Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 326(4): C1178-C1192, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38406825

RESUMO

K+ channel Kir7.1 expressed at the apical membrane of the retinal pigment epithelium (RPE) plays an essential role in retinal function. An isoleucine-to-threonine mutation at position 120 of the protein is responsible for blindness-causing vitreo-retinal dystrophy. We have studied the molecular mechanism of action of Kir7.1-I120T in vitro by heterologous expression and in vivo in CRISPR-generated knockin mice. Full-size Kir7.1-I120T reaches the plasma membrane but lacks any activity. Analysis of Kir7.1 and the I120T mutant in mixed transfection experiments, and that of tandem tetrameric constructs made by combining wild type (WT) and mutant protomers, leads us to conclude that they do not form heterotetramers in vitro. Homozygous I120T/I120T mice show cleft palate and tracheomalacia and do not survive beyond P0, whereas heterozygous WT/I120T develop normally. Membrane conductance of RPE cells isolated from WT/WT and heterozygous WT/I120T mice is dominated by Kir7.1 current. Using Rb+ as a charge carrier, we demonstrate that the Kir7.1 current of WT/I120T RPE cells corresponds to approximately 50% of that in cells from WT/WT animals, in direct proportion to WT gene dosage. This suggests a lack of compensatory effects or interference from the mutated allele product, an interpretation consistent with results obtained using WT/- hemizygous mouse. Electroretinography and behavioral tests also show normal vision in WT/I120T animals. The hypomorphic ion channel phenotype of heterozygous Kir7.1-I120T mutants is therefore compatible with normal development and retinal function. The lack of detrimental effect of this degree of functional deficit might explain the recessive nature of Kir7.1 mutations causing human eye disease.NEW & NOTEWORTHY Human retinal pigment epithelium K+ channel Kir7.1 is affected by generally recessive mutations leading to blindness. We investigate one such mutation, isoleucine-to-threonine at position 120, both in vitro and in vivo in knockin mice. The mutated channel is inactive and in heterozygosis gives a hypomorphic phenotype with normal retinal function. Mutant channels do not interfere with wild-type Kir7.1 channels which are expressed concomitantly without hindrance, providing an explanation for the recessive nature of the disease.


Assuntos
Isoleucina , Retina , Camundongos , Humanos , Animais , Isoleucina/metabolismo , Retina/metabolismo , Cegueira/metabolismo , Mutação/genética , Treonina/metabolismo
2.
Cell Mol Neurobiol ; 43(3): 1037-1048, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35792991

RESUMO

Retinitis pigmentosa (RP) is a group of genetic disorders resulting in inherited blindness due to the degeneration of rod and cone photoreceptors. The various mechanisms underlying rod degeneration primarily rely on genetic mutations, leading to night blindness initially. Cones gradually degenerate after rods are almost eliminated, resulting in varying degrees of visual disability and blindness. The mechanism of cone degeneration remains unclear. An understanding of the mechanisms underlying cone degeneration in RP, a highly heterogeneous disease, is essential to develop novel treatments of RP. Herein, we review recent advancements in the five hypotheses of cone degeneration, including oxidative stress, trophic factors, metabolic stress, light damage, and inflammation activation. We also discuss the connection among these theories to provide a better understanding of secondary cone degeneration in RP. Five current mechanisms of cone degenerations in RP Interactions among different pathways are involved in RP.


Assuntos
Células Fotorreceptoras Retinianas Cones , Retinose Pigmentar , Humanos , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Retinose Pigmentar/terapia , Cegueira/metabolismo , Estresse Oxidativo
3.
FASEB J ; 36(5): e22309, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35471581

RESUMO

RAB28 is a farnesylated, ciliary G-protein. Patient variants in RAB28 are causative of autosomal recessive cone-rod dystrophy (CRD), an inherited human blindness. In rodent and zebrafish models, the absence of Rab28 results in diminished dawn, photoreceptor, outer segment phagocytosis (OSP). Here, we demonstrate that Rab28 is also required for dusk peaks of OSP, but not for basal OSP levels. This study further elucidated the molecular mechanisms by which Rab28 controls OSP and inherited blindness. Proteomic profiling identified factors whose expression in the eye or whose expression at dawn and dusk peaks of OSP is dysregulated by loss of Rab28. Notably, transgenic overexpression of Rab28, solely in zebrafish cones, rescues the OSP defect in rab28 KO fish, suggesting rab28 gene replacement in cone photoreceptors is sufficient to regulate Rab28-OSP. Rab28 loss also perturbs function of the visual cycle as retinoid levels of 11-cRAL, 11cRP, and atRP are significantly reduced in larval and adult rab28 KO retinae (p < .05). These data give further understanding on the molecular mechanisms of RAB28-associated CRD, highlighting roles of Rab28 in both peaks of OSP, in vitamin A metabolism and in retinoid recycling.


Assuntos
Proteômica , Peixe-Zebra , Animais , Cegueira/metabolismo , Humanos , Fagocitose , Células Fotorreceptoras Retinianas Cones/metabolismo , Retinoides/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
4.
Int J Mol Sci ; 23(21)2022 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-36361918

RESUMO

Corneal blindness is the fifth leading cause of blindness worldwide, and therapeutic options are still often limited to corneal transplantation. The corneal epithelium has a strong barrier function, and regeneration is highly dependent on limbal stem cell proliferation and basement membrane remodeling. As a result of the lack of corneal donor tissues, regenerative medicine for corneal diseases affecting the epithelium is an area with quite advanced basic and clinical research. Surgery still plays a prominent role in the treatment of epithelial diseases; indeed, innovative surgical techniques have been developed to transplant corneal and non-corneal stem cells onto diseased corneas for epithelial regeneration applications. The main goal of applying regenerative medicine to clinical practice is to restore function by providing viable cells based on the use of a novel therapeutic approach to generate biological substitutes and improve tissue functions. Interest in corneal epithelium rehabilitation medicine is rapidly growing, given the exposure of the corneal outer layers to external insults. Here, we performed a review of basic, clinical and surgical research reports on regenerative medicine for corneal epithelial disorders, classifying therapeutic approaches according to their macro- or microscopic target, i.e., into cellular or subcellular therapies, respectively.


Assuntos
Doenças da Córnea , Epitélio Corneano , Humanos , Epitélio Corneano/metabolismo , Doenças da Córnea/terapia , Córnea , Células-Tronco/metabolismo , Cegueira/metabolismo , Células Epiteliais
5.
Chem Senses ; 45(7): 523-531, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32766717

RESUMO

It is well established that early blindness results in enhancement of the remaining nonvisual sensory modalities accompanied by functional and anatomical brain plasticity. While auditory and tactile functions have been largely investigated, the results regarding olfactory functions remained less explored and less consistent. In the present study, we investigated olfactory function in blind mice using 3 tests: the buried food test, the olfactory threshold test, and the olfactory performance test. The results indicated better performance of blind mice in the buried food test and odor performance test while there was no difference in the olfactory threshold test. Using histological measurements, we also investigated if there was anatomical plasticity in the olfactory bulbs (OB), the most salient site for olfactory processing. The results indicated a larger volume of the OB driven by larger glomerular and granular layers in blind mice compared with sighted mice. Structural plasticity in the OB may underlie the enhanced olfactory performance in blind mice.


Assuntos
Cegueira/patologia , Bulbo Olfatório/fisiologia , Percepção Olfatória/fisiologia , Animais , Cegueira/congênito , Cegueira/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Bulbo Olfatório/patologia , Olfato/fisiologia
6.
Proc Natl Acad Sci U S A ; 114(26): 6830-6835, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28607055

RESUMO

Sensory deprivation reorganizes neurocircuits in the human brain. The biological basis of such neuroplastic adaptations remains elusive. In this study, we applied two complementary graph theory-based functional connectivity analyses, one to evaluate whole-brain functional connectivity relationships and the second to specifically delineate distributed network connectivity profiles downstream of primary sensory cortices, to investigate neural reorganization in blind children compared with sighted controls. We also examined the relationship between connectivity changes and neuroplasticity-related gene expression profiles in the cerebral cortex. We observed that multisensory integration areas exhibited enhanced functional connectivity in blind children and that this reorganization was spatially associated with the transcription levels of specific members of the cAMP Response Element Binding protein gene family. Using systems-level analyses, this study advances our understanding of human neuroplasticity and its genetic underpinnings following sensory deprivation.


Assuntos
Cegueira/metabolismo , Regulação da Expressão Gênica , Rede Nervosa/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Plasticidade Neuronal , Córtex Somatossensorial/metabolismo , Cegueira/patologia , Criança , Feminino , Humanos , Masculino , Rede Nervosa/patologia , Córtex Somatossensorial/patologia
7.
Proc Natl Acad Sci U S A ; 114(32): E6536-E6545, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28739921

RESUMO

FKBP-domain proteins (FKBPs) are pivotal modulators of cellular signaling, protein folding, and gene transcription. Aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) is a distinctive member of the FKBP superfamily in terms of its biochemical properties, and it plays an important biological role as a chaperone of phosphodiesterase 6 (PDE6), an effector enzyme of the visual transduction cascade. Malfunction of mutant AIPL1 proteins triggers a severe form of Leber congenital amaurosis and leads to blindness. The mechanism underlying the chaperone activity of AIPL1 is largely unknown, but involves the binding of isoprenyl groups on PDE6 to the FKBP domain of AIPL1. We solved the crystal structures of the AIPL1-FKBP domain and its pathogenic mutant V71F, both in the apo form and in complex with isoprenyl moieties. These structures reveal a module for lipid binding that is unparalleled within the FKBP superfamily. The prenyl binding is enabled by a unique "loop-out" conformation of the ß4-α1 loop and a conformational "flip-out" switch of the key W72 residue. A second major conformation of apo AIPL1-FKBP was identified by NMR studies. This conformation, wherein W72 flips into the ligand-binding pocket and renders the protein incapable of prenyl binding, is supported by molecular dynamics simulations and appears to underlie the pathogenicity of the V71F mutant. Our findings offer critical insights into the mechanisms that underlie AIPL1 function in health and disease, and highlight the structural and functional diversity of the FKBPs.


Assuntos
Cegueira/genética , Cegueira/metabolismo , Proteínas de Transporte/química , Proteínas do Olho/química , Proteínas Adaptadoras de Transdução de Sinal , Substituição de Aminoácidos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Cristalografia por Raios X , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/química , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Humanos , Lipídeos , Mutação de Sentido Incorreto , Ressonância Magnética Nuclear Biomolecular , Domínios Proteicos , Estrutura Secundária de Proteína
8.
Mol Ther ; 26(6): 1581-1593, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29673930

RESUMO

Most genetically distinct inherited retinal degenerations are primary photoreceptor degenerations. We selected a severe early onset form of Leber congenital amaurosis (LCA), caused by mutations in the gene LCA5, in order to test the efficacy of gene augmentation therapy for a ciliopathy. The LCA5-encoded protein, Lebercilin, is essential for the trafficking of proteins and vesicles to the photoreceptor outer segment. Using the AAV serotype AAV7m8 to deliver a human LCA5 cDNA into an Lca5 null mouse model of LCA5, we show partial rescue of retinal structure and visual function. Specifically, we observed restoration of rod-and-cone-driven electroretinograms in about 25% of injected eyes, restoration of pupillary light responses in the majority of treated eyes, an ∼20-fold decrease in target luminance necessary for visually guided behavior, and improved retinal architecture following gene transfer. Using LCA5 patient-derived iPSC-RPEs, we show that delivery of the LCA5 cDNA restores lebercilin protein and rescues cilia quantity. The results presented in this study support a path forward aiming to develop safety and efficacy trials for gene augmentation therapy in human subjects with LCA5 mutations. They also provide the framework for measuring the effects of intervention in ciliopathies and other severe, early-onset blinding conditions.


Assuntos
Cegueira/metabolismo , Cegueira/terapia , Dependovirus/genética , Terapia Genética/métodos , Animais , Eletrorretinografia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Feminino , Humanos , Amaurose Congênita de Leber/metabolismo , Amaurose Congênita de Leber/terapia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo
9.
Ophthalmology ; 125(8): 1160-1171, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29625838

RESUMO

PURPOSE: Although most totally visually blind individuals exhibit nonentrained circadian rhythms due to an inability of light to entrain the circadian pacemaker, a small proportion retain photic circadian entrainment, melatonin suppression, and other nonimage-forming responses to light. It is thought that these responses to light persist because of the survival of melanospin-containing intrinsically photosensitive retinal ganglion cells (ipRGCs), which project primarily to the circadian pacemaker and are functionally distinct from the rod and cone photoreceptors that mediate vision. We aimed to assess the integrity of nonimage-forming photoreception in totally visually blind patients with a range of ocular disorders. DESIGN: Within-subject, dark-controlled design. PARTICIPANTS: A total of 18 totally visually blind individuals (7 females; mean age ± standard deviation = 49.8±11.0 years) with various causes of blindness, including 3 bilaterally enucleated controls. METHODS: Melatonin concentrations were compared during exposure to a 6.5-hour bright white light (∼7000 lux) with melatonin concentrations measured 24 hours earlier at the corresponding clock times under dim-light (4 lux) conditions. MAIN OUTCOME MEASURES: Area under the curve (AUC) for melatonin concentration. RESULTS: Melatonin concentrations were significantly suppressed (defined as ≥33% suppression) during the bright-light condition compared with the dim-light condition in 5 of 15 participants with eyes (retinitis pigmentosa, n = 2; retinopathy of prematurity [ROP], n = 2; bilateral retinal detachments, n = 1). Melatonin concentrations remained unchanged in response to light in the remaining 10 participants with eyes (ROP, n = 3; optic neuritis/neuropathy, n = 2; retinopathy unknown, n = 2; congenital glaucoma, n = 1; congenital rubella syndrome, n = 1; measles retinopathy, n = 1) and in all 3 bilaterally enucleated participants. CONCLUSIONS: These data confirm that light-induced suppression of melatonin remains functionally intact in a minority of totally visually blind individuals with eyes. None of the bilaterally enucleated individuals or those with phthisis bulbi was responsive to light; of the remainder, half were responsive to light. Although inner retinal damage is associated with a high likelihood that nonimage-forming photoreception is absent, the impact of outer retinal damage is more ambiguous, and therefore the assessment of the presence, attenuation, or absence of nonimage-forming light responses in totally blind patients requires careful individual confirmation and cannot simply be assumed from the type of blindness.


Assuntos
Cegueira/metabolismo , Ritmo Circadiano/fisiologia , Melatonina/metabolismo , Células Ganglionares da Retina/metabolismo , Percepção Visual/fisiologia , Pessoas com Deficiência Visual , Adulto , Idoso , Biomarcadores/metabolismo , Cegueira/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estimulação Luminosa
10.
J Sleep Res ; 27(1): 120-128, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28621018

RESUMO

We examined the structure, duration and quality of sleep, including non-rapid eye movement sleep and rapid eye movement sleep, in 11 blind individuals without conscious light perception and 11 age- and sex-matched sighted controls. Because blindness is associated with a greater incidence of free-running circadian rhythms, we controlled for circadian phase by a measure of melatonin onset timing. When circadian rhythm was entrained and melatonin onset occurred at normal times, sleep structure did not differ between blind and sighted individuals. On the other hand, an abnormal timing of the circadian phase, including delayed, shifted and unclassifiable melatonin onsets, led to larger rapid eye movement sleep latencies and increased wake times. No differences were observed for stages of non-rapid eye movement sleep, either between congenital and late blind and sighted individuals, or across the different circadian phases. Moreover, abnormal circadian phases were more common in the blind (n = 5) than the sighted (n = 2) sample. Our findings suggest that the sleep structure of blind individuals depends on entrainment of circadian phase, rather than on the absence of vision.


Assuntos
Cegueira/fisiopatologia , Ritmo Circadiano/fisiologia , Sono REM/fisiologia , Sono de Ondas Lentas/fisiologia , Adulto , Biomarcadores/metabolismo , Cegueira/metabolismo , Feminino , Humanos , Masculino , Melatonina/metabolismo , Pessoa de Meia-Idade , Latência do Sono/fisiologia
11.
Neural Plast ; 2018: 6120925, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30008742

RESUMO

Because the human brain consumes a disproportionate fraction of the resting body's energy, positron emission tomography (PET) measurements of absolute glucose metabolism (CMRglc) can serve as disease biomarkers. Global mean normalization (GMN) of PET data reveals disease-based differences from healthy individuals as fractional changes across regions relative to a global mean. To assess the impact of GMN applied to metabolic data, we compared CMRglc with and without GMN in healthy awake volunteers with eyes closed (i.e., control) against specific physiological/clinical states, including healthy/awake with eyes open, healthy/awake but congenitally blind, healthy/sedated with anesthetics, and patients with disorders of consciousness. Without GMN, global CMRglc alterations compared to control were detected in all conditions except in congenitally blind where regional CMRglc variations were detected in the visual cortex. However, GMN introduced regional and bidirectional CMRglc changes at smaller fractions of the quantitative delocalized changes. While global information was lost with GMN, the quantitative approach (i.e., a validated method for quantitative baseline metabolic activity without GMN) not only preserved global CMRglc alterations induced by opening eyes, sedation, and varying consciousness but also detected regional CMRglc variations in the congenitally blind. These results caution the use of GMN upon PET-measured CMRglc data in health and disease.


Assuntos
Cegueira/metabolismo , Encéfalo/metabolismo , Glucose/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Adulto , Cegueira/congênito , Cegueira/diagnóstico por imagem , Encéfalo/diagnóstico por imagem , Interpretação Estatística de Dados , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Processamento de Sinais Assistido por Computador , Adulto Jovem
12.
Int Ophthalmol ; 38(3): 1251-1260, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28602015

RESUMO

PURPOSE: NDP-related retinopathies are a group of X-linked disorders characterized by degenerative and proliferative changes of the neuroretina, occasionally accompanied with varying degrees of mental retardation and sensorineural hearing loss. NDP is the predominant gene associated with NDP-related retinopathies. The purpose of this study was to report the clinical and genetic findings in three unrelated patients diagnosed with NDP-related retinopathies. METHODS: The patients underwent complete ophthalmic examination followed by genetic analyses. NDP gene was screened by direct sequencing approach. Targeted resequencing of several other ocular genes was carried out in patient samples that either indicated NDP gene deletion or tested negative for NDP mutation. Gene quantitation analysis was performed using real-time PCR. RESULTS: The whole NDP gene was deleted in patient I, while a missense NDP mutation, c.205T>C, was identified in patient II, and both had classical Norrie disease ocular phenotype (with no other systemic defects). Patient III who was diagnosed with familial exudative vitreoretinopathy did not show any mutation in the known candidate genes as well as in other ocular genes tested. CONCLUSIONS: The patient with whole NDP gene deletion did not exhibit any apparent extraocular defects (like mental retardation or sensorineural hearing loss) during his first decade of life, and this is considered to be a notable finding. Our study also provides evidence emphasizing the need for genetic testing which could eliminate ambiguities in clinical diagnosis and detect carrier status, thereby aiding the patient and family members during genetic counseling.


Assuntos
Anormalidades Múltiplas , Cegueira/congênito , DNA/genética , Proteínas do Olho/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/genética , Doenças do Sistema Nervoso/genética , Retina/diagnóstico por imagem , Doenças Retinianas/genética , Espasmos Infantis/genética , Cegueira/diagnóstico , Cegueira/genética , Cegueira/metabolismo , Criança , Pré-Escolar , Análise Mutacional de DNA , Proteínas do Olho/metabolismo , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/diagnóstico , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Humanos , Lactente , Masculino , Microscopia Acústica , Proteínas do Tecido Nervoso/metabolismo , Doenças do Sistema Nervoso/diagnóstico , Doenças do Sistema Nervoso/metabolismo , Linhagem , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real , Retina/metabolismo , Degeneração Retiniana , Doenças Retinianas/diagnóstico , Doenças Retinianas/metabolismo , Estudos Retrospectivos , Espasmos Infantis/diagnóstico , Espasmos Infantis/metabolismo
13.
J Neurosci ; 36(21): 5808-19, 2016 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-27225770

RESUMO

UNLABELLED: RPE65, an abundant membrane-associate protein in the retinal pigment epithelium (RPE), is a key retinoid isomerase of the visual cycle necessary for generating 11-cis-retinal that functions not only as a molecular switch for activating cone and rod visual pigments in response to light stimulation, but also as a chaperone for normal trafficking of cone opsins to the outer segments. Many mutations in RPE65 are associated with Leber congenital amaurosis (LCA). A R91W substitution, the most frequent LCA-associated mutation, results in a severe decrease in protein level and enzymatic activity of RPE65, causing cone opsin mislocalization and early cone degeneration in the mutation knock-in mouse model of LCA. Here we show that R91W RPE65 undergoes ubiquitination-dependent proteasomal degradation in the knock-in mouse RPE due to misfolding. The 26S proteasome non-ATPase regulatory subunit 13 mediated degradation specifically of misfolded R91W RPE65. The mutation disrupted membrane-association and colocalization of RPE65 with lecithin:retinol acyltransferase (LRAT) that provides the hydrophobic substrate for RPE65. Systemic administration of sodium 4-phenylbutyrate (PBA), a chemical chaperone, increased protein stability, enzymatic activity, membrane-association, and colocalization of R91W RPE65 with LRAT. This rescue effect increased synthesis of 11-cis-retinal and 9-cis-retinal, a functional iso-chromophore of the visual pigments, led to alleviation of S-opsin mislocalization and cone degeneration in the knock-in mice. Importantly, PBA-treatment also improved cone-mediated vision in the mutant mice. These results indicate that PBA, a U.S. Food and Drug Administration-approved safe oral medication, may provide a noninvasive therapeutic intervention that delays daylight vision loss in patients with RPE65 mutations. SIGNIFICANCE STATEMENT: LCA is a severe early onset retinal dystrophy. Recent clinical trials of gene therapy have implicated the need of an alternative or combination therapy to improve cone survival and function in patients with LCA caused by RPE65 mutations. Using a mouse model carrying the most frequent LCA-associated mutation (R91W), we found that the mutant RPE65 underwent ubiquitination-dependent proteasomal degradation due to misfolding. Treatment of the mice with a chemical chaperone partially corrected stability, enzymatic activity, and subcellular localization of R91W RPE65, which was also accompanied by improvement of cone survival and vision. These findings identify an in vivo molecular pathogenic mechanism for R91W mutation and provide a feasible pharmacological approach that can delay vision loss in patients with RPE65 mutations.


Assuntos
Cegueira/prevenção & controle , Amaurose Congênita de Leber/tratamento farmacológico , Amaurose Congênita de Leber/metabolismo , Fenilbutiratos/administração & dosagem , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , cis-trans-Isomerases/metabolismo , Animais , Cegueira/metabolismo , Cegueira/patologia , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Amaurose Congênita de Leber/patologia , Masculino , Camundongos , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Resultado do Tratamento , Acuidade Visual/efeitos dos fármacos , cis-trans-Isomerases/genética
14.
J Biol Chem ; 291(47): 24504-24516, 2016 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-27703005

RESUMO

Substitutions of Arg838 in the dimerization domain of a human retinal membrane guanylyl cyclase 1 (RetGC1) linked to autosomal dominant cone-rod degeneration type 6 (CORD6) change RetGC1 regulation in vitro by Ca2+ In addition, we find that R838S substitution makes RetGC1 less sensitive to inhibition by retinal degeneration-3 protein (RD3). We selectively expressed human R838S RetGC1 in mouse rods and documented the decline in rod vision and rod survival. To verify that changes in rods were specifically caused by the CORD6 mutation, we used for comparison cones, which in the same mice did not express R838S RetGC1 from the transgenic construct. The R838S RetGC1 expression in rod outer segments reduced inhibition of cGMP production in the transgenic mouse retinas at the free calcium concentrations typical for dark-adapted rods. The transgenic mice demonstrated early-onset and rapidly progressed with age decline in visual responses from the targeted rods, in contrast to the longer lasting preservation of function in the non-targeted cones. The decline in rod function in the retina resulted from a progressive degeneration of rods between 1 and 6 months of age, with the severity and pace of the degeneration consistent with the extent to which the Ca2+ sensitivity of the retinal cGMP production was affected. Our study presents a new experimental model for exploring cellular mechanisms of the CORD6-related photoreceptor death. This mouse model provides the first direct biochemical and physiological in vivo evidence for the Arg838 substitutions in RetGC1 being the culprit behind the pathogenesis of the CORD6 congenital blindness.


Assuntos
Cegueira/metabolismo , Sinalização do Cálcio , GMP Cíclico/metabolismo , Guanilato Ciclase/metabolismo , Mutação de Sentido Incorreto , Receptores de Superfície Celular/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Substituição de Aminoácidos , Animais , Cegueira/genética , Cegueira/patologia , Cálcio/metabolismo , GMP Cíclico/genética , Modelos Animais de Doenças , Guanilato Ciclase/genética , Humanos , Camundongos , Camundongos Transgênicos , Receptores de Superfície Celular/genética , Células Fotorreceptoras Retinianas Bastonetes/patologia
15.
Genet Mol Res ; 15(2)2016 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-27323164

RESUMO

Diabetic retinopathy (DR) is a frequent microvascular complication of diabetes, and one of the most common causes of legal blindness in the world. Epigallocatechin-3-gallate (EGCG) produces an anti-oxidative and anti-inflammatory effect against various human diseases. In this study, we determined the effect of EGCG on a human retinal endothelial cell (HREC) line. The cell viability was determined by a standard MTT assay, while the cell cycle and apoptosis rate were analyzed by flow cytometry. Inflammatory marker expression was detected by enzyme-linked immunosorbent assay. Treatment of HRECs with EGCG (20 and 40 mM) led to a significant decrease in the apoptosis rate (2.35 ± 0.56 and 1.24 ± 0.32%). The culture supernatant of cells treated with high glucose concentrations showed significantly higher levels of TNF-α (598.7 ± 89.7 vs 193.2 ± 38.5 pg/mL; P < 0.001), IL-6 (6.16 ± 0.51 vs 1.61 ± 0.21 ng/mL; P < 0.001), and ICAM-1 (31.6 ± 4.4 vs 14.8 ± 2.9 ng/mL; P < 0.001) compared to the cells in the control group. EGCG decreased the expression level of phosphorylated p38-mitogen activated protein kinase (MAPK) and extracellular regulated kinase (ERK)1/2. Moreover, EGCG was shown to significantly inhibit the expression of vascular endothelial growth factor (VEGF). Therefore, EGCG treatment ameliorated the negative effect of high glucose concentrations on the cell viability and apoptotic rate. The protective effects of EGCG under high glucose conditions may be attributed to the regulation of inflammatory cytokines and inhibition of the MAPK/ERK-VEGF pathway.


Assuntos
Cegueira/tratamento farmacológico , Catequina/análogos & derivados , Retinopatia Diabética/tratamento farmacológico , Retina/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Cegueira/metabolismo , Cegueira/patologia , Catequina/administração & dosagem , Ciclo Celular/efeitos dos fármacos , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Interleucina-6/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/biossíntese , Retina/metabolismo , Retina/patologia , Fator de Necrose Tumoral alfa/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese
16.
Hum Mol Genet ; 22(5): 1005-16, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23201751

RESUMO

Norrie disease (ND) is a congenital disorder characterized by retinal hypovascularization and cognitive delay. ND has been linked to mutations in 'Norrie Disease Protein' (Ndp), which encodes the secreted protein Norrin. Norrin functions as a secreted angiogenic factor, although its role in neural development has not been assessed. Here, we show that Ndp expression is initiated in retinal progenitors in response to Hedgehog (Hh) signaling, which induces Gli2 binding to the Ndp promoter. Using a combination of genetic epistasis and acute RNAi-knockdown approaches, we show that Ndp is required downstream of Hh activation to induce retinal progenitor proliferation in the retina. Strikingly, Ndp regulates the rate of cell-cycle re-entry and not cell-cycle kinetics, thereby uncoupling the self-renewal and cell-cycle progression functions of Hh. Taken together, we have uncovered a cell autonomous function for Ndp in retinal progenitor proliferation that is independent of its function in the retinal vasculature, which could explain the neural defects associated with ND.


Assuntos
Cegueira/congênito , Proteínas do Olho/genética , Fatores de Transcrição Kruppel-Like/genética , Proteínas do Tecido Nervoso/genética , Doenças do Sistema Nervoso/genética , Retina/metabolismo , Espasmos Infantis/genética , Via de Sinalização Wnt , Animais , Cegueira/genética , Cegueira/metabolismo , Ciclo Celular/genética , Proliferação de Células , Epistasia Genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Doenças Genéticas Ligadas ao Cromossomo X , Proteínas Hedgehog/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Neovascularização Fisiológica , Proteínas do Tecido Nervoso/metabolismo , Doenças do Sistema Nervoso/metabolismo , Ligação Proteica , Retina/crescimento & desenvolvimento , Degeneração Retiniana , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/metabolismo , Espasmos Infantis/metabolismo , Via de Sinalização Wnt/genética , Proteína Gli2 com Dedos de Zinco
17.
FASEB J ; 28(4): 1854-69, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24421398

RESUMO

Patients with congenital disorder of glycosylation (CDG), type Ib (MPI-CDG or CDG-Ib) have mutations in phosphomannose isomerase (MPI) that impair glycosylation and lead to stunted growth, liver dysfunction, coagulopathy, hypoglycemia, and intestinal abnormalities. Mannose supplements correct hypoglycosylation and most symptoms by providing mannose-6-P (Man-6-P) via hexokinase. We generated viable Mpi hypomorphic mice with residual enzymatic activity comparable to that of patients, but surprisingly, these mice appeared completely normal except for modest (~15%) embryonic lethality. To overcome this lethality, pregnant dams were provided 1-2% mannose in their drinking water. However, mannose further reduced litter size and survival to weaning by 40 and 66%, respectively. Moreover, ~50% of survivors developed eye defects beginning around midgestation. Mannose started at birth also led to eye defects but had no effect when started after eye development was complete. Man-6-P and related metabolites accumulated in the affected adult eye and in developing embryos and placentas. Our results demonstrate that disturbing mannose metabolic flux in mice, especially during embryonic development, induces a highly specific, unanticipated pathological state. It is unknown whether mannose is harmful to human fetuses during gestation; however, mothers who are at risk for having MPI-CDG children and who consume mannose during pregnancy hoping to benefit an affected fetus in utero should be cautious.


Assuntos
Cegueira/etiologia , Suplementos Nutricionais/toxicidade , Manose-6-Fosfato Isomerase/metabolismo , Manose/toxicidade , Animais , Cegueira/genética , Cegueira/metabolismo , Western Blotting , Células Cultivadas , Defeitos Congênitos da Glicosilação/genética , Defeitos Congênitos da Glicosilação/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Olho/embriologia , Olho/crescimento & desenvolvimento , Olho/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Manose/sangue , Manose/metabolismo , Manose-6-Fosfato Isomerase/genética , Manosefosfatos/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Placenta/efeitos dos fármacos , Placenta/embriologia , Placenta/metabolismo , Gravidez
18.
PLoS Biol ; 10(3): e1001286, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22448144

RESUMO

Dorsal-ventral specification in the amphibian embryo is controlled by ß-catenin, whose activation in all dorsal cells is dependent on maternal Wnt11. However, it remains unknown whether other maternally secreted factors contribute to ß-catenin activation in the dorsal ectoderm. Here, we show that maternal Xenopus Norrin (xNorrin) promotes anterior neural tissue formation in ventralized embryos. Conversely, when xNorrin function is inhibited, early canonical Wnt signaling in the dorsal ectoderm and the early expression of the zygotic neural inducers Chordin, Noggin, and Xnr3 are severely suppressed, causing the loss of anterior structures. In addition, xNorrin potently inhibits BMP- and Nodal/Activin-related functions through direct binding to the ligands. Moreover, a subset of Norrin mutants identified in humans with Norrie disease retain Wnt activation but show defective inhibition of Nodal/Activin-related signaling in mesoderm induction, suggesting that this disinhibition causes Norrie disease. Thus, xNorrin is an unusual molecule that acts on two major signaling pathways, Wnt and TGF-ß, in opposite ways and is essential for early neuroectoderm specification.


Assuntos
Placa Neural/embriologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Via de Sinalização Wnt , Proteínas de Xenopus/metabolismo , Xenopus/metabolismo , Sequência de Aminoácidos , Animais , Cegueira/congênito , Cegueira/genética , Cegueira/metabolismo , Cegueira/patologia , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Sequência Conservada , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Doenças Genéticas Ligadas ao Cromossomo X , Humanos , Ligantes , Mesoderma/embriologia , Mesoderma/metabolismo , Mesoderma/patologia , Dados de Sequência Molecular , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Placa Neural/metabolismo , Placa Neural/fisiologia , Ligação Proteica , Degeneração Retiniana , Espasmos Infantis/genética , Espasmos Infantis/metabolismo , Espasmos Infantis/patologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/agonistas , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Xenopus/embriologia , Xenopus/genética , Proteínas de Xenopus/agonistas , Proteínas de Xenopus/genética
19.
Mol Ther ; 22(8): 1434-1440, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24821344

RESUMO

We previously showed that blind rats whose vision was restored by gene transfer of Chlamydomonas channelrhodopsin-2 (ChR2) could only detect wavelengths less than 540 nm because of the action spectrum of the transgene product. Volvox-derived channelrhodopsin-1, VChR1, has a broader spectrum than ChR2. However, the VChR1 protein was mainly localized in the cytoplasm and showed weak ion channel properties when the VChR1 gene was transfected into HEK293 cells. We generated modified Volvox channelrhodopsin-1 (mVChR1), which is a chimera of Volvox channelrhodopsin-1 and Chlamydomonas channelrhodopsin-1 and demonstrated increased plasma membrane integration and dramatic improvement in its channel properties. Under whole-cell patch clamp, mVChR1-expressing cells showed a photo-induced current upon stimulation at 468-640 nm. The evoked currents in mVChR1-expressing cells were ~30 times larger than those in VChR1-expressing cells. Genetically, blind rats expressing mVChR1 via an adeno-associated virus vector regained their visual responses to light with wavelengths between 468 and 640 nm and their recovered visual responses were maintained for a year. Thus, mVChR1 is a candidate gene for gene therapy for restoring vision, and gene delivery of mVChR1 may provide blind patients access to the majority of the visible light spectrum.


Assuntos
Cegueira/terapia , Terapia Genética/métodos , Retina/fisiopatologia , Rodopsina/metabolismo , Volvox/genética , Animais , Cegueira/metabolismo , Chlamydomonas/genética , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/uso terapêutico , Células HEK293 , Humanos , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Rodopsina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA