiple clinical examinations and the application of multiple regression analysis, we proved that patients with two "null" variants had a younger onset age and reached legal blindness earlier than patients with two "none-null" variants. Patients with one or more "none-null" variants tended to have better visual acuity and presented with milder fundus autofluorescence changes and more preserved rod functions on the full-field electroretinography than patients with two "null" variants. Furthermore, most patients with the p.(Phe2188Ser) variant shared a mild phenotype with a low fundus autofluorescence signal limited to the fovea and with normal full-field electroretinography responses. Our findings expand the variant spectrum of the ABCA4 gene and enhance the knowledge of Chinese patients with ABCA4-RD.


Subject(s)
ATP-Binding Cassette Transporters/genetics , DNA/genetics , Mutation , Stargardt Disease/genetics , ATP-Binding Cassette Transporters/metabolism , Adolescent , Adult , Aged , Child , Child, Preschool , China/epidemiology , DNA Mutational Analysis , Female , Fluorescein Angiography , Fundus Oculi , Genetic Testing , Genotype , Humans , Male , Middle Aged , Pedigree , Phenotype , Retrospective Studies , Rod Cell Outer Segment/pathology , Stargardt Disease/epidemiology , Stargardt Disease/metabolism , Visual Acuity , Young Adult
15.
Int J Mol Sci ; 22(1)2020 Dec 27.
Article in English | MEDLINE | ID: mdl-33375396

ABSTRACT

ABCA4 is an ATP-binding cassette (ABC) transporter expressed in photoreceptors, where it transports its substrate, N-retinylidene-phosphatidylethanolamine (N-Ret-PE), across outer segment membranes to facilitate the clearance of retinal from photoreceptors. Mutations in ABCA4 cause Stargardt macular degeneration (STGD1), an autosomal recessive disorder characterized by a loss of central vision and the accumulation of bisretinoid compounds. The purpose of this study was to determine the molecular properties of ABCA4 variants harboring disease-causing missense mutations in the transmembrane domains. Thirty-eight variants expressed in culture cells were analyzed for expression, ATPase activities, and substrate binding. On the basis of these properties, the variants were divided into three classes: Class 1 (severe variants) exhibited significantly reduced ABCA4 expression and basal ATPase activity that was not stimulated by its substrate N-Ret-PE; Class 2 (moderate variants) showed a partial reduction in expression and basal ATPase activity that was modestly stimulated by N-Ret-PE; and Class 3 (mild variants) displayed expression and functional properties comparable to normal ABCA4. The p.R653C variant displayed normal expression and basal ATPase activity, but lacked substrate binding and ATPase activation, suggesting that arginine 653 contributes to N-Ret-PE binding. Our classification provides a basis for better understanding genotype-phenotype correlations and evaluating therapeutic treatments for STGD1.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Biological Transport, Active/genetics , Stargardt Disease/genetics , Stargardt Disease/metabolism , ATP-Binding Cassette Transporters/chemistry , Animals , COS Cells , Chlorocebus aethiops , Fluorescent Antibody Technique , Gene Expression , Genetic Association Studies , HEK293 Cells , Humans , Models, Molecular , Mutation, Missense , Phosphatidylethanolamines/metabolism , Protein Binding , Protein Domains , Retinal Diseases/congenital , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinoids/metabolism , Stargardt Disease/enzymology
16.
Int J Mol Sci ; 21(22)2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33187113

ABSTRACT

Stargardt macular degeneration is an inherited retinal disease caused by mutations in the ATP-binding cassette subfamily A member 4 (ABCA4) gene. Here, we characterized the complement expression profile in ABCA4-/- retinae and aligned these findings with morphological markers of retinal degeneration. We found an enhanced retinal pigment epithelium (RPE) autofluorescence, cell loss in the inner retina of ABCA4-/- mice and demonstrated age-related differences in complement expression in various retinal cell types irrespective of the genotype. However, 24-week-old ABCA4-/- mice expressed more c3 in the RPE and fewer cfi transcripts in the microglia compared to controls. At the protein level, the decrease of complement inhibitors (complement factor I, CFI) in retinae, as well as an increased C3b/C3 ratio in the RPE/choroid and retinae of ABCA4-/-, mice was confirmed. We showed a corresponding increase of the C3d/C3 ratio in the serum of ABCA4-/- mice, while no changes were observed for CFI. Our findings suggest an overactive complement cascade in the ABCA4-/- retinae that possibly contributes to pathological alterations, including microglial activation and neurodegeneration. Overall, this underpins the importance of well-balanced complement homeostasis to maintain retinal integrity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Complement System Proteins/metabolism , Stargardt Disease/metabolism , Animals , Choroid/metabolism , Complement Activation/physiology , Disease Models, Animal , Female , Macular Degeneration/metabolism , Male , Mice , Mice, Inbred BALB C , Microglia/metabolism , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism
17.
Pharmacol Res Perspect ; 8(6): e00683, 2020 12.
Article in English | MEDLINE | ID: mdl-33164337

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of blindness in older people in the developed world while Stargardt's disease (SD) is a juvenile macular degeneration and an orphan disease. Both diseases are untreatable and are marked by accumulation of lipofuscin advancing to progressive deterioration of the retinal pigment epithelium (RPE) and retina and subsequent vision loss till blindness. We discovered that a small molecule belonging to the tetrahydropyridoether class of compounds, soraprazan renamed remofuscin, is able to remove existing lipofuscin from the RPE. This study investigated the drug penetration, distribution, and elimination into the eyes of a mouse model for increased lipofuscinogenesis, following a single intravitreal injection. We measured the time course of concentrations of remofuscin in different eye tissues using high-performance liquid chromatography combined with mass spectroscopy (HPLC-MS). We also visualized the penetration and distribution of 3 H-remofuscin in eye sections up to 20 weeks post-injection using transmission electron microscopic (TEM) autoradiography. The distribution of silver grains revealed that remofuscin accumulated specifically in the RPE by binding to the RPE pigments (melanin, lipofuscin and melanolipofuscin) and that it was still detected after 20 weeks. Importantly, the melanosomes in choroidal melanocytes only rarely bind remofuscin emphasizing its potential to serve as an active ingredient in the RPE for the treatment of SD and dry AMD. In addition, our study highlights the importance of electron microscopic autoradiography as it is the only method able to show drug binding with a high intracellular resolution.


Subject(s)
Autoradiography/methods , Imidazoles/pharmacokinetics , Intravitreal Injections/methods , Macular Degeneration/metabolism , Microscopy, Electron, Transmission/methods , Naphthyridines/pharmacokinetics , Stargardt Disease/metabolism , Animals , Female , Imidazoles/administration & dosage , Imidazoles/analysis , Macular Degeneration/drug therapy , Macular Degeneration/genetics , Male , Mice , Mice, Transgenic , Naphthyridines/administration & dosage , Naphthyridines/analysis , Stargardt Disease/drug therapy , Stargardt Disease/genetics , Treatment Outcome , Tritium/administration & dosage , Tritium/analysis , Tritium/pharmacokinetics
18.
Sci Rep ; 10(1): 16491, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020556

ABSTRACT

Stargardt disease is one of the most common forms of inherited retinal disease and leads to permanent vision loss. A diagnostic feature of the disease is retinal flecks, which appear hyperautofluorescent in fundus autofluorescence (FAF) imaging. The size and number of these flecks increase with disease progression. Manual segmentation of flecks allows monitoring of disease, but is time-consuming. Herein, we have developed and validated a deep learning approach for segmenting these Stargardt flecks (1750 training and 100 validation FAF patches from 37 eyes with Stargardt disease). Testing was done in 10 separate Stargardt FAF images and we observed a good overall agreement between manual and deep learning in both fleck count and fleck area. Longitudinal data were available in both eyes from 6 patients (average total follow-up time 4.2 years), with both manual and deep learning segmentation performed on all (n = 82) images. Both methods detected a similar upward trend in fleck number and area over time. In conclusion, we demonstrated the feasibility of utilizing deep learning to segment and quantify FAF lesions, laying the foundation for future studies using fleck parameters as a trial endpoint.


Subject(s)
Stargardt Disease/pathology , Zinc Phosphate Cement/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Child , Deep Learning , Electroretinography/methods , Female , Fluorescein Angiography/methods , Fundus Oculi , Humans , Longitudinal Studies , Male , Middle Aged , Ophthalmoscopy/methods , Optical Imaging/methods , Retina/metabolism , Retina/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Stargardt Disease/metabolism , Tomography, Optical Coherence/methods , Young Adult
19.
J Biol Chem ; 295(20): 6958-6971, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32265302

ABSTRACT

Disrupted clearance of all-trans-retinal (atRAL), a component of the visual (retinoid) cycle in the retina, may cause photoreceptor atrophy in autosomal recessive Stargardt disease (STGD1) and dry age-related macular degeneration (AMD). However, the mechanisms underlying atRAL-induced photoreceptor loss remain elusive. Here, we report that atRAL activates c-Jun N-terminal kinase (JNK) signaling at least partially through reactive oxygen species production, which promoted mitochondria-mediated caspase- and DNA damage-dependent apoptosis in photoreceptor cells. Damage to mitochondria in atRAL-exposed photoreceptor cells resulted from JNK activation, leading to decreased expression of Bcl2 apoptosis regulator (Bcl2), increased Bcl2 antagonist/killer (Bak) levels, and cytochrome c (Cyt c) release into the cytosol. Cytosolic Cyt c specifically provoked caspase-9 and caspase-3 activation and thereby initiated apoptosis. Phosphorylation of JNK in atRAL-loaded photoreceptor cells induced the appearance of γH2AX, a sensitive marker for DNA damage, and was also associated with apoptosis onset. Suppression of JNK signaling protected photoreceptor cells against atRAL-induced apoptosis. Moreover, photoreceptor cells lacking Jnk1 and Jnk2 genes were more resistant to atRAL-associated cytotoxicity. The Abca4-/-Rdh8-/- mouse model displays defects in atRAL clearance that are characteristic of STGD1 and dry AMD. We found that JNK signaling was activated in the neural retina of light-exposed Abca4-/-Rdh8-/- mice. Of note, intraperitoneal administration of JNK-IN-8, which inhibits JNK signaling, effectively ameliorated photoreceptor degeneration and apoptosis in light-exposed Abca4-/-Rdh8-/- mice. We propose that pharmacological inhibition of JNK signaling may represent a therapeutic strategy for preventing photoreceptor loss in retinopathies arising from atRAL overload.


Subject(s)
Apoptosis/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinaldehyde/pharmacology , Signal Transduction/drug effects , Stargardt Disease/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Apoptosis/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Photoreceptor Cells, Vertebrate/pathology , Signal Transduction/genetics , Stargardt Disease/genetics , Stargardt Disease/pathology
20.
Aging (Albany NY) ; 12(7): 6151-6171, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32255762

ABSTRACT

Atrophic A\age-related macular degeneration (AMD) and Stargardt disease (STGD) are major blinding diseases affecting millions of patients worldwide, but no treatment is available. In dry AMD and STGD oxidative stress and subretinal accumulation of N-retinylidene-N-retinylethanolamine (A2E), a toxic by-product of the visual cycle, causes retinal pigment epithelium (RPE) and photoreceptor degeneration leading to visual impairment. Acute and chronic retinal degeneration following blue light damage (BLD) in BALB/c mice and aging of Abca4-/- Rdh8-/- mice, respectively, reproduce features of AMD and STGD. Efficacy of systemic administrations of 9'-cis-norbixin (norbixin), a natural di-apocarotenoid, prepared from Bixa orellana seeds with anti-oxidative properties, was evaluated during BLD in BALB/c mice, and in Abca4-/- Rdh8-/- mice of different ages, following three experimental designs: "preventive", "early curative" and "late curative" supplementations. Norbixin injected intraperitoneally in BALB/c mice, maintained scotopic and photopic electroretinogram amplitude and was neuroprotective. Norbixin chronic oral administration for 6 months in Abca4-/- Rdh8-/- mice following the "early curative" supplementation showed optimal neuroprotection and maintenance of photoreceptor function and reduced ocular A2E accumulation. Thus, norbixin appears promising as a systemic drug candidate for both AMD and STGD treatment.


Subject(s)
Carotenoids/pharmacology , Macular Degeneration , Photoreceptor Cells, Vertebrate , Retinoids , Stargardt Disease , Animals , Drug Monitoring/methods , Electroretinography/methods , Injections, Intraperitoneal , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/prevention & control , Mice , Neuroprotective Agents/pharmacology , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Retinoids/antagonists & inhibitors , Retinoids/metabolism , Stargardt Disease/drug therapy , Stargardt Disease/metabolism , Stargardt Disease/prevention & control , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...