Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Bioengineering (Basel) ; 11(2)2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38391665

RESUMEN

Ocular diseases present a unique challenge and opportunity for therapeutic development. The eye has distinct advantages as a therapy target given its accessibility, compartmentalization, immune privilege, and size. Various methodologies for therapeutic delivery in ocular diseases are under investigation that impact long-term efficacy, toxicity, invasiveness, and delivery range. While gene, cell, and antibody therapy and nanoparticle delivery directly treat regions that have been damaged by disease, they can be limited in the duration of the therapeutic delivery and have a focal effect. In contrast, contact lenses and ocular implants can more effectively achieve sustained and widespread delivery of therapies; however, they can increase dilution of therapeutics, which may result in reduced effectiveness. Current therapies either offer a sustained release or a broad therapeutic effect, and future directions should aim toward achieving both. This review discusses current ocular therapy delivery systems and their applications, mechanisms for delivering therapeutic products to ocular tissues, advantages and challenges associated with each delivery system, current approved therapies, and clinical trials. Future directions for the improvement in existing ocular therapies include combination therapies, such as combined cell and gene therapies, as well as AI-driven devices, such as cortical implants that directly transmit visual information to the cortex.

2.
Gene Ther ; 31(5-6): 255-262, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38273095

RESUMEN

Retinitis pigmentosa (RP) is a heterogeneous disease and the main cause of vision loss within the group of inherited retinal diseases (IRDs). IRDs are a group of rare disorders caused by mutations in one or more of over 280 genes which ultimately result in blindness. Modifier genes play a key role in modulating disease phenotypes, and mutations in them can affect disease outcomes, rate of progression, and severity. Our previous studies have demonstrated that the nuclear hormone receptor 2 family e, member 3 (Nr2e3) gene reduced disease progression and loss of photoreceptor cell layers in RhoP23H-/- mice. This follow up, pharmacology study evaluates a longitudinal NR2E3 dose response in the clinically relevant heterozygous RhoP23H mouse. Reduced retinal degeneration and improved retinal morphology was observed 6 months following treatment evaluating three different NR2E3 doses. Histological and immunohistochemical analysis revealed regions of photoreceptor rescue in the treated retinas of RhoP23H+/- mice. Functional assessment by electroretinogram (ERG) showed attenuated photoreceptor degeneration with all doses. This study demonstrates the effectiveness of different doses of NR2E3 at reducing retinal degeneration and informs dose selection for clinical trials of RhoP23H-associated RP.


Asunto(s)
Modelos Animales de Enfermedad , Receptores Nucleares Huérfanos , Degeneración Retiniana , Retinitis Pigmentosa , Animales , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Ratones , Receptores Nucleares Huérfanos/genética , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/etiología , Electrorretinografía , Retina/metabolismo , Retina/patología , Terapia Genética/métodos
3.
Bioengineering (Basel) ; 11(1)2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38247923

RESUMEN

Alzheimer's Disease (AD) is a complex neurodegenerative disease resulting in progressive loss of memory, language and motor abilities caused by cortical and hippocampal degeneration. This review captures the landscape of understanding of AD pathology, diagnostics, and current therapies. Two major mechanisms direct AD pathology: (1) accumulation of amyloid ß (Aß) plaque and (2) tau-derived neurofibrillary tangles (NFT). The most common variants in the Aß pathway in APP, PSEN1, and PSEN2 are largely responsible for early-onset AD (EOAD), while MAPT, APOE, TREM2 and ABCA7 have a modifying effect on late-onset AD (LOAD). More recent studies implicate chaperone proteins and Aß degrading proteins in AD. Several tests, such as cognitive function, brain imaging, and cerebral spinal fluid (CSF) and blood tests, are used for AD diagnosis. Additionally, several biomarkers seem to have a unique AD specific combination of expression and could potentially be used in improved, less invasive diagnostics. In addition to genetic perturbations, environmental influences, such as altered gut microbiome signatures, affect AD. Effective AD treatments have been challenging to develop. Currently, there are several FDA approved drugs (cholinesterase inhibitors, Aß-targeting antibodies and an NMDA antagonist) that could mitigate AD rate of decline and symptoms of distress.

4.
Int Ophthalmol Clin ; 62(2): 197-214, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-35325919
5.
J Clin Med ; 10(3)2021 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-33513943

RESUMEN

NR2E3-associated recessive disease in humans is historically defined by congenital night blinding retinopathy, characterized by an initial increase in short-wavelength (S)-cone sensitivity and progressive loss of rod and cone function. The retinal degeneration 7 (rd7) murine model, harboring a recessive mutation in the mouse ortholog of NR2E3, has been a well-studied disease model and recently evaluated as a therapeutic model for NR2E3-associated retinal degenerations. This study aims to draw parallels between human and mouse NR2E3-related disease through examination of spectral domain optical coherence tomography (SD-OCT) imaging between different stage of human disease and its murine counterpart. We propose that SD-OCT is a useful non-invasive diagnostic tool to compare human clinical dystrophy presentation with that of the rd7 mouse and make inference that may be of therapeutically relevance. Additionally, a longitudinal assessment of rd7 disease progression, utilizing available clinical data from our patients as well as extensive retrospective analysis of visual acuity data from published cases of human NR2E3-related disease, was curated to identify further valuable correlates between human and mouse Nr2e3 disease. Results of this study validate the slow progression of NR2E3-associated disease in humans and the rd7 mice and identify SD-OCT characteristics in patients at or near the vascular arcades that correlate well with the whorls and rosettes that are seen also in the rd7 mouse and point to imaging features that appear to be associated with better preserved S-cone mediated retinal function. The correlation of histological findings between rd7 mice and human imaging provides a solid foundation for diagnostic use of pathophysiological and prognostic information to further define characteristics and a relevant timeline for therapeutic intervention in the field of NR2E3-associated retinopathies.

6.
Gene Ther ; 28(5): 223-241, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32123325

RESUMEN

Recent advances in viral vector engineering, as well as an increased understanding of the cellular and molecular mechanism of retinal diseases, have led to the development of novel gene therapy approaches. Furthermore, ease of accessibility and ocular immune privilege makes the retina an ideal target for gene therapies. In this study, the nuclear hormone receptor gene Nr2e3 was evaluated for efficacy as broad-spectrum therapy to attenuate early to intermediate stages of retinal degeneration in five unique mouse models of retinitis pigmentosa (RP). RP is a group of heterogenic inherited retinal diseases associated with over 150 gene mutations, affecting over 1.5 million individuals worldwide. RP varies in age of onset, severity, and rate of progression. In addition, ~40% of RP patients cannot be genetically diagnosed, confounding the ability to develop personalized RP therapies. Remarkably, Nr2e3 administered therapy resulted in reduced retinal degeneration as observed by increase in photoreceptor cells, improved electroretinogram, and a dramatic molecular reset of key transcription factors and associated gene networks. These therapeutic effects improved retinal homeostasis in diseased tissue. Results of this study provide evidence that Nr2e3 can serve as a broad-spectrum therapy to treat multiple forms of RP.


Asunto(s)
Degeneración Retiniana , Retinitis Pigmentosa , Animales , Modelos Animales de Enfermedad , Homeostasis , Humanos , Ratones , Receptores Nucleares Huérfanos , Células Fotorreceptoras , Retina , Degeneración Retiniana/genética , Degeneración Retiniana/terapia , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia
7.
Genet Med ; 21(9): 2103-2115, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30967659

RESUMEN

PURPOSE: To identify the molecular cause in five unrelated families with a distinct autosomal dominant ocular systemic disorder we called ROSAH syndrome due to clinical features of retinal dystrophy, optic nerve edema, splenomegaly, anhidrosis, and migraine headache. METHODS: Independent discovery exome and genome sequencing in families 1, 2, and 3, and confirmation in families 4 and 5. Expression of wild-type messenger RNA and protein in human and mouse tissues and cell lines. Ciliary assays in fibroblasts from affected and unaffected family members. RESULTS: We found the heterozygous missense variant in the ɑ-kinase gene, ALPK1, (c.710C>T, [p.Thr237Met]), segregated with disease in all five families. All patients shared the ROSAH phenotype with additional low-grade ocular inflammation, pancytopenia, recurrent infections, and mild renal impairment in some. ALPK1 was notably expressed in retina, retinal pigment epithelium, and optic nerve, with immunofluorescence indicating localization to the basal body of the connecting cilium of the photoreceptors, and presence in the sweat glands. Immunocytofluorescence revealed expression at the centrioles and spindle poles during metaphase, and at the base of the primary cilium. Affected family member fibroblasts demonstrated defective ciliogenesis. CONCLUSION: Heterozygosity for ALPK1, p.Thr237Met leads to ROSAH syndrome, an autosomal dominant ocular systemic disorder.


Asunto(s)
Nervio Óptico/patología , Proteínas Quinasas/genética , Retina/metabolismo , Distrofias Retinianas/genética , Exoma/genética , Femenino , Heterocigoto , Humanos , Hipohidrosis/genética , Hipohidrosis/patología , Masculino , Trastornos Migrañosos/genética , Trastornos Migrañosos/patología , Mutación Missense/genética , Nervio Óptico/metabolismo , Linaje , Fenotipo , Retina/patología , Distrofias Retinianas/patología , Esplenomegalia/genética , Esplenomegalia/patología
9.
Dev Biol ; 429(1): 343-355, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28551284

RESUMEN

Nuclear hormone receptors play a major role in the development of many tissues. This study uncovers a novel role for testicular receptor 2 (Tr2, Nr2c1) in defining the early phase of retinal development and regulating normal retinal cell patterning and topography. The mammalian retina undergoes an overlapping yet biphasic period of development to generate all seven retinal cell types. We discovered that Nr2c1 expression coincides with development of the early retinal cells. Loss of Nr2c1 causes a severe vision deficit and impacts early, but not late retina cell types. Retinal cone cell topography is disrupted with an increase in displaced amacrine cells. Additionally, genetic background significantly impacts phenotypic outcome of cone photoreceptor cells but not amacrine cells. Chromatin-IP experiments reveal NR2C1 regulates early cell transcription factors that regulate retinal progenitor cells during development, including amacrine (Satb2) and cone photoreceptor regulators thyroid and retinoic acid receptors. This study supports a role for Nr2c1 in defining the biphasic period of retinal development and specifically influencing the early phase of retinal cell fate.


Asunto(s)
Tipificación del Cuerpo/genética , Miembro 1 del Grupo C de la Subfamilia 2 de Receptores Nucleares/metabolismo , Retina/embriología , Retina/metabolismo , Células Amacrinas/citología , Células Amacrinas/metabolismo , Animales , Proliferación Celular , Forma de la Célula , Regulación del Desarrollo de la Expresión Génica , Fototransducción/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Miembro 1 del Grupo C de la Subfamilia 2 de Receptores Nucleares/genética , Unión Proteica/genética , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Sinapsis/metabolismo
10.
PLoS One ; 10(9): e0135927, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26352270

RESUMEN

Autism spectrum disorders (ASDs) are a range of complex neurodevelopmental conditions principally characterized by dysfunctions linked to mental development. Previous studies have shown that there are more than 1000 genes likely involved in ASD, expressed mainly in brain and highly interconnected among them. We applied whole exome sequencing in Colombian-South American trios. Two missense novel SNVs were found in the same child: ALDH1A3 (RefSeq NM_000693: c.1514T>C (p.I505T)) and FOXN1 (RefSeq NM_003593: c.146C>T (p.S49L)). Gene expression studies reveal that Aldh1a3 and Foxn1 are expressed in ~E13.5 mouse embryonic brain, as well as in adult piriform cortex (PC; ~P30). Conserved Retinoic Acid Response Elements (RAREs) upstream of human ALDH1A3 and FOXN1 and in mouse Aldh1a3 and Foxn1 genes were revealed using bioinformatic approximation. Chromatin immunoprecipitation (ChIP) assay using Retinoid Acid Receptor B (Rarb) as the immunoprecipitation target suggests RA regulation of Aldh1a3 and Foxn1 in mice. Our results frame a possible link of RA regulation in brain to ASD etiology, and a feasible non-additive effect of two apparently unrelated variants in ALDH1A3 and FOXN1 recognizing that every result given by next generation sequencing should be cautiously analyzed, as it might be an incidental finding.


Asunto(s)
Aldehído Oxidorreductasas/genética , Trastorno del Espectro Autista/genética , Exoma , Factores de Transcripción Forkhead/genética , Receptores de Ácido Retinoico/genética , Tretinoina/metabolismo , Adulto , Aldehído Oxidorreductasas/metabolismo , Animales , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/patología , Secuencia de Bases , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Encéfalo/patología , Niño , Estudios de Cohortes , Colombia , Embrión de Mamíferos , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Linaje , Pruebas Psicológicas , Receptores de Ácido Retinoico/metabolismo , Elementos de Respuesta , Transducción de Señal
11.
Hum Mutat ; 36(6): 599-610, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25703721

RESUMEN

NR2E3 encodes the photoreceptor-specific nuclear hormone receptor that acts as a repressor of cone-specific gene expression in rod photoreceptors, and as an activator of several rod-specific genes. Recessive variants located in the ligand-binding domain (LBD) of NR2E3 cause enhanced short wavelength sensitive- (S-) cone syndrome (ESCS), a retinal degeneration characterized by an excess of S-cones and non-functional rods. We analyzed the dimerization properties of NR2E3 and the effect of disease-causing LBD missense variants by bioluminescence resonance energy transfer (BRET(2) ) protein interaction assays. Homodimerization was not affected in presence of p.A256V, p.R039G, p.R311Q, and p.R334G variants, but abolished in presence of p.L263P, p.L336P, p.L353V, p.R385P, and p.M407K variants. Homology modeling predicted structural changes induced by NR2E3 LBD variants. NR2E3 LBD variants did not affect interaction with CRX, but with NRL and rev-erbα/NR1D1. CRX and NRL heterodimerized more efficiently together, than did either with NR2E3. NR2E3 did not heterodimerize with TLX/NR2E1 and RXRα/NR2C1. The identification of a new compound heterozygous patient with detectable rod function, who expressed solely the p.A256V variant protein, suggests a correlation between LBD variants able to form functional NR2E3 dimers and atypical mild forms of ESCS with residual rod function.


Asunto(s)
Enfermedades Hereditarias del Ojo/genética , Receptores Nucleares Huérfanos/química , Receptores Nucleares Huérfanos/genética , Dominios y Motivos de Interacción de Proteínas/genética , Multimerización de Proteína , Degeneración Retiniana/genética , Trastornos de la Visión/genética , Adolescente , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Línea Celular , Análisis Mutacional de ADN , Enfermedades Hereditarias del Ojo/diagnóstico , Proteínas del Ojo/metabolismo , Angiografía con Fluoresceína , Proteínas de Homeodominio/metabolismo , Humanos , Ligandos , Masculino , Modelos Moleculares , Mutación , Linaje , Unión Proteica , Conformación Proteica , Retina/metabolismo , Degeneración Retiniana/diagnóstico , Tomografía de Coherencia Óptica , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Trastornos de la Visión/diagnóstico
12.
J Exp Neurosci ; 9(Suppl 2): 93-121, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27168725

RESUMEN

The nuclear hormone receptor (NHR) superfamily is composed of a wide range of receptors involved in a myriad of important biological processes, including development, growth, metabolism, and maintenance. Regulation of such wide variety of functions requires a complex system of gene regulation that includes interaction with transcription factors, chromatin-modifying complex, and the proper recognition of ligands. NHRs are able to coordinate the expression of genes in numerous pathways simultaneously. This review focuses on the role of nuclear receptors in the central nervous system and, in particular, their role in regulating the proper development and function of the brain and the eye. In addition, the review highlights the impact of mutations in NHRs on a spectrum of human diseases from autism to retinal degeneration.

13.
PLoS One ; 9(9): e106507, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25188381

RESUMEN

BACKGROUND: Vision loss due to vascular disease of the retina is a leading cause of blindness in the world. Retinal angiomatous proliferation (RAP) is a subgroup of neovascular age-related macular degeneration (AMD), whereby abnormal blood vessels develop in the retina leading to debilitating vision loss and eventual blindness. The novel mouse strain, neoretinal vascularization 2 (NRV2), shows spontaneous fundus changes associated with abnormal neovascularization. The purpose of this study is to characterize the induction of pathologic angiogenesis in this mouse model. METHODS: The NRV2 mice were examined from postnatal day 12 (p12) to 3 months. The phenotypic changes within the retina were evaluated by fundus photography, fluorescein angiography, optical coherence tomography, and immunohistochemical and electron microscopic analysis. The pathological neovascularization was imaged by confocal microscopy and reconstructed using three-dimensional image analysis software. RESULTS: We found that NRV2 mice develop multifocal retinal depigmentation in the posterior fundus. Depigmented lesions developed vascular leakage observed by fluorescein angiography. The spontaneous angiogenesis arose from the retinal vascular plexus at postnatal day (p)15 and extended toward retinal pigment epithelium (RPE). By three months of age, histological analysis revealed encapsulation of the neovascular lesion by the RPE in the photoreceptor cell layer and subretinal space. CONCLUSIONS: The NRV2 mouse strain develops early neovascular lesions within the retina, which grow downward towards the RPE beginning at p15. This retinal neovascularization model mimics early stages of human retinal angiomatous proliferation (RAP) and will likely be a useful in elucidating targeted therapeutics for patients with ocular neovascular disease.


Asunto(s)
Neovascularización Retiniana/patología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Angiografía con Fluoresceína , Estudios Longitudinales , Ratones , Epitelio Pigmentado Ocular/patología , Retina/patología , Tomografía de Coherencia Óptica
14.
Invest Ophthalmol Vis Sci ; 55(6): 3543-54, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24812550

RESUMEN

PURPOSE: Current understanding of the genetic risk factors for age-related macular degeneration (AMD) is not sufficiently predictive of the clinical course. The VEGF pathway is a key therapeutic target for treatment of neovascular AMD; however, risk attributable to genetic variation within pathway genes is unclear. We sought to identify single nucleotide polymorphisms (SNPs) associated with AMD within the VEGF pathway. METHODS: Using a tagSNP, direct sequencing and meta-analysis approach within four ethnically diverse cohorts, we identified genetic risk present in FLT1, though not within other VEGF pathway genes KDR, VEGFA, or VASH1. We used ChIP and ELISA in functional analysis. RESULTS: The FLT1 SNPs rs9943922, rs9508034, rs2281827, rs7324510, and rs9513115 were significantly associated with increased risk of neovascular AMD. Each association was more significant after meta-analysis than in any one of the four cohorts. All associations were novel, within noncoding regions of FLT1 that do not tag for coding variants in linkage disequilibrium. Analysis of soluble FLT1 demonstrated higher expression in unaffected individuals homozygous for the FLT1 risk alleles rs9943922 (P = 0.0086) and rs7324510 (P = 0.0057). In silico analysis suggests that these variants change predicted splice sites and RNA secondary structure, and have been identified in other neovascular pathologies. These data were supported further by murine chromatin immunoprecipitation demonstrating that FLT1 is a target of Nr2e3, a nuclear receptor gene implicated in regulating an AMD pathway. CONCLUSIONS: Although exact variant functions are not known, these data demonstrate relevancy across ethnically diverse genetic backgrounds within our study and, therefore, hold potential for global efficacy.


Asunto(s)
Etnicidad , Predisposición Genética a la Enfermedad , Degeneración Macular/genética , Polimorfismo Genético , ARN/genética , Neovascularización Retiniana/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Genotipo , Grecia/etnología , Humanos , Inmunoprecipitación , Degeneración Macular/etnología , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Prevalencia , República de Corea/etnología , Neovascularización Retiniana/etnología , Neovascularización Retiniana/metabolismo , Factores de Riesgo , Reino Unido/etnología , Estados Unidos/epidemiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
PLoS One ; 9(1): e87942, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498227

RESUMEN

Nuclear hormone receptors play a major role in many important biological processes. Most nuclear hormone receptors are ubiquitously expressed and regulate processes such as metabolism, circadian function, and development. They function in these processes to maintain homeostasis through modulation of transcriptional gene networks. In this study we evaluate the effectiveness of a nuclear hormone receptor gene to modulate retinal degeneration and restore the integrity of the retina. Currently, there are no effective treatment options for retinal degenerative diseases leading to progressive and irreversible blindness. In this study we demonstrate that the nuclear hormone receptor gene Nr1d1 (Rev-Erbα) rescues Nr2e3-associated retinal degeneration in the rd7 mouse, which lacks a functional Nr2e3 gene. Mutations in human NR2E3 are associated with several retinal degenerations including enhanced S cone syndrome and retinitis pigmentosa. The rd7 mouse, lacking Nr2e3, exhibits an increase in S cones and slow, progressive retinal degeneration. A traditional genetic mapping approach previously identified candidate modifier loci. Here, we demonstrate that in vivo delivery of the candidate modifier gene, Nr1d1 rescues Nr2e3 associated retinal degeneration. We observed clinical, histological, functional, and molecular restoration of the rd7 retina. Furthermore, we demonstrate that the mechanism of rescue at the molecular and functional level is through the re-regulation of key genes within the Nr2e3-directed transcriptional network. Together, these findings reveal the potency of nuclear receptors as modulators of disease and specifically of NR1D1 as a novel therapeutic for retinal degenerations.


Asunto(s)
Enfermedades Hereditarias del Ojo , Terapia Genética/métodos , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares , Receptores Nucleares Huérfanos , Degeneración Retiniana , Retinitis Pigmentosa , Trastornos de la Visión , Animales , Enfermedades Hereditarias del Ojo/genética , Enfermedades Hereditarias del Ojo/metabolismo , Enfermedades Hereditarias del Ojo/patología , Enfermedades Hereditarias del Ojo/terapia , Humanos , Ratones , Ratones Transgénicos , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Receptores Nucleares Huérfanos/biosíntesis , Receptores Nucleares Huérfanos/genética , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Degeneración Retiniana/terapia , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/terapia , Trastornos de la Visión/genética , Trastornos de la Visión/metabolismo , Trastornos de la Visión/patología , Trastornos de la Visión/terapia
16.
Hum Genomics ; 5(6): 538-68, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22155603

RESUMEN

Vitamin D has been shown to have anti-angiogenic properties and to play a protective role in several types of cancer, including breast, prostate and cutaneous melanoma. Similarly, vitamin D levels have been shown to be protective for risk of a number of conditions, including cardiovascular disease and chronic kidney disease, as well as numerous autoimmune disorders such as multiple sclerosis, inflammatory bowel diseases and type 1 diabetes mellitus. A study performed by Parekh et al. was the first to suggest a role for vitamin D in age-related macular degeneration (AMD) and showed a correlation between reduced serum vitamin D levels and risk for early AMD. Based on this study and the protective role of vitamin D in diseases with similar pathophysiology to AMD, we examined the role of vitamin D in a family-based cohort of 481 sibling pairs. Using extremely phenotypically discordant sibling pairs, initially we evaluated the association of neovascular AMD and vitamin D/sunlight-related epidemiological factors. After controlling for established AMD risk factors, including polymorphisms of the genes encoding complement factor H (CFH) and age-related maculopathy susceptibility 2/HtrA serine peptidase (ARMS2/HTRA1), and smoking history, we found that ultraviolet irradiance was protective for the development of neovascular AMD (p = 0.001). Although evaluation of serum vitamin D levels (25-hydroxyvitamin D [25(OH)D]) was higher in unaffected individuals than in their affected siblings, this finding did not reach statistical significance. Based on the relationship between ultraviolet irradiance and vitamin D production, we employed a candidate gene approach for evaluating common variation in key vitamin D pathway genes (the genes encoding the vitamin D receptor [VDR]; cytochrome P450, family 27, subfamily B, polypeptide 1 [CYP27B1]; cytochrome P450, family 24, subfamily A, polypeptide 1 [CYP24A1]; and CYP27A1) in this same family-based cohort. Initial findings were then validated and replicated in the extended family cohort, an unrelated case-control cohort from central Greece and a prospective nested case-control population from the Nurse's Health Study and Health Professionals Follow-Up Studies, which included patients with all subtypes of AMD for a total of 2,528 individuals. Single point variants in CYP24A1 (the gene encoding the catabolising enzyme of the vitamin D pathway) were demonstrated to influence AMD risk after controlling for smoking history, sex and age in all populations, both separately and, more importantly, in a meta-analysis. This is the first report demonstrating a genetic association between vitamin D metabolism and AMD risk. These findings were also supplemented with expression data from human donor eyes and human retinal cell lines. These data not only extend previous biological studies in the AMD field, but further emphasise common antecedents between several disorders with an inflammatory/immunogenic component such as cardiovascular disease, cancer and AMD.


Asunto(s)
Predisposición Genética a la Enfermedad , Degeneración Macular/etiología , Degeneración Macular/patología , Polimorfismo Genético/genética , Biología de Sistemas , Deficiencia de Vitamina D/complicaciones , Vitamina D/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Factor H de Complemento/genética , Estudios Epidemiológicos , Femenino , Estudios de Seguimiento , Genotipo , Grecia/epidemiología , Humanos , Degeneración Macular/epidemiología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Receptores de Calcitriol/genética , Factores de Riesgo , Hermanos , Deficiencia de Vitamina D/epidemiología , Deficiencia de Vitamina D/genética
17.
PLoS One ; 6(10): e25775, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21998696

RESUMEN

ROBO1 is a strong candidate gene for age-related macular degeneration (AMD) based upon its location under a linkage peak on chromosome 3p12, its expression pattern, and its purported function in a pathway that includes RORA, a gene previously associated with risk for neovascular AMD. Previously, we observed that expression of ROBO1 and RORA is down-regulated among wet AMD cases, as compared to their unaffected siblings. Thus, we hypothesized that contribution of association signals in ROBO1, and interaction between these two genes may be important for both wet and dry AMD. We evaluated association of 19 single nucleotide polymorphisms (SNPs) in ROBO1 with wet and dry stages of AMD in a sibling cohort and a Greek case-control cohort containing 491 wet AMD cases, 174 dry AMD cases and 411 controls. Association signals and interaction results were replicated in an independent prospective cohort (1070 controls, 164 wet AMD cases, 293 dry AMD cases). The most significantly associated ROBO1 SNPs were rs1387665 under an additive model (meta P = 0.028) for wet AMD and rs9309833 under a recessive model (meta P = 6 × 10(-4)) for dry AMD. Further analyses revealed interaction between ROBO1 rs9309833 and RORA rs8034864 for both wet and dry AMD (interaction P<0.05). These studies were further supported by whole transcriptome expression profile studies from 66 human donor eyes and chromatin immunoprecipitation assays from mouse retinas. These findings suggest that distinct ROBO1 variants may influence the risk of wet and dry AMD, and the effects of ROBO1 on AMD risk may be modulated by RORA variants.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Degeneración Macular/genética , Degeneración Macular/fisiopatología , Proteínas del Tejido Nervioso/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Fenotipo , Receptores Inmunológicos/genética , Anciano , Animales , Inmunoprecipitación de Cromatina , Epistasis Genética/genética , Ojo/metabolismo , Ojo/fisiopatología , Femenino , Atrofia Geográfica/genética , Atrofia Geográfica/fisiopatología , Humanos , Masculino , Ratones , Polimorfismo de Nucleótido Simple/genética , Transcriptoma , Degeneración Macular Húmeda/genética , Degeneración Macular Húmeda/fisiopatología , Proteínas Roundabout
18.
PLoS One ; 6(7): e21858, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21779340

RESUMEN

Variation in genetic background can significantly influence the phenotypic outcome of both disease and non-disease associated traits. Additionally, differences in temporal and strain specific gene expression can also contribute to phenotypes in the mammalian retina. This is the first report of microarray based cross-strain analysis of gene expression in the retina investigating genetic background effects. Microarray analyses were performed on retinas from the following mouse strains: C57BL6/J, AKR/J, CAST/EiJ, and NOD.NON-H2(-nb1) at embryonic day 18.5 (E18.5) and postnatal day 30.5 (P30.5). Over 3000 differentially expressed genes were identified between strains and developmental stages. Differential gene expression was confirmed by qRT-PCR, Western blot, and immunohistochemistry. Three major gene networks were identified that function to regulate retinal or photoreceptor development, visual perception, cellular transport, and signal transduction. Many of the genes in these networks are implicated in retinal diseases such as bradyopsia, night-blindness, and cone-rod dystrophy. Our analysis revealed strain specific variations in cone photoreceptor cell patterning and retinal function. This study highlights the substantial impact of genetic background on both development and function of the retina and the level of gene expression differences tolerated for normal retinal function. These strain specific genetic variations may also be present in other tissues. In addition, this study will provide valuable insight for the development of more accurate models for human retinal diseases.


Asunto(s)
Variación Genética/genética , Retina/metabolismo , Animales , Western Blotting , Inmunohistoquímica , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Retina/citología , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
PLoS One ; 6(3): e17494, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21408158

RESUMEN

The majority of diseases in the retina are caused by genetic mutations affecting the development and function of photoreceptor cells. The transcriptional networks directing these processes are regulated by genes such as nuclear hormone receptors. The nuclear hormone receptor gene Rev-erb alpha/Nr1d1 has been widely studied for its role in the circadian cycle and cell metabolism, however its role in the retina is unknown. In order to understand the role of Rev-erb alpha/Nr1d1 in the retina, we evaluated the effects of loss of Nr1d1 to the developing retina and its co-regulation with the photoreceptor-specific nuclear receptor gene Nr2e3 in the developing and mature retina. Knock-down of Nr1d1 expression in the developing retina results in pan-retinal spotting and reduced retinal function by electroretinogram. Our studies show that NR1D1 protein is co-expressed with NR2E3 in the outer neuroblastic layer of the developing mouse retina. In the adult retina, NR1D1 is expressed in the ganglion cell layer and is co-expressed with NR2E3 in the outer nuclear layer, within rods and cones. Several genes co-targeted by NR2E3 and NR1D1 were identified that include: Nr2c1, Recoverin, Rgr, Rarres2, Pde8a, and Nupr1. We examined the cyclic expression of Nr1d1 and Nr2e3 over a twenty-four hour period and observed that both nuclear receptors cycle in a similar manner. Taken together, these studies reveal a novel role for Nr1d1, in conjunction with its cofactor Nr2e3, in regulating transcriptional networks critical for photoreceptor development and function.


Asunto(s)
Redes Reguladoras de Genes/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Retina/metabolismo , Envejecimiento/metabolismo , Envejecimiento/efectos de la radiación , Animales , Recuento de Células , Elementos de Facilitación Genéticos/genética , Inyecciones , Luz , Fototransducción/genética , Fototransducción/efectos de la radiación , Ratones , Especificidad de Órganos/efectos de la radiación , Transporte de Proteínas/efectos de la radiación , ARN Interferente Pequeño/metabolismo , Retina/citología , Retina/crecimiento & desarrollo , Retina/efectos de la radiación , Coloración y Etiquetado , Activación Transcripcional/genética , Activación Transcripcional/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA