Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 162(3): 607-21, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26232227

RESUMEN

We identified a dominant missense mutation in the SCN transcription factor Zfhx3, termed short circuit (Zfhx3(Sci)), which accelerates circadian locomotor rhythms in mice. ZFHX3 regulates transcription via direct interaction with predicted AT motifs in target genes. The mutant protein has a decreased ability to activate consensus AT motifs in vitro. Using RNA sequencing, we found minimal effects on core clock genes in Zfhx3(Sci/+) SCN, whereas the expression of neuropeptides critical for SCN intercellular signaling was significantly disturbed. Moreover, mutant ZFHX3 had a decreased ability to activate AT motifs in the promoters of these neuropeptide genes. Lentiviral transduction of SCN slices showed that the ZFHX3-mediated activation of AT motifs is circadian, with decreased amplitude and robustness of these oscillations in Zfhx3(Sci/+) SCN slices. In conclusion, by cloning Zfhx3(Sci), we have uncovered a circadian transcriptional axis that determines the period and robustness of behavioral and SCN molecular rhythms.


Asunto(s)
Ritmo Circadiano , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Neuropéptidos/genética , Núcleo Supraquiasmático/metabolismo , Secuencia de Aminoácidos , Animales , Regulación hacia Abajo , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutación , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Alineación de Secuencia , Transcripción Genética
2.
PLoS Genet ; 16(4): e1008583, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32236127

RESUMEN

The precise control of eye size is essential for normal vision. TMEM98 is a highly conserved and widely expressed gene which appears to be involved in eye size regulation. Mutations in human TMEM98 are found in patients with nanophthalmos (very small eyes) and variants near the gene are associated in population studies with myopia and increased eye size. As complete loss of function mutations in mouse Tmem98 result in perinatal lethality, we produced mice deficient for Tmem98 in the retinal pigment epithelium (RPE), where Tmem98 is highly expressed. These mice have greatly enlarged eyes that are very fragile with very thin retinas, compressed choroid and thin sclera. To gain insight into the mechanism of action we used a proximity labelling approach to discover interacting proteins and identified MYRF as an interacting partner. Mutations of MYRF are also associated with nanophthalmos. The protein is an endoplasmic reticulum-tethered transcription factor which undergoes autoproteolytic cleavage to liberate the N-terminal part which then translocates to the nucleus where it acts as a transcription factor. We find that TMEM98 inhibits the self-cleavage of MYRF, in a novel regulatory mechanism. In RPE lacking TMEM98, MYRF is ectopically activated and abnormally localised to the nuclei. Our findings highlight the importance of the interplay between TMEM98 and MYRF in determining the size of the eye.


Asunto(s)
Ojo/anatomía & histología , Ojo/metabolismo , Proteínas de la Membrana/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Animales , Electrorretinografía , Anomalías del Ojo/genética , Femenino , Eliminación de Gen , Mutación con Pérdida de Función , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Tamaño de los Órganos/genética , Unión Proteica , Transporte de Proteínas , Epitelio Pigmentado de la Retina/anomalías , Epitelio Pigmentado de la Retina/metabolismo , Retinaldehído/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo
3.
FASEB J ; 35(9): e21802, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34383984

RESUMEN

Mutations in transcription factors often exhibit pleiotropic effects related to their complex expression patterns and multiple regulatory targets. One such mutation in the zinc finger homeobox 3 (ZFHX3) transcription factor, short circuit (Sci, Zfhx3Sci/+ ), is associated with significant circadian deficits in mice. However, given evidence of its retinal expression, we set out to establish the effects of the mutation on retinal function using molecular, cellular, behavioral and electrophysiological measures. Immunohistochemistry confirms the expression of ZFHX3 in multiple retinal cell types, including GABAergic amacrine cells and retinal ganglion cells including intrinsically photosensitive retinal ganglion cells (ipRGCs). Zfhx3Sci/+ mutants display reduced light responsiveness in locomotor activity and circadian entrainment, relatively normal electroretinogram and optomotor responses but exhibit an unexpected pupillary reflex phenotype with markedly increased sensitivity. Furthermore, multiple electrode array recordings of Zfhx3Sci/+ retina show an increased sensitivity of ipRGC light responses.


Asunto(s)
Ritmo Circadiano/fisiología , Proteínas de Homeodominio/metabolismo , Retina/metabolismo , Células Amacrinas/metabolismo , Animales , Luz , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Estimulación Luminosa/métodos , Células Ganglionares de la Retina/metabolismo , Visión Ocular/fisiología
4.
Gene Ther ; 28(5): 265-276, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33750925

RESUMEN

Dry age-related macular degeneration (AMD) is characterised by loss of central vision and currently has no approved medical treatment. Dysregulation of the complement system is thought to play an important role in disease pathology and supplementation of Complement Factor I (CFI), a key regulator of the complement system, has the potential to provide a treatment option for AMD. In this study, we demonstrate the generation of AAV constructs carrying the human CFI sequence and expression of CFI in cell lines and in the retina of C57BL/6 J mice. Four codon optimised constructs were compared to the most common human CFI sequence. All constructs expressed CFI protein; however, most codon optimised sequences resulted in significantly reduced CFI secretion compared to the non-optimised CFI sequence. In vivo expression analysis showed that CFI was predominantly expressed in the RPE and photoreceptors. Secreted protein in vitreous humour was demonstrated to be functionally active. The findings presented here have led to the formulation of an AAV-vectored gene therapy product currently being tested in a first-in-human clinical trial in subjects with geographic atrophy secondary to dry AMD (NCT03846193).


Asunto(s)
Factor I de Complemento , Degeneración Macular , Animales , Factor I de Complemento/genética , Terapia Genética , Humanos , Degeneración Macular/genética , Degeneración Macular/terapia , Ratones , Ratones Endogámicos C57BL , Retina
5.
Mol Ther ; 28(3): 830-844, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32027843

RESUMEN

The authors describe retinal reconstruction and restoration of visual function in heritably blind mice missing the rhodopsin gene using a novel method of ex vivo gene therapy and cell transplantation. Photoreceptor precursors with the same chromosomal genetic mutation were treated ex vivo using minicircle DNA, a non-viral technique that does not present the packaging limitations of adeno-associated virus (AAV) vectors. Following transplantation, genetically modified cells reconstructed a functional retina and supported vision in blind mice harboring the same founder gene mutation. Gene delivery by minicircles showed comparable long-term efficiency to AAV in delivering the missing gene, representing the first non-viral system for robust treatment of photoreceptors. This important proof-of-concept finding provides an innovative convergence of cell and gene therapies for the treatment of hereditary neurodegenerative disease and may be applied in future studies toward ex vivo correction of patient-specific cells to provide an autologous source of tissue to replace lost photoreceptors in inherited retinal blindness. This is the first report using minicircles in photoreceptor progenitors and the first to transplant corrected photoreceptor precursors to restore vision in blind animals.


Asunto(s)
ADN/administración & dosificación , Terapia Genética , Células-Madre Neurales/metabolismo , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/terapia , Trasplante de Células Madre , Animales , Diferenciación Celular , Células Cultivadas , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Orden Génico , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Ratones , Ratones Noqueados , Plásmidos/genética , Rodopsina/genética , Trasplante de Células Madre/métodos , Transducción Genética , Transgenes
6.
Exp Eye Res ; 190: 107895, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31816293

RESUMEN

Imaging techniques have revolutionised the assessment of retinal disease in humans and animal models. Here we describe a novel technique for the in vivo visualisation of rod photoreceptors which permits semiquantitative assessment of outer retinal degeneration, and validate this approach in two mouse models of retinitis pigmentosa (RP). Transgenic mice carrying an Nrl-EGFP allele and homozygous for either knock-out of rhodopsin (Nrl-EGFP, Rho-/-) or heterozygous for knock-in of P23H mutant rhodopsin (Nrl-EGFP, RhoP23H/+) were used in this study. These novel strains have green fluorescent rods which undergo a progressive degeneration. Fundus imaging was performed at three-weekly intervals by near infrared reflectance (NIR) and blue light autofluorescence (BAF) confocal scanning laser ophthalmoscopy (cSLO). Mean grey values (mGV), which quantify fluorescence levels within such images, were compared for degenerate and age-matched non-degenerate (Nrl-EGFP, Rho+/+) controls. Mean grey value significantly decreased over time in the Rho-/- and RhoP23H/+ groups but was maintained in Rho+/+ mice (P < 0.001, two-way ANOVA). This corresponded to outer nuclear layer (ONL) thinning as observed by histology. The mGV of superior retina was significantly greater than that of inferior retina in RhoP23H/+ (P = 0.0024) but not in age-matched Rho+/+ (P = 0.45) or Rho-/- (P = 0.65) mice reflecting histological findings. Focal loss of rods could be visualised and mapped in vivo with this technique following a toxic insult, with thinning of the ONL being confirmed in hypofluorescent regions by spectral domain ocular coherence tomography (OCT). Fluorescence labelling of rods permits in vivo characterisation of models of RP and may provide new insights into patterns of degeneration, or rescue effect after treatment. mGV can be used in such cases as a semiquantitative metric of ONL degeneration, and can be used to identify regional variations in photoreceptor loss.


Asunto(s)
Modelos Animales de Enfermedad , Genes Reporteros/genética , Proteínas Fluorescentes Verdes/genética , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/patología , Animales , Dependovirus , Expresión Génica/fisiología , Vectores Genéticos , Inyecciones Intraoculares , Ratones , Ratones Noqueados , Ratones Transgénicos , Oftalmoscopía , Parvovirinae/genética , Degeneración Retiniana/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinitis Pigmentosa/metabolismo , Rodopsina/genética , Coloración y Etiquetado , Tomografía de Coherencia Óptica
7.
Proc Natl Acad Sci U S A ; 114(42): 11211-11216, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-28973921

RESUMEN

Optogenetic strategies to restore vision in patients who are blind from end-stage retinal degenerations aim to render remaining retinal cells light sensitive once photoreceptors are lost. Here, we assessed long-term functional outcomes following subretinal delivery of the human melanopsin gene (OPN4) in the rd1 mouse model of retinal degeneration using an adeno-associated viral vector. Ectopic expression of OPN4 using a ubiquitous promoter resulted in cellular depolarization and ganglion cell action potential firing. Restoration of the pupil light reflex, behavioral light avoidance, and the ability to perform a task requiring basic image recognition were restored up to 13 mo following injection. These data suggest that melanopsin gene therapy via a subretinal route may be a viable and stable therapeutic option for the treatment of end-stage retinal degeneration in humans.


Asunto(s)
Terapia Genética/métodos , Degeneración Retiniana/terapia , Opsinas de Bastones/genética , Animales , Dependovirus , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C3H , Visión Ocular
8.
Int J Mol Sci ; 21(3)2020 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-31991730

RESUMEN

RNA editing aims to treat genetic disease through altering gene expression at the transcript level. Pairing site-directed RNA-targeting mechanisms with engineered deaminase enzymes allows for the programmable correction of G>A and T>C mutations in RNA. This offers a promising therapeutic approach for a range of genetic diseases. For inherited retinal degenerations caused by point mutations in large genes not amenable to single-adeno-associated viral (AAV) gene therapy such as USH2A and ABCA4, correcting RNA offers an alternative to gene replacement. Genome editing of RNA rather than DNA may offer an improved safety profile, due to the transient and potentially reversible nature of edits made to RNA. This review considers the current site-directing RNA editing systems, and the potential to translate these to the clinic for the treatment of inherited retinal degeneration.


Asunto(s)
Edición Génica , Terapia Genética , Edición de ARN , Retina/metabolismo , Transgenes , Adenosina Desaminasa/metabolismo , Animales , Sistemas CRISPR-Cas , Técnica del Anticuerpo Fluorescente , Marcación de Gen , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/terapia , Humanos , Proteínas de Unión al ARN/metabolismo , Retina/patología , Enfermedades de la Retina/genética , Enfermedades de la Retina/terapia
9.
Int J Mol Sci ; 21(17)2020 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-32842706

RESUMEN

Retinitis pigmentosa (RP) is a generic term for a group of genetic diseases characterized by loss of rod and cone photoreceptor cells. Although the genetic causes of RP frequently only affect the rod photoreceptor cells, cone photoreceptors become stressed in the absence of rods and undergo a secondary degeneration. Changes in the gene expression profile of cone photoreceptor cells are likely to occur prior to observable physiological changes. To this end, we sought to achieve greater understanding of the changes in cone photoreceptor cells early in the degeneration process of the Rho-/- mouse model. To account for gene expression changes attributed to loss of cone photoreceptor cells, we normalized PCR in the remaining number of cones to a cone cell reporter (OPN1-GFP). Gene expression profiles of key components involved in the cone phototransduction cascade were correlated with tests of retinal cone function prior to cell loss. A significant downregulation of the photoreceptor transcription factor Crx was observed, which preceded a significant downregulation in cone opsin transcripts that coincided with declining cone function. Our data add to the growing understanding of molecular changes that occur prior to cone dysfunction in a model of rod-cone dystrophy. It is of interest that gene supplementation of CRX by adeno-associated viral vector delivery prior to cone cell loss did not prevent cone photoreceptor degeneration in this mouse model.


Asunto(s)
Distrofias de Conos y Bastones/genética , Distrofias de Conos y Bastones/fisiopatología , Animales , Distrofias de Conos y Bastones/terapia , Modelos Animales de Enfermedad , Electrorretinografía , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/farmacología , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/farmacología , Humanos , Ratones Transgénicos , Oftalmoscopía , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Conos/fisiología , Rodopsina/genética , Opsinas de Bastones/genética , Tomografía de Coherencia Óptica , Transactivadores/genética , Transactivadores/farmacología , Visión Ocular/genética
10.
Int J Mol Sci ; 21(14)2020 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-32674481

RESUMEN

Effective treatment of retinal diseases with adeno-associated virus (AAV)-mediated gene therapy is highly dependent on the proportion of successfully transduced cells. However, due to inflammatory reactions at high vector doses, adjunctive treatment may be necessary to enhance the therapeutic outcome. Hydroxychloroquine and chloroquine are anti-malarial drugs that have been successfully used in the treatment of autoimmune diseases. Evidence suggests that at high concentrations, hydroxychloroquine and chloroquine can impact viral infection and replication by increasing endosomal and lysosomal pH. This effect has led to investigations into the potential benefits of these drugs in the treatment of viral infections, including human immunodeficiency virus and severe acute respiratory syndrome coronavirus-2. However, at lower concentrations, hydroxychloroquine and chloroquine appear to exert immunomodulatory effects by inhibiting nucleic acid sensors, including toll-like receptor 9 and cyclic GMP-AMP synthase. This dose-dependent effect on their mechanism of action supports observations of increased viral infections associated with lower drug doses. In this review, we explore the immunomodulatory activity of hydroxychloroquine and chloroquine, their impact on viral infections, and their potential to improve the efficacy and safety of retinal gene therapy by reducing AAV-induced immune responses. The safety and practicalities of delivering hydroxychloroquine into the retina will also be discussed.


Asunto(s)
Cloroquina/uso terapéutico , Terapia Genética , Hidroxicloroquina/uso terapéutico , Enfermedades de la Retina/terapia , Virosis/tratamiento farmacológico , Animales , Betacoronavirus/efectos de los fármacos , Cloroquina/farmacología , Dependovirus/genética , Humanos , Hidroxicloroquina/farmacología , Inmunomodulación/efectos de los fármacos , Enfermedades de la Retina/patología , SARS-CoV-2
11.
FASEB J ; 32(8): 4302-4314, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29561690

RESUMEN

Cryptochromes 1 and 2 (CRY1/2) are key components of the negative limb of the mammalian circadian clock. Like many peripheral tissues, Cry1 and -2 are expressed in the retina, where they are thought to play a role in regulating rhythmic physiology. However, studies differ in consensus as to their localization and function, and CRY1 immunostaining has not been convincingly demonstrated in the retina. Here we describe the expression and function of CRY1 and -2 in the mouse retina in both sexes. Unexpectedly, we show that CRY1 is expressed throughout all retinal layers, whereas CRY2 is restricted to the photoreceptor layer. Retinal period 2::luciferase recordings from CRY1-deficient mice show reduced clock robustness and stability, while those from CRY2-deficient mice show normal, albeit long-period, rhythms. In functional studies, we then investigated well-defined rhythms in retinal physiology. Rhythms in the photopic electroretinogram, contrast sensitivity, and pupillary light response were all severely attenuated or abolished in CRY1-deficient mice. In contrast, these physiological rhythms are largely unaffected in mice lacking CRY2, and only photopic electroretinogram rhythms are affected. Together, our data suggest that CRY1 is an essential component of the mammalian retinal clock, whereas CRY2 has a more limited role.-Wong, J. C. Y., Smyllie, N. J., Banks, G. T., Pothecary, C. A., Barnard, A. R., Maywood, E. S., Jagannath, A., Hughes, S., van der Horst, G. T. J., MacLaren, R. E., Hankins, M. W., Hastings, M. H., Nolan, P. M., Foster, R. G., Peirson, S. N. Differential roles for cryptochromes in the mammalian retinal clock.


Asunto(s)
Criptocromos/metabolismo , Mamíferos/metabolismo , Mamíferos/fisiología , Retina/metabolismo , Retina/fisiología , Animales , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Electrorretinografía/métodos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/fisiología
12.
Proc Natl Acad Sci U S A ; 113(10): 2756-61, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26903623

RESUMEN

The suprachiasmatic nucleus (SCN) defines 24 h of time via a transcriptional/posttranslational feedback loop in which transactivation of Per (period) and Cry (cryptochrome) genes by BMAL1-CLOCK complexes is suppressed by PER-CRY complexes. The molecular/structural basis of how circadian protein complexes function is poorly understood. We describe a novel N-ethyl-N-nitrosourea (ENU)-induced mutation, early doors (Edo), in the PER-ARNT-SIM (PAS) domain dimerization region of period 2 (PER2) (I324N) that accelerates the circadian clock of Per2(Edo/Edo) mice by 1.5 h. Structural and biophysical analyses revealed that Edo alters the packing of the highly conserved interdomain linker of the PER2 PAS core such that, although PER2(Edo) complexes with clock proteins, its vulnerability to degradation mediated by casein kinase 1ε (CSNK1E) is increased. The functional relevance of this mutation is revealed by the ultrashort (<19 h) but robust circadian rhythms in Per2(Edo/Edo); Csnk1e(Tau/Tau) mice and the SCN. These periods are unprecedented in mice. Thus, Per2(Edo) reveals a direct causal link between the molecular structure of the PER2 PAS core and the pace of SCN circadian timekeeping.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/genética , Mutación Missense , Proteínas Circadianas Period/genética , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Caseína Cinasa 1 épsilon/genética , Caseína Cinasa 1 épsilon/metabolismo , Chlorocebus aethiops , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Femenino , Células HEK293 , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Datos de Secuencia Molecular , Actividad Motora/genética , Actividad Motora/fisiología , Proteínas Circadianas Period/química , Proteínas Circadianas Period/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiopatología
13.
Mol Ther ; 25(8): 1854-1865, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28549772

RESUMEN

X-linked retinitis pigmentosa (XLRP) is generally a severe form of retinitis pigmentosa, a neurodegenerative, blinding disorder of the retina. 70% of XLRP cases are due to mutations in the retina-specific isoform of the gene encoding retinitis pigmentosa GTPase regulator (RPGRORF15). Despite successful RPGRORF15 gene replacement with adeno-associated viral (AAV) vectors being established in a number of animal models of XLRP, progression to human trials has not yet been possible. The inherent sequence instability in the purine-rich region of RPGRORF15 (which contains highly repetitive nucleotide sequences) leads to unpredictable recombination errors during viral vector cloning. While deleted RPGR may show some efficacy in animal models, which have milder disease, the therapeutic effect of a mutated RPGR variant in patients with XLRP cannot be predicted. Here, we describe an optimized gene replacement therapy for human XLRP disease using an AAV8 vector that reliably and consistently produces the full-length correct RPGR protein. The glutamylation pattern in the RPGR protein derived from the codon-optimized sequence is indistinguishable from the wild-type variant, implying that codon optimization does not significantly alter post-translational modification. The codon-optimized sequence has superior stability and expression levels in vitro. Significantly, when delivered by AAV8 vector and driven by the rhodopsin kinase promoter, the codon-optimized RPGR rescues the disease phenotype in two relevant animal models (Rpgr-/y and C57BL/6JRd9/Boc) and shows good safety in C57BL6/J wild-type mice. This work provides the basis for clinical trial development to treat patients with XLRP caused by RPGR mutations.


Asunto(s)
Proteínas Portadoras/genética , Codón , Dependovirus/genética , Proteínas del Ojo/genética , Genes Ligados a X , Terapia Genética , Vectores Genéticos/genética , Retinitis Pigmentosa/genética , Animales , Modelos Animales de Enfermedad , Expresión Génica , Ratones , Mutación , Fenotipo , Biosíntesis de Proteínas , Procesamiento Proteico-Postraduccional , Estabilidad del ARN , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/terapia , Transducción Genética , Transgenes
14.
Proc Natl Acad Sci U S A ; 112(27): 8415-20, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26106163

RESUMEN

Stargardt disease, an ATP-binding cassette, subfamily A, member 4 (ABCA4)-related retinopathy, is a genetic condition characterized by the accelerated accumulation of lipofuscin in the retinal pigment epithelium, degeneration of the neuroretina, and loss of vision. No approved treatment exists. Here, using a murine model of Stargardt disease, we show that the propensity of vitamin A to dimerize is responsible for triggering the formation of the majority of lipofuscin and transcriptional dysregulation of genes associated with inflammation. Data further demonstrate that replacing vitamin A with vitamin A deuterated at the carbon 20 position (C20-D3-vitamin A) impedes the dimerization rate of vitamin A--by approximately fivefold for the vitamin A dimer A2E--and subsequent lipofuscinogenesis and normalizes the aberrant transcription of complement genes without impairing retinal function. Phenotypic rescue by C20-D3-vitamin A was also observed noninvasively by quantitative autofluorescence, an imaging technique used clinically, in as little as 3 months after the initiation of treatment, whereas upon interruption of treatment, the age-related increase in autofluorescence resumed. Data suggest that C20-D3-vitamin A is a clinically amiable tool to inhibit vitamin A dimerization, which can be used to determine whether slowing the dimerization of vitamin A can prevent vision loss caused by Stargardt disease and other retinopathies associated with the accumulation of lipofuscin in the retina.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Degeneración Macular/congénito , Epitelio Pigmentado de la Retina/efectos de los fármacos , Vitamina A/farmacología , Transportadoras de Casetes de Unión a ATP/genética , Animales , Deuterio , Dimerización , Electrorretinografía , Lipofuscina/biosíntesis , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/genética , Ratones de la Cepa 129 , Ratones Noqueados , Microscopía Electrónica de Transmisión , Fenotipo , Retina/efectos de los fármacos , Retina/metabolismo , Retina/fisiopatología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Enfermedad de Stargardt , Resultado del Tratamiento , Vitamina A/química , Vitaminas/química , Vitaminas/farmacología
15.
J Neurosci ; 36(1): 222-34, 2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26740663

RESUMEN

The Neuroplastin gene encodes two synapse-enriched protein isoforms, Np55 and Np65, which are transmembrane glycoproteins that regulate several cellular processes, including the genesis, maintenance, and plasticity of synapses. We found that an absence of Np65 causes early-onset sensorineural hearing loss and prevented the normal synaptogenesis in inner hair cells (IHCs) in the newly identified mouse mutant pitch. In wild-type mice, Np65 is strongly upregulated in the cochlea from around postnatal day 12 (P12), which corresponds to the onset of hearing. Np65 was specifically localized at the presynaptic region of IHCs. We found that the colocalization of presynaptic IHC ribbons and postsynaptic afferent terminals is greatly reduced in pitch mutants. Moreover, IHC exocytosis is also reduced with mutant mice showing lower rates of vesicle release. Np65 appears to have a nonessential role in vision. We propose that Np65, by regulating IHC synaptogenesis, is critical for auditory function in mammals. SIGNIFICANCE STATEMENT: In the mammalian cochlea, the sensory inner hair cells (IHCs) encode auditory information. They do this by converting sound wave-induced mechanical motion of their hair bundles into an electrical current. This current generates a receptor potential that controls release of glutamate neurotransmitter from their ribbon synapses onto the auditory afferent fiber. We show that the synapse-enriched protein Np65, encoded by the Neuroplastin gene, is localized at the IHC presynaptic region. In mutant mice, absence of Np65 causes early-onset sensorineural hearing loss and prevents normal neurotransmitter release in IHCs and colocalization of presynaptic ribbons with postsynaptic afferents. We identified Neuroplastin as a novel deafness gene required for ribbon synapse formation and function, which is critical for sound perception in mammals.


Asunto(s)
Sordera/fisiopatología , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patología , Glicoproteínas de Membrana/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis
16.
Yale J Biol Med ; 90(4): 543-551, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29259520

RESUMEN

Achromatopsia is a rare congenital cause of vision loss due to isolated cone photoreceptor dysfunction. The most common underlying genetic mutations are autosomal recessive changes in CNGA3, CNGB3, GNAT2, PDE6H, PDE6C, or ATF6. Animal models of Cnga3, Cngb3, and Gnat2 have been rescued using AAV gene therapy; showing partial restoration of cone electrophysiology and integration of this new photopic vision in reflexive and behavioral visual tests. Three gene therapy phase I/II trials are currently being conducted in human patients in the USA, the UK, and Germany. This review details the AAV gene therapy treatments of achromatopsia to date. We also present novel data showing rescue of a Cnga3-/- mouse model using an rAAV.CBA.CNGA3 vector. We conclude by synthesizing the implications of this animal work for ongoing human trials, particularly, the challenge of restoring integrated cone retinofugal pathways in an adult visual system. The evidence to date suggests that gene therapy for achromatopsia will need to be applied early in childhood to be effective.


Asunto(s)
Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/terapia , Terapia Genética/métodos , Animales , Ritmo Circadiano , Ensayos Clínicos como Asunto , Defectos de la Visión Cromática/patología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Perros , Humanos , Ratones , Retina/patología
17.
Gene Ther ; 23(11): 767-774, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27416076

RESUMEN

Gene therapy using adeno-associated viral (AAV) vectors for the treatment of retinal degenerations has shown safety and efficacy in clinical trials. However, very high levels of vector expression may be necessary for the treatment of conditions such as Stargardt disease where a dual vector approach is potentially needed, or in optogenetic strategies for end-stage degeneration in order to achieve maximal light sensitivity. In this study, we assessed two vectors with single capsid mutations, rAAV2/2(Y444F) and rAAV2/8(Y733F) in their ability to transduce retina in the Abca4-/- and rd1 mouse models of retinal degeneration. We noted significantly increased photoreceptor transduction using rAAV2/8(Y733F) in the Abca4-/- mouse, in contrast to previous work where vectors tested in this model have shown low levels of photoreceptor transduction. Bipolar cell transduction was achieved following subretinal delivery of both vectors in the rd1 mouse, and via intravitreal delivery of rAAV2/2(Y444F). The successful use of rAAV2/8(Y733F) to target bipolar cells was further validated on human tissue using an ex vivo culture system of retinal explants. Capsid mutant AAV vectors transduce human retinal cells and may be particularly suited to treat retinal degenerations in which high levels of transgene expression are required.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Proteínas de la Cápside/genética , Dependovirus/genética , Terapia Genética , Mutación Missense , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/terapia , Animales , Línea Celular Tumoral , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL , Degeneración Retiniana/genética
18.
Mol Ther ; 23(8): 1308-1319, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25896245

RESUMEN

The long-term outcome of neuroprotection as a therapeutic strategy for preventing cell death in neurodegenerative disorders remains unknown, primarily due to slow disease progression and the inherent difficulty of assessing neuronal survival in vivo. Employing a murine model of retinal disease, we demonstrate that ciliary neurotrophic factor (CNTF) confers life-long protection against photoreceptor degeneration. Repetitive retinal imaging allowed the survival of intrinsically fluorescent cone photoreceptors to be quantified in vivo. Imaging of the visual cortex and assessment of visually-evoked behavioral responses demonstrated that surviving cones retain function and signal correctly to the brain. The mechanisms underlying CNTF-mediated neuroprotection were explored through transcriptome analysis, revealing widespread upregulation of proteolysis inhibitors, which may prevent cellular/extracellular matrix degradation and complement activation in neurodegenerative diseases. These findings provide insights into potential novel therapeutic avenues for diseases such as retinitis pigmentosa and amyotrophic lateral sclerosis, for which CNTF has been evaluated unsuccessfully in clinical trials.


Asunto(s)
Factor Neurotrófico Ciliar/genética , Terapia Genética/métodos , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/prevención & control , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Electrofisiología , Electrorretinografía , Matriz Extracelular/metabolismo , Fondo de Ojo , Vectores Genéticos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Neuroprotección , Retina/patología , Células Fotorreceptoras Retinianas Conos/patología , Retinitis Pigmentosa/fisiopatología , Análisis de Secuencia de ARN , Transcriptoma , Resultado del Tratamiento , Corteza Visual/patología
19.
Proc Natl Acad Sci U S A ; 110(3): 1101-6, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23288902

RESUMEN

One strategy to restore vision in retinitis pigmentosa and age-related macular degeneration is cell replacement. Typically, patients lose vision when the outer retinal photoreceptor layer is lost, and so the therapeutic goal would be to restore vision at this stage of disease. It is not currently known if a degenerate retina lacking the outer nuclear layer of photoreceptor cells would allow the survival, maturation, and reconnection of replacement photoreceptors, as prior studies used hosts with a preexisting outer nuclear layer at the time of treatment. Here, using a murine model of severe human retinitis pigmentosa at a stage when no host rod cells remain, we show that transplanted rod precursors can reform an anatomically distinct and appropriately polarized outer nuclear layer. A trilaminar organization was returned to rd1 hosts that had only two retinal layers before treatment. The newly introduced precursors were able to resume their developmental program in the degenerate host niche to become mature rods with light-sensitive outer segments, reconnecting with host neurons downstream. Visual function, assayed in the same animals before and after transplantation, was restored in animals with zero rod function at baseline. These observations suggest that a cell therapy approach may reconstitute a light-sensitive cell layer de novo and hence repair a structurally damaged visual circuit. Rather than placing discrete photoreceptors among preexisting host outer retinal cells, total photoreceptor layer reconstruction may provide a clinically relevant model to investigate cell-based strategies for retinal repair.


Asunto(s)
Degeneración Retiniana/cirugía , Células Fotorreceptoras Retinianas Bastones/trasplante , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Células Fotorreceptoras Retinianas Bastones/fisiología , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/fisiopatología , Retinitis Pigmentosa/cirugía , Segmento Externo de la Célula en Bastón/patología , Trasplante de Células Madre , Visión Ocular
20.
Lancet ; 383(9923): 1129-37, 2014 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-24439297

RESUMEN

BACKGROUND: Choroideremia is an X-linked recessive disease that leads to blindness due to mutations in the CHM gene, which encodes the Rab escort protein 1 (REP1). We assessed the effects of retinal gene therapy with an adeno-associated viral (AAV) vector encoding REP1 (AAV.REP1) in patients with this disease. METHODS: In a multicentre clinical trial, six male patients (aged 35-63 years) with choroideremia were administered AAV.REP1 (0·6-1·0×10(10) genome particles, subfoveal injection). Visual function tests included best corrected visual acuity, microperimetry, and retinal sensitivity tests for comparison of baseline values with 6 months after surgery. This study is registered with ClinicalTrials.gov, number NCT01461213. FINDINGS: Despite undergoing retinal detachment, which normally reduces vision, two patients with advanced choroideremia who had low baseline best corrected visual acuity gained 21 letters and 11 letters (more than two and four lines of vision). Four other patients with near normal best corrected visual acuity at baseline recovered to within one to three letters. Mean gain in visual acuity overall was 3·8 letters (SE 4·1). Maximal sensitivity measured with dark-adapted microperimetry increased in the treated eyes from 23·0 dB (SE 1·1) at baseline to 25·3 dB (1·3) after treatment (increase 2·3 dB [95% CI 0·8-3·8]). In all patients, over the 6 months, the increase in retinal sensitivity in the treated eyes (mean 1·7 [SE 1·0]) was correlated with the vector dose administered per mm(2) of surviving retina (r=0·82, p=0·04). By contrast, small non-significant reductions (p>0·05) were noted in the control eyes in both maximal sensitivity (-0·8 dB [1·5]) and mean sensitivity (-1·6 dB [0·9]). One patient in whom the vector was not administered to the fovea re-established variable eccentric fixation that included the ectopic island of surviving retinal pigment epithelium that had been exposed to vector. INTERPRETATION: The initial results of this retinal gene therapy trial are consistent with improved rod and cone function that overcome any negative effects of retinal detachment. These findings lend support to further assessment of gene therapy in the treatment of choroideremia and other diseases, such as age-related macular degeneration, for which intervention should ideally be applied before the onset of retinal thinning. FUNDING: UK Department of Health and Wellcome Trust.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/administración & dosificación , Coroideremia/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Proteínas Adaptadoras Transductoras de Señales/genética , Adenoviridae/genética , Adulto , Anciano , Coroideremia/fisiopatología , Fluorescencia , Técnicas de Transferencia de Gen , Humanos , Inyecciones Intraoculares , Masculino , Persona de Mediana Edad , Desprendimiento de Retina/fisiopatología , Desprendimiento de Retina/terapia , Transgenes/genética , Agudeza Visual/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA