Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Invest Ophthalmol Vis Sci ; 61(5): 30, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32428231

RESUMO

Purpose: To determine the therapeutic window for gene augmentation for Leber congenital amaurosis (LCA) associated with mutations in LCA5. Methods: Five patients (ages 6-31) with LCA and biallelic LCA5 mutations underwent an ophthalmic examination including optical coherence tomography (SD-OCT), full-field stimulus testing (FST), and pupillometry. The time course of photoreceptor degeneration in the Lca5gt/gt mouse model and the efficacy of subretinal gene augmentation therapy with AAV8-hLCA5 delivered at postnatal day 5 (P5) (early, n = 11 eyes), P15 (mid, n = 14), and P30 (late, n = 13) were assessed using SD-OCT, histologic study, electroretinography (ERG), and pupillometry. Comparisons were made with the human disease. Results: Patients with LCA5-LCA showed a maculopathy with detectable outer nuclear layer (ONL) in the pericentral retina and at least 4 log units of dark-adapted sensitivity loss. The Lca5gt/gt mouse has a similarly severe and rapid photoreceptor degeneration. The ONL became progressively thinner and was undetectable by P60. Rod- and cone-mediated ERGs were severely reduced in amplitudes at P30 and became nondetectable by P60. Subretinal AAV8-hLCA5 administered to Lca5gt/gt mice at P5 and P15, but not at P30, resulted in structural and functional rescue. Conclusions: LCA5-LCA is a particularly severe form of LCA that was recapitulated in the Lca5gt/gt mouse. Gene augmentation resulted in structural and functional rescue in the Lca5gt/gt mouse if delivered before P30. Retained photoreceptors were visible within the central retina in all patients with LCA5-LCA, at a level equivalent to that observed in rescued Lca5gt/gt mice, suggesting a window of opportunity for the treatment of patients with LCA5-LCA.


Assuntos
Dependovirus/genética , Proteínas do Olho/genética , Terapia Genética , Amaurose Congênita de Leber/terapia , Proteínas Associadas aos Microtúbulos/genética , Retina/fisiopatologia , Adulto , Animais , Criança , Modelos Animais de Doenças , Eletrorretinografia , Feminino , Terapia Genética/métodos , Vetores Genéticos , Humanos , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Imagem Óptica , Fenótipo , Pupila/fisiologia , Tomografia de Coerência Óptica , Acuidade Visual/fisiologia , Testes de Campo Visual , Campos Visuais/fisiologia , Adulto Jovem
2.
Stem Cells Int ; 2019: 7281912, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30800164

RESUMO

Recombinant adeno-associated virus (rAAV), produced from a nonpathogenic parvovirus, has become an increasing popular vector for gene therapy applications in human clinical trials. However, transduction and transgene expression of rAAVs can differ across in vitro and ex vivo cellular transduction strategies. This study compared 11 rAAV serotypes, carrying one reporter transgene cassette containing a cytomegalovirus immediate-early enhancer (eCMV) and chicken beta actin (CBA) promoter driving the expression of an enhanced green-fluorescent protein (eGFP) gene, which was transduced into four different cell types: human iPSC, iPSC-derived RPE, iPSC-derived cortical, and dissociated embryonic day 18 rat cortical neurons. Each cell type was exposed to three multiplicity of infections (MOI: 1E4, 1E5, and 1E6 vg/cell). After 24, 48, 72, and 96 h posttransduction, GFP-expressing cells were examined and compared across dosage, time, and cell type. Retinal pigmented epithelium showed highest AAV-eGFP expression and iPSC cortical the lowest. At an MOI of 1E6 vg/cell, all serotypes show measurable levels of AAV-eGFP expression; moreover, AAV7m8 and AAV6 perform best across MOI and cell type. We conclude that serotype tropism is not only capsid dependent but also cell type plays a significant role in transgene expression dynamics.

3.
Sci Rep ; 8(1): 17286, 2018 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-30470797

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

4.
Invest Ophthalmol Vis Sci ; 59(12): 5225-5236, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30372751

RESUMO

Purpose: To describe the retinal phenotype of pediatric patients with mutations in the retinol dehydrogenase 12 (RDH12) gene. Methods: Twenty-one patients from 14 families (ages 2-17 years) with RDH12-associated inherited retinal degeneration (RDH12-IRD) underwent a complete ophthalmic exam and imaging with spectral domain optical coherence tomography (SD-OCT) and near infrared and short-wavelength fundus autofluorescence. Visual field extent was measured with Goldmann kinetic perimetry, visual thresholds with dark-adapted static perimetry or with dark-adapted chromatic full-field stimulus testing (FST) and transient pupillometry. Results: Visual acuity ranged from 20/40 to light perception. There was parafoveal depigmentation or atrophic maculopathies accompanied by midperipheral intraretinal pigment migration. SD-OCT revealed foveal thinning in all patients and detectable but thinned outer nuclear layer (ONL) at greater eccentricities from the fovea. Photoreceptor outer segment (POS) signals were only detectable in small pockets within the central retina. Measurable kinetic visual fields were limited to small (<5-10°) central islands of vision. Electroretinograms were reported as undetectable or severely reduced in amplitude. FST sensitivities to a 467 nm stimulus were rod-mediated and reduced on average by ∼2.5 log units. A thinned central ONL colocalized with severely reduced to nondetectable cone-mediated sensitivities. Pupillometry confirmed the psychophysically measured abnormalities. Conclusions: RDH12-IRD causes an early-onset, retina-wide disease with particularly severe central retinal abnormalities associated with relatively less severe rod photoreceptor dysfunction, a pattern consistent with an early-onset cone-rod dystrophy. Severely abnormal POS but detectable ONL in the pericentral and peripapillary retina suggest these regions may become targets for gene therapy.


Assuntos
Oxirredutases do Álcool/genética , Mutação , Degeneração Retiniana/genética , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Adolescente , Criança , Pré-Escolar , Adaptação à Escuridão , Eletrorretinografia , Feminino , Humanos , Masculino , Fenótipo , Degeneração Retiniana/diagnóstico , Degeneração Retiniana/fisiopatologia , Tomografia de Coerência Óptica , Acuidade Visual/fisiologia , Testes de Campo Visual , Campos Visuais/fisiologia
5.
Stem Cell Res ; 27: 140-150, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29414605

RESUMO

Choroideremia (CHM) is a rare monogenic, X-linked recessive inherited retinal degeneration resulting from mutations in the Rab Escort Protein-1 (REP1) encoding CHM gene. The primary retinal cell type leading to CHM is unknown. In this study, we explored the utility of induced pluripotent stem cell-derived models of retinal pigmented epithelium (iPSC-RPE) to study disease pathogenesis and a potential gene-based intervention in four different genetically distinct forms of CHM. A number of abnormal cell biologic, biochemical, and physiologic functions were identified in the CHM mutant cells. We then identified a recombinant adeno-associated virus (AAV) serotype, AAV7m8, that is optimal for both delivering transgenes to iPSC-RPEs as well as to appropriate target cells (RPE cells and rod photoreceptors) in the primate retina. To establish the proof of concept of AAV7m8 mediated CHM gene therapy, we developed AAV7m8.hCHM, which delivers the human CHM cDNA under control of CMV-enhanced chicken ß-actin promoter (CßA). Delivery of AAV7m8.hCHM to CHM iPSC-RPEs restored protein prenylation, trafficking and phagocytosis. The results confirm that AAV-mediated delivery of the REP1-encoding gene can rescue defects in CHM iPSC-RPE regardless of the type of disease-causing mutation. The results also extend our understanding of mechanisms involved in the pathophysiology of choroideremia.


Assuntos
Coroideremia/metabolismo , Coroideremia/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Epitélio Pigmentado da Retina/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Dependovirus/genética , Imunofluorescência , Humanos , Fagocitose/fisiologia , Primatas
6.
Cell Stem Cell ; 18(6): 688-689, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27257755

RESUMO

Human retinal degeneration can cause blindness, and the lack of relevant model systems has made identifying underlying mechanisms challenging. Parfitt et al. (2016) generate three-dimensional retinal tissue from patient-derived induced pluripotent stem cells to identify how CEP290 mutations cause retinal degeneration, and show an antisense approach can correct disease-associated phenotypes.


Assuntos
Retina , Degeneração Retiniana/genética , Humanos , Células-Tronco Pluripotentes Induzidas , Mutação
7.
Sci Rep ; 5: 17105, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26597678

RESUMO

The future of treating inherited and acquired genetic diseases will be defined by our ability to introduce transgenes into cells and restore normal physiology. Here we describe an autogenous transgene regulatory system (ARES), based on the bacterial lac repressor, and demonstrate its utility for controlling the expression of a transgene in bacteria, eukaryotic cells, and in the retina of mice. This ARES system is inducible by the small non-pharmacologic molecule, Isopropyl ß-D-1-thiogalactopyranoside (IPTG) that has no off-target effects in mammals. Following subretinal injection of an adeno-associated virus (AAV) vector encoding ARES, luciferase expression can be reversibly controlled in the murine retina by oral delivery of IPTG over three induction-repression cycles. The ability to induce transgene expression repeatedly via administration of an oral inducer in vivo, suggests that this type of regulatory system holds great promise for applications in human gene therapy.


Assuntos
Expressão Gênica , Terapia Genética , Ativação Transcricional/efeitos dos fármacos , Administração Oral , Animais , Dependovirus/genética , Genes Reporter , Células HEK293 , Humanos , Isopropiltiogalactosídeo/administração & dosagem , Luciferases/biossíntese , Luciferases/genética , Camundongos , Retina/metabolismo , Transgenes
9.
J Gene Med ; 16(5-6): 122-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24962736

RESUMO

BACKGROUND: Choroideremia (CHM) is a slowly progressive X-linked retinal degeneration that results in a loss of photoreceptors, retinal pigment epithelium and choroid. CHM, the gene implicated in choroideremia, encodes Rab escort protein-1 (REP-1), which is involved in the post-translational activation via prenylation of Rab proteins. METHODS: We evaluated AAV8.CBA.hCHM, a recombinant adeno-associated virus serotype 8 (rAAV8) vector, which targets retinal cells efficiently, for both therapeutic effect and safety in vitro and in vivo in a murine model. In vitro studies included western blot analyses and prenylation assays. In vivo studies included ophthalmoscopy, pupillometry, histology and immunofluorescence analysis. RESULTS: Infection with AAV8.CBA.hCHM induced the expression of REP-1 protein in a dose-responsive fashion. Transduction with AAV8.CBA.hCHM reverses the biochemical and pathogenetic defects in CHM both in vitro and in vivo and showed no safety concerns in the in vivo investigations performed in the present study. CONCLUSIONS: AAV8 is a promising vector for human clinical gene therapy trials for choroideremia.


Assuntos
Coroideremia/genética , Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular , Células Cultivadas , Coroideremia/terapia , Modelos Animais de Doenças , Feminino , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Heterozigoto , Humanos , Camundongos , Camundongos Knockout , Retina/metabolismo , Retina/patologia , Retina/fisiopatologia , Transdução Genética , Transgenes
10.
PLoS One ; 8(5): e61396, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667438

RESUMO

Choroideremia (CHM) is an X- linked retinal degeneration that is symptomatic in the 1(st) or 2(nd) decade of life causing nyctalopia and loss of peripheral vision. The disease progresses through mid-life, when most patients become blind. CHM is a favorable target for gene augmentation therapy, as the disease is due to loss of function of a protein necessary for retinal cell health, Rab Escort Protein 1 (REP1).The CHM cDNA can be packaged in recombinant adeno-associated virus (rAAV), which has an established track record in human gene therapy studies, and, in addition, there are sensitive and quantitative assays to document REP1 activity. An animal model that accurately reflects the human condition is not available. In this study, we tested the ability to restore REP1 function in personalized in vitro models of CHM: lymphoblasts and induced pluripotent stems cells (iPSCs) from human patients. The initial step of evaluating safety of the treatment was carried out by evaluating for acute retinal histopathologic effects in normal-sighted mice and no obvious toxicity was identified. Delivery of the CHM cDNA to affected cells restores REP1 enzymatic activity and also restores proper protein trafficking. The gene transfer is efficient and the preliminary safety data are encouraging. These studies pave the way for a human clinical trial of gene therapy for CHM.


Assuntos
Coroideremia/genética , Coroideremia/terapia , Dependovirus/genética , Terapia Genética/métodos , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular , Feminino , Terapia Genética/efeitos adversos , Humanos , Masculino , Camundongos , Plasmídeos/genética , Medicina de Precisão , Transporte Proteico/genética , Segurança , Proteínas rab de Ligação ao GTP/metabolismo
12.
PLoS One ; 6(6): e21193, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21738619

RESUMO

The P23H mutation in the rhodopsin gene causes rhodopsin misfolding, altered trafficking and formation of insoluble aggregates leading to photoreceptor degeneration and autosomal dominant retinitis pigmentosa (RP). There are no effective therapies to treat this condition. Compounds that enhance dissociation of protein aggregates may be of value in developing new treatments for such diseases. Anti-protein aggregating activity of curcumin has been reported earlier. In this study we present that treatment of COS-7 cells expressing mutant rhodopsin with curcumin results in dissociation of mutant protein aggregates and decreases endoplasmic reticulum stress. Furthermore we demonstrate that administration of curcumin to P23H-rhodopsin transgenic rats improves retinal morphology, physiology, gene expression and localization of rhodopsin. Our findings indicate that supplementation of curcumin improves retinal structure and function in P23H-rhodopsin transgenic rats. This data also suggest that curcumin may serve as a potential therapeutic agent in treating RP due to the P23H rhodopsin mutation and perhaps other degenerative diseases caused by protein trafficking defects.


Assuntos
Curcumina/farmacologia , Curcumina/uso terapêutico , Degeneração Retiniana/tratamento farmacológico , Degeneração Retiniana/metabolismo , Rodopsina/metabolismo , Animais , Células COS , Chlorocebus aethiops , Imuno-Histoquímica , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Retina/efeitos dos fármacos , Retina/metabolismo , Retina/patologia , Rodopsina/química , Rodopsina/genética
13.
Prog Retin Eye Res ; 29(3): 191-207, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20096366

RESUMO

Stargardt-like macular degeneration (STGD3) is an early onset, autosomal dominant macular degeneration. STGD3 is characterized by a progressive pathology, the loss of central vision, atrophy of the retinal pigment epithelium, and accumulation of lipofuscin, clinical features that are also characteristic of age-related macular degeneration. The onset of clinical symptoms in STGD3, however, is typically observed within the second or third decade of life (i.e., starting in the teenage years). The clinical profile at any given age among STGD3 patients can be variable suggesting that, although STGD3 is a single gene defect, other genetic or environmental factors may play a role in moderating the final disease phenotype. Genetic studies localized the STGD3 disease locus to a small region on the short arm of human chromosome 6, and application of a positional candidate gene approach identified protein truncating mutations in the elongation of very long chain fatty acids-4 gene (ELOVL4) in patients with this disease. The ELOVL4 gene encodes a protein homologous to the ELO group of proteins that participate in fatty acid elongation in yeast. Pathogenic mutations found in the ELOVL4 gene result in altered trafficking of the protein and behave with a dominant negative effect. Mice carrying an Elovl4 mutation developed photoreceptor degeneration and depletion of very long chain fatty acids (VLCFA). ELOVL4 protein participates in the synthesis of fatty acids with chain length longer than 26 carbons. Studies on ELOVL4 indicate that VLCFA may be necessary for normal function of the retina, and the defective protein trafficking and/or altered VLCFA elongation underlies the pathology associated with STGD3. Determining the role of VLCFA in the retina and discerning the implications of abnormal trafficking of mutant ELOVL4 and depleted VLCFA content in the pathology of STGD3 will provide valuable insight in understanding the retinal structure, function, and pathology underlying STGD3 and may lead to a better understanding of the process of macular disease in general.


Assuntos
Proteínas do Olho/genética , Degeneração Macular/genética , Degeneração Macular/patologia , Proteínas de Membrana/genética , Mutação/genética , Patologia Molecular , Animais , Cromossomos Humanos Par 6 , Ácidos Graxos/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos
14.
Exp Eye Res ; 89(6): 905-12, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19682985

RESUMO

The mechanism underlying photoreceptor degeneration in autosomal dominant Stargardt-like macular degeneration (STGD3) due to mutations in the elongation of very long chain fatty acids-4 (ELOVL4) gene is not fully understood. To evaluate the pathological events associated with STGD3, we used a mouse model that mimics the human STGD3 phenotype and studied the progression of retinal degeneration. Morphological changes in the retina of Elovl4 5-bp deletion knock-in mice (E_mut(+/-)) were evaluated at 22 months of age. The localization of ELOVL4, and the expression pattern of inner retinal tissue marker proteins, and ubiquitin were determined by immunofluorescence labeling of retinal sections. Levels of the retinal pigment epithelium (RPE) lipofuscin fluorophores were measured by quantitative HPLC. Morphological evaluation of the retina revealed an accumulation of RPE debris in the subretinal space. A significant increase in the amount of ELOVL4 was observed in the outer plexiform layer in E_mut(+/-) mice compared to controls. Apart from the accumulation of ELOVL4, E_mut(+/-) mice also exhibited high expression of ubiquitin in the retina. Analysis of lipofuscin fluorophores in the RPE showed a significant elevation of A2E and compounds of the all-trans-retinal dimer series in retinas from four and ten month old E_mut(+/-) mice compared to wild-type littermates. These observations suggest that abnormal accumulation of ELOVL4 protein and lipofuscin may lead to photoreceptor degeneration in E_mut(+/-) mice.


Assuntos
Proteínas do Olho/metabolismo , Lipofuscina/metabolismo , Degeneração Macular/metabolismo , Proteínas de Membrana/metabolismo , Retina/metabolismo , Animais , Cromatografia Líquida de Alta Pressão/métodos , Modelos Animais de Doenças , Proteínas do Olho/genética , Técnicas de Introdução de Genes , Degeneração Macular/genética , Degeneração Macular/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Retina/ultraestrutura , Epitélio Pigmentado da Retina/metabolismo , Ubiquitina/metabolismo
15.
Invest Ophthalmol Vis Sci ; 50(7): 3456-63, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19369235

RESUMO

PURPOSE: A strong association between retinal degeneration and obesity has been shown in humans. However, the molecular basis of increased risk for retinal degeneration in obesity is unknown. Thus, an animal model with obesity and retinal degeneration would greatly aid the understanding of obesity-associated retinal degeneration. The retinal abnormalities in a novel rat model (WNIN-Ob) with spontaneously developed obesity are described. METHODS: Histologic and immunohistochemical examination were performed on retinal sections of 2- to 12-month-old WNIN-Ob rats, and findings were compared with those of lean littermate controls. RNA from retinas of 12-month-old WNIN-Ob and lean littermate rats was used for microarray and qRT-PCR analysis. RESULTS: The WNIN-Ob rats developed severe obesity, with an onset at approximately 35 days. Evaluation of retinal morphology in 2- to 12-month-old WNIN-Ob and age-matched lean littermate controls revealed progressive retinal degeneration, with an onset between 4 to 6 months of age. Immunohistochemical analysis with anti-rhodopsin, anti-cone opsin, and PSD-95 antibodies further confirmed retinal degeneration, particularly rod cell loss and thinner outer plexiform layer, in the obese rat retina. Gene expression by microarray analysis and qRT-PCR established activation of stress response, tissue remodeling, impaired phototransduction, and photoreceptor degeneration in WNIN-Ob rat retina. CONCLUSIONS: WNIN-Ob rats develop increased stress in retinal tissue and progressive retinal degeneration after the onset of severe obesity. The WNIN-Ob rat is the first rat model to develop retinal degeneration after the onset of obesity. This novel rat model may be a valuable tool for investigating retinal degeneration associated with obesity in humans.


Assuntos
Modelos Animais de Doenças , Obesidade/fisiopatologia , Retina/fisiopatologia , Degeneração Retiniana/fisiopatologia , Animais , Proteína 4 Homóloga a Disks-Large , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Obesidade/genética , Obesidade/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Opsinas/metabolismo , Ratos , Ratos Wistar , Retina/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rodopsina/metabolismo
17.
Hum Mol Genet ; 16(5): 471-82, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17208947

RESUMO

Mutations in elongation of very long-chain fatty acid-4 (ELOVL4) are associated with autosomal dominant Stargardt-like macular degeneration (STGD3), with a five base-pair (5 bp) deletion mutation resulting in the loss of 51 carboxy-terminal amino acids and truncation of the protein. In addition to the retina, Elovl4 is expressed in a limited number of mammalian tissues, including skin, with unknown function(s). We generated a knock-in mouse model with the 5-bp deletion in the Elovl4 gene. As anticipated, mice carrying this mutation in the heterozygous state (Elovl4(+/del)) exhibit progressive photoreceptor degeneration. Unexpectedly, homozygous mice (Elovl4(del/del)) display scaly, wrinkled skin, have severely compromised epidermal permeability barrier function, and die within a few hours after birth. Histopathological evaluation of the Elovl4(del/del) pups revealed no apparent abnormality(ies) in vital internal organs. However, skin histology showed an abnormally-compacted outer epidermis [stratum corneum (SC)], while electron microscopy revealed deficient epidermal lamellar body contents, and lack of normal SC lamellar membranes that are essential for permeability barrier function. Lipid analyses of epidermis from Elovl4(del/del) mice revealed a global decrease in very long-chain fatty acids (VLFAs) (i.e., carbon chain > or =C28) in both the ceramide/glucosylceramide and the free fatty-acid fractions. Strikingly, Elovl4(del/del) skin was devoid of the epidermal-unique omega-O-acylceramides, that are key hydrophobic components of the extracellular lamellar membranes in mammalian SC. These findings demonstrate that ELOVL4 is required for generating VLFA critical for epidermal barrier function, and that the lack of epidermal omega-O-acylceramides is incompatible with survival in a desiccating environment.


Assuntos
Ceramidas/deficiência , Proteínas do Olho/metabolismo , Ácidos Graxos/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Anormalidades da Pele/mortalidade , Anormalidades da Pele/patologia , Pele/metabolismo , Animais , Animais Recém-Nascidos , Pareamento de Bases , Biomarcadores , Diferenciação Celular , Cromatografia Líquida , Epiderme/química , Epiderme/patologia , Epiderme/ultraestrutura , Proteínas do Olho/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Espectrometria de Massas , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Deleção de Sequência , Pele/patologia
18.
Invest Ophthalmol Vis Sci ; 47(12): 5505-13, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17122142

RESUMO

PURPOSE: In a prior study, a S163R mutation in the complement-1q tumor necrosis factor-related protein 5 (CTRP5/ C1QTNF5) was reported to be associated with early-onset long anterior zonules (LAZ) and late-onset retinal degeneration (L-ORD). The ocular tissues involved in the phenotype are the retinal pigment epithelium (RPE) in the posterior segment and ciliary epithelium (CE) and lens in the anterior segment. The purpose of this study was to characterize the spatial and temporal expression of the mouse Ctrp5 gene, determine tissue and subcellular localization, and study the effect of the S163R mutation. METHODS: The expression of the Ctrp5 gene in the mouse was studied by quantitative (q)RT-PCR and in situ hybridization. CTRP5 protein expression and distribution were studied by Western blot analysis, immunohistochemistry, and immunoelectron microscopy. Cellular location of wild-type and mutant CTRP5 in MDCK and COS-7 cells was determined by immunofluorescence and immunoblot analysis. RESULTS: A significant level of Ctrp5 expression was detected in the adult mouse in the ciliary body (CB) and RPE, and expression started at a very early stage of embryogenesis. Immunohistochemical analysis showed CTRP5 protein in the apical processes of the RPE and forming a hexagonal lattice associated with the RPE lateral membranes. In the ciliary body, CTRP5 was localized to the apical aspects of the CE, the region between the bilayered ciliary epithelial cells. The membrane association of CTRP5 in the RPE and CE was further confirmed by immunoelectron microscopy. Furthermore, cultured cells were used to show that the CTRP5 is a secretory protein and that its secretion is impaired by the S163R mutation. CONCLUSIONS: CTRP5, a secretory and membrane-associated protein, is localized to the lateral and apical membranes of the RPE and CB. Impaired secretion of the mutant protein may underlie the pathophysiology of L-ORD and LAZ.


Assuntos
Corpo Ciliar/metabolismo , Proteínas de Membrana/genética , Epitélio Pigmentado Ocular/metabolismo , Mutação Puntual , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Técnica Indireta de Fluorescência para Anticorpo , Expressão Gênica/fisiologia , Hibridização In Situ , Doenças do Cristalino/genética , Ligamentos/anormalidades , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Imunoeletrônica , Mutagênese Sítio-Dirigida , Degeneração Retiniana/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
19.
Invest Ophthalmol Vis Sci ; 47(12): 5514-21, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17122143

RESUMO

PURPOSE: Mutations in the membrane frizzled-related protein (MFRP) gene cause nanophthalmos in humans, and a splice site mutation causes recessive retinal degeneration in the rd6 mouse. In human and mouse genomes, the MFRP gene lies adjoining to the complement 1q tumor necrosis factor-related protein 5 (CTRP5/C1QTNF5) gene involved in causing retinal degeneration and abnormal lens zonules in human. The purpose of this study was to characterize the spatial and temporal expression of the mouse Mfrp gene, determine tissue and subcellular localization of MFRP protein, and study its interaction with CTRP5. METHODS: Expression of the Mfrp gene in the mouse was studied by quantitative (q)RT-PCR. MFRP protein expression and distribution were studied by Western blot analysis, immunohistochemistry, and immunoelectron microscopy. Interaction with CTRP5 was studied by immunoprecipitation and immunoblot analysis, using mouse eye and human retinal pigmented epithelium (RPE) choroid extracts and by expressing full-length CTRP5 and MFRP in a heterologous system. RESULTS: The Mfrp gene is specifically expressed in RPE and ciliary body (CB), and its expression starts during early stages of embryogenesis. In the albino mouse eye, MFRP is localized to the apical and basal membranes of RPE and ciliary epithelium (CE). In addition, MFRP and CTRP5 were found to colocalize in RPE, CE, and MDCK cells, a general model of polarized epithelia. These proteins interact with each other in ocular tissues and also in a heterologous system. CONCLUSIONS: MFRP is localized to the plasma membrane of CE and RPE, and colocalizes and interacts with CTRP5 indicating a functional relationship between these two proteins.


Assuntos
Corpo Ciliar/metabolismo , Proteínas do Olho/genética , Expressão Gênica/fisiologia , Proteínas de Membrana/genética , Epitélio Pigmentado Ocular/metabolismo , Animais , Western Blotting , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Corpo Ciliar/embriologia , Proteínas do Olho/metabolismo , Imuno-Histoquímica , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Imunoeletrônica , Epitélio Pigmentado Ocular/embriologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
20.
Invest Ophthalmol Vis Sci ; 47(10): 4558-68, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17003453

RESUMO

PURPOSE: To develop and characterize a heterozygous knock-in mouse model carrying the 5-bp deletion in Elovl4 (E_mut+/-) and to study the pathology underlying Stargardt-like macular degeneration (STGD3). METHODS: E_mut+/- mice were generated by targeting a 5-bp deletion (AACTT) in the Elovl4 gene by homologous recombination. E_mut+/- mice of age 2 to 18 months and age-matched wild-type (Wt) littermate control animals were analyzed for the expression of Elovl4 transcript, ELOVL4 protein, photoreceptor-specific genes, and retinal fatty acid composition. Functional retinal changes were evaluated by electroretinography (ERG) and by morphologic and ultrastructural criteria. RESULTS: E_mut+/- mice retinas showed the presence of both Wt and mutant Elovl4 transcripts and proteins. Morphologic evaluation revealed cone photoreceptor ultrastructural abnormalities as early as 2 months of age, accumulation of lipofuscin in retinal pigment epithelium (RPE), and subretinal deposits at later ages. Shortening of rod outer segments (OS) was observed at approximately 10 months of age. Both cone and rod changes progressed with age. Unlike rod-specific genes, expression of selected cone specific genes was significantly reduced by 7 months of age. Mixed rod-cone and light-adapted b-waves were higher than normal at both 8 and 15 months. Levels of the fatty acids 20:5 (P = 0.027), 22:5 (P = 0.040) and 24:6 (P = 0.005) were found to be significantly lower in the retinas of E_mut+/- mice than in retinas of control subjects. CONCLUSIONS: E_mut+/- animals display characteristic features associated with Stargardt-like macular degeneration and serve as a model for the study of the mechanism underlying STGD3.


Assuntos
Modelos Animais de Doenças , Proteínas do Olho/genética , Proteínas de Membrana/genética , Mutação , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/genética , Animais , Sequência de Bases , Western Blotting , Cromatografia Gasosa , Progressão da Doença , Eletrorretinografia , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Fotorreceptoras de Vertebrados/metabolismo , Reação em Cadeia da Polimerase , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...