Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Gene Ther ; 26(9): 386-398, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31308478

RESUMEN

Recombinant adeno-associated virus (rAAV) has been widely used for gene delivery in animal models and successfully applied in clinical trials for treating inherited retinal disease. Although subretinal delivery of AAVs can effectively transduce photoreceptors and/or retinal pigmental epithelium (RPE), cells most affected by inherited retinal diseases, the procedure is invasive and complicated, and only delivers the gene to a limited retinal area. AAVs can also be delivered intravitreally to the retina, a much less invasive nonsurgical procedure. However, intravitreal administration of non-modified AAV serotypes tends to transduce only ganglion cells and inner nuclear layer cells. To date, most non-modified AAV serotypes that have been identified are incapable of efficiently transducing photoreceptors and/or RPE when delivered intravitreally. In this study, we investigate the retinal tropism of AAVrh10 vector administered by intravitreal injection to mouse, rat, and rabbit eyes. Our results demonstrate that AAVrh10 is capable of transducing not only inner retinal cells, but also outer retinal cells in all three species, though the transduction efficiency in rabbit was low. In addition, AAVrh10 preferentially transduced outer retinal cells in mouse models of retinal disease. Therefore, AAVrh10 vector could be a useful candidate to intravitreally deliver genes to photoreceptor and RPE cells.


Asunto(s)
Dependovirus/genética , Retina , Transducción Genética/métodos , Animales , Citomegalovirus/genética , Dependovirus/fisiología , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Inyecciones Intravítreas , Masculino , Ratones , Células Fotorreceptoras/virología , Conejos , Ratas , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Retina/virología , Enfermedades de la Retina/terapia , Tropismo Viral
2.
Mol Ther ; 26(9): 2282-2294, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30196853

RESUMEN

This study evaluated the safety and tolerability of ocular RS1 adeno-associated virus (AAV8-RS1) gene augmentation therapy to the retina of participants with X-linked retinoschisis (XLRS). XLRS is a monogenic trait affecting only males, caused by mutations in the RS1 gene. Retinoschisin protein is secreted principally in the outer retina, and its absence results in retinal cavities, synaptic dysfunction, reduced visual acuity, and susceptibility to retinal detachment. This phase I/IIa single-center, prospective, open-label, three-dose-escalation clinical trial administered vector to nine participants with pathogenic RS1 mutations. The eye of each participant with worse acuity (≤63 letters; Snellen 20/63) received the AAV8-RS1 gene vector by intravitreal injection. Three participants were assigned to each of three dosage groups: 1e9 vector genomes (vg)/eye, 1e10 vg/eye, and 1e11 vg/eye. The investigational product was generally well tolerated in all but one individual. Ocular events included dose-related inflammation that resolved with topical and oral corticosteroids. Systemic antibodies against AAV8 increased in a dose-related fashion, but no antibodies against RS1 were observed. Retinal cavities closed transiently in one participant. Additional doses and immunosuppressive regimens are being explored to pursue evidence of safety and efficacy (ClinicalTrials.gov: NCT02317887).


Asunto(s)
Proteínas del Ojo/metabolismo , Terapia Genética/métodos , Retinosquisis/terapia , Adulto , Anciano , Proteínas del Ojo/genética , Femenino , Humanos , Inyecciones Intravítreas , Masculino , Persona de Mediana Edad , Mutación/genética , Retina/metabolismo , Retina/patología , Retinosquisis/genética , Retinosquisis/metabolismo , Adulto Joven
3.
Adv Exp Med Biol ; 801: 559-66, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24664744

RESUMEN

Light-activated movement of transducin-α (Gαt1) from rod photoreceptor outer segments (ROS) into inner segments (IS) enables rods to rapidly adapt to changes in light intensity. The threshold light intensity at which Gαt1 translocates from ROS into IS is primarily determined by the rates of activation and inactivation of Gαt1. Loss- of- expression of the retina specific cell surface protein, retinoschsin (Rs1-KO), led to a dramatic 3-10 fold increase, depending on age, in the luminance threshold for transducin translocation from ROS into IS compared with wild-type control. In contrast, arrestin translocated from IS into ROS at the same light intensity both in WT and Rs1-KO mice. Biochemical changes, including reduced transducin protein levels and enhanced transducin GTPase activity, explain the shift in light intensity threshold for Gαt1 translocation in Rs1-KO mice. These changes in Rs1-KO mice were also associated with age related alterations in photoreceptor morphology and transcription factor expression that suggest delayed photoreceptor maturation.


Asunto(s)
Moléculas de Adhesión Celular/genética , Proteínas del Ojo/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Retinosquisis/genética , Retinosquisis/patología , Transducina/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Humanos , Luz , Ratones , Ratones Noqueados , Segmento Interno de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Interno de las Células Fotorreceptoras Retinianas/patología , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinosquisis/metabolismo
4.
J Neurosci ; 32(38): 13010-21, 2012 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-22993419

RESUMEN

Loss of retinoschisin (RS1) in Rs1 knock-out (Rs1-KO) retina produces a post-photoreceptor phenotype similar to X-linked retinoschisis in young males. However, Rs1 is expressed strongly in photoreceptors, and Rs1-KO mice have early reduction in the electroretinogram a-wave. We examined light-activated transducin and arrestin translocation in young Rs1-KO mice as a marker for functional abnormalities in maturing rod photoreceptors. We found a progressive reduction in luminance threshold for transducin translocation in wild-type (WT) retinas between postnatal days P18 and P60. At P21, the threshold in Rs1-KO retinas was 10-fold higher than WT, but it decreased to <2.5-fold higher by P60. Light-activated arrestin translocation and re-translocation of transducin in the dark were not affected. Rs1-KO rod outer segment (ROS) length was significantly shorter than WT at P21 but was comparable with WT at P60. These findings suggested a delay in the structural and functional maturation of Rs1-KO ROS. Consistent with this, transcription factors CRX and NRL, which are fundamental to maturation of rod protein expression, were reduced in ROS of Rs1-KO mice at P21 but not at P60. Expression of transducin was 15-30% lower in P21 Rs1-KO ROS and transducin GTPase hydrolysis was nearly twofold faster, reflecting a 1.7- to 2.5-fold increase in RGS9 (regulator of G-protein signaling) level. Transduction protein expression and activity levels were similar to WT at P60. Transducin translocation threshold elevation indicates photoreceptor functional abnormalities in young Rs1-KO mice. Rapid reduction in threshold coupled with age-related changes in transduction protein levels and transcription factor expression are consistent with delayed maturation of Rs1-KO photoreceptors.


Asunto(s)
Arrestina/metabolismo , Moléculas de Adhesión Celular/deficiencia , Adaptación a la Oscuridad/genética , Fototransducción/genética , Células Fotorreceptoras Retinianas Bastones/fisiología , Transducina/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta en la Radiación , Electrorretinografía , Proteínas del Ojo , GTP Fosfohidrolasas/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Luminiscencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estimulación Luminosa , Fotones , Fotoperiodo , Transporte de Proteínas/genética , Retina/citología , Rodopsina/metabolismo , Factores de Tiempo , Vías Visuales/fisiología
5.
Proc Natl Acad Sci U S A ; 107(28): 12710-5, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20616020

RESUMEN

Inward rectifying potassium (Kir) channels participate in regulating potassium concentration (K(+)) in the central nervous system (CNS), including in the retina. We explored the contribution of Kir channels to retinal function by delivering Kir antibodies (Kir-Abs) into the rat eye in vivo to interrupt channel activity. Kir-Abs were coupled to a peptide carrier to reach intracellular epitopes. Functional effects were evaluated by recording the scotopic threshold response (STR) and photopic negative response (PhNR) of the electroretinogram (ERG) noninvasively with an electrode on the cornea to determine activity of the rod and cone pathways, respectively. Intravitreal delivery of Kir2.1-Ab coupled to the peptide carrier diminished these ERG responses equivalent to dimming the stimulus 10- to 100-fold. Immunohistochemistry (IHC) showed Kir2.1 immunostaining of retinal bipolar cells (BCs) matching the labeling pattern obtained with conventional IHC of applying Kir2.1-Ab to fixed retinal sections postmortem. Whole-cell voltage-clamp BC recordings in rat acute retinal slices showed suppression of barium-sensitive Kir2.1 currents upon inclusion of Kir2.1-Ab in the patch pipette. The in vivo functional and structural results implicate a contribution of Kir2.1 channel activity in these electronegative ERG potentials. Studies with Kir4.1-Ab administered in vivo also suppressed the ERG components and showed immunostaining of Müller cells. The strategy of administering Kir antibodies in vivo, coupled to a peptide carrier to facilitate intracellular delivery, identifies roles for Kir2.1 and Kir4.1 in ERG components arising in the proximal retina and suggests this approach could be of further value in research.


Asunto(s)
Canales de Potasio/metabolismo , Retina/metabolismo , Animales , Anticuerpos/metabolismo , Anticuerpos/farmacología , Anticuerpos/fisiología , Bario/metabolismo , Bario/farmacología , Bario/fisiología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/fisiología , Citoplasma/metabolismo , Citoplasma/fisiología , Electrorretinografía , Degeneración Nerviosa/metabolismo , Neuroglía/metabolismo , Neuroglía/fisiología , Técnicas de Placa-Clamp , Potasio/metabolismo , Potasio/farmacología , Potasio/fisiología , Canales de Potasio/farmacología , Canales de Potasio/fisiología , Canales de Potasio de Rectificación Interna , Ratas , Ratas Mutantes , Retina/efectos de los fármacos , Retina/fisiología , Retinaldehído/metabolismo , Retinaldehído/farmacología , Retinaldehído/fisiología
6.
Proc Natl Acad Sci U S A ; 106(23): 9397-402, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19470639

RESUMEN

In nonphagocytic cells, Rac1 is a component of NADPH oxidase that produces reactive oxygen species [Ushio-Fukai M (2006) Sci STKE 2006:re8]. Rac1 is expressed abundantly in mammalian retinal photoreceptors, where it is activated in response to light stimuli [Balasubramanian N, Slepak VZ (2003) Curr Biol 13:1306-1310]. We used Cre-LoxP conditional gene targeting to knock down Rac1 expression in mouse rod photoreceptors and found protection against light-induced photoreceptor death compared with WT litter-mates. We also found a similar protective effect on rods using apocynin, which inhibits NADPH oxidase activity. These results implicate both neuronal Rac1 and NADPH oxidase in cell death in this model of CNS degeneration. Studies in which dominant-mutants of Rac1 were expressed in transgenic Drosophila species demonstrated that Rac1 is a key regulator of photoreceptor morphogenesis and polarity [Chang HY, Ready DF (2000) Science 290:1978-1980]. However, we found that diminished Rac1 expression in mouse rods had no effect on retinal structure or function examined by light microscopy, electron microscopy, rhodopsin measurement, electroretinogram activity, and visual acuity, indicating rod outer segment morphogenesis proceeded normally in Rac1 conditional knockout mice. The lack of structural or functional effect of Rac1 depletion on photoreceptors, but protection under conditions of stress, indicate that the Rac1 pathway warrants exploration as a target for therapy in retinal neurodegenerative diseases.


Asunto(s)
Neuropéptidos/metabolismo , Estrés Oxidativo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Luz , Ratones , Ratones Noqueados , Enfermedades Neurodegenerativas/metabolismo , Células Fotorreceptoras Retinianas Bastones/citología , Proteína de Unión al GTP rac1
7.
Transl Vis Sci Technol ; 9(7): 28, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32844051

RESUMEN

Purpose: Electric micro-current has been shown to enhance penetration and transduction of adeno-associated viral (AAV) vectors in mouse retina after intravitreal administration. We termed this: "electric-current vector mobility (ECVM)." The present study considered whether ECVM could augment retinal transduction efficiency of intravitreal AAV8-CMV-EGFP in normal rabbit and nonhuman primate (NHP) macaque. Potential mechanisms underlying enhanced retinal transduction by ECVM were also studied. Methods: We applied an electric micro-current across the intact eye of normal rabbit and monkey in vivo for a brief period immediately after intravitreal injection of AAV8-CMV-EGFP. Retinal GFP expression was evaluated by fundus imaging in vivo. Retinal immunohistochemistry was performed to assess the distribution of retinal cells transduced by the AAV8-EGFP. Basic fibroblast growth factor (bFGF) was analyzed by quantitative RT-polymerase chain reaction (PCR). Müller glial reactivity and inner limiting membrane (ILM) were examined by the glial fibrillary acidic protein (GFAP) and vimentin staining in mouse retina, respectively. Results: ECVM significantly increased the efficiency of AAV reaching and transducing the rabbit retina following intravitreal injection, with gene expression in inner nuclear layer, ganglion cells, and Müller cells. Similar trend of improvement was observed in the ECVM-treated monkey eye. The electric micro-current upregulated bFGF expression in Müller cells and vimentin showed ILM structural changes in mouse retina. Conclusions: ECVM promotes the transduction efficiency of AAV8-CMV-GFP in normal rabbit and monkey retinas following intravitreal injection. Translational Relevance: This work has potential translational relevance to human ocular gene therapy by increasing retinal expression of therapeutic vectors given by intravitreal administration.


Asunto(s)
Dependovirus , Vectores Genéticos , Animales , Dependovirus/genética , Expresión Génica , Vectores Genéticos/genética , Conejos , Retina , Transducción Genética
8.
PLoS Genet ; 2(10): e177, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17069463

RESUMEN

The Ran-binding protein 2 (RanBP2) is a large multimodular and pleiotropic protein. Several molecular partners with distinct functions interacting specifically with selective modules of RanBP2 have been identified. Yet, the significance of these interactions with RanBP2 and the genetic and physiological role(s) of RanBP2 in a whole-animal model remain elusive. Here, we report the identification of two novel partners of RanBP2 and a novel physiological role of RanBP2 in a mouse model. RanBP2 associates in vitro and in vivo and colocalizes with the mitochondrial metallochaperone, Cox11, and the pacemaker of glycolysis, hexokinase type I (HKI) via its leucine-rich domain. The leucine-rich domain of RanBP2 also exhibits strong chaperone activity toward intermediate and mature folding species of Cox11 supporting a chaperone role of RanBP2 in the cytosol during Cox11 biogenesis. Cox11 partially colocalizes with HKI, thus supporting additional and distinct roles in cell function. Cox11 is a strong inhibitor of HKI, and RanBP2 suppresses the inhibitory activity of Cox11 over HKI. To probe the physiological role of RanBP2 and its role in HKI function, a mouse model harboring a genetically disrupted RanBP2 locus was generated. RanBP2(-/-) are embryonically lethal, and haploinsufficiency of RanBP2 in an inbred strain causes a pronounced decrease of HKI and ATP levels selectively in the central nervous system. Inbred RanBP2(+/-) mice also exhibit deficits in growth rates and glucose catabolism without impairment of glucose uptake and gluconeogenesis. These phenotypes are accompanied by a decrease in the electrophysiological responses of photosensory and postreceptoral neurons. Hence, RanBP2 and its partners emerge as critical modulators of neuronal HKI, glucose catabolism, energy homeostasis, and targets for metabolic, aging disorders and allied neuropathies.


Asunto(s)
Glucosa/metabolismo , Haploidia , Hexoquinasa/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Proteínas Transportadoras de Cobre , Proteínas del Complejo de Cadena de Transporte de Electrón , Electrorretinografía , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de la Membrana/química , Ratones , Ratones Mutantes , Proteínas Mitocondriales/química , Modelos Biológicos , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Datos de Secuencia Molecular , Mutagénesis Insercional , Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/genética , Fenotipo , Células Fotorreceptoras/citología , Células Fotorreceptoras/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Relación Estructura-Actividad
9.
Invest Ophthalmol Vis Sci ; 60(10): 3680-3688, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31469895

RESUMEN

Purpose: The b-wave of the cone ERG increases in amplitude and speed during the first few minutes of adaptation to a rod-suppressing background light. Earlier studies implicate rod pathway input to the cone pathway in these changes. Methods: The timing and amplitude of the cone b-wave and isolated oscillatory potentials (OP) during the first 10 minutes of light adaptation in wild-type (WT) mice and two mutant lines without functional rods was examined: rhodopsin knockout (Rho-/-), lacking rod outer segments, and NRL knockout (Nrl-/-), in which rods are replaced by S-cones. Expression of the immediate-early gene c-fos, which is increased in the inner retina by light-induced activity, was evaluated by immunohistochemistry in dark- and light-adapted retinas. Results: WT b-wave and OP amplitudes increased, and implicit times decreased during light adaptation. Subtracting OP did not alter b-wave changes. Rho-/- b-wave and OP amplitudes did not increase during adaptation. B-wave timing and amplitude and the timing of the major OP at 1 minute of adaptation were equivalent to WT at 10 minutes. The light-adapted ERG b-wave in Nrl-/- mice, which originates in both the rod and cone pathways, changed in absolute amplitude and timing similar to WT. C-fos expression was present in the inner retinas of dark-adapted Rho-/- but not WT or Nrl-/- mice. Conclusions: Activity in the distal rod pathway produces changes in the cone ERG during light adaptation. Rods in Rho-/- mice constitutively activate this rod-cone pathway interaction. The rod pathway S-cones in Nrl-/- mice may maintain the WT interaction.


Asunto(s)
Adaptación Ocular/fisiología , Células Fotorreceptoras Retinianas Conos/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Electrorretinografía , Proteínas del Ojo/genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estimulación Luminosa , Proteínas Proto-Oncogénicas c-fos/genética , Retina/fisiología , Rodopsina/genética
10.
Mol Ther Methods Clin Dev ; 13: 77-85, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-30719486

RESUMEN

Adeno-associated virus (AAV) vector-mediated gene delivery is a promising approach for therapy, but implementation in the eye currently is hampered by the need for delivering the vector underneath the retina, using surgical application into the subretinal space. This limits the extent of the retina that is treated and may cause surgical injury. Vector delivery into the vitreous cavity would be preferable because it is surgically less invasive and would reach more of the retina. Unfortunately, most conventional, non-modified AAV vector serotypes penetrate the retina poorly from the vitreous; this limits efficient transduction and expression by target cells (retinal pigment epithelium and photoreceptors). We developed a method of applying a small and safe electric current across the intact eye in vivo for a brief period following intravitreal vector administration. This significantly improved AAV-mediated transduction of retinal cells in wild-type mice following intravitreal delivery, with gene expression in retinal pigment epithelium and photoreceptor cells. The low-level current had no adverse effects on retinal structure and function. This method should be generally applicable for other AAV serotypes and may have broad application in both basic research and clinical studies.

11.
J Neurosci ; 27(33): 8805-15, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699662

RESUMEN

Previous experiments indicate that congenital human retinal degeneration caused by genetic mutations that change the Ca(2+) sensitivity of retinal guanylyl cyclase (retGC) can result from an increase in concentration of free intracellular cGMP and Ca(2+) in the photoreceptors. To rescue degeneration in transgenic mouse models having either the Y99C or E155G mutations of the retGC modulator guanylyl cyclase-activating protein 1 (GCAP-1), which produce elevated cGMP synthesis in the dark, we used the G90D rhodopsin mutation, which produces constitutive stimulation of cGMP hydrolysis. The effects of the G90D transgene were evaluated by measuring retGC activity biochemically, by recording single rod and electroretinogram (ERG) responses, by intracellular free Ca(2+) measurement, and by retinal morphological analysis. Although the G90D rhodopsin did not alter the abnormal Ca(2+) sensitivity of retGC in the double-mutant animals, the intracellular free cGMP and Ca(2+) concentrations returned close to normal levels, consistent with constitutive activation of the phosphodiesterase PDE6 cascade in darkness. G90D decreased the light sensitivity of rods but spared them from severe retinal degeneration in Y99C and E155G GCAP-1 mice. More than half of the photoreceptors remained alive, appeared morphologically normal, and produced electrical responses, at the time when their siblings lacking the G90D rhodopsin transgene lost the entire retinal outer nuclear layer and no longer responded to illumination. These experiments indicate that mutations that lead to increases in cGMP and Ca(2+) can trigger photoreceptor degeneration but that constitutive activation of the transduction cascade in these animals can greatly enhance cell survival.


Asunto(s)
Ácido Aspártico/genética , GMP Cíclico/metabolismo , Glicina/genética , Proteínas Activadoras de la Guanilato-Ciclasa/metabolismo , Degeneración Retiniana/metabolismo , Células Fotorreceptoras Retinianas Bastones/fisiopatología , Rodopsina/fisiología , Adaptación Ocular/genética , Animales , Calcio/metabolismo , GMP Cíclico/genética , Cisteína/genética , Adaptación a la Oscuridad/genética , Adaptación a la Oscuridad/fisiología , Modelos Animales de Enfermedad , Electrorretinografía/métodos , Regulación de la Expresión Génica/efectos de la radiación , Proteínas Activadoras de la Guanilato-Ciclasa/genética , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión/métodos , Estimulación Física/métodos , Retina/patología , Retina/fisiopatología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Rodopsina/genética , Tirosina/genética
12.
Invest Ophthalmol Vis Sci ; 49(1): 442-52, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18172124

RESUMEN

PURPOSE: The authors investigated photopic electroretinographic changes during degeneration in the Royal College of Surgeons (RCS) and transgenic P23H rhodopsin rat models, including the cellular origins of a large corneal-negative component that persists in the RCS rat. METHODS: Photopic and scotopic electroretinograms (ERGs) were recorded from dystrophic RCS (RCS-p(+)/Lav) rats (4-18 weeks old) and transgenic rhodopsin Pro23His line 1 (P23H) rats (4-30 weeks old). Age-matched congenic (RCS-rdy(+)p(+)/Lav) and Sprague-Dawley rats were used as controls. N-methyl-DL-aspartic acid (NMA), dopamine, and gamma-aminobutyric acid (GABA) were injected intravitreally, and optic nerve sectioning (ONS) was performed to suppress or remove inner retinal neuron activity. Retinal morphology for cone cell counts and immunohistochemistry for quantification of Kir4.1 channels were performed at various stages of degeneration. RESULTS: As degeneration progressed, the photopic ERG of RCS dystrophic rats was distinctly different from that of P23H rats, primarily because of the growth of a corneal-negative response (RCS-NPR) after the b-wave in RCS rats. This response had a peak time similar to the photopic negative response (PhNR) in controls but with a more gradual recovery phase, and it was not affected by ONS. The PhNR in P23H rats declined linearly with the b-wave. NMA and GABA eliminated the RCS-NPR and uncovered a larger b-wave in RCS rats at late stages of degeneration, but NMA had little effect on the ERG in P23H rats. The NMA-sensitive negative response in RCS rats declined with age more slowly than did the NMA-isolated b-wave. The density of Kir4.1 channels at the endfeet of Müller cells and in the proximal retina increased significantly between 6 to 10 weeks and 14 weeks of age in the RCS rat retina but not in the P23H rat retina. CONCLUSIONS: The photopic ERG of the dystrophic RCS rat retina becomes increasingly electronegative because of an aberrant negative response, originating from amacrine cell activity, which declines more slowly than the b-wave with degeneration. The absence of this response in the P23H rat indicates that the inner retinal cone pathway pathology is different in the two models. A relative increase in Kir4.1 channels on Müller cells of RCS retina may contribute to the enhanced negative ERG response in the RCS rat.


Asunto(s)
Células Amacrinas/fisiología , Electrorretinografía , Retina/fisiología , Degeneración Retiniana/fisiopatología , Transducción de Señal/fisiología , Animales , Animales Modificados Genéticamente , Recuento de Células , Dopamina/farmacología , Luz , Microscopía Confocal , N-Metilaspartato/análogos & derivados , N-Metilaspartato/farmacología , Canales de Potasio de Rectificación Interna/metabolismo , Ratas , Ratas Long-Evans , Ratas Mutantes , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Degeneración Retiniana/genética , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/fisiología , Rodopsina/genética , Ácido gamma-Aminobutírico/farmacología
13.
Mol Vis ; 14: 2227-36, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19057657

RESUMEN

PURPOSE: To determine whether cadherin 23 and protocadherin 15 can substitute for one another in the maintenance of the retina and other tissues in the mouse. Does homozygosity for both v and av mutant alleles (i.e., a double homozygous mouse) cause retinal degeneration or an obvious retinal histopathology? METHODS: We generated mice homozygous for both Cdh23(v-6J) and Pcdh15(av-Jfb) alleles. The retinal phenotypes of double heterozygous and double homozygous mutant mice were determined by light microscopy and electroretinography (ERG). Histology on 32 different tissues, scanning electron microscopy of organ of Corti hair cells as well as serum biochemical and hematological examinations were evaluated. RESULTS: ERG waves of double heterozygous and double homozygous mice showed similar shape, growth of the amplitude with intensity, and implicit time for both rod and cone pathway mediated responses. Mice homozygous for both Cdh23(v-6J) and Pcdh15(av-Jfb) mutations showed no sign of retinitis pigmentosa or photoreceptor degeneration but, as expected, were deaf and had disorganized hair cell sensory bundles. CONCLUSIONS: The simultaneous presence of homozygous mutant alleles of cadherin 23 and protocadherin 15 results only in deafness, not retinal degeneration or any other additional obvious phenotype of the major organ systems. We conclude that in the mouse cadherin 23 or protocadherin 15 appear not to compensate for one another to maintain the retina.


Asunto(s)
Alelos , Homocigoto , Degeneración Retiniana/genética , Empalme Alternativo , Animales , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Núcleo Celular/patología , Cilios/ultraestructura , Electrorretinografía , Ojo/patología , Ojo/ultraestructura , Heterocigoto , Ratones , Ratones Mutantes , Fenotipo , Precursores de Proteínas/genética
14.
J Neurosci ; 26(52): 13523-30, 2006 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-17192435

RESUMEN

Ciliary neurotrophic factor (CNTF) promotes photoreceptor survival but also suppresses electroretinogram (ERG) responses. This has caused concerns about whether CNTF is detrimental to the function of photoreceptors because it is considered to be a potential treatment for retinal degenerative disorders. Here we report that the suppression of ERG responses is attributable to negative regulation of the phototransduction machinery in rod photoreceptors. Intravitreal injection of recombinant human CNTF protein in rat results in a series of biochemical and morphological changes in rod photoreceptors. CNTF induces a decrease in rhodopsin expression and an increase in arrestin level. Morphologically, CNTF treatment causes a shortening of rod outer segments (ROS). All of these changes are fully reversible. The lower rhodopsin level and shortened ROS reduce the photon catch of rods. Less rhodopsin and more arrestin dramatically increase the arrestin-to-rhodopsin ratio so that more arrestin molecules are available to quench the photoexcited rhodopsin. The overall effect of CNTF is to negatively regulate the phototransduction machinery, which reduces the photoresponsiveness of rods, resulting in lower ERG amplitude at a given intensity of light stimulus. The CNTF-induced changes in rods are similar to those in light-induced photoreceptor plasticity. Whether CNTF-induced changes in rods are through the same mechanism that mediates light-induced photoreceptor plasticity remains to be answered.


Asunto(s)
Factor Neurotrófico Ciliar/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Visión Ocular/fisiología , Animales , Adaptación a la Oscuridad/fisiología , Electrorretinografía/métodos , Humanos , Ratas , Ratas Long-Evans , Células Fotorreceptoras Retinianas Bastones/ultraestructura
15.
Invest Ophthalmol Vis Sci ; 48(8): 3837-45, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17652759

RESUMEN

PURPOSE: The authors characterized the natural history of a retinoschisin gene knockout (Rs1h-KO) mouse model and evaluated the long-term effects of retinal rescue after AAV(2/2)-CMV-Rs1h gene delivery. METHODS: Full-field scotopic electroretinograms (ERGs) were recorded from 44 male hemizygous Rs1h-KO and 44 male wild-type (WT) C57BL/6J mice at six ages between 1 and 16 months. Retinal morphometry included outer segment layer (OSL) width, photoreceptor cell count, and grading of schisis cavity severity. One eye each of seven Rs1h-KO mice at age 14 days was injected with AAV(2/2)-CMV-Rs1h, and retinal histology and ERG findings at 14 months were analyzed. RESULTS: The outer nuclear layer (ONL) of 1-month-old Rs1h-KO mice was disorganized but had nearly normal cell counts. The OSL was thinned, rod outer segments were misaligned, and abundant schisis cavities spanned the inner nuclear and outer plexiform layers in all retinas. ERG a- and b-wave amplitudes at this age were reduced by 33% and 50%, respectively. ERG and ONL cell numbers decreased further between 1 and 16 months, with unequal changes in the a- and b-waves with age. The a-wave reduction correlated well with the steady decline in ONL cell number, whereas a rapid decline in the b-wave and a (b/a-wave) ratio less than in WT were associated with increasing severity of schisis cavities at young ages. At 4 months, the cavities were maximal, but they coalesced and disappeared at older ages. The (b/a-wave) ratio was inversely correlated with cavity severity across all ages (r = -0.74; P < 0.0001; n = 22). Considerable heterogeneity was observed at each age in the ERG amplitudes and retinal morphology. Mice injected with AAV-Rs1h at 14 days showed considerable structural and functional rescue at age 14 months, including improved rod outer and inner segment integrity, less photoreceptor cell loss, and larger ERG amplitudes compared with untreated fellow eyes. CONCLUSIONS: The ERG of the Rs1h-KO mouse at early ages reflects disruption of photoreceptor and second-order neuron function. In mid to late ages, the ERG decline reflects primarily photoreceptor degeneration. The Rs1h-KO mouse is consistent with human clinical X-linked juvenile retinoschisis (XLRS) in showing schisis cavities, which affect primarily the b-wave, the regression of schisis cavities at older ages, and a considerable range in phenotypic severity across individuals. This mouse model also indicates the critical roll of RS-protein in photoreceptor survival consistent with decreased a-waves in some patients with XLRS. Long-term rescue of retinal morphology and function by AAV-Rs1h gene transfer may provide a basis for considering intervention in the homologous human XLRS condition.


Asunto(s)
Moléculas de Adhesión Celular/genética , Proteínas del Ojo/genética , Terapia Genética/métodos , Retinosquisis/genética , Retinosquisis/terapia , Factores de Edad , Animales , Recuento de Células , Modelos Animales de Enfermedad , Electrorretinografía , Técnicas de Transferencia de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/patología , Retina/fisiología , Retinosquisis/patología , Segmento Externo de la Célula en Bastón/patología , Segmento Externo de la Célula en Bastón/fisiología
16.
Invest Ophthalmol Vis Sci ; 48(3): 991-1000, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17325137

RESUMEN

PURPOSE: Retinoschisin (RS) is a retina-specific, secreted protein implicated in X-linked juvenile retinoschisis and essential for the structural and functional integrity of the retina. This biochemical characterization and ultrastructural localization of RS in intact murine retina was performed to further understanding of the molecular basis of its function. METHODS: Subcellular fractions and fractions enriched in photoreceptor inner and outer segments were prepared from mouse retina by differential or density gradient ultracentrifugation. Immunoblot analysis was used to assess the expression of RS in various subcellular compartments and its fractionation into soluble phase on treatment of retinal cell membranes with several solubilizing reagents. RS-lipid interactions were evaluated by a protein-lipid overlay assay that used wild-type and mutant forms of RS discoidin domain glutathione S-transferase (GST) fusion proteins. The subcellular localization of RS in mouse retina was visualized by pre-embedding immunogold electron microscopy. Ultrastructure was evaluated by transmission electron microscopy. RESULTS: RS was intimately associated with cell membranes of the retina. It was found to cluster on the outer leaflet of the plasma membrane of the photoreceptor inner segments, which synthesize and secrete it. It was released from the membrane at high pH, which is characteristic of a peripheral membrane protein. It was extracted from the membrane by the nonionic detergent NP-40, together with glycerophospholipids. Protein-lipid overlay assays indicated a preferential interaction between RS and anioic phospholipids. Extraction of RS from the membrane was inhibited by divalent cations. Photoreceptor inner segment morphology was markedly affected in RS(-)(/y) mice, which failed to express RS protein. CONCLUSIONS: RS in intact retina is a peripheral membrane protein. Although distributed over the two membrane faces, RS is associated primarily with the outer leaflet of the inner segment plasma membrane through anionic phospholipids and divalent cations. RS's localization in photoreceptors and its biochemical properties suggest a functional role locally, at the site of secretion and membrane adhesion, in maintaining the photoreceptor inner segment stability and architecture.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Membrana Celular/metabolismo , Proteínas del Ojo/metabolismo , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Fosfolípidos/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Animales , Aniones , Western Blotting , Moléculas de Adhesión Celular/ultraestructura , Membrana Celular/ultraestructura , Centrifugación por Gradiente de Densidad , Cromatografía en Capa Delgada , Electroforesis en Gel de Poliacrilamida , Proteínas del Ojo/ultraestructura , Glutatión Transferasa/metabolismo , Proteínas de la Membrana/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microscopía Inmunoelectrónica , Células Fotorreceptoras de Vertebrados/ultraestructura , Proteínas Recombinantes de Fusión/metabolismo , Retinosquisis/metabolismo , Fracciones Subcelulares
17.
Curr Eye Res ; 32(11): 917-22, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18027167

RESUMEN

PURPOSE: The complement system plays important roles in a variety of chronic ocular diseases such as age-related macular degeneration. Here we examined the deposition of complement components in mouse eyes damaged by various mechanisms. METHODS: Mouse eyes were damaged by light or by three models of inflammation, i.e., local transgenic expression of cytokines, interleukin-1 or -7, or by induction of experimental autoimmune uveitis. Eye tissues obtained from each model were immunostained with antibodies against complement components C1q, C3, and C4. RESULTS: No complement deposition was seen in light damaged eyes, while in inflamed eyes we found complement deposition at sites of tissue damage and cellular infiltration. In addition to affected tissues, intense immunoreactivity against complement was unexpectedly observed in corneal tissues and lens capsule, despite lack of inflammation in these tissues. CONCLUSION: Our observations suggest that ocular tissues adjacent to inflammatory sites undergo changes that facilitate complement deposition.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Complemento C1q/metabolismo , Complemento C3/metabolismo , Complemento C4/metabolismo , Inflamación/metabolismo , Uveítis/metabolismo , Animales , Enfermedades Autoinmunes/inducido químicamente , Córnea/metabolismo , Proteínas del Ojo/toxicidad , Femenino , Expresión Génica/fisiología , Interleucina-1/genética , Interleucina-7/genética , Cápsula del Cristalino/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Microscopía Fluorescente , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Traumatismos Experimentales por Radiación/metabolismo , Enfermedades de la Retina/metabolismo , Proteínas de Unión al Retinol/toxicidad , Uveítis/inducido químicamente
18.
Invest Ophthalmol Vis Sci ; 58(3): 1656-1664, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28297725

RESUMEN

Purpose: To test the effects of rearing light intensity on retinal function and morphology in the retinoschisis knockout (Rs1-KO) mouse model of X-linked retinoschisis, and whether it affects functional outcome of RS1 gene replacement. Methods: Seventy-six Rs1-KO mice were reared in either cyclic low light (LL, 20 lux) or moderate light (ML, 300 lux) and analyzed at 1 and 4 months. Retinal function was assessed by electroretinogram and cavity size by optical coherence tomography. Expression of inward-rectifier K+ channel (Kir4.1), water channel aquaporin-4 (AQP4), and glial fibrillary acidic protein (GFAP) were analyzed by Western blotting. In a separate study, Rs1-KO mice reared in LL (n = 29) or ML (n = 27) received a unilateral intravitreal injection of scAAV8-hRs-IRBP at 21 days, and functional outcome was evaluated at 4 months by electroretinogram. Results: At 1 month, no functional or structural differences were found between LL- or ML-reared Rs1-KO mice. At 4 months, ML-reared Rs1-KO mice showed significant reduction of b-wave amplitude and b-/a-wave ratio with no changes in a-wave, and a significant increase in cavity size, compared to LL-reared animals. Moderate light rearing increased Kir4.1 expression in Rs1-KO mice by 4 months, but not AQP4 and GFAP levels. Administration of scAAV8-hRS1-IRBP to Rs1-KO mice showed similar improvement of inner retinal ERG function independent of LL or ML rearing. Conclusions: Rearing light conditions affect the development of retinal cavities and post-photoreceptor function in Rs1-KO mice. However, the effect of rearing light intensity does not interact with the efficacy of RS1 gene replacement in Rs1-KO mice.


Asunto(s)
Terapia Genética/métodos , Luz , Segmento Interno de las Células Fotorreceptoras Retinianas/patología , Retinosquisis/terapia , Animales , Western Blotting , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Electrorretinografía , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Estudios de Seguimiento , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Canales de Potasio de Rectificación Interna/biosíntesis , Canales de Potasio de Rectificación Interna/genética , ARN/genética , Segmento Interno de las Células Fotorreceptoras Retinianas/efectos de la radiación , Retinosquisis/diagnóstico , Retinosquisis/genética , Retinosquisis/fisiopatología , Factores de Tiempo , Tomografía de Coherencia Óptica
19.
Invest Ophthalmol Vis Sci ; 47(7): 3074-84, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16799054

RESUMEN

PURPOSE: Mutations of PCDH15, the gene encoding protocadherin 15, cause either nonsyndromic deafness DFNB23 or Usher syndrome type 1F (USH1F) in humans and deafness with balance problems in Ames waltzer (av) mice. Persons with USH1 usually begin to exhibit signs of retinitis pigmentosa (RP) in early adolescence, but av mice are reported to have functional retinas. In this study, the auditory, visual and molecular biological phenotype of Pcdh15av-5J and Pcdh15av-Jfb mice is characterized, and their usefulness as animal models of USH1 is evaluated. METHODS: Hearing thresholds of mice between 6 and 10 weeks of age were measured by auditory brain stem response (ABR). Immunohistochemistry and histology were used to examine the effect of homozygosity of Pcdh15av-5J on stereocilia bundles of inner ear hair cells and on the photoreceptor cells of the retina. Scotopic and photopic Ganzfeld ERGs were recorded from homozygous Pcdh15av-5J and Pcdh15av-Jfb mice at different ages. Heterozygous littermates served as control subjects. Measurements of the width of the outer nuclear layer (ONL) and the length of rod photoreceptor outer segment (ROS) were made. RESULTS: Homozygous Pcdh15av-5J mice have profound hearing loss and disorganized stereocilia bundles of inner ear hair cells. Compared with heterozygous littermates, homozygous Pcdh15av-5J and Pcdh15av-Jfb mutant mice had scotopic ERG amplitudes consistently reduced by approximately 40% at all light intensities. The b-to-a-wave ratio confirmed that the a- and b-waves were reduced proportionally in homozygous mutant mice. Histologic measurements of retinal sections revealed no significant differences in either the ONL width or the ROS length as a function of genotype. The protocadherin 15 labeling pattern with antisera PB303 in the retina of both heterozygous and homozygous Pcdh15av-5J mice was indistinguishable from the wild type. Wild-type Pcdh15 have many alternatively spliced isoforms. A novel isoform was found in the retina of homozygous Pcdh15av-5J mice, which appears to circumvent the effect of the mutant allele (IVS14-2A-->G), which causes skipping of exon 14, a shift in the translation reading frame and a premature stop codon in exon 15. CONCLUSIONS: Pcdh15(av-5J) and Pcdh15(av-Jfb) mice do not faithfully mimic the RP found in USH1 due to mutations of PCDH15, but have significantly attenuated ERG function in the absence of histologic change. The decline in ERG amplitude with a preserved b-to-a-wave ratio suggests a role for Pcdh15 in retinal function and/or generation of the ERG potentials. Understanding the molecular mechanism by which av mice circumvent degeneration of the retina might offer insights into potential therapies for USH1.


Asunto(s)
Empalme Alternativo/genética , Cadherinas/genética , Sordera/congénito , Precursores de Proteínas/genética , Retina/fisiopatología , Retinitis Pigmentosa/genética , Enfermedades Vestibulares/genética , Animales , Umbral Auditivo , Proteínas Relacionadas con las Cadherinas , Modelos Animales de Enfermedad , Electrorretinografía , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Células Ciliadas Auditivas Internas/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Mutación , Fenotipo , Células Fotorreceptoras de Vertebrados/metabolismo , Retinitis Pigmentosa/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Invest Ophthalmol Vis Sci ; 47(8): 3603-11, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16877435

RESUMEN

PURPOSE: Autosomal dominant Stargardt-like (STGD3) disease results from mutations in the ELOVL4 gene (elongation of very-long-chain fatty acids). This study was undertaken to characterize a mouse model with a targeted deletion of Elovl4 and to explore the role of this gene in retinal/macular degeneration. METHODS: A construct targeted to exon 2 of the Elovl4 gene was used to suppress expression of the gene. Elovl4 homozygous pups were nonviable and were not available for study. Hence, the analysis was performed on heterozygous Elovl4(+/-) mice 16 to 22 month of age and littermate wild-type (WT) control mice of the same age. Characterization included examining gene message and protein levels, electroretinogram (ERG), retinal morphology and ultrastructure, and plasma and retinal fatty acid composition. RESULTS: Although the level of Elovl4 mRNA was reduced in Elovl4(+/-) retinas, only minimal morphologic abnormalities were found, and the retinal (ERG) function was essentially normal in Elovl4(+/-) retinas compared with the WT control retinas. Systemic fatty acid profiles of Elovl4(+/-) mice were unremarkable, although the concentration of several fatty acids was significantly lower in Elovl4(+/-) retinas, particularly the monounsaturated fatty acids. CONCLUSIONS: The detailed characterization of this animal model provides the first in vivo evidence that Elovl4 haploinsufficiency is not the underlying key disease mechanism in STGD3. The results are consistent with a dominant negative mechanism for the deletion mutation. The Elovl4 knockout mouse is one of three complementary animal models that will help elucidate the disease mechanism.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Regulación de la Expresión Génica/fisiología , Degeneración Macular/genética , Proteínas de la Membrana/genética , Animales , Western Blotting , Electrorretinografía , Ácidos Grasos/metabolismo , Femenino , Eliminación de Gen , Genotipo , Haplotipos , Degeneración Macular/metabolismo , Degeneración Macular/patología , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Retina/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rodopsina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA