Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Cell Mol Med ; 25(21): 10020-10027, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34617687

RESUMEN

Biallelic mutations in the RCBTB1 gene cause retinal dystrophy. Here, we characterized the effects of RCBTB1 gene deficiency in retinal pigment epithelial (RPE) cells derived from a patient with RCBTB1-associated retinopathy and restored RCBTB1 expression in these cells using adeno-associated viral (AAV) vectors. Induced pluripotent stem cells derived from a patient with compound heterozygous RCBTB1 mutations (c.170delG and c.707delA) and healthy control subjects were differentiated into RPE cells. RPE cells were treated with AAV vectors carrying a RCBTB1 transgene. Patient-derived RPE cells showed reduced expression of RCBTB1. Expression of NFE2L2 showed a non-significant reduction in patient RPE cells compared with controls, while expression of its target genes (RXRA, IDH1 and SLC25A25) was significantly reduced. Trans-epithelial electrical resistance, surface microvillus densities and primary cilium lengths were reduced in patient-derived RPE cells, compared with controls. Treatment of patient RPE with AAV vectors significantly increased RCBTB1, NFE2L2 and RXRA expression and cilium lengths. Our study provides the first report examining the phenotype of RPE cells derived from a patient with RCBTB1-associated retinopathy. Furthermore, treatment of patient-derived RPE with AAV-RCBTB1 vectors corrected deficits in gene expression and RPE ultrastructure, supporting the use of gene replacement therapy for treating this inherited retinal disease.


Asunto(s)
Cilios/metabolismo , Expresión Génica , Terapia Genética , Factores de Intercambio de Guanina Nucleótido/genética , Epitelio Pigmentado de la Retina/metabolismo , Transgenes , Diferenciación Celular , Células Cultivadas , Cilios/ultraestructura , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Distrofias Retinianas/genética , Distrofias Retinianas/terapia , Epitelio Pigmentado de la Retina/ultraestructura , Transducción Genética
2.
Genes (Basel) ; 12(10)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34680937

RESUMEN

Retinitis pigmentosa 11 (RP11) is caused by dominant mutations in PRPF31, however a significant proportion of mutation carriers do not develop retinopathy. Here, we investigated the relationship between CNOT3 polymorphism, MSR1 repeat copy number and disease penetrance in RP11 patients and non-penetrant carriers (NPCs). We further characterized PRPF31 and CNOT3 expression in fibroblasts from eight RP11 patients and one NPC from a family carrying the c.1205C>T variant. Retinal organoids (ROs) and retinal pigment epithelium (RPE) were differentiated from induced pluripotent stem cells derived from RP11 patients, an NPC and a control subject. All RP11 patients were homozygous for the 3-copy MSR1 repeat in the PRPF31 promoter, while 3/5 NPCs carried a 4-copy MSR1 repeat. The CNOT3 rs4806718 genotype did not correlate with disease penetrance. PRFP31 expression declined with age in adult cadaveric retina. PRPF31 and CNOT3 expression was reduced in RP11 fibroblasts, RO and RPE compared with controls. Both RP11 and NPC RPE displayed shortened primary cilia compared with controls, however a subpopulation of cells with normal cilia lengths was present in NPC RPE monolayers. Our results indicate that RP11 non-penetrance is associated with the inheritance of a 4-copy MSR1 repeat, but not with CNOT3 polymorphisms.


Asunto(s)
Proteínas del Ojo/genética , Penetrancia , Retinitis Pigmentosa/genética , Adolescente , Adulto , Anciano , Células Cultivadas , Niño , Proteínas del Ojo/metabolismo , Femenino , Genes Modificadores , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo Genético , Retina/metabolismo , Retina/patología , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase A/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Ophthalmic Genet ; 42(3): 266-275, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33624564

RESUMEN

Background: Mutations in the RCC1 and BTB domain-containing protein 1 (RCBTB1) gene have been implicated in a rare form of retinal dystrophy. Herein, we report the clinical features of a 45-year-old Singaporean-Chinese female and her presymptomatic sibling, who each possesses compound heterozygous mutations in RCBTB1. Expression of RCBTB1 in patient-derived cells was evaluated.Materials and Methods: The natural history was documented by a series of ophthalmic examinations including electroretinography, fundus autofluorescence imaging, spectral-domain optical coherence tomography, visual field, microperimetry, and adaptive optics retinal imaging. Patient DNA was genetically analysed using a 537-gene Next Generation Sequencing panel and targeted Sanger sequencing. Expression of RCBTB1 in lymphocytes, fibroblasts, and induced pluripotent stem cells (iPSC) derived from the proband and healthy controls was characterized by quantitative PCR, Sanger sequencing, and western blotting.Results: The proband presented with left visual distortion at age 40 due to extrafoveal chorioretinal atrophy. Atrophy expanded at 1.3 (OD) and 1.0 (OS) mm2/year. Total macular volume declined by 0.09 (OD) and 0.13 (OS) mm3/year. Microperimetry demonstrated enlarging scotoma in both eyes. Generalised cone dysfunction was demonstrated by electroretinography. A retinal dystrophy panel testing revealed biallelic frameshifting mutations, c.170delG (p.Gly57Glufs*12) and c.707delA (p.Asn236Thrfs*11) in RCBTB1. The level of RCBTB1 mRNA expression was reduced in patient-derived lymphocytes compared to controls. RCBTB1 protein was detected in control fibroblasts and iPSC but was absent in patient-derived cells.Conclusions: Atrophy expansion rate and macular volume change are feasible endpoints for monitoring RCBTB1-associated retinopathy. We provide further functional evidence of pathogenicity for two disease-causing variants using patient-derived iPSCs.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Mutación/genética , Distrofias Retinianas/genética , Pueblo Asiatico/etnología , Pueblo Asiatico/genética , Western Blotting , Electrorretinografía , Femenino , Fibroblastos/metabolismo , Angiografía con Fluoresceína , Expresión Génica , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Linfocitos/metabolismo , Persona de Mediana Edad , Fenotipo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Singapur/epidemiología
4.
Mol Genet Genomic Med ; 9(3): e1601, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33497524

RESUMEN

BACKGROUND: Mutations in CLN3 cause Batten disease, however non-syndromic CLN3 disease, characterized by retinal-specific degeneration, has been also described. Here, we characterized an induced pluripotent stem cell (iPSC)-derived disease model derived from a patient with non-syndromic CLN3-associated retinopathy. METHODS: Patient-iPSC, carrying the 1 kb-deletion and c.175G>A variants in CLN3, coisogenic iPSC, in which the c.175G>A variant was corrected, and control iPSC were differentiated into neural retinal organoids (NRO) and cardiomyocytes. CLN3 transcripts were analyzed by Sanger sequencing. Gene expression was characterized by qPCR and western blotting. NRO were characterized by immunostaining and electron microscopy. RESULTS: Novel CLN3 transcripts were detected in adult human retina and control-NRO. The major transcript detected in patient-NRO displayed skipping of exons 2 and 4-9. Accumulation of subunit-C of mitochondrial ATPase (SCMAS) protein was demonstrated in patient-derived cells. Photoreceptor progenitor cells in patient-NRO displayed accumulation of peroxisomes and vacuolization of inner segments. Correction of the c.175G>A variant restored CLN3 mRNA and protein expression and prevented SCMAS and inner segment vacuolization. CONCLUSION: Our results demonstrate the expression of novel CLN3 transcripts in human retinal tissues. The c.175G>A variant alters splicing of the CLN3 pre-mRNA, leading to features consistent with CLN3 deficiency, which were prevented by gene correction.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Glicoproteínas de Membrana/genética , Chaperonas Moleculares/genética , Lipofuscinosis Ceroideas Neuronales/genética , Organoides/metabolismo , Retina/metabolismo , Diferenciación Celular , Células Cultivadas , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Lipofuscinosis Ceroideas Neuronales/patología , Organoides/patología , Peroxisomas/metabolismo , Mutación Puntual , Empalme del ARN , Retina/patología
5.
Mol Genet Genomic Med ; 8(11): e1489, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32931148

RESUMEN

BACKGROUND: Mutations in the human crumbs homologue 1 (CRB1) gene are associated with a spectrum of inherited retinal diseases. However, functional studies demonstrating the impact of individual CRB1 mutations on gene expression are lacking for most variants. Here, we investigated the effect of two CRB1 variants on pre-mRNA splicing using neural retinal organoids (NRO) derived from a patient with recessive rod-cone dystrophy caused by compound heterozygous mutations in CRB1 (c.1892A>G and c.2548G>A). METHODS: The patient received ophthalmological examinations including multimodal imaging. NRO were differentiated from induced pluripotent stem cells (iPSCs) derived from the patient and a control subject. CRB1 transcripts were characterized by RT-PCR and Sanger sequencing. RESULTS: The Patient displayed retinal thickening with disorganization of retinal layers and preservation of para-arteriolar retinal pigment epithelium. Both patient and control iPSC produced NRO containing photoreceptor progenitor cells expressing CRB1 mRNA. Patient NRO expressed a novel CRB1 transcript displaying skipping of exon 6. CRB1 transcripts containing the c.2548G>A substitution in exon 7 were expressed in patient NRO. CONCLUSIONS: Together, these results confirm the pathogenicity of the c.1892A>G and c.2548G>A CRB1 variants in a family with recessive adult-onset rod-cone dystrophy and further demonstrate the effects of these variants on pre-mRNA splicing. This data provide important insights into the pathogenic mechanisms associated with these variants.


Asunto(s)
Distrofias de Conos y Bastones/genética , Proteínas del Ojo/genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Empalme del ARN , Células Cultivadas , Distrofias de Conos y Bastones/patología , Proteínas del Ojo/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Organoides/citología , Organoides/metabolismo , Retina/citología , Retina/metabolismo
6.
Stem Cell Res ; 48: 101947, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32810830

RESUMEN

Autosomal recessive Stargardt disease is the most common cause of inherited retinal disease. In this report, we describe the generation and characterization of two human induced pluripotent stem cell (iPSC) lines from a patient with compound heterozygous mutations in the ABCA4 gene (c.[768G>T];[6079C>T]). Patient dermal fibroblasts were reprogrammed using episomal plasmids encoding OCT4, SOX2, KLF4, L-MYC, LIN28, mir302/367 microRNA and shRNA for P53. The clonal iPSC lines LEIi012-A and LEIi012-B were established. Both lines had a normal karyotype, displayed iPSC morphology, expressed pluripotency genes at similar levels to control iPSC and displayed trilineage differentiation potential during embryoid body differentiation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Células Madre Pluripotentes Inducidas , Enfermedad de Stargardt , Transportadoras de Casetes de Unión a ATP/genética , Diferenciación Celular , Línea Celular , Humanos , Factor 4 Similar a Kruppel , Mutación , Enfermedad de Stargardt/genética
7.
Glia ; 66(11): 2316-2323, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30098078

RESUMEN

Oxidative stress and oxidative DNA damage are early features of mild cognitive impairment and Alzheimer's disease (AD), occurring before the formation of classical AD neuropathology, and resulting from an imbalance between pro- and anti-oxidants. Astrocytes play a major neuroprotective role, producing high levels of anti-oxidants including metallothionein-I and -II (MT-I/II). In the present study we characterized the immunoreactive profile of MT-I/II in the temporal cortex of the Cognitive Function and Ageing Study (CFAS) aging population-representative neuropathology cohort, and examined H2 O2 -modulation of MT transcription by human astrocytes. MT-I/II is primarily expressed by astrocytes in the aging brain, but is also associated with pyramidal neurons in a small proportion of cases. Astrocyte expression of MT-I/II does not correlate with Alzheimer-type pathology (Aß plaques and neurofibrillary tangles) but does relate to astrocyte oxidative DNA damage (rs = .312, p = .006) and the astrocyte response to oxidative DNA damage in vivo (rs = .238, p = .04), and MT gene expression is significantly induced in human astrocytes response to oxidative stress in vitro (p = .01). In contrast, neuronal MT-I/II does not relate to oxidative DNA damage or the neuronal DNA damage response, but is significantly higher in cases with high levels of local tangle pathology (p = .007). As MT-I/II is neuroprotective against oxidative stress, modulation of MT-I/II expression is a potential therapeutic target to treat the onset and progression of cognitive impairment.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Encéfalo/metabolismo , Daño del ADN/fisiología , Metalotioneína/metabolismo , Anciano , Anciano de 80 o más Años , Astrocitos/efectos de los fármacos , Astrocitos/patología , Encéfalo/patología , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Metalotioneína/genética , Neuronas/metabolismo , Tauopatías/metabolismo , Tauopatías/patología , Factores de Tiempo
8.
Neuroscience ; 390: 46-59, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30056117

RESUMEN

The insulin/insulin-like growth factor 1 (IGF1) signaling pathways are implicated in longevity and in progression of Alzheimer's disease. Previously, we showed that insulin-like growth factor 1 receptor (IGF1R) and downstream signaling transcripts are reduced in astrocytes in human brain with progression of Alzheimer's neuropathology and developed a model of IGF1 signaling impairment in human astrocytes using an IGF1R-specific monoclonal antibody, MAB391. Here, we have established a novel human astrocyte-neuron co-culture system to determine whether loss of astrocytic IGF1R affects their support for neurons. Astrocyte-neuron co-cultures were developed using human primary astrocytes and differentiated Lund Human Mesencephalic Cells (LUHMES). Neurite outgrowth assays, performed to measure astrocytic support for neurons, showed astrocytes provided contact-mediated support for neurite outgrowth. Loss of IGF1R did not affect neurite outgrowth under control conditions but when challenged with hydrogen peroxide IGF1R-impaired astrocytes were less able to protect LUHMES. To determine how loss of IGF1R affects neuronal support MAB391-treated astrocytes were FACS sorted from GFP-LUHMES and their transcriptomic profile was investigated using microarrays. Changes in transcripts involved in astrocyte energy metabolism were identified, particularly NDUFA2 and NDUFB6, which are related to complex I assembly. Loss of complex I activity in MAB391-treated astrocytes validated these findings. In conclusion, reduced IGF1 signaling in astrocytes impairs their support for neurons under conditions of stress and this is associated with defects in the mitochondrial respiratory chain in astrocytes.


Asunto(s)
Astrocitos/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Neuronas/metabolismo , Receptores de Somatomedina/metabolismo , Anticuerpos Monoclonales/administración & dosificación , Técnicas de Cocultivo/métodos , Metabolismo Energético , Humanos , Proyección Neuronal , Estrés Oxidativo , Cultivo Primario de Células , Receptor IGF Tipo 1 , Receptores de Somatomedina/inmunología , Transcriptoma
9.
Eur J Neurosci ; 47(12): 1444-1456, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29738614

RESUMEN

Occludin is a component of tight junctions, which are essential structural components of the blood-brain barrier. However, occludin is expressed in cells without tight junctions, implying additional functions. We determined the expression and localisation of occludin in astrocytes in cell culture and in human brain tissue, and sought novel binding partners using a proteomic approach. Expression was investigated by immunocytochemistry and immunoblotting in the 1321N1 astrocytoma cell line and ScienCell human primary astrocytes, and by immunohistochemistry in human autopsy brain tissue. Recombinant N- and C-terminal occludin was used to pull-down proteins from 1321N1 cell lysates and protein-binding partners identified by mass spectrometry analysis. Occludin was expressed in both the cytoplasm and nucleus of astrocytes in vitro and in vivo. Mass spectrometry identified binding to nuclear and cytoplasmic proteins, particularly those related to RNA metabolism and nuclear function. Occludin is expressed in several subcellular compartments of brain cell-types that do not form tight junctions and the expression patterns in cell culture reflect those in human brain tissue, indicating they are suitable model systems. Proteomic analysis suggests that occludin has novel functions in neuroepithelial cells that are unrelated to tight junction formation. Further research will establish the roles of these functions in both cellular physiology and in disease states.


Asunto(s)
Astrocitos/metabolismo , Astrocitoma/metabolismo , Encéfalo/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Endoteliales/metabolismo , Ocludina/metabolismo , ARN/metabolismo , Línea Celular Tumoral , Células Cultivadas , Técnicas Citológicas , Feto , Humanos , Espectrometría de Masas , Proteómica
10.
Tissue Eng Part C Methods ; 22(12): 1108-1117, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27846777

RESUMEN

Tissue-engineered oral mucosal equivalents (OME) are being increasingly used to measure toxicity, drug delivery, and to model oral diseases. Current OME mainly comprise normal oral keratinocytes (NOK) cultured on top of a normal oral fibroblasts-containing matrix. However, the commercial supply of NOK is limited, restricting widespread use of these mucosal models. In addition, NOK suffer from poor longevity and donor-to-donor variability. Therefore, we constructed, characterized, and tested the functionality of OME based on commercial TERT2-immortalized oral keratinocytes (FNB6) to produce a more readily available alternative to NOK-based OME. FNB6 OME cultured at an air-to-liquid interface for 14 days exhibited expression of differentiation markers cytokeratin 13 in the suprabasal layers and cytokeratin 14 in basal layer of the epithelium. Proliferating cells were restricted to the basal epithelium, and there was immuno-positive expression of E-cadherin confirming the presence of established cell-to-cell contacts. The histology and expression of these structural markers paralleled those observed in the normal oral mucosa and NOK-based models. On stimulation with TNFα and IL-1, FNB6 OME displayed a similar global gene expression profile to NOK-based OME, with increased expression of many common pro-inflammatory molecules such as chemokines (CXCL8), cytokines (IL-6), and adhesion molecules (ICAM-1) when analyzed by gene array and quantitative PCR. Similarly, pathway analysis showed that both FNB6 and NOK models initiated similar intracellular signaling on stimulation. Gene expression in FNB6 OME was more consistent than NOK-based OME that suffered from donor variation in response to stimuli. Mucosal equivalents based on immortalized FNB6 cells are accessible, reproducible and will provide an alternative animal experimental system for studying mucosal drug delivery systems, host-pathogen interactions, and drug-induced toxicity.


Asunto(s)
Biomarcadores/metabolismo , Fibroblastos/citología , Queratinocitos/citología , Mucosa Bucal/fisiología , Cadherinas/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Técnicas In Vitro , Queratinocitos/metabolismo , Queratinas/metabolismo , Mucosa Bucal/citología
11.
J Med Chem ; 58(23): 9309-33, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26580420

RESUMEN

A number of indole-3-glyoxylamides have previously been reported as tubulin polymerization inhibitors, although none has yet been successfully developed clinically. We report here a new series of related compounds, modified according to a strategy of reducing aromatic ring count and introducing a greater degree of saturation, which retain potent tubulin polymerization activity but with a distinct SAR from previously documented libraries. A subset of active compounds from the reported series is shown to interact with tubulin at the colchicine binding site, disrupt the cellular microtubule network, and exert a cytotoxic effect against multiple cancer cell lines. Two compounds demonstrated significant tumor growth inhibition in a mouse xenograft model of head and neck cancer, a type of the disease which often proves resistant to chemotherapy, supporting further development of the current series as potential new therapeutics.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Indoles/química , Indoles/uso terapéutico , Moduladores de Tubulina/química , Moduladores de Tubulina/uso terapéutico , Animales , Antineoplásicos/farmacocinética , Células CACO-2 , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Xenoinjertos , Humanos , Indoles/farmacocinética , Masculino , Ratones , Ratones Desnudos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Microtúbulos/patología , Trasplante de Neoplasias , Relación Estructura-Actividad , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/ultraestructura , Moduladores de Tubulina/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA