Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Invest Ophthalmol Vis Sci ; 58(11): 4545-4558, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28877319

RESUMEN

Purpose: Sigma 1 Receptor (Sig1R) is a novel therapeutic target in neurodegenerative diseases, including retinal disease. Sig1R-/- mice have late-onset retinal degeneration with ganglion cell loss that worsens under stress. Whether Sig1R plays a role in maintaining other retinal neurons is unknown, but was investigated here using rd10 mice, a model of severe photoreceptor degeneration. Methods: Wild-type, rd10, and rd10/Sig1R-/- mice were subjected to ERG and spectral-domain optical coherence tomography (SD-OCT) to assess visual function/structure in situ. Retinas imaged microscopically were subjected to morphometric analysis, immunodetection of cones, and analysis of gliosis. Oxidative and endoplasmic reticulum (ER) stress was evaluated at mRNA/protein levels. Results: Photopic ERG responses were reduced significantly in rd10/Sig1R-/- versus rd10 mice at P28 (31 ± 6 vs. 56 ± 7 µV), indicating accelerated cone loss when Sig1R was absent. At P28, SD-OCT revealed reduced retinal thickness in rd10/Sig1R-/- mice (60% of WT) versus rd10 (80% of WT). Morphometric analysis disclosed profound photoreceptor nuclei loss in rd10/Sig1R-/- versus rd10 mice. rd10/Sig1R-/- mice had 35% and 60% fewer photoreceptors, respectively, at P28 and P35, than rd10. Peanut agglutinin cone labeling decreased significantly; gliosis increased significantly in rd10/Sig1R-/- versus rd10 mice. At P21, NRF2 levels increased in rd10/Sig1R-/- mice versus rd10 and downstream antioxidants increased indicating oxidative stress. At P28, ER stress genes/proteins, especially XBP1, a potent transcriptional activator of the unfolded protein response and CHOP, a proapoptotic transcription factor, increased significantly in rd10/Sig1R-/- mice versus rd10. Conclusions: Photoreceptor cell degeneration accelerates and cone function diminishes much earlier in rd10/Sig1R-/- than rd10 mice emphasizing the importance of Sig1R as a modulator of retinal cell survival.


Asunto(s)
Apoptosis , Modelos Animales de Enfermedad , Receptores sigma/fisiología , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/patología , Animales , Electrorretinografía , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Retina/fisiología , Tomografía de Coherencia Óptica , Receptor Sigma-1
2.
Invest Ophthalmol Vis Sci ; 58(4): 1954-1963, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28384716

RESUMEN

Purpose: Hyperhomocysteinemia (Hhcy) is implicated in certain retinal neurovascular diseases, although whether it is causative remains uncertain. In isolated ganglion cells (GCs), mild Hhcy induces profound death, whereas retinal phenotypes in Hhcy mice caused by mutations in remethylation (methylene tetrahydrofolatereductase [Mthfr+/-]) or transsulfuration pathways (cystathionine ß-synthase [Cbs+/-]) demonstrate mild GC loss and mild vasculopathy. The current work investigated compensation in vivo of one pathway for the other, and, because the transsulfuration pathway yields cysteine necessary for formation of glutathione (GSH), taurine, and hydrogen sulfide (H2S), they were analyzed also. Methods: Retinas isolated from wild-type (WT), Mthfr+/-, and Cbs+/- mice (12 and 22 weeks) were analyzed for methylene tetrahydrofolate reductase (MTHFR), cystathionine-ß-synthase (CBS), and cystathionase (CTH) RNA/protein levels. Retinas were evaluated for levels of reduced:oxidized GSH (GSH:GSSG), Slc7a11 (xCT), taurine, taurine transporter (TAUT), and H2S. Results: Aside from decreased CBS RNA/protein levels in Cbs+/- retinas, there were minimal alterations in remethylation/transsulfuration pathways in the two mutant mice strains. Glutathione and taurine levels in Mthfr+/- and Cbs+/- retinas were similar to WT, which may be due to robust levels of xCT and TAUT in mutant retinas. Interestingly, levels of H2S were markedly increased in retinas of Mthfr+/- and Cbs+/- mice compared with WT. Conclusions: Ganglion cell loss and vasculopathy observed in Mthfr+/- and Cbs+/- mouse retinas may be milder than expected, not because of compensatory increases of enzymes in remethylation/transsulfuration pathways, but because downstream transsulfuration pathway products GSH, taurine, and H2S are maintained at robust levels. Elevation of H2S is particularly intriguing owing to neuroprotective properties reported for this gasotransmitter.


Asunto(s)
Regulación de la Expresión Génica , Glutatión/metabolismo , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Células Ganglionares de la Retina/metabolismo , Taurina/metabolismo , Animales , Modelos Animales de Enfermedad , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Metilenotetrahidrofolato Reductasa (NADPH2)/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/metabolismo , Retina/patología , Enfermedades de la Retina/etiología , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Células Ganglionares de la Retina/patología
3.
Adv Exp Med Biol ; 964: 267-284, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28315277

RESUMEN

This review article focuses on studies of Sigma 1 Receptor (Sigma1R) and retina . It provides a brief overview of the earliest pharmacological studies performed in the late 1990s that provided evidence of the presence of Sigma1R in various ocular tissues. It then describes work from a number of labs concerning the location of Sigma1R in several retinal cell types including ganglion, Müller glia , and photoreceptors . The role of Sigma1R ligands in retinal neuroprotection is emphasized. Early studies performed in vitro clearly showed that targeting Sigma1R could attenuate stress-induced retinal cell loss. These studies were followed by in vivo experiments. Data about the usefulness of targeting Sigma1R to prevent ganglion cell loss associated with diabetic retinopathy are reviewed. Mechanisms of Sigma1R-mediated retinal neuroprotection involving Müller cells , especially in modulating oxidative stress are described along with information about the retinal phenotype of mice lacking Sigma1R (Sigma1R -/- mice). The retina develops normally in Sigma1R -/- mice, but after many months there is evidence of apoptosis in the optic nerve head, decreased ganglion cell function and eventual loss of these cells. Additional studies using the Sigma1R -/- mice provide strong evidence that in the retina, Sigma1R plays a key role in modulating cellular stress. Recent work has shown that targeting Sigma1R may extend beyond protection of ganglion cells to include photoreceptor cell degeneration as well.


Asunto(s)
Receptores sigma/metabolismo , Retina/metabolismo , Animales , Células Ependimogliales/metabolismo , Humanos , Estrés Oxidativo/fisiología , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Receptor Sigma-1
4.
Proc Natl Acad Sci U S A ; 113(26): E3764-72, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27298364

RESUMEN

Retinal degenerative diseases are major causes of untreatable blindness, and novel approaches to treatment are being sought actively. Here we explored the activation of a unique protein, sigma 1 receptor (Sig1R), in the treatment of PRC loss because of its multifaceted role in cellular survival. We used Pde6ß(rd10) (rd10) mice, which harbor a mutation in the rod-specific phosphodiesterase gene Pde6ß and lose rod and cone photoreceptor cells (PRC) within the first 6 wk of life, as a model for severe retinal degeneration. Systemic administration of the high-affinity Sig1R ligand (+)-pentazocine [(+)-PTZ] to rd10 mice over several weeks led to the rescue of cone function as indicated by electroretinographic recordings using natural noise stimuli and preservation of cone cells upon spectral domain optical coherence tomography and retinal histological examination. The protective effect appears to result from the activation of Sig1R, because rd10/Sig1R(-/-) mice administered (+)-PTZ exhibited no cone preservation. (+)-PTZ treatment was associated with several beneficial cellular phenomena including attenuated reactive gliosis, reduced microglial activation, and decreased oxidative stress in mutant retinas. To our knowledge, this is the first report that activation of Sig1R attenuates inherited PRC loss. The findings may have far-reaching therapeutic implications for retinal neurodegenerative diseases.


Asunto(s)
Receptores sigma/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Degeneración Retiniana/metabolismo , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Receptores sigma/genética , Degeneración Retiniana/congénito , Degeneración Retiniana/tratamiento farmacológico , Degeneración Retiniana/genética , Receptor Sigma-1
5.
Invest Ophthalmol Vis Sci ; 57(6): 2770-81, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27206247

RESUMEN

PURPOSE: Sigma receptor 1 (Sigma1R), a nonopioid putative molecular chaperone, has neuroprotective properties in retina. This study sought to determine whether delaying administration of (+)-pentazocine, a high-affinity Sigma1R ligand after onset of diabetes in Ins2Akita/+ diabetic mice would afford retinal neuroprotection and to determine consequences on retinal phenotype in Ins2Akita/+ diabetic mice in the absence of Sigma1R. METHODS: Ins2Akita/+ diabetic and WT mice received intraperitoneal injections of (+)-pentazocine beginning 4 or 8 weeks after onset of diabetes; eyes were harvested at 25 weeks. Retinal histologic sections were analyzed to determine thicknesses of retinal layers, number of ganglion cells, and evidence of gliosis (increased glial fibrillary acidic protein levels). Ins2Akita/+/Sig1R-/-mice were generated and subjected to in vivo assessment of retinal architecture (optical coherence tomography [OCT]) and retinal vasculature using fluorescein angiography (FA) at 12 and 16 weeks compared with age-matched Ins2Akita/+ mice. Eyes were then harvested for retinal morphometric assessment and gliosis assessment. RESULTS: Wild-type mice had 13 ± 0.06 cells/100 µm retinal length; cell bodies in Ins2Akita/+ mice injected 4 and 8 weeks after onset of diabetes with (+)-pentazocine retained significantly more ganglion cells compared with Ins2Akita/+ mice (9 ± 0.04) and demonstrated significant attenuation of gliosis. Ins2Akita/+/Sig1R-/-mouse retinas, analyzed to determine whether the Ins2Akita/+ phenotype was accelerated when lacking Sigma1R, revealed increased nerve fiber layer thickness (OCT), evidence of vitreal opacities, and vessel beading (FA) compared with Ins2Akita/+ mice. Morphometric analysis revealed significantly fewer ganglion cells in Ins2Akita/+/Sig1R-/-mice compared with Ins2Akita/+ mice. CONCLUSIONS: Sigma1R may be a novel retinal stress modulator, and targeting it even after disease onset may afford retinal neuroprotection.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética/complicaciones , Regulación de la Expresión Génica , ARN/genética , Receptores sigma/genética , Degeneración Retiniana/etiología , Células Ganglionares de la Retina/metabolismo , Animales , Muerte Celular , Retinopatía Diabética/metabolismo , Femenino , Angiografía con Fluoresceína , Fondo de Ojo , Insulina/genética , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Receptores sigma/biosíntesis , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/patología , Tomografía de Coherencia Óptica , Receptor Sigma-1
6.
Genetics ; 201(2): 631-49, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26265702

RESUMEN

Dynactin is a multi-subunit complex that functions as a regulator of the Dynein motor. A central component of this complex is Dynamitin/p50 (Dmn). Dmn is required for endosome motility in mammalian cell lines. However, the extent to which Dmn participates in the sorting of cargo via the endosomal system is unknown. In this study, we examined the endocytic role of Dmn using the Drosophila melanogaster oocyte as a model. Yolk proteins are internalized into the oocyte via clathrin-mediated endocytosis, trafficked through the endocytic pathway, and stored in condensed yolk granules. Oocytes that were depleted of Dmn contained fewer yolk granules than controls. In addition, these oocytes accumulated numerous endocytic intermediate structures. Particularly prominent were enlarged endosomes that were relatively devoid of Yolk proteins. Ultrastructural and genetic analyses indicate that the endocytic intermediates are produced downstream of Rab5. Similar phenotypes were observed upon depleting Dynein heavy chain (Dhc) or Lis1. Dhc is the motor subunit of the Dynein complex and Lis1 is a regulator of Dynein activity. We therefore propose that Dmn performs its function in endocytosis via the Dynein motor. Consistent with a role for Dynein in endocytosis, the motor colocalized with the endocytic machinery at the oocyte cortex in an endocytosis-dependent manner. Our results suggest a model whereby endocytic activity recruits Dynein to the oocyte cortex. The motor along with its regulators, Dynactin and Lis1, functions to ensure efficient endocytic uptake and maturation.


Asunto(s)
Endocitosis/genética , Endosomas/genética , Proteínas Asociadas a Microtúbulos/genética , Oocitos/metabolismo , Animales , Citoesqueleto/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Complejo Dinactina , Dineínas/biosíntesis , Dineínas/genética , Endosomas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Oocitos/crecimiento & desarrollo , Transporte de Proteínas/genética
7.
Invest Ophthalmol Vis Sci ; 56(4): 2684-95, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25766590

RESUMEN

PURPOSE: Methylenetetrahydrofolate reductase (Mthfr) is a key enzyme in homocysteine-methionine metabolism. We investigated Mthfr expression in retina and asked whether mild hyperhomocysteinemia, due to Mthfr deficiency, alters retinal neurovascular structure and function. METHODS: Expression of Mthfr was investigated at the gene and protein level using quantitative (q) RT-PCR, in situ hybridization, immunoblotting, and immunohistochemistry (IHC). The Mthfr+/+ and Mthfr+/- mice were subjected to comprehensive evaluation using ERG, funduscopy, fluorescein angiography (FA), spectral-domain optical coherence tomography (SD-OCT), HPLC, and morphometric and IHC analysis of glial fibrillary acidic protein (GFAP) at 8 to 24 weeks. RESULTS: Gene and protein analyses disclosed widespread retinal expression of Mthfr. Electroretinography (ERG) revealed a significant decrease in positive scotopic threshold response in retinas of Mthfr+/- mice at 24 weeks. Fundus examination in mice from both groups was normal; FA revealed areas of focal vascular leakage in 20% of Mthfr+/- mice at 12 to 16 weeks and 60% by 24 weeks. The SD-OCT revealed a significant decrease in nerve fiber layer (NFL) thickness at 24 weeks in Mthfr+/- compared to Mthfr+/+ mice. There was a 2-fold elevation in retinal hcy at 24 weeks in Mthfr+/- mice by HPLC and IHC. Morphometric analysis revealed an approximately 20% reduction in cells in the ganglion cell layer of Mthfr+/- mice at 24 weeks. The IHC indicated significantly increased GFAP labeling suggestive of Müller cell activation. CONCLUSIONS: Mildly hyperhomocysteinemic Mthfr+/- mice demonstrate reduced ganglion cell function, thinner NFL, and mild vasculopathy by 24 weeks. The retinal phenotype is similar to that of hyperhomocysteinemic mice with deficiency of cystathionine-ß-synthase (Cbs) reported earlier. The data support the hypothesis that hyperhomocysteinemia may be causative in certain retinal neurovasculopathies.


Asunto(s)
ADN/genética , Regulación de la Expresión Génica , Homocistinuria/patología , Hiperhomocisteinemia/patología , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Espasticidad Muscular/patología , Células Ganglionares de la Retina/patología , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Angiografía con Fluoresceína , Fondo de Ojo , Homocistinuria/genética , Homocistinuria/metabolismo , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Immunoblotting , Inmunohistoquímica , Metilenotetrahidrofolato Reductasa (NADPH2)/biosíntesis , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Ratones , Ratones Transgénicos , Espasticidad Muscular/genética , Espasticidad Muscular/metabolismo , Trastornos Psicóticos/genética , Trastornos Psicóticos/metabolismo , Trastornos Psicóticos/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Tomografía de Coherencia Óptica
8.
Invest Ophthalmol Vis Sci ; 54(2): 939-49, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23307965

RESUMEN

PURPOSE: Mice with moderate/severe hyperhomocysteinemia due to deficiency or absence of the cbs gene encoding cystathionine-beta-synthase (CBS) have marked retinal disruption, ganglion cell loss, optic nerve mitochondrial dysfunction, and ERG defects; those with mild hyperhomocysteinemia have delayed retinal morphological/functional phenotype. Excess homocysteine is a risk factor for cardiovascular diseases; however, it is not known whether excess homocysteine alters retinal vasculature. METHODS: Cbs(+/+), cbs(+/-), and cbs(-/-) mice (age ∼3 weeks) were subjected to angiography; retinas were harvested for cryosections, flat-mount preparations, or trypsin digestion and subjected to immunofluorescence microscopy to visualize vessels using isolectin-B4, to detect angiogenesis using anti-VEGF and anti-endoglin (anti-CD105) and activated glial cells (anti-glial fibrillary acidic protein [anti-GFAP]) and to investigate the blood-retinal barrier using the tight junction markers zonula occludens-1 (ZO-1) and occludin. Expression of vegf was determined by quantitative RT-PCR (qRT-PCR) and immunoblotting. Human retinal endothelial cells (HRECs) were treated with excess homocysteine to analyze permeability. RESULTS: Angiography revealed vascular leakage in cbs(-/-) mice; immunohistochemical analysis demonstrated vascular patterns consistent with ischemia; isolectin-B4 labeling revealed a capillary-free zone centrally and new vessels with capillary tufts midperipherally. This was associated with increased vegf mRNA and protein, CD105, and GFAP in cbs(-/-) retinas concomitant with a marked decrease in ZO-1 and occludin. Homocysteine-treated HRECs showed increased permeability. CONCLUSIONS: Severe elevation of homocysteine in cbs(-/-) mutant mice is accompanied by alterations in retinal vasculature (ischemia, neovascularization, and incompetent blood-retinal barrier). The marked disruption of retinal structure and decreased visual function reported in cbs(-/-) mice may reflect vasculopathy as well as neuropathy.


Asunto(s)
Regulación de la Expresión Génica , Homocisteína/metabolismo , Hiperhomocisteinemia/genética , ARN Mensajero/genética , Retina/patología , Enfermedades de la Retina/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Barrera Hematorretinal/metabolismo , Barrera Hematorretinal/fisiopatología , Permeabilidad Capilar , Cistationina betasintasa/deficiencia , Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Angiografía con Fluoresceína , Fondo de Ojo , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Inmunohistoquímica , Ratones , Ratones Mutantes , Microscopía Fluorescente , Retina/metabolismo , Retina/fisiopatología , Enfermedades de la Retina/etiología , Enfermedades de la Retina/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/biosíntesis
9.
Invest Ophthalmol Vis Sci ; 52(8): 5551-8, 2011 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-21642619

RESUMEN

PURPOSE: To evaluate the effect of excess homocysteine on the regulation of retinal ganglion cell mitochondrial dynamics. METHODS: Mice deficient in cystathionine-ß-synthase (cbs) were used as a model of hyperhomocysteinemia. Gene and protein expression analyses of Opa1 and Fis1 were performed on cbs⁺/⁻ neural retinas. Mitochondria within retinal ganglion cell axons underwent systematic ultrastructural analysis to measure area, length, width, and the distance between the mitochondria and the axon wall. Primary mouse ganglion cells were cultured, treated with homocysteine, and assessed for levels of Opa1 and Fis1 protein, the number of mitochondria per length of neurite, and levels of cleaved caspase-3. RESULTS: Opa1 and Fis1 protein levels in cbs⁺/⁻ neural retinas were elevated to 191.00% ± 26.40% and 226.20% ± 4.57%, respectively, compared with wild-type. Mitochondria of cbs⁺/⁻ retinas were smaller in all parameters studied, including area (0.32 ± 0.01 µm² vs. 0.42 ± 0.02 µm²), compared with wild-type. Primary ganglion cells treated with homocysteine had elevations in Opa1 and Fis1 proteins, a significantly higher number of mitochondria per length of neurite (0.1781 ± 0.017 vs. 0.1156 ± 0.012), and significantly higher levels of cleaved caspase-3 compared with control. CONCLUSIONS: This study provides the first evidence that homocysteine-induced ganglion cell loss involves the dysregulation of mitochondrial dynamics, both in vivo and in vitro. The present data suggest increased mitochondrial fission as a novel mechanism of homocysteine toxicity to neurons. Of particular relevance are glaucoma and Alzheimer's disease, neurodegenerative diseases that are associated with hyperhomocysteinemia and, more recently, have implicated increased mitochondrial fission in their pathogeneses.


Asunto(s)
Homocisteína/toxicidad , Hiperhomocisteinemia/metabolismo , Mitocondrias/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Animales Recién Nacidos , Axones/efectos de los fármacos , Axones/metabolismo , Axones/ultraestructura , Caspasa 3/genética , Caspasa 3/metabolismo , Células Cultivadas , Cartilla de ADN , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente Indirecta , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/patología , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , ARN Mensajero/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Invest Ophthalmol Vis Sci ; 51(6): 3226-35, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20053979

RESUMEN

PURPOSE: To analyze the mechanisms of folate uptake in retinal Müller cells. METHODS: RT-PCR and Western blot analysis were performed in freshly isolated neural retina and RPE/eyecup, primary mouse Müller cells, and rMC-1 cells for the three known folate transport proteins folate receptor alpha (FRalpha), proton-coupled folate transporter (PCFT), and reduced folate carrier (RFC). Laser scanning confocal microscopy (LSCM) and immunoelectron microscopy were used to determine the subcellular location of FRalpha and PCFT in primary Müller cells. The pH dependence of the uptake of [(3)H]-methyltetrahydrofolate ([(3)H]-MTF) was assayed in Müller cells in the presence/absence of thiamine pyrophosphate, an inhibitor of RFC. RESULTS: FRalpha and PCFT are expressed abundantly in the retina in several cell layers, including the inner nuclear layer; they are present in primary mouse Müller cells and rMC-1 cells. LSCM localized these proteins to the plasma membrane, nuclear membrane, and perinuclear region. Immunoelectron microscopic studies revealed the colocalization of FRalpha and PCFT on the plasma membrane and nuclear membrane and within endosomal structures. Müller cell uptake of [(3)H]-MTF was robust at pH 5.0 to 6.0, consistent with PCFT activity, but also at neutral pH, reflecting RFC function. RFC was expressed in mouse Müller cells that had been allowed to proliferate in culture, but not in freshly isolated primary cells. CONCLUSIONS: FRalpha and PCFT are expressed in retinal Müller cells and colocalize in the endosomal compartment, suggesting that the two proteins may work coordinately to mediate folate uptake. The unexpected finding of RFC expression and activity in cultured Müller cells may reflect the upregulation of this protein under proliferative conditions.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Neuroglía/metabolismo , Receptores de Superficie Celular/metabolismo , Tetrahidrofolatos/metabolismo , Animales , Western Blotting , Células Cultivadas , Endosomas/metabolismo , Receptor 1 de Folato , Regulación de la Expresión Génica/fisiología , Concentración de Iones de Hidrógeno , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/ultraestructura , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Microscopía Inmunoelectrónica , Neuroglía/efectos de los fármacos , Neuroglía/ultraestructura , Transportador de Folato Acoplado a Protón , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/ultraestructura , Proteína Portadora de Folato Reducido , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiamina Pirofosfato/farmacología
11.
Invest Ophthalmol Vis Sci ; 50(9): 4460-70, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19357353

RESUMEN

PURPOSE: To determine the effects of endogenous elevation of homocysteine on the retina using the cystathionine beta-synthase (cbs) mutant mouse. METHODS: Retinal homocysteine in cbs mutant mice was measured by high-performance liquid chromatography (HPLC). Retinal cryosections from cbs(-/-) mice and cbs(+/-) mice were examined for histologic changes by light and electron microscopy. Morphometric analysis was performed on retinas of cbs(+/-) mice maintained on a high-methionine diet (cbs(+/-) HM). Changes in retinal gene expression were screened by microarray. RESULTS: HPLC analysis revealed an approximate twofold elevation in retinal homocysteine in cbs(+/-) mice and an approximate sevenfold elevation in cbs(-/-) mice. Distinct alterations in the ganglion, inner plexiform, inner nuclear, and epithelial layers were observed in retinas of cbs(-/-) and 1-year-old cbs(+/-) mice. Retinas of cbs(+/-) HM mice demonstrated an approximate 20% decrease in cells of the ganglion cell layer (GCL), which occurred as early as 5-weeks after onset of the HM diet. Microarray analysis revealed alterations in expression of several genes, including increased expression of Aven, Egr1, and Bat3 in retinas of cbs(+/-) HM mice. CONCLUSIONS: This study provides the first analysis of morphologic and molecular effects of endogenous elevations of retinal homocysteine in an in vivo model. Increased retinal homocysteine alters inner and outer retinal layers in cbs homozygous mice and older cbs heterozygous mice, and it primarily affects the cells of the GCL in younger heterozygous mice. Elevated retinal homocysteine alters expression of genes involved in endoplasmic reticular stress, N-methyl-d-aspartate (NMDA) receptor activation, cell cycle, and apoptosis.


Asunto(s)
Apoptosis , Cistationina betasintasa/genética , Homocisteína/fisiología , Hiperhomocisteinemia/patología , Mutación , Enfermedades de la Retina/patología , Células Ganglionares de la Retina/patología , Animales , Cromatografía Líquida de Alta Presión , Cistationina betasintasa/deficiencia , Cistationina betasintasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica/fisiología , Glutatión/metabolismo , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Masculino , Ratones , Ratones Noqueados , Análisis por Micromatrices , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Células Ganglionares de la Retina/metabolismo
12.
Mol Vis ; 15: 362-72, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19223991

RESUMEN

PURPOSE: GPR109A has been identified as a G-protein-coupled receptor for niacin. beta-hydroxybutyrate (beta-HB) is a physiologic ligand for the receptor. beta-HB, the predominate ketone body in circulation, is an important energy source for neurons, including retinal neurons, under various physiologic and pathologic conditions. The identification of GPR109A as the receptor for beta-HB suggests additional, hitherto unknown, functions for this metabolite. The circulating levels of beta-HB increase in diabetes. Since retinopathy is a serious complication associated with diabetes, we investigated GPR109A expression in retina and in different retinal cell types to determine if the receptor may have a role in the pathophysiology of diabetic retinopathy. METHODS: RT-PCR, fluorescent in situ hybridization, and immunofluorescent techniques were used to analyze GPR109A expression in mouse retina and in three transformed retinal cell lines: ARPE-19 (RPE), RGC-5 (ganglion), and rMC-1 (Müller). Activation of GPR109A by niacin and beta-HB was demonstrated in ARPE-19 cells by cAMP assay. RESULTS: Studies conducted using mouse retinal tissues demonstrated that GPR109A is expressed in retina with its expression restricted to RPE, where it differentially polarizes to the basolateral membrane. These results were confirmed with cell lines, which demonstrated GPR109A expression in ARPE-19, but not in rMC-1 and RGC-5 cells. Primary cultures of mouse RPE also showed robust expression of GPR109A. cAMP assay demonstrated that GPR109A expressed in RPE is functional. CONCLUSIONS: These data represent the first report on GPR109A expression in retina. The exclusive expression of GPR109A in RPE basolateral membrane, which has access to beta-HB in blood, may have biologic importance in diabetic retinopathy.


Asunto(s)
Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Línea Celular Transformada , AMP Cíclico/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica/fisiología , Humanos , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Ophthalmol Eye Dis ; 1: 3-11, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20407615

RESUMEN

Hyperhomocysteinemia has been implicated in visual dysfunction. We reported recently that mice with endogenous hyperhomocysteinemia, due to mutation of the cystathionine-ß-synthase (cbs) gene, demonstrate loss of neurons in the retinal ganglion cell (RGC) layer and other retinal layers as homocysteine levels increase. Some clinical studies implicate hyperhomocysteinemia in the pathogenesis of diabetic retinopathy, which is also characterized by RGC loss. The present study used cbs(+/-) mice to determine whether modest elevation of plasma homocysteine, in the presence of diabetes, accelerates neuronal cell loss. Diabetes (DB) was induced in 3 wk old cbs(+/-) and wildtype mice using streptozotocin; four groups of mice were studied: DB cbs(+/-); non-DB cbs(+/-); DB cbs(+/+); non-DB cbs(+/+). One group of diabetic cbs(+/-) mice was maintained on a high methionine diet (HMD, 0.5% methionine drinking water) to increase plasma homocysteine slightly. Eyes were harvested at 5, 10 and 15 weeks post-onset of diabetes; retinal cryosections were examined by light microscopy and subjected to systematic morphometric analysis. Diabetic cbs(+/-) had significantly fewer RGCs at 5 weeks compared to age-matched, non-diabetic cbs(+/-) and wildtype controls (10.0 ± 0.5 versus 14.9 ± 0.5 and 15.8 ± 0.6 cells/100 µm retina length, respectively). Significant differences in retinas of DB/high homocysteine versus controls were obtained 15 wks post-onset of diabetes including fewer RGCS and decreased thickness of inner nuclear and plexiform layers. Moderate increases in plasma homocysteine coupled with diabetes cause a more dramatic alteration of retinal phenotype than elevated homocysteine or diabetes alone and suggest that diabetes accelerates the retinal neuronal death in hyperhomocysteinemic mice.

14.
Invest Ophthalmol Vis Sci ; 49(11): 5151-60, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18689705

RESUMEN

PURPOSE: Glutamine transport is essential for the glutamate-glutamine cycle, which occurs between neurons and glia. System N, consisting of SN1 (SNAT3) and SN2 (SNAT5), is the principal mediator of glutamine transport in retinal Müller cells. Mediators of glutamine transport in retinal ganglion cells were investigated. METHODS: The relative contributions of various transport systems for glutamine uptake (systems N, A, L, y+L, ASCT, and ATB(0,+)) were examined in RGC-5 cells based on differential features of the individual transport systems. mRNA for the genes encoding members of these transport systems were analyzed by RT-PCR. Based on these data, SN1 and SN2 were analyzed in mouse retina, RGC-5 cells, and primary mouse ganglion cells (GCs) by in situ hybridization (ISH), immunofluorescence (IF), and Western blotting. RESULTS: Three transport systems--N, A, and L--participated in glutamine uptake in RGC-5 cells. System N was the principal contributor; systems A and L contributed considerably less. ISH and IF revealed SN1 and SN2 expression in the ganglion, inner nuclear, and photoreceptor cell layers. SN1 and SN2 colocalized with the ganglion cell marker Thy 1.2 and with the Müller cell marker vimentin, confirming their presence in both retinal cell types. SN1 and SN2 proteins were detected in primary mouse GCs. CONCLUSIONS: These findings suggest that in addition to its role in glutamine uptake in retinal glial cells, system N contributes significantly to glutamine uptake in ganglion cells and, hence, contributes to the retinal glutamate-glutamine cycle.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Expresión Génica , ARN Mensajero/genética , Células Ganglionares de la Retina/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Animales , Western Blotting , Células Cultivadas , Hibridación in Situ , Ratones , Neurotransmisores , Células Ganglionares de la Retina/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
15.
Dev Biol ; 319(2): 359-69, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18514182

RESUMEN

The innermost layer of the Drosophila eggshell, the vitelline membrane, provides structural support and positional information to the embryo. It is assembled in an incompletely understood manner from four major proteins to form a homogeneous, transparent extracellular matrix. Here we show that RNAi knockdown or genetic deletion of a minor constituent of this matrix, Palisade, results in structural disruptions during the initial synthesis of the vitelline membrane by somatic follicle cells surrounding the oocyte, including wide size variation among the precursor vitelline bodies and disorganization of follicle cell microvilli. Loss of Palisade or the microvillar protein Cad99C results in abnormal uptake into the oocyte of sV17, a major vitelline membrane protein, and defects in non-disulfide cross-linking of sV17 and sV23, while loss of Palisade has additional effects on processing and disulfide cross-linking of these proteins. Embryos surrounded by the abnormal vitelline membranes synthesized when Palisade is reduced are fertilized but undergo developmental arrest, usually during the first 13 nuclear divisions, with a nuclear phenotype of chromatin margination similar to that described for wild-type embryos subjected to anoxia. Our results demonstrate that Palisade is involved in coordinating assembly of the vitelline membrane and is required for functional properties of the eggshell.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/embriología , Ovario/fisiología , Membrana Vitelina/fisiología , Animales , Animales Modificados Genéticamente , Anticuerpos , Western Blotting , Proteínas de Drosophila/análisis , Proteínas de Drosophila/química , Femenino , Interferencia de ARN , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/química , Transfección
16.
Invest Ophthalmol Vis Sci ; 49(9): 4154-61, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18469181

RESUMEN

PURPOSE: To evaluate the neuroprotective properties of the sigma receptor 1 (sigmaR1) ligand, (+)-pentazocine in an in vivo model of retinal neurodegeneration. METHODS: Spontaneously diabetic Ins2(Akita/+) and wild-type mice received intraperitoneal injections of (+)-pentazocine for 22 weeks beginning at diabetes onset. Retinal mRNA and protein were analyzed by RT-PCR and Western blot analysis. Retinal histologic sections were measured to determine total retinal thickness, thicknesses of inner-outer nuclear and plexiform layers (INL, ONL, IPL, INL), and the number of cell bodies in the ganglion cell layer (GCL). Immunolabeling experiments were performed using antibodies specific for 4-hydroxynonenal and nitrotyrosine, markers of lipid peroxidation, and reactive nitrogen species, respectively, and an antibody specific for vimentin to view radial Müller fibers. RESULTS: sigmaR1 mRNA and protein levels in the Ins2(Akita/+) retina were comparable to those in the wild-type, indicating that sigmaR1 is an available target during the disease process. Histologic evaluation of eyes of Ins2(Akita/+) mice showed disruption of retinal architecture. By 17 to 25 weeks after birth, Ins2(Akita/+) mice demonstrated approximately 30% and 25% decreases in IPL and INL thicknesses, respectively, and a 30% reduction in ganglion cells. In the (+)-pentazocine-treated group, retinas of Ins2(Akita/+) mice showed remarkable preservation of retinal architecture; IPL and INL thicknesses of (+)-pentazocine-treated Ins2(Akita/+) mouse retinas were within normal limits. The number of ganglion cells was 15.6 +/- 1.5 versus 10.4 +/- 1.2 cells/100 mum retinal length in (+)-pentazocine-treated versus nontreated mutant mice. Levels of nitrotyrosine and 4-hydroxynonenal increased in Ins2(Akita/+) retinas, but were reduced in (+)-pentazocine-treated mice. Retinas of Ins2(Akita/+) mice showed loss of the uniform organization of radial Müller fibers. Retinas of (+)-pentazocine-treated mice maintained the radial organization of glial processes. CONCLUSION: Sustained (+)-pentazocine treatment in an in vivo model of retinal degeneration conferred significant neuroprotection, reduced evidence of oxidative stress, and preserved retinal architecture, suggesting that sigmaR1 ligands are promising therapeutic agents for intervention in neurodegenerative diseases of the retina.


Asunto(s)
Retinopatía Diabética/prevención & control , Pentazocina/uso terapéutico , Receptores sigma/fisiología , Degeneración Retiniana/prevención & control , Animales , Glucemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/fisiopatología , Ligandos , Ratones , Ratones Endogámicos C57BL , Receptores sigma/genética , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Sigma-1
17.
J Neurochem ; 104(4): 970-8, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17976164

RESUMEN

D-Serine, the endogenous ligand for the glycine modulatory binding site of the NMDA receptor, and serine racemase, the enzyme that converts L-serine to D-serine, have been reported in vertebrate retina; initial reports suggested that localization was restricted to Müller glial cells. Recent reports, in which D-serine and serine racemase were detected in neurons of the brain, prompted the present investigation of neuronal expression of D-serine and serine racemase in retina and whether expression patterns were developmentally regulated. RT-PCR, in situ hybridization, western blotting, immunohistochemistry, and immunocytochemical methods were used to localize D-serine and serine racemase in intact retina obtained from 1 to 3 day, 3 week, and 18 week mouse retinas and in primary ganglion cells harvested by immunopanning from neonatal mouse retina. Results of these analyses revealed robust expression of D-serine and serine racemase in ganglion cells, both in intact retina and in cultured cells. The levels appear to be developmentally regulated with D-serine levels being quite high in ganglion cells of neonatal retinas and decreasing rapidly postnatally. Serine racemase levels are also developmentally regulated, with high levels detected during the early postnatal period, but diminishing considerably in the mature retina. This represents the first report of neuronal expression of D-serine and serine racemase in the vertebrate retina and suggests an important contribution of neuronal D-serine during retinal development.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Racemasas y Epimerasas/biosíntesis , Retina/crecimiento & desarrollo , Células Ganglionares de la Retina/fisiología , Serina/biosíntesis , Animales , Animales Recién Nacidos , Ratones , Ratones Endogámicos C57BL , Racemasas y Epimerasas/genética , Retina/metabolismo , Serina/química , Serina/genética
18.
Biochem J ; 411(1): 79-88, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18042040

RESUMEN

Hepcidin is a hormone central to the regulation of iron homeostasis in the body. It is believed to be produced exclusively by the liver. Ferroportin, an iron exporter, is the receptor for hepcidin. This transporter/receptor is expressed in Müller cells, photoreceptor cells and the RPE (retinal pigment epithelium) within the retina. Since the retina is protected by the retinal-blood barriers, we asked whether ferroportin in the retina is regulated by hepcidin in the circulation or whether the retina produces hepcidin for regulation of its own iron homeostasis. Here we show that hepcidin is expressed robustly in Müller cells, photoreceptor cells and RPE cells, closely resembling the expression pattern of ferroportin. We also show that bacterial LPS (lipopolysaccharide) is a regulator of hepcidin expression in Müller cells and the RPE, both in vitro and in vivo, and that the regulation occurs at the transcriptional level. The action of LPS on hepcidin expression is mediated by the TLR4 (Toll-like receptor-4). The upregulation of hepcidin by LPS occurs independent of Hfe (human leukocyte antigen-like protein involved in Fe homeostasis). The increase in hepcidin levels in retinal cells in response to LPS treatment is associated with a decrease in ferroportin levels. The LPS-induced upregulation of hepcidin and consequent down-regulation of ferroportin is associated with increased oxidative stress and apoptosis within the retina in vivo. We conclude that retinal iron homeostasis may be regulated in an autonomous manner by hepcidin generated within the retina and that chronic bacterial infection/inflammation of the retina may disrupt iron homeostasis and retinal function.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I , Lipopolisacáridos/farmacología , Proteínas de la Membrana , Retina/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/inmunología , Apoptosis , Proteínas de Transporte de Catión/genética , Proteína de la Hemocromatosis , Hepcidinas , Homeostasis , Hierro/metabolismo , Ratones , Estrés Oxidativo
19.
Mol Genet Metab ; 92(4): 315-24, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17884651

RESUMEN

Carnitine is essential for transport of long-chain fatty acids into mitochondria for their subsequent beta-oxidation, but its role in the gastrointestinal tract has not been well described. Recently several genetic epidemiologic studies have shown strong association between mutations in carnitine transporter genes OCTN1 and OCTN2 and a propensity to develop Crohn's disease. This study aims to investigate role of carnitine and beta-oxidation in the GI tract. We have studied the gastrointestinal tract effects of carnitine deficiency in a mouse model with loss-of-function mutation in the OCTN2 carnitine transporter. juvenile visceral steatosis (OCTN2(-/-)) mouse spontaneously develops intestinal villous atrophy, breakdown and inflammation with intense lymphocytic and macrophage infiltration, leading to ulcer formation and gut perforation. There is increased apoptosis of jvs (OCTN2(-/-)) gut epithelial cells. We observed an up-regulation of heat shock factor-1 (HSF-1) and several heat shock proteins (HSPs) which are known to regulate OCTN2 gene expression. Intestinal and colonic epithelial cells in wild type mice showed high expression and activity of the enzymes of beta-oxidation pathway. These studies provide evidence of an obligatory role for carnitine in the maintenance of normal intestinal and colonic structure and morphology. Fatty acid oxidation, a metabolic pathway regulated by carnitine-dependent entry of long-chain fatty acids into mitochondrial matrix, is likely essential for normal gut function. Our studies suggest that carnitine supplementation, as a means of boosting fatty acid oxidation, may be therapeutically beneficial in patients with inflammation of the intestinal tract.


Asunto(s)
Carnitina/deficiencia , Enfermedad de Crohn/fisiopatología , Proteínas de Unión al ADN/metabolismo , Inflamación/etiología , Proteínas de Transporte de Catión Orgánico , Factores de Transcripción/metabolismo , Sistemas de Transporte de Aminoácidos , Animales , Atrofia/etiología , Carnitina/metabolismo , Colon/anomalías , Enfermedad de Crohn/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Factores de Transcripción del Choque Térmico , Intestinos/anomalías , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Mutantes , Mutación
20.
Invest Ophthalmol Vis Sci ; 48(7): 3356-63, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17591909

RESUMEN

PURPOSE: Monocarboxylates are primary energy substrates in the retina. Recently, the authors identified two sodium-coupled monocarboxylate transporters (SMCTs), SMCT1 (a high-affinity transporter) and SMCT2 (a low-affinity transporter). Expression of SMCT1 and SMCT2 has been studied in several tissues; however, little is known about their expression in retina. In the present study, the authors asked whether SMCT1 and SMCT2 are also expressed in retina and, if so, in which particular retinal cell types. METHODS: SMCT1 and SMCT2 expression was analyzed in intact mouse retina and cultured retinal cells (ganglion, Müller, RPE) by RT-PCR, in situ hybridization, and immunofluorescence. Uptake assays were performed to demonstrate SMCT1 (RGC-5 and ARPE-19 cells) and SMCT2 (rMC-1 cells) expression at the functional level. RESULTS: SMCT1 mRNA and protein were detected in the ganglion cell layer, inner nuclear layer, inner/outer plexiform layers, photoreceptor inner segments, and RPE. In RPE, the expression of SMCT1 was restricted to the basolateral membrane. SMCT2 mRNA and protein were detected only in neural retina, with a pattern of protein localization consistent with labeling of Müller cells. In vitro studies confirmed the cell type-specific expression of SMCT1 and SMCT2. Uptake assays demonstrated Na(+)-coupled monocarboxylate transport in RGC-5, ARPE-19, and rMC-1 cells. CONCLUSIONS: These data provide the first evidence for the expression of SMCT1 and SMCT2 in the retina and for the cell-type specific distribution of these transporters within the retina. These studies suggest that SMCT1 and SMCT2 play a differential role in monocarboxylate transport in the retina in a cell type-specific manner.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Células del Tejido Conectivo/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Transportadores de Ácidos Monocarboxílicos/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...