Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Extracell Biol ; 2(10)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-38108061

RESUMEN

The retinal pigmented epithelium (RPE) constitutes the outer blood-retinal barrier, enables photoreceptor function of the eye, and is constantly exposed to oxidative stress. As such, dysfunction of the RPE underlies pathology leading to development of age-related macular degeneration (AMD), the leading cause of vision loss among the elderly in industrialized nations. A major responsibility of the RPE is to process photoreceptor outer segments, which relies on the proper functioning of its endocytic pathways and endosomal trafficking. Exosomes and other extracellular vesicles (EVs) from RPE are an essential part of these pathways and may be early indicators of cellular stress. To test the role of small EVs (sEVs) including exosomes, that may underlie the early stages of AMD, we used a polarized primary RPE cell culture model under chronic subtoxic oxidative stress. Unbiased proteomic analyses of highly purified basolateral sEVs from oxidatively stressed RPE cultures revealed changes in proteins involved in epithelial barrier integrity. There were also significant changes in proteins accumulating in the basal-side sub-RPE extracellular matrix during oxidative stress, that could be prevented with an inhibitor of sEV release. Thus, chronic subtoxic oxidative stress in primary RPE cultures induces changes in sEV content, including basal-side specific desmosome and hemidesmosome shedding via sEVs. These findings provide novel biomarkers of early cellular dysfunction and opportunity for therapeutic intervention in age-related retinal diseases (e.g., AMD).

2.
bioRxiv ; 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37398366

RESUMEN

The retinal pigmented epithelium (RPE) constitutes the outer blood-retinal barrier, enables photoreceptor function of the eye, and is constantly exposed to oxidative stress. As such, dysfunction of the RPE underlies pathology leading to development of age-related macular degeneration (AMD), the leading cause of vision loss among the elderly in industrialized nations. A major responsibility of the RPE is to process photoreceptor outer segments, which relies on the proper functioning of its endocytic pathways and endosomal trafficking. Exosomes and other extracellular vesicles from RPE are an essential part of these pathways and may be early indicators of cellular stress. To test the role of exosomes that may underlie the early stages of AMD, we used a polarized primary RPE cell culture model under chronic subtoxic oxidative stress. Unbiased proteomic analyses of highly purified basolateral exosomes from oxidatively stressed RPE cultures revealed changes in proteins involved in epithelial barrier integrity. There were also significant changes in proteins accumulating in the basal-side sub-RPE extracellular matrix during oxidative stress, that could be prevented with an inhibitor of exosome release. Thus, chronic subtoxic oxidative stress in primary RPE cultures induces changes in exosome content, including basal-side specific desmosome and hemidesmosome shedding via exosomes. These findings provide novel biomarkers of early cellular dysfunction and opportunity for therapeutic intervention in age-related retinal diseases, (e.g., AMD) and broadly from blood-CNS barriers in other neurodegenerative diseases.

3.
Invest Ophthalmol Vis Sci ; 64(10): 25, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37471073

RESUMEN

Purpose: Complement dysregulation in the eye has been implicated in the pathogenesis of age-related macular degeneration (AMD), and genetic variants of complement factor H (CFH) are strongly associated with AMD risk. We therefore aimed to untangle the role of CFH and its splice variant, factor H-like 1 (FHL-1), in ocular complement regulation derived from local versus circulating sources. We assessed the therapeutic efficacy of adeno-associated viruses (AAVs) expressing human FHL-1 and a truncated version of CFH (tCFH), which retains the functional N- and C-terminal ends of the CFH protein, in restoring the alternative complement pathway in Cfh-/- mouse eyes and plasma. Methods: Using Cfh-/- mice as a model of complement dysregulation, AAV vectors expressing tCFH or FHL-1 were injected subretinally or via tail vein, and the efficacy of the constructs was evaluated. Results: Following subretinal injections, tCFH expression rescued factor B (FB) retention in the eye, but FHL-1 expression did not. By contrast, both constructs restored FB detection in plasma following tail vein injections. Both tCFH and FHL-1 proteins accumulated in the posterior eyecup from the circulation following liver transduction; however, neither was able to significantly regulate local ocular complement. Conclusions: Our findings demonstrate that the C-terminus of human CFH is necessary for complement regulation in the murine eye. Furthermore, exogenous CFH must be synthesized locally to maximize complement regulation in the retina. These findings establish a critical foundation for development of CFH augmentation-based gene therapies for the eye.


Asunto(s)
Factor H de Complemento , Degeneración Macular , Animales , Humanos , Ratones , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Hígado/metabolismo , Degeneración Macular/genética , Polimorfismo de Nucleótido Simple , Retina/metabolismo , Ratones Noqueados
4.
J Biol Chem ; 295(39): 13601-13616, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32737203

RESUMEN

Strong evidence suggests that dysregulated lipid metabolism involving dysfunction of the retinal pigmented epithelium (RPE) underlies the pathogenesis of age-related macular degeneration (AMD), the leading cause of irreversible blindness in the elderly. A hallmark of AMD is the overproduction of lipid- and protein-rich extracellular deposits that accumulate in the extracellular matrix (Bruch's membrane (BrM)) adjacent to the RPE. We analyzed apolipoprotein A-1 (ApoA-1)-containing lipoproteins isolated from BrM of elderly human donor eyes and found a unique proteome, distinct from high-density lipoprotein (HDL) isolated from donor plasma of the same individuals. The most striking difference is higher concentrations of ApoB and ApoE, which bind to glycosaminoglycans. We hypothesize that this interaction promotes lipoprotein deposition onto BrM glycosaminoglycans, initiating downstream effects that contribute to RPE dysfunction/death. We tested this hypothesis using two potential therapeutic strategies to alter the lipoprotein/protein profile of these extracellular deposits. First, we used short heparan sulfate oligosaccharides to remove lipoproteins already deposited in both the extracellular matrix of RPE cells and aged donor BrM tissue. Second, an ApoA-1 mimetic, 5A peptide, was demonstrated to modulate the composition and concentration of apolipoproteins secreted from primary porcine RPE cells. Significantly, in a mouse model of AMD, this 5A peptide altered the proteomic profile of circulating HDL and ameliorated some of the potentially harmful changes to the protein composition resulting from the high-fat, high-cholesterol diet in this model. Together, these results suggest that targeting HDL interactions with BrM represents a new strategy to slow AMD progression in humans.


Asunto(s)
Lipoproteínas HDL/metabolismo , Degeneración Macular/metabolismo , Animales , Apolipoproteína A-I/análisis , Apolipoproteína A-I/metabolismo , Lámina Basal de la Coroides/metabolismo , Células Cultivadas , Humanos , Lipoproteínas HDL/sangre , Lipoproteínas HDL/aislamiento & purificación , Ratones , Epitelio Pigmentado de la Retina/metabolismo , Porcinos
5.
Proc Natl Acad Sci U S A ; 116(9): 3703-3711, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808757

RESUMEN

One of the strongest susceptibility genes for age-related macular degeneration (AMD) is complement factor H (CFH); however, its impact on AMD pathobiology remains unresolved. Here, the effect of the principal AMD-risk-associated CFH variant (Y402H) on the development and progression of age-dependent AMD-like pathologies was determined in vivo. Transgenic mice expressing equal amounts of the full-length normal human CFH Y402 (CFH-Y/0) or the AMD-risk associated CFH H402 (CFH-H/H) variant on a Cfh-/- background were aged to 90 weeks and switched from normal diet (ND) to a high fat, cholesterol-enriched (HFC) diet for 8 weeks. The resulting phenotype was compared with age-matched controls maintained on ND. Remarkably, an AMD-like phenotype consisting of vision loss, increased retinal pigmented epithelium (RPE) stress, and increased basal laminar deposits was detected only in aged CFH-H/H mice following the HFC diet. These changes were not observed in aged CFH-Y/0 mice or in younger (36- to 40-week-old) CFH mice of both genotypes fed either diet. Biochemical analyses of aged CFH mice after HFC diet revealed genotype-dependent changes in plasma and eyecup lipoproteins, but not complement activation, which correlated with the AMD-like phenotype in old CFH-H/H mice. Specifically, apolipoproteins B48 and A1 are elevated in the RPE/choroid of the aged CFH-H/H mice compared with age-matched control CFH-Y/0 fed a HFC diet. Hence, we demonstrate a functional consequence of the Y402H polymorphism in vivo, which promotes AMD-like pathology development and affects lipoprotein levels in aged mice. These findings support targeting lipoproteins as a viable therapeutic strategy for treating AMD.


Asunto(s)
Activación de Complemento/genética , Factor H de Complemento/genética , Lipoproteínas/genética , Degeneración Macular/genética , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Genotipo , Humanos , Lipoproteínas/metabolismo , Degeneración Macular/patología , Masculino , Ratones , Ratones Transgénicos/genética , Polimorfismo de Nucleótido Simple/genética , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
6.
Sci Rep ; 8(1): 17327, 2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30455433

RESUMEN

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.

7.
Invest Ophthalmol Vis Sci ; 59(2): 662-673, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29392311

RESUMEN

Purpose: A large body of evidence supports a central role for complement activation in the pathobiology of age-related macular degeneration (AMD), including plasma complement component 5a (C5a). Interestingly, C5a is a chemotactic agent for monocytes, a cell type also shown to contribute to AMD. However, the role monocytes play in the pathogenesis of "dry" AMD and the pharmacologic potential of targeting C5a to regulate these cells are unclear. We addressed these questions via C5a blockade in a unique model of early/intermediate dry AMD and large panel flow cytometry to immunophenotype monocytic involvement. Methods: Heterozygous complement factor H (Cfh+/-) mice aged to 90 weeks were fed a high-fat, cholesterol-enriched diet (Cfh+/-∼HFC) for 8 weeks and were given weekly intraperitoneal injections of 30 mg/kg anti-C5a (4C9, Pfizer). Flow cytometry, retinal pigmented epithelium (RPE) flat mounts, and electroretinograms were used to characterize anti-C5a treatment. Results: Aged Cfh+/- mice developed RPE damage, sub-RPE basal laminar deposits, and attenuation of visual function and immune cell recruitment to the choroid that was accompanied by expression of inflammatory and extracellular matrix remodeling genes following 8 weeks of HFC diet. Concomitant systemic administration of an anti-C5a antibody successfully inhibited local recruitment of mononuclear phagocytes to the choroid-RPE interface but did not ameliorate these AMD-like pathologies in this mouse model. Conclusions: These results show that immunotherapy targeting C5a is not sufficient to block the development of the AMD-like pathologies observed in Cfh+/-∼HFC mice and suggest that other complement components or molecules/mechanisms may be driving "early" and "intermediate" AMD pathologies.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Neovascularización Coroidal/terapia , Complemento C5a/antagonistas & inhibidores , Modelos Animales de Enfermedad , Atrofia Geográfica/terapia , Inmunoterapia , Animales , Colesterol en la Dieta/administración & dosificación , Neovascularización Coroidal/inmunología , Neovascularización Coroidal/patología , Activación de Complemento , Complemento C5a/inmunología , Electrorretinografía , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Atrofia Geográfica/inmunología , Atrofia Geográfica/patología , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Epitelio Pigmentado de la Retina/patología
8.
Sci Rep ; 7(1): 4901, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28687758

RESUMEN

The retinal pigmented epithelium (RPE) forms the outer blood-retinal barrier in the eye and its polarity is responsible for directional secretion and uptake of proteins, lipoprotein particles and extracellular vesicles (EVs). Such a secretional division dictates directed interactions between the systemic circulation (basolateral) and the retina (apical). Our goal is to define the polarized proteomes and physical characteristics of EVs released from the RPE. Primary cultures of porcine RPE cells were differentiated into polarized RPE monolayers on permeable supports. EVs were isolated from media bathing either apical or basolateral RPE surfaces, and two subpopulations of small EVs including exosomes, and dense EVs, were purified and processed for proteomic profiling. In parallel, EV size distribution and concentration were determined. Using protein correlation profiling mass spectrometry, a total of 631 proteins were identified in exosome preparations, 299 of which were uniquely released apically, and 94 uniquely released basolaterally. Selected proteins were validated by Western blot. The proteomes of these exosome and dense EVs preparations suggest that epithelial polarity impacts directional release. These data serve as a foundation for comparative studies aimed at elucidating the role of exosomes in the molecular pathophysiology of retinal diseases and help identify potential therapeutic targets and biomarkers.


Asunto(s)
Células Epiteliales/metabolismo , Exosomas/química , Proteínas del Ojo/genética , Proteoma/genética , Epitelio Pigmentado de la Retina/metabolismo , Animales , Barrera Hematorretinal/metabolismo , Diferenciación Celular , Polaridad Celular , Impedancia Eléctrica , Células Epiteliales/citología , Exosomas/metabolismo , Proteínas del Ojo/clasificación , Proteínas del Ojo/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Espectrometría de Masas , Anotación de Secuencia Molecular , Cultivo Primario de Células , Proteoma/clasificación , Proteoma/metabolismo , Epitelio Pigmentado de la Retina/citología , Porcinos
9.
Proc Natl Acad Sci U S A ; 112(23): E3040-9, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-25991857

RESUMEN

Complement factor H (CFH) is a major susceptibility gene for age-related macular degeneration (AMD); however, its impact on AMD pathobiology is unresolved. Here, the role of CFH in the development of AMD pathology in vivo was interrogated by analyzing aged Cfh(+/-) and Cfh(-/-) mice fed a high-fat, cholesterol-enriched diet. Strikingly, decreased levels of CFH led to increased sub-retinal pigmented epithelium (sub-RPE) deposit formation, specifically basal laminar deposits, following high-fat diet. Mechanistically, our data show that deposits are due to CFH competition for lipoprotein binding sites in Bruch's membrane. Interestingly and despite sub-RPE deposit formation occurring in both Cfh(+/-) and Cfh(-/-) mice, RPE damage accompanied by loss of vision occurred only in old Cfh(+/-) mice. We demonstrate that such pathology is a function of excess complement activation in Cfh(+/-) mice versus complement deficiency in Cfh(-/-) animals. Due to the CFH-dependent increase in sub-RPE deposit height, we interrogated the potential of CFH as a previously unidentified regulator of Bruch's membrane lipoprotein binding and show, using human Bruch's membrane explants, that CFH removes endogenous human lipoproteins in aged donors. Thus, advanced age, high-fat diet, and decreased CFH induce sub-RPE deposit formation leading to complement activation, which contributes to RPE damage and visual function impairment. This new understanding of the complicated interactions of CFH in AMD-like pathology provides an improved foundation for the development of targeted therapies for AMD.


Asunto(s)
Factor H de Complemento/fisiología , Degeneración Macular/fisiopatología , Animales , Coroides/metabolismo , Coroides/patología , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Dieta Alta en Grasa , Degeneración Macular/patología , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
10.
Am J Pathol ; 185(1): 29-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25447048

RESUMEN

Complement factor H (CFH) is an important regulatory protein in the alternative pathway of the complement system, and CFH polymorphisms increase the genetic risk of age-related macular degeneration dramatically. These same human CFH variants have also been associated with dense deposit disease. To mechanistically study the function of CFH in the pathogenesis of these diseases, we created transgenic mouse lines using human CFH bacterial artificial chromosomes expressing full-length human CFH variants and crossed these to Cfh knockout (Cfh(-/-)) mice. Human CFH protein inhibited cleavage of mouse complement component 3 and factor B in plasma and in retinal pigment epithelium/choroid/sclera, establishing that human CFH regulates activation of the mouse alternative pathway. One of the mouse lines, which express relatively higher levels of CFH, demonstrated functional and structural protection of the retina owing to the Cfh deletion. Impaired visual function, detected as a deficit in the scotopic electroretinographic response, was improved in this transgenic mouse line compared with Cfh(-/-) mice, and transgenics had a thicker outer nuclear layer and less sub-retinal pigment epithelium deposit accumulation. In addition, expression of human CFH also completely protected the mice from developing kidney abnormalities associated with loss of CFH. These humanized CFH mice present a valuable model for study of the molecular mechanisms of age-related macular degeneration and dense deposit disease and for testing therapeutic targets.


Asunto(s)
Enfermedades Renales/genética , Degeneración Macular/genética , Enfermedades de la Retina/genética , Animales , Coroides/patología , Complemento C3/metabolismo , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Cruzamientos Genéticos , Electrorretinografía , Humanos , Enfermedades Renales/patología , Degeneración Macular/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Retina/metabolismo , Enfermedades de la Retina/patología , Epitelio Pigmentado de la Retina/patología , Esclerótica/patología , Ovinos
11.
Adv Exp Med Biol ; 801: 213-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24664701

RESUMEN

Variations in several complement genes are now known to be significant risk factors for the development of age-related macular degeneration (AMD). Despite dramatic effects on disease susceptibility, the underlying mechanisms by which common polymorphisms in complement proteins alter disease risk have remained unclear. Genetically modified mice in which the activity of the complement has been altered are available and can be used to investigate the role of complement in the pathogenesis of AMD. In this mini review, we will discuss some existing complement models of AMD and our efforts to develop and characterize the ocular phenotype in a variety of mice in which complement is either chronically activated or inhibited. A spectrum of complement dysregulation was modeled on the APOE4 AMD mouse model by crossing these mice to complement factor H knockout (cfh-/-) mice to test the impact of excess complement activation, and by crossing them to soluble-complement-receptor-1-related protein y (sCrry) mice, in which sCrry acts as a potent inhibitor of mouse complement acting in a manner similar to CFH. In addition, we have also generated humanized CFH mice expressing normal and risk variants of CFH.


Asunto(s)
Factor H de Complemento/deficiencia , Factor H de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Enfermedades Renales/inmunología , Degeneración Macular/inmunología , Animales , Factor H de Complemento/genética , Modelos Animales de Enfermedad , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Ratones , Ratones Noqueados
12.
Proc Natl Acad Sci U S A ; 108(28): E279-87, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21690377

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of visual dysfunction worldwide. Amyloid ß (Aß) peptides, Aß1-40 (Aß40) and Aß1-42 (Aß42), have been implicated previously in the AMD disease process. Consistent with a pathogenic role for Aß, we show here that a mouse model of AMD that invokes multiple factors that are known to modify AMD risk (aged human apolipoprotein E 4 targeted replacement mice on a high-fat, cholesterol-enriched diet) presents with Aß-containing deposits basal to the retinal pigmented epithelium (RPE), histopathologic changes in the RPE, and a deficit in scotopic electroretinographic response, which is reflective of impaired visual function. Strikingly, these electroretinographic deficits are abrogated in a dose-dependent manner by systemic administration of an antibody targeting the C termini of Aß40 and Aß42. Concomitant reduction in the levels of Aß and activated complement components in sub-RPE deposits and structural preservation of the RPE are associated with anti-Aß40/42 antibody immunotherapy and visual protection. These observations are consistent with the reduction in amyloid plaques and improvement of cognitive function in mouse models of Alzheimer's disease treated with anti-Aß antibodies. They also implicate Aß in the pathogenesis of AMD and identify Aß as a viable therapeutic target for its treatment.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Degeneración Macular/terapia , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/metabolismo , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/uso terapéutico , Apolipoproteína E4/genética , Proteínas del Sistema Complemento/metabolismo , Grasas de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Inmunoterapia , Degeneración Macular/etiología , Degeneración Macular/patología , Degeneración Macular/fisiopatología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Baja Visión/fisiopatología , Baja Visión/prevención & control
13.
J Immunol ; 185(9): 5486-94, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20876352

RESUMEN

An imbalance between activation and inhibition of the complement system has been implicated in the etiologies of numerous common diseases. Allotypic variants of a key complement fluid-phase regulatory protein, complement factor H (CFH), are strongly associated with age-related macular degeneration (AMD), a leading cause of worldwide visual dysfunction, although its specific role in AMD pathogenesis is still not clear. CFH was isolated from individuals carrying combinations of two of the nonsynonymous coding variants most strongly associated with AMD risk, V62/H402 (risk haplotype variants), I62/Y402 (nonrisk haplotype variants), and V62/Y402. These proteins were used in two functional assays (cell surface- and fluid-phase-based) measuring cofactor activity of CFH in the factor I-mediated cleavage of C3b. Although no variant-specific differences in the cofactor activity were detected, when heparan sulfate (HS) was added to these assays, it accelerated the rate of C3b cleavage, and this effect could be modulated by degree of HS sulfation. Bruch's membrane/choroid, a site of tissue damage in AMD, contains high concentrations of glycosaminoglycans, including HS. Addition of human Bruch's membrane/choroid to the fluid-phase assay accelerated the C3b cleavage, and this effect was lost posttreatment of the tissue with heparinase III. Binding of CFH variants to Bruch's membrane/choroid isolated from elderly, non-AMD donor eyes, was similar, as was the functional activity of bound CFH. These findings refine our understanding of interactions of HS and complement and support the hypothesis that these interactions play a role in the transition between normal aging and AMD in Bruch's membrane/choroid.


Asunto(s)
Lámina Basal de la Coroides/inmunología , Vía Alternativa del Complemento/inmunología , Heparitina Sulfato/inmunología , Degeneración Macular/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Lámina Basal de la Coroides/química , Lámina Basal de la Coroides/metabolismo , Complemento C3b/inmunología , Complemento C3b/metabolismo , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Femenino , Heparitina Sulfato/metabolismo , Humanos , Degeneración Macular/metabolismo , Degeneración Macular/patología , Masculino , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo
14.
Invest Ophthalmol Vis Sci ; 50(4): 1540-5, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19029036

RESUMEN

PURPOSE: Variations in the complement factor H (CFH) gene are tightly associated with age-related macular degeneration (AMD) across diverse populations. Of the many nonsynonymous coding variants in CFH, two are most strongly associated with increased risk of AMD: isoleucine 62 to valine (I62V) and tyrosine 402 to histidine (Y402H). Detection of these variations in a patient's blood is important for a risk assessment of AMD and disease prognosis. However, traditional methods of genetic analysis cannot be used for measuring CFH allotypes in some sources of human plasma and other biological fluids not containing DNA. The purpose was to develop a protein-based method of detecting CFH allotypes. METHODS: A combination of a single-step affinity enrichment of CFH, gel separation, and mass spectrometry identification of the CFH peptides spanning amino acids at positions 62 and 402 was used to identify individual CFH allotypes. RESULTS: The CFH isoforms V62, I62, H402, and Y402 were reliably detected based on identification of tryptic peptides with masses of 1148.59 Da, 1162.60 Da, 2031.88 Da, and 2057.88 Da, respectively, using MALDI-TOF-TOF. The presence or absence pattern of these peptides in mass spectra of different CFH samples robustly correlated with all nine genotypes of CFH, as a result of variations at positions 62 and 402. CONCLUSIONS: A rapid and sensitive method has been developed for detection of V62, I62, H402, and Y402 variants of CFH in human plasma samples using mass spectrometry. This method can be used in clinical laboratories equipped with a basic inexpensive mass spectrometer capable of performing peptide fingerprinting.


Asunto(s)
Polimorfismo de Nucleótido Simple/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Cromatografía de Afinidad , Factor H de Complemento/genética , Electroforesis en Gel de Poliacrilamida , Genotipo , Humanos , Fragmentos de Péptidos/análisis
15.
Biochim Biophys Acta ; 1773(9): 1473-82, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17673310

RESUMEN

The phosphatase of regenerating liver-1, PRL-1, gene was detected in a screen for foveal cone photoreceptor-associated genes. It encodes a small protein tyrosine phosphatase that was previously immunolocalized to the photoreceptors in primate retina. Here we report that in cones and cone-derived cultured cells both PRL-1 activity and PRL-1 gene expression are modulated under oxidative stress. Oxidation reversibly inhibited the phosphatase activity of PRL-1 due to the formation of an intramolecular disulfide bridge between Cys104 within the active site and another conserved Cys, Cys49. This modulation was observed in vitro, in cell culture and in isolated retinas exposed to hydrogen peroxide. The same treatment caused a rapid increase in PRL-1 expression levels in cultured cells which could be blocked by the protein translation inhibitor, cycloheximide. Increased PRL-1 expression was also observed in living rats subjected to constant light exposure inducing photooxidative stress. We further demonstrated that both oxidation and overexpression of PRL-1 upon oxidative stress are greatly enhanced by inhibition of the glutathione system responsible for cellular redox regulation. These findings suggest that PRL-1 is a molecular component of the photoreceptor's response to oxidative stress acting upstream of the glutathione system.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de la Membrana/metabolismo , Estrés Oxidativo , Proteínas Tirosina Fosfatasas/metabolismo , Retina/metabolismo , Animales , Dominio Catalítico , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular , Células Cultivadas , Cicloheximida/farmacología , Cisteína/metabolismo , Ditiotreitol/análogos & derivados , Ditiotreitol/farmacología , Relación Dosis-Respuesta a Droga , Glutatión/análisis , Peróxido de Hidrógeno/farmacología , Proteínas Inmediatas-Precoces/análisis , Proteínas Inmediatas-Precoces/química , Proteínas Inmediatas-Precoces/genética , Proteínas de la Membrana/análisis , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Oxidantes/farmacología , Oxidación-Reducción , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Tirosina Fosfatasas/análisis , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/genética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Sustancias Reductoras/farmacología , Retina/citología , Retina/efectos de los fármacos , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/enzimología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos
16.
Invest Ophthalmol Vis Sci ; 44(10): 4515-21, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14507900

RESUMEN

PURPOSE: To analyze the patterns of expression of the cryptochromes, CRY1 and CRY2, in the human retina and to correlate expression of these putative blue-light receptors with nonvisual photoreceptor localization. METHODS: CRY1 and CRY2 mRNA expression was analyzed in 4-mm diameter punches of macula and midperipheral human retina by quantitative RT-PCR. CRY2 protein expression was examined by immunohistochemistry in cross sections of human retina, and its subcellular localization was determined by immunoblot analysis of fractionated human retinal extracts. RESULTS: CRY2 mRNA was 11 times more abundant than CRY1 throughout adult human retina. CRY2 immunoreactivity was detected in most cells in the ganglion cell layer (GCL) and in a subset of cells in the inner nuclear layer (INL) in both the macula and periphery. Immunoperoxidase staining further revealed that CRY2 was localized throughout the cytoplasm of cells in the GCL as well as within nuclei. This intracellular localization of CRY2 was confirmed by immunoblot analysis of fractionated human retinal extracts. CONCLUSIONS: Photopigments governing circadian photoreception have been localized to the inner retina. The relative abundance of CRY2 transcripts, coupled with CRY2 localization to the inner retina, supports a photoreceptive role for CRY2 in human retina. Furthermore, the discovery that CRY2 is also localized within the cytoplasm of some cells in the GCL, suggests it may perform a function separate from its known nuclear role in the transcriptional feedback loop underlying the molecular circadian clock.


Asunto(s)
Proteínas de Drosophila , Proteínas del Ojo , Flavoproteínas/genética , Células Fotorreceptoras de Invertebrados , Retina/metabolismo , Pigmentos Retinianos/genética , Adulto , Anciano , Criptocromos , Femenino , Flavoproteínas/metabolismo , Flavoproteínas/efectos de la radiación , Humanos , Inmunohistoquímica , Luz , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G , Células Ganglionares de la Retina/metabolismo , Pigmentos Retinianos/metabolismo , Pigmentos Retinianos/efectos de la radiación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fracciones Subcelulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...