Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Biol Chem ; 289(30): 20570-82, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24898254

RESUMEN

Every day, shortly after light onset, photoreceptor cells shed approximately a tenth of their outer segment. The adjacent retinal pigment epithelial (RPE) cells phagocytize and digest shed photoreceptor outer segment, which provides a rich source of fatty acids that could be utilized as an energy substrate. From a microarray analysis, we found that RPE cells express particularly high levels of the mitochondrial HMG-CoA synthase 2 (Hmgcs2) compared with all other tissues (except the liver and colon), leading to the hypothesis that RPE cells, like hepatocytes, can produce ß-hydroxybutyrate (ß-HB) from fatty acids. Using primary human fetal RPE (hfRPE) cells cultured on Transwell filters with separate apical and basal chambers, we demonstrate that hfRPE cells can metabolize palmitate, a saturated fatty acid that constitutes .15% of all lipids in the photoreceptor outer segment, to produce ß-HB. Importantly, we found that hfRPE cells preferentially release ß-HB into the apical chamber and that this process is mediated primarily by monocarboxylate transporter isoform 1 (MCT1). Using a GC-MS analysis of (13)C-labeled metabolites, we showed that retinal cells can take up and metabolize (13)C-labeled ß-HB into various TCA cycle intermediates and amino acids. Collectively, our data support a novel mechanism of RPE-retina metabolic coupling in which RPE cells metabolize fatty acids to produce ß-HB, which is transported to the retina for use as a metabolic substrate.


Asunto(s)
Ácido 3-Hidroxibutírico/metabolismo , Proteínas del Ojo/metabolismo , Ácidos Grasos/metabolismo , Hidroximetilglutaril-CoA Sintasa/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Simportadores/metabolismo , Animales , Células Cultivadas , Ciclo del Ácido Cítrico/fisiología , Femenino , Humanos , Masculino , Ratones , Epitelio Pigmentado de la Retina/citología
2.
Exp Eye Res ; 126: 77-84, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24485945

RESUMEN

Mitochondrial dysfunction has been shown to contribute to age-related and proliferative retinal diseases. Over the past decade, the primary human fetal RPE (hfRPE) culture model has emerged as an effective tool for studying RPE function and mechanisms of retinal diseases. This model system has been rigorously characterized and shown to closely resemble native RPE cells at the genomic and protein level, and that they are capable of accomplishing the characteristic functions of a healthy native RPE (e.g., rod phagocytosis, ion and fluid transport, and retinoid cycle). In this review, we demonstrated that the metabolic activity of the RPE is an indicator of its health and state of differentiation, and present the hfRPE culture model as a valuable in vitro system for evaluating RPE metabolism in the context of RPE differentiation and retinal disease.


Asunto(s)
Metabolismo Energético/fisiología , Modelos Biológicos , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Células Epiteliales/fisiología , Feto/citología , Humanos
3.
Am J Med Genet A ; 164A(9): 2294-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24898194

RESUMEN

X-linked intellectual disability is the most common form of cognitive disability in males. Syndromic intellectual disability encompasses cognitive deficits with other medical and behavioral manifestations. Recently, a large family with a novel form of syndromic X-linked intellectual disability was characterized. Eight of 24 members of the family are male and had cognitive dysfunction, short stature, aphasia, skeletal abnormalities, and minor anomalies. To identify the causative gene(s), we performed exome sequencing in three affected boys, both parents, and an unaffected sister. We identified a haplotype consisting of eight variants located in cis within the linkage region that segregated with affected members in the family. Of these variants, two were novel. The first was at the splice-donor site of intron 7 (c.974+1G>T) in the cullin-RING ubiquitin ligase (E3) gene, CUL4B. This variant is predicted to result in failure to splice and remove intron 7 from the primary transcript. The second variant mapped to the 3'-UTR region of the KAISO gene (c.1127T>G). Sanger sequencing validated the variants in these relatives as well as in three affected males and five carriers. The KAISO gene variant was predicted to create a binding site for the microRNAs miR-4999 and miR-4774; however, luciferase expression assays failed to validate increased targeting of these miRNAs to the variant 3'-UTR. This SNP may affect 3'-UTR structure leading to decreased mRNA stability. Our results suggest that the intellectual disability phenotype in this family is caused by aberrant splicing and removal of intron 7 from CUL4B gene primary transcript.


Asunto(s)
Proteínas Cullin/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Mutación/genética , Sitios de Empalme de ARN/genética , Regiones no Traducidas 3'/genética , Sitios de Unión , Análisis Mutacional de ADN , Exoma/genética , Femenino , Ligamiento Genético , Genoma Humano/genética , Haplotipos/genética , Humanos , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Linaje , Fenotipo , Factores de Transcripción/genética
4.
J Biol Chem ; 287(24): 20491-503, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22523078

RESUMEN

The retinal pigment epithelium (RPE) plays a fundamental role in maintaining visual function and dedifferentiation of RPE contributes to the pathophysiology of several ocular diseases. To identify microRNAs (miRNAs) that may be involved in RPE differentiation, we compared the miRNA expression profiles of differentiated primary human fetal RPE (hfRPE) cells to dedifferentiated hfRPE cells. We found that miR-204/211, the two most highly expressed miRNAs in the RPE, were significantly down-regulated in dedifferentiated hfRPE cells. Importantly, transfection of pre-miR-204/211 into hfRPE cells promoted differentiation whereas adding miR-204/211 inhibitors led to their dedifferentiation. Microphthalmia-associated transcription factor (MITF) is a key regulator of RPE differentiation that was also down-regulated in dedifferentiated hfRPE cells. MITF knockdown decreased miR-204/211 expression and caused hfRPE dedifferentiation. Significantly, co-transfection of MITF siRNA with pre-miR-204/211 rescued RPE phenotype. Collectively, our data show that miR-204/211 promote RPE differentiation, suggesting that miR-204/211-based therapeutics may be effective treatments for diseases that involve RPE dedifferentiation such as proliferative vitreoretinopathy.


Asunto(s)
Diferenciación Celular/fisiología , Regulación hacia Abajo/fisiología , MicroARNs/biosíntesis , Factor de Transcripción Asociado a Microftalmía/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , MicroARNs/genética , Factor de Transcripción Asociado a Microftalmía/genética , Epitelio Pigmentado de la Retina/citología , Vitreorretinopatía Proliferativa/genética , Vitreorretinopatía Proliferativa/metabolismo , Vitreorretinopatía Proliferativa/terapia
5.
Curr Top Membr ; 70: 275-311, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23177990

RESUMEN

The SLC16A family of monocarboxylate transporters (MCTs) is composed of 14 members. MCT1 through MCT4 (MCTs 1-4) are H(+)-coupled monocarboxylate transporters, MCT8 and MCT10 transport thyroid hormone and aromatic amino acids, while the substrate specificity and function of other MCTs have yet to be determined. The focus of this review is on MCTs 1-4 because their role in lactate transport is intrinsically linked to cellular metabolism in various biological systems, including skeletal muscle, brain, retina, and testis. Although MCTs 1-4 all transport lactate, they differ in their transport kinetics and vary in tissue and subcellular distribution, where they facilitate "lactate-shuttling" between glycolytic and oxidative cells within tissues and across blood-tissue barriers. However, the role of MCTs 1-4 is not confined to cellular metabolism. By interacting with bicarbonate transport proteins and carbonic anhydrases, MCTs participate in the regulation of pH homeostasis and fluid transport in renal proximal tubule and corneal endothelium, respectively. Here, we provide a comprehensive review of MCTs 1-4, linking their cellular distribution to their functions in various parts of the human body, so that we can better understand the physiological roles of MCTs at the systemic level.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos/química , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/química , Simportadores/metabolismo , Animales , Transporte Biológico/fisiología , Endotelio Corneal/metabolismo , Glucólisis , Humanos , Concentración de Iones de Hidrógeno , Túbulos Renales Proximales/metabolismo , Cinética
6.
Eur J Pharm Biopharm ; 95(Pt B): 353-67, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25592325

RESUMEN

For patients with hereditary retinal diseases, retinal gene therapy offers significant promise for the prevention of retinal degeneration. While adeno-associated virus (AAV)-based systems remain the most popular gene delivery method due to their high efficiency and successful clinical results, other delivery systems, such as non-viral nanoparticles (NPs) are being developed as additional therapeutic options. NP technologies come in several categories (e.g., polymer, liposomes, peptide compacted DNA), several of which have been tested in mouse models of retinal disease. Here, we discuss the key biochemical features of the different NPs that influence how they are internalized into cells, escape from endosomes, and are delivered into the nucleus. We review the primary mechanism of NP uptake by retinal cells and highlight various NPs that have been successfully used for in vivo gene delivery to the retina and RPE. Finally, we consider the various strategies that can be implemented in the plasmid DNA to generate persistent, high levels of gene expression.


Asunto(s)
Terapia Genética/métodos , Nanopartículas , Enfermedades de la Retina/terapia , Animales , ADN/administración & dosificación , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Ratones , Nanotecnología/métodos , Retina/citología , Retina/metabolismo , Retina/fisiopatología , Enfermedades de la Retina/genética
7.
Cell Metab ; 17(6): 976-987, 2013 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-23747253

RESUMEN

Mitochondrial Ca(2+) uptake via the uniporter is central to cell metabolism, signaling, and survival. Recent studies identified MCU as the uniporter's likely pore and MICU1, an EF-hand protein, as its critical regulator. How this complex decodes dynamic cytoplasmic [Ca(2+)] ([Ca(2+)]c) signals, to tune out small [Ca(2+)]c increases yet permit pulse transmission, remains unknown. We report that loss of MICU1 in mouse liver and cultured cells causes mitochondrial Ca(2+) accumulation during small [Ca(2+)]c elevations but an attenuated response to agonist-induced [Ca(2+)]c pulses. The latter reflects loss of positive cooperativity, likely via the EF-hands. MICU1 faces the intermembrane space and responds to [Ca(2+)]c changes. Prolonged MICU1 loss leads to an adaptive increase in matrix Ca(2+) binding, yet cells show impaired oxidative metabolism and sensitization to Ca(2+) overload. Collectively, the data indicate that MICU1 senses the [Ca(2+)]c to establish the uniporter's threshold and gain, thereby allowing mitochondria to properly decode different inputs.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Animales , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Células Cultivadas , Células HEK293 , Células HeLa , Hepatocitos/metabolismo , Humanos , Ratones , Proteínas de Transporte de Membrana Mitocondrial/deficiencia , Proteínas de Transporte de Membrana Mitocondrial/genética , Membranas Mitocondriales/metabolismo , Interferencia de ARN , ARN Interferente Pequeño
8.
J Gen Physiol ; 133(6): 603-22, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19468075

RESUMEN

In the intact eye, the transition from light to dark alters pH, [Ca2+], and [K] in the subretinal space (SRS) separating the photoreceptor outer segments and the apical membrane of the retinal pigment epithelium (RPE). In addition to these changes, oxygen consumption in the retina increases with a concomitant release of CO2 and H2O into the SRS. The RPE maintains SRS pH and volume homeostasis by transporting these metabolic byproducts to the choroidal blood supply. In vitro, we mimicked the transition from light to dark by increasing apical bath CO2 from 5 to 13%; this maneuver decreased cell pH from 7.37 +/- 0.05 to 7.14 +/- 0.06 (n = 13). Our analysis of native and cultured fetal human RPE shows that the apical membrane is significantly more permeable (approximately 10-fold; n = 7) to CO2 than the basolateral membrane, perhaps due to its larger exposed surface area. The limited CO2 diffusion at the basolateral membrane promotes carbonic anhydrase-mediated HCO3 transport by a basolateral membrane Na/nHCO3 cotransporter. The activity of this transporter was increased by elevating apical bath CO2 and was reduced by dorzolamide. Increasing apical bath CO2 also increased intracellular Na from 15.7 +/- 3.3 to 24.0 +/- 5.3 mM (n = 6; P < 0.05) by increasing apical membrane Na uptake. The CO2-induced acidification also inhibited the basolateral membrane Cl/HCO3 exchanger and increased net steady-state fluid absorption from 2.8 +/- 1.6 to 6.7 +/- 2.3 microl x cm(-2) x hr(-1) (n = 5; P < 0.05). The present experiments show how the RPE can accommodate the increased retinal production of CO2 and H(2)O in the dark, thus preventing acidosis in the SRS. This homeostatic process would preserve the close anatomical relationship between photoreceptor outer segments and RPE in the dark and light, thus protecting the health of the photoreceptors.


Asunto(s)
Bicarbonatos/metabolismo , Dióxido de Carbono/farmacología , Epitelio Pigmentado de la Retina/metabolismo , Sodio/metabolismo , Transporte Biológico Activo , Anhidrasas Carbónicas/metabolismo , Feto/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Transporte Iónico , Iones/metabolismo , Modelos Biológicos , Simportadores de Sodio-Bicarbonato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA