Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 151(1): 25-40, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23021213

RESUMEN

Astrocytes release glutamate upon activation of various GPCRs to exert important roles in synaptic functions. However, the molecular mechanism of release has been controversial. Here, we report two kinetically distinct modes of nonvesicular, channel-mediated glutamate release. The fast mode requires activation of G(αi), dissociation of G(ßγ), and subsequent opening of glutamate-permeable, two-pore domain potassium channel TREK-1 through direct interaction between G(ßγ) and N terminus of TREK-1. The slow mode is Ca(2+) dependent and requires G(αq) activation and opening of glutamate-permeable, Ca(2+)-activated anion channel Best1. Ultrastructural analyses demonstrate that TREK-1 is preferentially localized at cell body and processes, whereas Best1 is mostly found in microdomains of astrocytes near synapses. Diffusion modeling predicts that the fast mode can target neuronal mGluR with peak glutamate concentration of 100 µM, whereas slow mode targets neuronal NMDA receptors at around 1 µM. Our results reveal two distinct sources of astrocytic glutamate that can differentially influence neighboring neurons.


Asunto(s)
Astrocitos/metabolismo , Proteínas del Ojo/metabolismo , Ácido Glutámico/metabolismo , Canales Iónicos/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Bestrofinas , Células Cultivadas , Exocitosis , Proteínas del Ojo/genética , Células HEK293 , Humanos , Canales Iónicos/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Canales de Potasio de Dominio Poro en Tándem/genética , Alineación de Secuencia , Transducción de Señal
2.
Mol Vis ; 23: 228-241, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28465655

RESUMEN

PURPOSE: To examine the effects of autophagy deficiency induced by RPE-specific deletion of Atg5 or Atg7 in mice as a function of age. METHODS: Conditional knockout mice with a floxed allele of Atg5 or Atg7 were crossed with inducible VMD2-rtTA/Cre transgenic mice. VMD2-directed RPE-specific Cre recombinase expression was induced with doxycycline feeding in the resulting mice. Cre-mediated deletion of floxed Atg5 or Atg7 resulted in RPE-specific inactivation of the Atg5 or Atg7 gene. Plastic and thin retinal sections were analyzed with light and electron microscopy for histological changes. Photoreceptor outer segment (POS) thickness in plastic sections was measured using the Adobe Photoshop CS4 extended ruler tool. Autophagic adaptor p62/SQSTM1 and markers for oxidatively damaged lipids, proteins, and DNA were examined with immunofluorescence staining of cryosections. Fluorescence signals were quantified using Image J software. RESULTS: Accumulation of p62/SQSTM1 reflecting autophagy deficiency was observed in the RPE of the Atg5ΔRPE and Atg7ΔRPE mice. 3-nitrotyrosine, advanced glycation end products (AGEs), and 8-hydroxy-2'-deoxyguanosine (8-OHdG), markers for oxidatively damaged proteins and DNA, were also found to accumulate in the RPE of these mice. We observed retinal degeneration in 35% of the Atg5ΔRPE mice and 45% of the Atg7ΔRPE mice at 8 to 24 months old. Degeneration severity and the number of mice with degeneration increased with age. The mean POS thickness of these mice was 25 µm at 8-12 months, 15 µm at 13-18 months, and 3 µm at 19-24 months, compared to 35 µm, 30 µm, and 24 µm in the wild-type mice, respectively. Early age-related macular degeneration (AMD)-like RPE defects were found in all the Atg5ΔRPE and Atg7ΔRPE mice 13 months old or older, including vacuoles, uneven RPE thickness, diminished basal infoldings, RPE hypertrophy/hypotrophy, pigmentary irregularities, and necrosis. The severity of the RPE defects increased with age and in the mice with retinal degeneration. RPE atrophy and choroidal neovascularization (CNV) were occasionally observed in the Atg5ΔRPE and Atg7ΔRPE mice with advanced age. CONCLUSIONS: Autophagy deficiency induced by RPE-specific deletion of Atg5 or Atg7 predisposes but does not necessarily drive the development of AMD-like phenotypes or retinal degeneration.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/genética , Autofagia , Eliminación de Gen , Degeneración Macular/genética , Degeneración Retiniana/genética , Epitelio Pigmentado de la Retina/patología , Alelos , Animales , Biomarcadores/metabolismo , Electrorretinografía , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Degeneración Macular/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Biochim Biophys Acta ; 1842(12 Pt B): 2579-83, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25108282

RESUMEN

Adenylyl cyclases (ACs) are a family of enzymes which convert ATP to cAMP, an essential intermediate in many signal transduction pathways. Of the 10 AC genes in man, 9 fall into the category of transmembrane ACs (tmACs), which associate with G-protein coupled receptors (GPCRs) and are activated by forskolin. The 10th AC, termed soluble AC (sAC) is neither activated by forskolin nor does it interact with GPCRs. Rather, sAC can be found in many compartments within the cell and is activated by bicarbonate. As such, sAC is considered a major sensor of bicarbonate in many tissues. The pathways involving sAC vary in different tissues and organ systems, and are as diverse as facilitating sperm capacitation and regulating pressure in the eye. The role of sAC in the eye has only recently begun to receive significant attention. Here we summarize what is known about the roles of sAC in the eye. This article is part of a Special Issue entitled: The role of soluble adenylyl cyclase in health and disease.


Asunto(s)
Adenilil Ciclasas/metabolismo , Ojo/enzimología , Animales , Humanos
4.
Hum Mol Genet ; 22(23): 4688-97, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23825107

RESUMEN

Mutations in bestrophin-1 (Best1) cause Best vitelliform macular dystrophy (BVMD), a dominantly inherited retinal degenerative disease. Best1 is a homo-oligomeric anion channel localized to the basolateral surface of retinal pigment epithelial (RPE) cells. A number of Best1 mutants mislocalize in Madin-Darby canine kidney (MDCK) cells. However, many proteins traffic differently in MDCK and RPE cells, and MDCK cells do not express endogenous Best1. Thus, effects of Best1 mutations on localization in MDCK cells may not translate to RPE cells. To determine whether BVMD causing mutations affect Best1 localization, we compared localization and oligomerization of Best1 with Best1 mutants V9M, W93C, and R218C. In MDCK cells, Best1 and Best1(R218C) were basolaterally localized. Best1(W93C) and Best1(V9M) accumulated in cells. In cultured fetal human retinal pigment epithelium cells (fhRPE) expressing endogenous Best1, Best1(R218C) and Best1(W93C) were basolateral. Best1(V9M) was intracellular. All three mutants exhibited similar fluorescence resonance energy transfer (FRET) efficiencies to, and co-immunoprecipitated with Best1, indicating unimpaired oligomerization. When human Best1 was expressed in RPE in mouse eyes it was basolaterally localized. However, Best1(V9M) accumulated in intracellular compartments in mouse RPE. Co-expression of Best1 and Best1(W93C) in MDCK cells resulted in basolateral localization of both Best1 and Best1(W93C), but co-expression of Best1 with Best1(V9M) resulted in mislocalization of both proteins. We conclude that different mutations in Best1 cause differential effects on its localization and that this effect varies with the presence or absence of wild-type (WT) Best1. Furthermore, MDCK cells can substitute for RPE when examining the effects of BVMD causing mutations on Best1 localization if co-expressed with WT Best1.


Asunto(s)
Canales de Cloruro/metabolismo , Proteínas del Ojo/metabolismo , Canales Iónicos/metabolismo , Distrofia Macular Viteliforme/patología , Animales , Bestrofinas , Señalización del Calcio , Membrana Celular/metabolismo , Células Cultivadas , Canales de Cloruro/genética , Ojo/metabolismo , Proteínas del Ojo/genética , Regulación de la Expresión Génica , Humanos , Canales Iónicos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Transporte de Proteínas/genética , Distrofia Macular Viteliforme/genética
5.
N Engl J Med ; 376(12): 1195, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28332386
6.
Mol Vis ; 21: 347-59, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25878489

RESUMEN

PURPOSE: Mutations in BEST1, encoding Bestrophin-1 (Best1), cause Best vitelliform macular dystrophy (BVMD) and other inherited retinal degenerative diseases. Best1 is an integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium (RPE). Data from numerous in vitro and in vivo models have demonstrated that Best1 regulates intracellular Ca2+ levels. Although it is known from in vitro and crystal structure data that Best1 is also a calcium-activated anion channel, evidence for Best1 functioning as a channel in human RPE is lacking. To assess Best1-associated channel activity in the RPE, we examined the transepithelial electrical properties of fetal human RPE (fhRPE) cells, which express endogenous Best1. METHODS: Using adenovirus-mediated gene transfer, we overexpressed Best1 and the BVMD mutant Best1W93C in fhRPE cells and assessed resting transepithelial potential (TEP), transepithelial resistance, short circuit current (Isc), and intracellular Ca2+ levels. Cl- currents were directly measured in transfected HEK293 cells using whole-cell patch clamp. RESULTS: Best1W93C showed ablated Cl- currents and, when co-expressed, suppressed the channel activity of Best1 in HEK293 cells. In fhRPE, overexpression of Best1 increased TEP and Isc, while Best1W93C diminished TEP and Isc. Substitution of Cl- in the bath media resulted in a significant reduction of Isc in monolayers overexpressing Best1, but no significant Isc change in monolayers expressing Best1W93C. We removed Ca2+ as a limit on transepithelial electrical properties by treating cells with ionomycin, and found that changes in Isc and TEP for monolayers expressing Best1 were absent in monolayers expressing Best1W93C. Similarly, inhibition of calcium-activated anion channels with niflumic acid reduced both Isc and TEP of control and Best1 monolayers, but did not notably affect Best1W93C monolayers. Stimulation with extracellular ATP induced an increase in TEP in control monolayers that was greater than that observed in those expressing Best1(W93C). Examination of [Ca2+]i following ATP stimulation demonstrated that the expression of Best1W93C impaired intracellular Ca2+ signaling. CONCLUSIONS: These data indicate that Best1 activity strongly influences electrophysiology and Ca2+ signaling in RPE cells, and that a common BVMD mutation disrupts both of these parameters. Our findings support the hypothesis that Best1 functions as an anion channel in human RPE.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Membrana Celular/metabolismo , Canales de Cloruro/metabolismo , Células Epiteliales/metabolismo , Proteínas del Ojo/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Adenosina Trifosfato/farmacología , Adenovirus Humanos/genética , Bestrofinas , Membrana Celular/efectos de los fármacos , Canales de Cloruro/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Proteínas del Ojo/genética , Feto , Expresión Génica , Vectores Genéticos , Células HEK293 , Humanos , Transporte Iónico/efectos de los fármacos , Ionomicina/farmacología , Potenciales de la Membrana/efectos de los fármacos , Mutación , Ácido Niflúmico/farmacología , Técnicas de Placa-Clamp , Cultivo Primario de Células , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Transfección , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo , Distrofia Macular Viteliforme/patología
7.
Proc Natl Acad Sci U S A ; 109(4): 1199-203, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22223659

RESUMEN

Body temperature is maintained in a narrow range in mammals, primarily controlled by sweating. In humans, the dynamic thermoregulatory organ, comprised of 2-4 million sweat glands distributed over the body, can secrete up to 4 L of sweat per day, thereby making it possible to withstand high temperatures and endure prolonged physical stress (e.g., long-distance running). The genetic basis for sweat gland function, however, is largely unknown. We find that the forkhead transcription factor, FoxA1, is required to generate mouse sweating capacity. Despite continued sweat gland morphogenesis, ablation of FoxA1 in mice results in absolute anihidrosis (lack of sweating). This inability to sweat is accompanied by down-regulation of the Na-K-Cl cotransporter 1 (Nkcc1) and the Ca(2+)-activated anion channel Bestrophin 2 (Best2), as well as glycoprotein accumulation in gland lumens and ducts. Furthermore, Best2-deficient mice display comparable anhidrosis and glycoprotein accumulation. These findings link earlier observations that both sodium/potassium/chloride exchange and Ca(2+) are required for sweat production. FoxA1 is inferred to regulate two corresponding features of sweat secretion. One feature, via Best2, catalyzes a bicarbonate gradient that could help to drive calcium-associated ionic transport; the other, requiring Nkcc1, facilitates monovalent ion exchange into sweat. These mechanistic components can be pharmaceutical targets to defend against hyperthermia and alleviate defective thermoregulation in the elderly, and may provide a model relevant to more complex secretory processes.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Canales de Cloruro/metabolismo , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica/fisiología , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Sudoración/fisiología , Análisis de Varianza , Animales , Bestrofinas , Western Blotting , Cruzamientos Genéticos , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Galactósidos , Perfilación de la Expresión Génica , Genotipo , Factor Nuclear 3-alfa del Hepatocito/genética , Indoles , Ratones , Modelos Biológicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Miembro 2 de la Familia de Transportadores de Soluto 12 , Sudoración/genética
8.
Exp Eye Res ; 121: 74-85, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24560797

RESUMEN

BEST1 encodes Bestrophin-1 (Best1), a homo-oligomeric, integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium. Mutations in BEST1 cause five distinct retinal degenerative diseases, including adult vitelliform macular dystrophy (AVMD), autosomal recessive bestrophinopathy (ARB), autosomal dominant vitreoretinochoroidopathy (ADVIRC), and retinitis pigmentosa (RP). The mechanisms underlying these diseases and why mutations cause one disease over another are, for the most part, unknown. To gain insights into these four diseases, we expressed 28 Best1 mutants fused to YFP in polarized MDCK monolayers and, via confocal microscopy and immunofluorescence, live-cell FRET, and reciprocal co-immunoprecipitation experiments, screened these mutants for defects in localization and oligomerization. All 28 mutants exhibited comparable FRET efficiencies to and co-immunoprecipitated with WT Best1, indicating unimpaired oligomerization. RP- and ADVIRC-associated mutants were properly localized to the basolateral plasma membrane of cells, while two AVMD and most ARB mutants were mislocalized. When co-expressed, all mislocalized mutants caused mislocalization of WT Best1 to intracellular compartments. Our current and past results indicate that mislocalization of Best1 is not an absolute feature of any individual bestrophinopathy, occurring in AVMD, BVMD, and ARB. Furthermore, some ARB mutants that do not also cause dominant disease cause mislocalization of Best1, indicating that mislocalization is not a cause of disease, and that absence of Best1 activity from the plasma membrane is tolerated. Lastly, we find that the ARB truncation mutants L174Qfs*57 and R200X can form oligomers with WT Best1, indicating that the first ∼174 amino acids of Best1 are sufficient for oligomerization to occur.


Asunto(s)
Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Enfermedades Hereditarias del Ojo/genética , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Mutación Missense , Multimerización de Proteína/fisiología , Enfermedades de la Retina/genética , Adenoviridae/genética , Animales , Proteínas Bacterianas/metabolismo , Bestrofinas , Western Blotting , Enfermedades de la Coroides/genética , Enfermedades de la Coroides/metabolismo , Perros , Electrofisiología , Enfermedades Hereditarias del Ojo/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Proteínas Luminiscentes/metabolismo , Células de Riñón Canino Madin Darby/metabolismo , Microscopía Confocal , Técnicas de Placa-Clamp , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Enfermedades de la Retina/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Transfección , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo
9.
Invest Ophthalmol Vis Sci ; 65(4): 8, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38568620

RESUMEN

Purpose: No large-mammal surgical models exist for geographic atrophy (GA), choroidal neovascularization (CNV), and pachychoroidal vascular remodeling. Our goal was to develop a porcine RPE debridement model of advanced macular degeneration to study photoreceptor cell loss and choroidal remodeling. Methods: Seven 2-month-old female domestic pigs were used for this study. After 25G vitrectomy, the area centralis was detached via subretinal bleb. A nitinol wire (Finesse Flex Loop) was used to debride RPE cells across a 3- to 5-mm diameter region. Fluid-air exchange was performed, and 20% SF6 gas injected. Animals underwent fundus photography, fluorescein angiography, optical coherence tomography (OCT), and OCT-angiography (OCTA) at 2 weeks, 1 month, 2 months, 3 months, and 6 months postoperatively. Retinal histology was obtained at euthanasia, 2 months (n = 3), 3 months (n = 2), or 6 months (n = 2) after surgery. Results: RPE debridement resulted in GA with rapid loss of choriocapillaris, progressive loss of photoreceptors, and pachychoroidal changes in Sattler's and Haller's layers in all seven eyes undergoing debridement within 2 months. OCT and histological findings included subretinal disciform scar with overlying outer retinal atrophy; outer retinal tubulations and subretinal hyper-reflective material. OCTA revealed type 2 CNV (n = 4) at the edges of the debridement zone by 2 months, but there was no significant exudation noted at any time point. Conclusions: Surgical debridement of the RPE results in GA, CNV, and pachychoroid and reproduced all forms of advanced macular degeneration. This surgical model may be useful in examining the role of RPE and other cell replacement in treating advanced macular disease.


Asunto(s)
Neovascularización Coroidal , Atrofia Geográfica , Degeneración Macular , Femenino , Porcinos , Animales , Desbridamiento , Degeneración Macular/diagnóstico , Degeneración Macular/cirugía , Atrofia Geográfica/diagnóstico , Sus scrofa , Retina , Coroides , Neovascularización Coroidal/diagnóstico , Neovascularización Coroidal/cirugía
10.
J Ocul Pharmacol Ther ; 39(5): 317-323, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37097314

RESUMEN

Purpose: We investigated whether a clinically used carbonic anhydrase inhibitor (CAIs) can modulate intraocular pressure (IOP) through soluble adenylyl cyclase (sAC) signaling. Methods: IOP was measured 1 h after topical treatment with brinzolamide, a topically applied and clinically used CAIs, using direct cannulation of the anterior chamber in sAC knockout (KO) mice or C57BL/6J mice in the presence or absence of the sAC inhibitor (TDI-10229). Results: Mice treated with the sAC inhibitor TDI-10229 had elevated IOP. CAIs treatment significantly decreased increased intraocular pressure (IOP) in wild-type, sAC KO mice, as well as TDI-10229-treated mice. Conclusions: Inhibiting carbonic anhydrase reduces IOP independently from sAC in mice. Our studies suggest that the signaling cascade by which brinzolamide regulates IOP does not involve sAC.


Asunto(s)
Glaucoma , Presión Intraocular , Animales , Ratones , Inhibidores de Anhidrasa Carbónica , Adenilil Ciclasas/uso terapéutico , Ratones Endogámicos C57BL , Glaucoma/tratamiento farmacológico
11.
Melanoma Res ; 33(6): 482-491, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37650708

RESUMEN

To investigate downstream molecular changes caused by mitogen-activated protein kinase (MEK) inhibitor treatment and further explore the impact of direct knockdown of early growth response-1 ( EGR1 ) in melanoma cell culture. RNA-sequencing (RNA-Seq) was performed to determine gene expression changes with MEK inhibitor treatment. Treatment with MEK inhibitor (trametinib) was then assessed in two cutaneous (MEL888, MEL624) and one conjunctival (YUARGE 13-3064) melanoma cell line. Direct knockdown of EGR1 was accomplished using lentiviral vectors containing shRNA. Cell viability was measured using PrestoBlueHS Cell Viability Reagent. Total RNA and protein were assessed by qPCR and SimpleWestern. RNA-Seq demonstrated a profound reduction in EGR1 with MEK inhibitor treatment, prompting further study of melanoma cell lines. Following trametinib treatment of melanoma cells, viability was reduced in both cutaneous (MEL888 26%, P  < 0.01; MEL624 27%, P  < 0.001) and conjunctival (YUARGE 13-3064 33%, P  < 0.01) melanoma compared with DMSO control, with confirmed EGR1 knockdown to 0.04-, 0.01-, and 0.16-fold DMSO-treated levels (all P  < 0.05) in MEL888, MEL624, and YUARGE 13-3064, respectively. Targeted EGR1 knockdown using shRNA reduced viability in both cutaneous (MEL624 78%, P  = 0.05) and conjunctival melanoma (YUARGE-13-3064 67%, P  = 0.02). RNA-Sequencing in MEK inhibitor-treated cells identified EGR1 as a candidate effector molecule of interest. In a malignant melanoma cell population, MEK inhibition reduced viability in both cutaneous and conjunctival melanoma with a profound downstream reduction in EGR1 expression. Targeted knockdown of EGR1 reduced both cutaneous and conjunctival melanoma cell viability independent of MEK inhibition, suggesting a key role for EGR1 in melanoma pathobiology.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Mitógenos , Dimetilsulfóxido , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos , ARN Interferente Pequeño , Línea Celular Tumoral , Proteínas Proto-Oncogénicas B-raf/genética
12.
J Biol Chem ; 286(48): 41353-41358, 2011 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-21994938

RESUMEN

Glaucoma is a leading cause of blindness affecting as many as 2.2 million Americans. All current glaucoma treatment strategies aim to reduce intraocular pressure (IOP). IOP results from the resistance to drainage of aqueous humor (AH) produced by the ciliary body in a process requiring bicarbonate. Once secreted into the anterior chamber, AH drains from the eye via two pathways: uveoscleral and pressure-dependent or conventional outflow (C(t)). Modulation of "inflow" and "outflow" pathways is thought to occur via distinct, local mechanisms. Mice deficient in the bicarbonate channel bestrophin-2 (Best2), however, exhibit a lower IOP despite an increase in AH production. Best2 is expressed uniquely in nonpigmented ciliary epithelial (NPE) cells providing evidence for a bicarbonate-dependent communicative pathway linking inflow and outflow. Here, we show that bicarbonate-sensitive soluble adenylyl cyclase (sAC) is highly expressed in the ciliary body in NPE cells, but appears to be absent from drainage tissues. Pharmacologic inhibition of sAC in mice causes a significant increase in IOP due to a decrease in C(t) with no effect on inflow. In mice deficient in sAC IOP is elevated, and C(t) is decreased relative to wild-type mice. Pharmacologic inhibition of sAC did not alter IOP or C(t) in sAC-deficient mice. Based on these data we propose that the ciliary body can regulate C(t) and that sAC serves as a critical sensor of bicarbonate in the ciliary body regulating the secretion of substances into the AH that govern outflow facility independent of pressure.


Asunto(s)
Adenilil Ciclasas/metabolismo , Bicarbonatos/metabolismo , Cuerpo Ciliar/enzimología , Glaucoma/enzimología , Presión Intraocular , Adenilil Ciclasas/genética , Animales , Humor Acuoso/enzimología , Bestrofinas , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Glaucoma/epidemiología , Glaucoma/genética , Humanos , Ratones , Ratones Noqueados , Porcinos
13.
Hum Mol Genet ; 19(6): 1108-18, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20053664

RESUMEN

Mutations in BEST1, encoding bestrophin-1 (Best1), cause Best vitelliform macular dystrophy (BVMD), a dominantly inherited macular degeneration characterized by a diminished electrooculogram light peak (LP), lipofuscin in retinal pigment epithelial cells (RPE), and fluid- and debris-filled retinal detachments. To understand the pathogenesis of BVMD we generated knock-in mice carrying the BVMD-causing mutation W93C in Best1. Both Best1(+/W93C)and Best1(W93C/W93C) mice had normal ERG a- and b-waves, but exhibited an altered LP luminance response reminiscent of that observed in BVMD patients. Morphological analysis identified fluid- and debris-filled retinal detachments in mice as young as 6 months of age. By 18-24 months of age Best1(+/W93C)and Best1(W93C/W93C) mice exhibited enhanced accumulation of lipofuscin in the RPE, and a significant deposition of debris composed of unphagocytosed photoreceptor outer segments and lipofuscin granules in the subretinal space. Although Best1 is thought to function as a Ca(2+)-activated Cl(-) channel, RPE cells from Best1(W93C) mice exhibited normal Cl(-) conductances. We have previously shown that Best1(-/-) mice exhibit increased [Ca(2+)](i) in response to ATP stimulation. However, ATP-stimulated changes in [Ca(2+)](i) in RPE cells from Best1(+/W93C) and Best1(W93C/W93C) mice were suppressed relative to Best1(+/+) littermates. Based on these data we conclude that mice carrying the Best1(W93C) mutation are a valid model for BVMD. Furthermore, these data suggest that BVMD is not because of Best1 deficiency, as the phenotypes of Best1(+/W93C) and Best1(W93C/W93C) mice are distinct from that of Best1(-/-) mice with regard to lipofuscin accumulation, and changes in the LP and ATP Ca(2+) responses.


Asunto(s)
Señalización del Calcio , Modelos Animales de Enfermedad , Degeneración Macular/metabolismo , Adenosina Trifosfato/farmacología , Sustitución de Aminoácidos/genética , Animales , Bestrofinas , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/efectos de la radiación , Cloruros/metabolismo , Electrooculografía , Electrorretinografía , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Técnicas de Sustitución del Gen , Genotipo , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/efectos de la radiación , Canales Iónicos , Luz , Degeneración Macular/genética , Degeneración Macular/patología , Degeneración Macular/fisiopatología , Ratones , Proteínas Mutantes/metabolismo , Mutación/genética , Epitelio Pigmentado Ocular/metabolismo , Epitelio Pigmentado Ocular/patología , Epitelio Pigmentado Ocular/efectos de la radiación , Epitelio Pigmentado Ocular/ultraestructura
14.
Transl Vis Sci Technol ; 11(4): 24, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35486039

RESUMEN

Purpose: To improve outcomes for subretinal implantation surgery in pigs. Methods: Analysis of variables affecting the success of subretinal implantation surgery was performed on videos of 37 surgeries. Ex vivo experiments were conducted to measure intraocular pressure (IOP) and test various prototyped implanters for effectiveness at maintaining IOP. Results: A video analysis revealed a prolonged sclerotomy open time owing to a combination of uncontrolled bleeding and excessive fluid outflow often resulting in retinal prolapse. Precauterization of the choroid before full-thickness sclerotomy (n = 10) resulted in a reduced incidence of uncontrolled bleeding from 39.1% (9/23) versus 0% (0/10) (P = 0.005) and improved implantation success from 73% to 90%. An ex vivo analysis of the IOP revealed a mean decrease in the IOP from 30.2 ± 3.0 mm Hg to 5.0 ± 2.1 mm Hg after a fully penetrating sclerotomy. To address this situation, we produced a series of plugs that integrated with a custom implant insertion device to seal the sclerotomy during implantation. The use of the plugs was cumbersome, however, and so we opted instead to increase the width of the inserter tip to fill the open sclerotomy. This improved device restored and maintained IOP during implantation (27.1 ± 1.9 mm Hg). Combined with precauterization the improved inserter resulted in 100% successful implantation (n = 4). Conclusions: For subretinal implantation in pigs, a modified procedure to precauterize the choroid before sclerotomy combined with an instrument that better fills the scleral opening decreases bleeding, hypotony, and open sclerotomy time, improving the success rate. Translational Relevance: Better management of IOP and bleeding from a sclerotomy will improve implant-based therapies.


Asunto(s)
Implantes de Drenaje de Glaucoma , Glaucoma de Ángulo Abierto , Animales , Glaucoma de Ángulo Abierto/cirugía , Presión Intraocular , Porcinos , Tonometría Ocular , Resultado del Tratamiento
15.
J Biol Chem ; 285(17): 12990-3001, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20177062

RESUMEN

Activation of an apical Ca(2+)-dependent Cl(-) channel (CaCC) is the rate-limiting step for fluid secretion in many exocrine tissues. Here, we compared the properties of native CaCC in mouse submandibular salivary gland acinar cells to the Ca(2+)-gated Cl(-) currents generated by Tmem16A and Best2, members from two distinct families of Ca(2+)-activated Cl(-) channels found in salivary glands. Heterologous expression of Tmem16A and Best2 transcripts in HEK293 cells produced Ca(2+)-activated Cl(-) currents with time and voltage dependence and inhibitor sensitivity that resembled the Ca(2+)-activated Cl(-) current found in native salivary acinar cells. Best2(-/-) and Tmem16A(-/-) mice were used to further characterize the role of these channels in the exocrine salivary gland. The amplitude and the biophysical footprint of the Ca(2+)-activated Cl(-) current in submandibular gland acinar cells from Best2-deficient mice were the same as in wild type cells. Consistent with this observation, the fluid secretion rate in Best2 null mice was comparable with that in wild type mice. In contrast, submandibular gland acinar cells from Tmem16A(-/-) mice lacked a Ca(2+)-activated Cl(-) current and a Ca(2+)-mobilizing agonist failed to stimulate Cl(-) efflux, requirements for fluid secretion. Furthermore, saliva secretion was abolished by the CaCC inhibitor niflumic acid in wild type and Best2(-/-) mice. Our results demonstrate that both Tmem16A and Best2 generate Ca(2+)-activated Cl(-) current in vitro with similar properties to those expressed in native cells, yet only Tmem16A appears to be a critical component of the acinar Ca(2+)-activated Cl(-) channel complex that is essential for saliva production by the submandibular gland.


Asunto(s)
Calcio/metabolismo , Canales de Cloruro/metabolismo , Cloruros/metabolismo , Glándula Submandibular/metabolismo , Animales , Anoctamina-1 , Canales de Cloruro/genética , Regulación de la Expresión Génica/fisiología , Transporte Iónico/fisiología , Ratones , Ratones Noqueados , Especificidad de Órganos/fisiología , Glándula Submandibular/citología
16.
Retin Cases Brief Rep ; 15(4): 356-358, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-30300315

RESUMEN

PURPOSE: To report a case of adult-onset vitelliform macular dystrophy in a patient who was found to have a previously unreported variant of the IMPG2 gene. METHODS: Case report. RESULTS: A 65-year-old white woman with no significant medical or ocular history presented with a complaint of persistent wavy vision for 10 months. On funduscopic examination, bilateral vitelliform lesions of approximately 1 mm in the right eye and 0.5 mm in the left eye were evident, with no choroidal neovascularization in either eye. The patient was diagnosed with adult-onset vitelliform macular dystrophy. Genetic testing revealed a single likely pathogenic variant of the IMPG2 gene that may explain the examination findings. CONCLUSION: Adult-onset vitelliform macular dystrophy is a common and relatively benign condition occurring in approximately 1 in 8,000 individuals. Although vitelliform lesions can be a manifestation of systemic diseases or be idiopathic, in a minority of patients, genetic predisposition may play a role. Mutations in four particular genes BEST1, PRPH2, IMPG1, and IMPG2 have been associated with some cases of adult-onset vitelliform macular dystrophy, with this particular gene variant of IMPG2 being previously unreported.


Asunto(s)
Mutación , Proteoglicanos , Distrofia Macular Viteliforme , Edad de Inicio , Anciano , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Proteoglicanos/genética , Distrofia Macular Viteliforme/genética
17.
J Biomed Mater Res A ; 109(11): 2357-2368, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33973708

RESUMEN

Fibrin is a degradable biopolymer with an excellent clinical safety profile. Use of higher mechanical strength fibrin hydrogels is limited by the rapid rate of fibrin polymerization. We recently demonstrated the use of higher mechanical strength (fibrinogen concentrations >30 mg/ml) fibrin scaffolds for surgical implantation of cells. The rapid polymerization of fibrin at fibrinogen concentrations impaired our ability to scale production of these fibrin scaffolds. We serendipitously discovered that the azo dye Trypan blue (TB) slowed fibrin gelation kinetics allowing for more uniform mixing of fibrinogen and thrombin at high concentrations. A screen of closely related compounds identified similar activity for Evans blue (EB), an isomer of TB. Both TB and EB exhibited a concentration dependent increase in clot time, though EB had a larger effect. While gelation time was increased by TB or EB, overall polymerization time was unaffected. Scanning electron microscopy showed similar surface topography, but transmission electron microscopy showed a higher cross-linking density for gels formed with TB or EB versus controls. Based on these data we conclude that addition of TB or EB during thrombin mediated fibrin polymerization slows the initial gelation time permitting generation of larger more uniform fibrin hydrogels with high-mechanical strength.


Asunto(s)
Compuestos Azo/química , Fibrina/química , Hidrogeles/química , Modelos Químicos , Cinética
18.
Trends Genet ; 23(5): 225-31, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17368622

RESUMEN

Macular degenerations (MD), age-related or inherited, interfere with the ability to read, drive and recognize faces. Understanding this class of diseases has been challenging because the mouse, the mammal most amenable to genetic manipulation, lacks a macula. Here we discuss whether we can model MD in the mouse, present criteria for an 'ideal' mouse model of MD and discuss how mouse models have contributed to our knowledge of MD by contrasting how well they meet the 'ideal' criteria with how informative they have actually been. By modeling MD in mice, we can learn about aspects of MD that an animal with a macula would be unable to teach us.


Asunto(s)
Modelos Animales de Enfermedad , Degeneración Macular/genética , Envejecimiento/patología , Animales , Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo/genética , Predicción , Proteínas de Filamentos Intermediarios/genética , Degeneración Macular/patología , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Periferinas , Proteínas Recombinantes/genética , Degeneración Retiniana/genética , Degeneración Retiniana/patología
19.
Mol Vis ; 16: 200-6, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-20157619

RESUMEN

PURPOSE: Mice in which bestrophin 2 (Best2) is disrupted exhibit changes in aqueous flow and drainage, resulting in a reduction in intraocular pressure in comparison to wild-type mice. Best2 encodes a putative anion channel localized uniquely to the basolateral plasma membrane of non-pigmented epithelium cells in mice. In this study, we examine the localization of Best2 in the human eye. METHODS: Rabbit polyclonal antibodies recognizing human Best2 (hBest2) were generated and characterized for use in western blot, immunoprecipitation, and immunofluorescence assays. The expression of hBest2 using these antibodies was examined using human donor eye tissues. RESULTS: We could not detect hBest2 in human ciliary bodies or other ocular tissues by western blot. However, when enriched by immunoprecipitation, hBest2 was identified in ciliary bodies, but not in the retinal pigment epithelium. Using immunofluorescence, we located hBest2 in the basolateral plasma membrane of non-pigmented epithelial cells. CONCLUSIONS: We found expression of hBest2 similar to mice only in NPE cells. These data suggest that Best2 may play a functional role in the regulation of aqueous flow and drainage in humans. We conclude that Best2 represents a new potential target for glaucoma therapy.


Asunto(s)
Canales de Cloruro/metabolismo , Cuerpo Ciliar/metabolismo , Proteínas del Ojo/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Pigmentación , Especificidad de Anticuerpos , Bestrofinas , Línea Celular , Humanos , Inmunoprecipitación , Transporte de Proteínas , Donantes de Tejidos , Transfección
20.
Ophthalmol Retina ; 4(5): 535-544, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31956075

RESUMEN

PURPOSE: To assess the frequency and impact of abnormal foveal avascular zone (FAZ) topography (i.e., a fragmented FAZ) on visual acuity and foveal anatomic features. DESIGN: Prospective, cross-sectional study from March 2018 through July 2019. PARTICIPANTS: Two-hundred fifty participants were screened from a normative OCT angiography database. Of those, 12 participants were found to have at least 1 eye with a fragmented FAZ. Eight returned for follow-up imaging, along with an additional 3 participants with ocular disease (amblyopia, autosomal recessive bestrophinopathy, premature birth) having a similar FAZ phenotype. METHODS: Follow-up OCT imaging and monocular best-corrected visual acuity (BCVA) were performed for these 11 participants. Twenty-four participants with a clearly defined FAZ were recruited for comparison. A normative database was created measuring parafoveal intercapillary area (PICA) to determine if an FAZ was fragmented. MAIN OUTCOME MEASURES: Monocular BCVA, foveal pit depth, foveal pit area, PICA, outer nuclear layer thickness, foveal inner retinal area, and peak cone density. RESULTS: The frequency of a fragmented FAZ was 4.8% of individuals (12 of 250) or 3.6% of eyes (18 of 500 eyes). A significant difference was found between the control eyes and eyes with fragmented FAZs for foveal pit depth, pit area, and total PICA (P < 0.001, P = 0.002, and P < 0.001, respectively). The presence of a fragmented FAZ did not affect visual acuity. CONCLUSIONS: The presence of a fragmented FAZ seems not to be a rare phenotype in individuals with normal vision. The presence of altered FAZ topography in patients with retinal or systemic disease could negatively impact the accuracy and sensitivity of biomarkers dependent on FAZ identification.


Asunto(s)
Angiografía con Fluoresceína/métodos , Fóvea Central/irrigación sanguínea , Vasos Retinianos/diagnóstico por imagen , Tomografía de Coherencia Óptica/métodos , Agudeza Visual , Adolescente , Adulto , Niño , Estudios Transversales , Femenino , Fondo de Ojo , Humanos , Masculino , Estudios Prospectivos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA