Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 149(4): 899-911, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22579290

RESUMEN

Fragile X syndrome (FXS), the leading monogenic cause of intellectual disability and autism, results from loss of function of the RNA-binding protein FMRP. Here, we show that FMRP regulates translation of neuronal nitric oxide synthase 1 (NOS1) in the developing human neocortex. Whereas NOS1 mRNA is widely expressed, NOS1 protein is transiently coexpressed with FMRP during early synaptogenesis in layer- and region-specific pyramidal neurons. These include midfetal layer 5 subcortically projecting neurons arranged into alternating columns in the prospective Broca's area and orofacial motor cortex. Human NOS1 translation is activated by FMRP via interactions with coding region binding motifs absent from mouse Nos1 mRNA, which is expressed in mouse pyramidal neurons, but not efficiently translated. Correspondingly, neocortical NOS1 protein levels are severely reduced in developing human FXS cases, but not FMRP-deficient mice. Thus, alterations in FMRP posttranscriptional regulation of NOS1 in developing neocortical circuits may contribute to cognitive dysfunction in FXS.


Asunto(s)
Corteza Cerebral/embriología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/embriología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Corteza Cerebral/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/fisiopatología , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Neurogénesis , Células Piramidales/metabolismo , Procesamiento Postranscripcional del ARN , Especificidad de la Especie
2.
Hum Mol Genet ; 30(17): 1632-1648, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34077515

RESUMEN

Fragile X-associated tremor and ataxia syndrome (FXTAS) is a late-onset, progressive neurodegenerative disorder characterized by tremors, ataxia and neuropsychological problems. This disease is quite common in the general population with approximately 20 million carriers worldwide. The risk of developing FXTAS increases dramatically with age, with about 45% of male carriers over the age of 50 being affected. FXTAS is caused by a CGG-repeat expansion (CGGexp) in the fragile X mental retardation 1 (FMR1) gene. CGGexp RNA is translated into the FMRpolyG protein by a mechanism called RAN translation. Although both gene and pathogenic trigger are known, no therapeutic interventions are available at this moment. Here, we present, for the first time, primary hippocampal neurons derived from the ubiquitous inducible mouse model which is used as a screening tool for targeted interventions. A promising candidate is the repeat binding, RAN translation blocking, small molecule 1a. Small molecule 1a shields the disease-causing CGGexp from being translated into the toxic FMRpolyG protein. Primary hippocampal neurons formed FMRpolyG-positive inclusions, and upon treatment with 1a, the numbers of FMRpolyG-positive inclusions are reduced. We also describe for the first time the formation of FMRpolyG-positive inclusions in the liver of this mouse model. Treatment with 1a reduced the insoluble FMRpolyG protein fraction in the liver but not the number of inclusions. Moreover, 1a treatment had a reducing effect on the number of Rad23b-positive inclusions and insoluble Rad23b protein levels. These data suggest that targeted small molecule therapy is effective in an FXTAS mouse model and has the potential to treat CGGexp-mediated diseases, including FXTAS.


Asunto(s)
Ataxia/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Temblor/genética , Animales , Ataxia/fisiopatología , Comunicación Celular , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/fisiopatología , Humanos , Masculino , Ratones , Neuronas/metabolismo , Temblor/fisiopatología , Expansión de Repetición de Trinucleótido
3.
Acta Neuropathol ; 146(2): 353-368, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37119330

RESUMEN

Hereditary spastic paraplegias (HSP) are rare, inherited neurodegenerative or neurodevelopmental disorders that mainly present with lower limb spasticity and muscle weakness due to motor neuron dysfunction. Whole genome sequencing identified bi-allelic truncating variants in AMFR, encoding a RING-H2 finger E3 ubiquitin ligase anchored at the membrane of the endoplasmic reticulum (ER), in two previously genetically unexplained HSP-affected siblings. Subsequently, international collaboration recognized additional HSP-affected individuals with similar bi-allelic truncating AMFR variants, resulting in a cohort of 20 individuals from 8 unrelated, consanguineous families. Variants segregated with a phenotype of mainly pure but also complex HSP consisting of global developmental delay, mild intellectual disability, motor dysfunction, and progressive spasticity. Patient-derived fibroblasts, neural stem cells (NSCs), and in vivo zebrafish modeling were used to investigate pathomechanisms, including initial preclinical therapy assessment. The absence of AMFR disturbs lipid homeostasis, causing lipid droplet accumulation in NSCs and patient-derived fibroblasts which is rescued upon AMFR re-expression. Electron microscopy indicates ER morphology alterations in the absence of AMFR. Similar findings are seen in amfra-/- zebrafish larvae, in addition to altered touch-evoked escape response and defects in motor neuron branching, phenocopying the HSP observed in patients. Interestingly, administration of FDA-approved statins improves touch-evoked escape response and motor neuron branching defects in amfra-/- zebrafish larvae, suggesting potential therapeutic implications. Our genetic and functional studies identify bi-allelic truncating variants in AMFR as a cause of a novel autosomal recessive HSP by altering lipid metabolism, which may potentially be therapeutically modulated using precision medicine with statins.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Paraplejía Espástica Hereditaria , Animales , Humanos , Paraplejía Espástica Hereditaria/tratamiento farmacológico , Paraplejía Espástica Hereditaria/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Pez Cebra , Mutación , Neuronas Motoras , Receptores del Factor Autocrino de Motilidad/genética
4.
Exp Eye Res ; 230: 109437, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36924981

RESUMEN

Myopia, or nearsightedness, is the most common type of refractive error and is characterized by a mismatch between the optical power and ocular axial length. Light, and more specifically the spectral composition of light, has been known to influence myopic axial growth. In this pilot study, we exposed zebrafish to illuminations that vary in spectral composition and screened for changes in axial length. The illumination spectra included narrow band ultra-violet A (UVA) (peak wavelength 369 nm), violet (425 nm), cyan (483 nm), green/yellow (557 nm), and red (633 nm) light, as well as broad band white light (2700 K and 6500 K), dim white light and broad spectrum (day) light. We found that rearing zebrafish in cyan or red light leads to a reduction of the ocular axial length. The results of this pilot study may contribute to new perspectives on the role of light and lighting as an intervention strategy for myopia control.


Asunto(s)
Miopía , Errores de Refracción , Animales , Pez Cebra , Proyectos Piloto , Ojo , Miopía/prevención & control , Refracción Ocular , Longitud Axial del Ojo
5.
J Am Soc Nephrol ; 32(5): 1210-1226, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33782168

RESUMEN

BACKGROUND: Urinary extracellular vesicles (uEVs) are a promising source for biomarker discovery, but optimal approaches for normalization, quantification, and characterization in spot urines are unclear. METHODS: Urine samples were analyzed in a water-loading study, from healthy subjects and patients with kidney disease. Urine particles were quantified in whole urine using nanoparticle tracking analysis (NTA), time-resolved fluorescence immunoassay (TR-FIA), and EVQuant, a novel method quantifying particles via gel immobilization. RESULTS: Urine particle and creatinine concentrations were highly correlated in the water-loading study (R2 0.96) and in random spot urines from healthy subjects (R2 0.47-0.95) and patients (R2 0.41-0.81). Water loading reduced aquaporin-2 but increased Tamm-Horsfall protein (THP) and particle detection by NTA. This finding was attributed to hypotonicity increasing uEV size (more EVs reach the NTA size detection limit) and reducing THP polymerization. Adding THP to urine also significantly increased particle count by NTA. In both fluorescence NTA and EVQuant, adding 0.01% SDS maintained uEV integrity and increased aquaporin-2 detection. Comparison of intracellular- and extracellular-epitope antibodies suggested the presence of reverse topology uEVs. The exosome markers CD9 and CD63 colocalized and immunoprecipitated selectively with distal nephron markers. Conclusions uEV concentration is highly correlated with urine creatinine, potentially replacing the need for uEV quantification to normalize spot urines. Additional findings relevant for future uEV studies in whole urine include the interference of THP with NTA, excretion of larger uEVs in dilute urine, the ability to use detergent to increase intracellular-epitope recognition in uEVs, and CD9 or CD63 capture of nephron segment-specific EVs.


Asunto(s)
Vesículas Extracelulares/metabolismo , Enfermedades Renales/diagnóstico , Enfermedades Renales/orina , Adulto , Biomarcadores/orina , Estudios de Casos y Controles , Creatinina/orina , Femenino , Humanos , Masculino , Reproducibilidad de los Resultados , Urinálisis
6.
Hum Mol Genet ; 27(12): 2039-2051, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29590342

RESUMEN

Fragile X syndrome (FXS) is a monogenic form of intellectual disability and autism spectrum disorder caused by the absence of the fragile X mental retardation protein (FMRP). In biological models for the disease, this leads to upregulated mRNA translation and as a consequence, deficits in synaptic architecture and plasticity. Preclinical studies revealed that pharmacological interventions restore those deficits, which are thought to mediate the FXS cognitive and behavioral symptoms. Here, we characterized the de novo rate of protein synthesis in patients with FXS and their relationship with clinical severity. We measured the rate of protein synthesis in fibroblasts derived from 32 individuals with FXS and from 17 controls as well as in fibroblasts and primary neurons of 27 Fmr1 KO mice and 20 controls. Here, we show that levels of protein synthesis are increased in fibroblasts of individuals with FXS and Fmr1 KO mice. However, this cellular phenotype displays a broad distribution and a proportion of fragile X individuals and Fmr1 KO mice do not show increased levels of protein synthesis, having measures in the normal range. Because the same Fmr1 KO animal measures in fibroblasts predict those in neurons we suggest the validity of this peripheral biomarker. Our study offers a potential explanation for the comprehensive drug development program undertaken thus far yielding negative results and suggests that a significant proportion, but not all individuals with FXS, may benefit from the reduction of excessive levels of protein synthesis.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Adolescente , Adulto , Anciano , Animales , Trastorno del Espectro Autista/fisiopatología , Niño , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/fisiopatología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología , Adulto Joven
7.
Hum Mol Genet ; 26(11): 2133-2145, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28369393

RESUMEN

A CGG-repeat expansion in the premutation range in the Fragile X mental retardation 1 gene (FMR1) has been identified as the genetic cause of Fragile X-associated tremor/ataxia syndrome (FXTAS), a late-onset neurodegenerative disorder that manifests with action tremor, gait ataxia and cognitive impairments. In this study, we used a bigenic mouse model, in which expression of a 90CGG premutation tract is activated in neural cells upon doxycycline administration-P90CGG mouse model. We, here, demonstrate the behavioural manifestation of clinically relevant features of FXTAS patients and premutation carrier individuals in this inducible mouse model. P90CGG mice display heightened anxiety, deficits in motor coordination and impaired gait and represent the first FXTAS model that exhibits an ataxia phenotype as observed in patients. The behavioural phenotype is accompanied by the formation of ubiquitin/FMRpolyglycine-positive intranuclear inclusions, as another hallmark of FXTAS, in the cerebellum, hippocampus and amygdala. Strikingly, upon cessation of transgene induction the anxiety phenotype of mice recovers along with a reduction of intranuclear inclusions in dentate gyrus and amygdala. In contrast, motor function deteriorates further and no reduction in intranuclear inclusions can be observed in the cerebellum. Our data thus demonstrate that expression of a 90CGG premutation expansion outside of the FMR1 context is sufficient to evoke an FXTAS-like behavioural phenotype. Brain region-specific neuropathology and (partial) behavioural reversibility make the inducible P90CGG a valuable mouse model for testing pathogenic mechanisms and therapeutic intervention methods.


Asunto(s)
Ataxia/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Temblor/genética , Animales , Ansiedad/genética , Ansiedad/metabolismo , Ataxia/metabolismo , Encéfalo/patología , Ataxia Cerebelosa/genética , Trastornos del Conocimiento/genética , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Marcha , Ataxia de la Marcha/genética , Ataxia de la Marcha/metabolismo , Cuerpos de Inclusión Intranucleares/genética , Ratones , Trastornos del Movimiento/genética , Neuronas/patología , Temblor/metabolismo , Expansión de Repetición de Trinucleótido/genética
8.
Hum Mol Genet ; 24(17): 4948-57, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26060190

RESUMEN

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder affecting carriers of the fragile X-premutation, who have an expanded CGG repeat in the 5'-UTR of the FMR1 gene. FXTAS is characterized by progressive development of intention tremor, ataxia, parkinsonism and neuropsychological problems. The disease is thought to be caused by a toxic RNA gain-of-function mechanism, and the major hallmark of the disease is ubiquitin-positive intranuclear inclusions in neurons and astrocytes. We have developed a new transgenic mouse model in which we can induce expression of an expanded repeat in the brain upon doxycycline (dox) exposure (i.e. Tet-On mice). This Tet-On model makes use of the PrP-rtTA driver and allows us to study disease progression and possibilities of reversibility. In these mice, 8 weeks of dox exposure was sufficient to induce the formation of ubiquitin-positive intranuclear inclusions, which also stain positive for the RAN translation product FMRpolyG. Formation of these inclusions is reversible after stopping expression of the expanded CGG RNA at an early developmental stage. Furthermore, we observed a deficit in the compensatory eye movements of mice with inclusions, a functional phenotype that could be reduced by stopping expression of the expanded CGG RNA early in the disease development. Taken together, this study shows, for the first time, the potential of disease reversibility and suggests that early intervention might be beneficial for FXTAS patients.


Asunto(s)
Ataxia/genética , Ataxia/fisiopatología , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/fisiopatología , Temblor/genética , Temblor/fisiopatología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Movimientos Oculares/genética , Expresión Génica , Genes Reporteros , Humanos , Cuerpos de Inclusión Intranucleares/patología , Ratones , Ratones Transgénicos , Péptidos/metabolismo , Unión Proteica , Transporte de Proteínas , Expansión de Repetición de Trinucleótido , Ubiquitina/metabolismo
9.
Hum Mol Genet ; 23(5): 1320-32, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24150847

RESUMEN

Primary open-angle glaucoma (POAG) is a hereditary neurodegenerative disease, characterized by optic nerve changes including increased excavation, notching and optic disc hemorrhages. The excavation can be described by the vertical cup-disc ratio (VCDR). Previously, genome-wide significant evidence for the association of rs10483727 in SIX1-SIX6 locus with VCDR and subsequent POAG was found. Using 1000 genomes-based imputation of four independent population-based cohorts in the Netherlands, we identified a missense variant rs33912345 (His141Asn) in SIX6 associated with VCDR (Pmeta = 7.74 × 10(-7), n = 11 473) and POAG (Pmeta = 6.09 × 10(-3), n = 292). Exome sequencing analysis revealed another missense variant rs146737847 (Glu129Lys) also in SIX6 associated with VCDR (P = 5.09 × 10(-3), n = 1208). These two findings point to SIX6 as the responsible gene for the previously reported association signal. Functional characterization of SIX6 in zebrafish revealed that knockdown of six6b led to a small eye phenotype. Histological analysis showed retinal lamination, implying an apparent normal development of the eye, but an underdeveloped lens, and reduced optic nerve diameter. Expression analysis of morphants at 3 dpf showed a 5.5-fold up-regulation of cdkn2b, a cyclin-dependent kinase inhibitor, involved in cell cycle regulation and previously associated with VCDR and POAG in genome-wide association studies (GWASs). Since both six6b and cdkn2b play a key role in cell proliferation, we assessed the proliferative activity in the eye of morphants and found an alteration in the proliferative pattern of retinal cells. Our findings in humans and zebrafish suggest a functional involvement of six6b in early eye development, and open new insights into the genetic architecture of POAG.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Homeodominio/genética , Degeneración Nerviosa/genética , Nervio Óptico/metabolismo , Nervio Óptico/patología , Transactivadores/genética , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Proliferación Celular , Mapeo Cromosómico , Exoma , Ojo/embriología , Ojo/metabolismo , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/metabolismo , Glaucoma de Ángulo Abierto/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de Homeodominio/metabolismo , Humanos , Persona de Mediana Edad , Modelos Biológicos , Organogénesis/genética , Fenotipo , Sitios de Carácter Cuantitativo , Transactivadores/metabolismo , Adulto Joven , Pez Cebra
10.
Proc Natl Acad Sci U S A ; 110(32): 13109-13, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23878230

RESUMEN

Recent evidence indicates there is a role for small membrane vesicles, including exosomes, as vehicles for intercellular communication. Exosomes secreted by most cell types can mediate transfer of proteins, mRNAs, and microRNAs, but their role in the transmission of infectious agents is less established. Recent studies have shown that hepatocyte-derived exosomes containing hepatitis C virus (HCV) RNA can activate innate immune cells, but the role of exosomes in the transmission of HCV between hepatocytes remains unknown. In this study, we investigated whether exosomes transfer HCV in the presence of neutralizing antibodies. Purified exosomes isolated from HCV-infected human hepatoma Huh7.5.1 cells were shown to contain full-length viral RNA, viral protein, and particles, as determined by RT-PCR, mass spectrometry, and transmission electron microscopy. Exosomes from HCV-infected cells were capable of transmitting infection to naive human hepatoma Huh7.5.1 cells and establishing a productive infection. Even with subgenomic replicons, lacking structural viral proteins, exosome-mediated transmission of HCV RNA was observed. Treatment with patient-derived IgGs showed a variable degree of neutralization of exosome-mediated infection compared with free virus. In conclusion, this study showed that hepatic exosomes can transmit productive HCV infection in vitro and are partially resistant to antibody neutralization. This discovery sheds light on neutralizing antibodies resistant to HCV transmission by exosomes as a potential immune evasion mechanism.


Asunto(s)
Exosomas/virología , Hepacivirus/genética , ARN Viral/genética , Virión/genética , Anticuerpos Neutralizantes/inmunología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Claudina-1/inmunología , Claudina-1/metabolismo , Exosomas/metabolismo , Exosomas/ultraestructura , Hepacivirus/inmunología , Hepacivirus/fisiología , Hepatitis C/inmunología , Hepatitis C/virología , Interacciones Huésped-Patógeno , Humanos , Inmunoglobulina G/inmunología , Espectrometría de Masas , Microscopía Confocal , Microscopía Electrónica de Transmisión , ARN Viral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptores Depuradores de Clase B/inmunología , Receptores Depuradores de Clase B/metabolismo , Tetraspanina 28/inmunología , Tetraspanina 28/metabolismo , Virión/fisiología , Virión/ultraestructura
11.
Hum Mol Genet ; 22(10): 1971-82, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23390134

RESUMEN

While FMR1 is silenced in Fragile X syndrome (FXS) patients carrying the full mutation, its expression is elevated (2-8 fold) in premutated individuals. These people may develop the Fragile X-associated Tremor/Ataxia syndrome (FXTAS), a late onset neurodegenerative disorder characterized by ataxia and parkinsonism. In addition, people carrying the premutation can be affected by a set of neurological and behavioral disorders during young age. Problems of memory have been detected in these patients as well as in the mouse models for FXTAS. To date little is known concerning the metabolism of FMR1 mRNA, notwithstanding the importance of the finely tuned regulation of the expression of this gene. In the present study, we identified three microRNAs that specifically target the 3' UTR of FMR1 and can modulate its expression throughout the brain particularly at the synapse where their expression is very high. The expression level of miR-221 is reduced in synaptosomal preparations of young FXTAS mice suggesting a general deregulation of transcripts located at the synapse of these mice. By transcriptome analysis, we show here a robust deregulation of the expression levels of genes involved in learning, memory and autistic behavior, Parkinson disease and neurodegeneration. These findings suggest the presence of a synaptopathy in these animals. Interestingly, many of those deregulated mRNAs are target of the same microRNAs that modulate the expression of FMR1 at the synapse.


Asunto(s)
Regiones no Traducidas 3' , Ataxia/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/metabolismo , MicroARNs/metabolismo , Sinapsis/metabolismo , Temblor/metabolismo , Animales , Ataxia/genética , Ataxia/patología , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Células HeLa , Humanos , Ratones , MicroARNs/genética , Células 3T3 NIH , Sinapsis/genética , Sinapsis/patología , Temblor/genética , Temblor/patología
13.
Hum Genet ; 134(11-12): 1211-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26385552

RESUMEN

Electrocardiogram (ECG) measurements are a powerful tool for evaluating cardiac function and are widely used for the diagnosis and prediction of a variety of conditions, including myocardial infarction, cardiac arrhythmias, and sudden cardiac death. Recently, genome-wide association studies (GWASs) identified a large number of genes related to ECG parameter variability, specifically for the QT, QRS, and PR intervals. The aims of this study were to establish the heritability of ECG traits, including indices of left ventricular hypertrophy, and to directly assess the proportion of those heritabilities explained by GWAS variants. These analyses were conducted in a large, Dutch family-based cohort study, the Erasmus Rucphen Family study using variance component methods implemented in the SOLAR (Sequential Oligogenic Linkage Analysis Routines) software package. Heritability estimates ranged from 34% for QRS and Cornell voltage product to 49% for 12-lead sum. Trait-specific GWAS findings for each trait explained a fraction of their heritability (17% for QRS, 4% for QT, 2% for PR, 3% for Sokolow-Lyon index, and 4% for 12-lead sum). The inclusion of all ECG-associated single nucleotide polymorphisms explained an additional 6% of the heritability of PR. In conclusion, this study shows that, although GWAS explain a portion of ECG trait variability, a large amount of heritability remains to be explained. In addition, larger GWAS for PR are likely to detect loci already identified, particularly those observed for QRS and 12-lead sum.


Asunto(s)
Frecuencia Cardíaca/genética , Corazón/fisiología , Carácter Cuantitativo Heredable , Adulto , Estudios de Cohortes , Electrocardiografía , Femenino , Ligamiento Genético , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Hipertrofia Ventricular Izquierda/genética , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple
14.
Am J Hum Genet ; 90(3): 467-77, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22341971

RESUMEN

Manganese is essential for several metabolic pathways but becomes toxic in excessive amounts. Manganese levels in the body are therefore tightly regulated, but the responsible protein(s) remain incompletely known. We studied two consanguineous families with neurologic disorders including juvenile-onset dystonia, adult-onset parkinsonism, severe hypermanganesemia, polycythemia, and chronic hepatic disease, including steatosis and cirrhosis. We localized the genetic defect by homozygosity mapping and then identified two different homozygous frameshift SLC30A10 mutations, segregating with disease. SLC30A10 is highly expressed in the liver and brain, including in the basal ganglia. Its encoded protein belongs to a large family of membrane transporters, mediating the efflux of divalent cations from the cytosol. We show the localization of SLC30A10 in normal human liver and nervous system, and its depletion in liver from one affected individual. Our in silico analyses suggest that SLC30A10 possesses substrate specificity different from its closest (zinc-transporting) homologs. We also show that the expression of SLC30A10 and the levels of the encoded protein are markedly induced by manganese in vitro. The phenotype associated with SLC30A10 mutations is broad, including neurologic, hepatic, and hematologic disturbances. Intrafamilial phenotypic variability is also present. Chelation therapy can normalize the manganesemia, leading to marked clinical improvements. In conclusion, we show that SLC30A10 mutations cause a treatable recessive disease with pleomorphic phenotype, and provide compelling evidence that SLC30A10 plays a pivotal role in manganese transport. This work has broad implications for understanding of the manganese biology and pathophysiology in multiple human organs.


Asunto(s)
Proteínas de Transporte de Catión/genética , Intoxicación por Manganeso/genética , Proteínas de Transporte de Membrana/genética , Enfermedades Metabólicas/genética , Trastornos Parkinsonianos/genética , Anciano , Secuencia de Aminoácidos , Encéfalo/metabolismo , Proteínas de Transporte de Catión/metabolismo , Mapeo Cromosómico/métodos , Femenino , Mutación del Sistema de Lectura/genética , Genes Recesivos , Predisposición Genética a la Enfermedad , Células Hep G2 , Homocigoto , Humanos , Inmunohistoquímica/métodos , Hígado/metabolismo , Masculino , Manganeso/metabolismo , Intoxicación por Manganeso/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Enfermedades Metabólicas/metabolismo , Persona de Mediana Edad , Datos de Secuencia Molecular , Fenotipo , Alineación de Secuencia/métodos , Células Tumorales Cultivadas , Transportador 8 de Zinc
15.
Brain ; 137(Pt 5): 1361-73, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24722252

RESUMEN

Pathological accumulation of intermediate filaments can be observed in neurodegenerative disorders, such as Alzheimer's disease, frontotemporal dementia and Parkinson's disease, and is also characteristic of neuronal intermediate filament inclusion disease. Intermediate filaments type IV include three neurofilament proteins (light, medium and heavy molecular weight neurofilament subunits) and α-internexin. The phosphorylation of intermediate filament proteins contributes to axonal growth, and is regulated by protein kinase A. Here we describe a family with a novel late-onset neurodegenerative disorder presenting with dementia and/or parkinsonism in 12 affected individuals. The disorder is characterized by a unique neuropathological phenotype displaying abundant neuronal inclusions by haematoxylin and eosin staining throughout the brain with immunoreactivity for intermediate filaments. Combining linkage analysis, exome sequencing and proteomics analysis, we identified a heterozygous c.149T>G (p.Leu50Arg) missense mutation in the gene encoding the protein kinase A type I-beta regulatory subunit (PRKAR1B). The pathogenicity of the mutation is supported by segregation in the family, absence in variant databases, and the specific accumulation of PRKAR1B in the inclusions in our cases associated with a specific biochemical pattern of PRKAR1B. Screening of PRKAR1B in 138 patients with Parkinson's disease and 56 patients with frontotemporal dementia did not identify additional novel pathogenic mutations. Our findings link a pathogenic PRKAR1B mutation to a novel hereditary neurodegenerative disorder and suggest an altered protein kinase A function through a reduced binding of the regulatory subunit to the A-kinase anchoring protein and the catalytic subunit of protein kinase A, which might result in subcellular dislocalization of the catalytic subunit and hyperphosphorylation of intermediate filaments.


Asunto(s)
Subunidad RIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Polimorfismo de Nucleótido Simple/genética , Anciano , Péptidos beta-Amiloides/metabolismo , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/química , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Tomografía con Microscopio Electrónico , Salud de la Familia , Femenino , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Lóbulo Frontal/ultraestructura , Estudios de Asociación Genética , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Proteínas del Tejido Nervioso/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
16.
Nat Genet ; 38(11): 1348-54, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17033623

RESUMEN

The spatial organization of DNA in the cell nucleus is an emerging key contributor to genomic function. We developed 4C technology (chromosome conformation capture (3C)-on-chip), which allows for an unbiased genome-wide search for DNA loci that contact a given locus in the nuclear space. We demonstrate here that active and inactive genes are engaged in many long-range intrachromosomal interactions and can also form interchromosomal contacts. The active beta-globin locus in fetal liver preferentially contacts transcribed, but not necessarily tissue-specific, loci elsewhere on chromosome 7, whereas the inactive locus in fetal brain contacts different transcriptionally silent loci. A housekeeping gene in a gene-dense region on chromosome 8 forms long-range contacts predominantly with other active gene clusters, both in cis and in trans, and many of these intra- and interchromosomal interactions are conserved between the tissues analyzed. Our data demonstrate that chromosomes fold into areas of active chromatin and areas of inactive chromatin and establish 4C technology as a powerful tool to study nuclear architecture.


Asunto(s)
Núcleo Celular/química , Ensamble y Desensamble de Cromatina , Cromatina/química , Cromosomas de los Mamíferos/química , Conformación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Encéfalo/citología , Encéfalo/embriología , Mapeo Cromosómico/métodos , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Globinas/genética , Hibridación Fluorescente in Situ/métodos , Hígado/citología , Hígado/embriología , Ratones , Modelos Biológicos
17.
Neurobiol Dis ; 65: 43-54, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24418349

RESUMEN

Male premutation carriers presenting between 55 and 200 CGG repeats in the Fragile-X-associated (FMR1) gene are at risk of developing Fragile X Tremor/Ataxia Syndrome (FXTAS), and females undergo Premature Ovarian Failure (POF1). Here, we have evaluated gene expression profiles from blood in male FMR1 premutation carriers and detected a strong deregulation of genes enriched in FXTAS relevant biological pathways, including inflammation, neuronal homeostasis and viability. Gene expression profiling distinguished between control individuals, carriers with FXTAS and carriers without FXTAS, with levels of expanded FMR1 mRNA being increased in FXTAS patients. In vitro studies in a neuronal cell model indicate that expression levels of expanded FMR1 5'-UTR are relevant in modulating the transcriptome. Thus, perturbations of the transcriptome may be an interplay between the CGG expansion size and FMR1 expression levels. Several deregulated genes (DFFA, BCL2L11, BCL2L1, APP, SOD1, RNF10, HDAC5, KCNC3, ATXN7, ATXN3 and EAP1) were validated in brain samples of a FXTAS mouse model. Downregulation of EAP1, a gene involved in the female reproductive system physiology, was confirmed in female carriers. Decreased levels were detected in female carriers with POF1 compared to those without POF1, suggesting that EAP1 levels contribute to ovarian insufficiency. In summary, gene expression profiling in blood has uncovered mechanisms that may underlie different pathological aspects of the premutation. A better understanding of the transcriptome dynamics in relation with expanded FMR1 mRNA expression levels and CGG expansion size may provide mechanistic insights into the disease process and a more accurate FXTAS diagnosis to the myriad of phenotypes associated with the premutation.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Regulación de la Expresión Génica/genética , Infertilidad/genética , Enfermedades Neurodegenerativas/sangre , Expansión de Repetición de Trinucleótido/genética , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Análisis por Micromatrices , Proteínas de Microfilamentos , Persona de Mediana Edad , Neuroblastoma/patología , Enfermedades Neurodegenerativas/genética , Fenotipo , Proteínas/genética , Proteínas/metabolismo , Securina/genética , Securina/metabolismo
18.
EMBO J ; 29(7): 1248-61, 2010 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-20186122

RESUMEN

Fragile X-associated Tremor/Ataxia Syndrome (FXTAS) is a neurodegenerative disorder caused by expansion of 55-200 CGG repeats in the 5'-UTR of the FMR1 gene. FXTAS is characterized by action tremor, gait ataxia and impaired executive cognitive functioning. It has been proposed that FXTAS is caused by titration of RNA-binding proteins by the expanded CGG repeats. Sam68 is an RNA-binding protein involved in alternative splicing regulation and its ablation in mouse leads to motor coordination defects. Here, we report that mRNAs containing expanded CGG repeats form large and dynamic intranuclear RNA aggregates that recruit several RNA-binding proteins sequentially, first Sam68, then hnRNP-G and MBNL1. Importantly, Sam68 is sequestered by expanded CGG repeats and thereby loses its splicing-regulatory function. Consequently, Sam68-responsive splicing is altered in FXTAS patients. Finally, we found that regulation of Sam68 tyrosine phosphorylation modulates its localization within CGG aggregates and that tautomycin prevents both Sam68 and CGG RNA aggregate formation. Overall, these data support an RNA gain-of-function mechanism for FXTAS neuropathology, and suggest possible target routes for treatment options.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo , Proteínas de Unión al ADN/metabolismo , Síndrome del Cromosoma X Frágil/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/análisis , Animales , Ataxia/genética , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Proteínas de Unión al ADN/análisis , Inhibidores Enzimáticos/farmacología , Síndrome del Cromosoma X Frágil/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Ratones , Fosforilación , Piranos/farmacología , ARN Mensajero/química , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/análisis , Secuencias Repetitivas de Ácidos Nucleicos , Compuestos de Espiro/farmacología , Tirosina/metabolismo
19.
Neurobiol Learn Mem ; 109: 160-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24462720

RESUMEN

Environmental enrichment results in increased levels of Fmrp in brain and increased dendritic complexity. The present experiment evaluated activity-dependent increases in Fmrp levels in the motor cortex in response to training on a skilled forelimb reaching task in the CGG KI mouse model of the fragile X premutation. Fmrp, Arc, and c-Fos protein levels were quantified by Western blot in the contralateral motor cortex of mice following training to reach for sucrose pellets with a non-preferred paw and compared to levels in the ipsilateral motor cortex. After training, all mice showed increases in Fmrp, Arc, and c-Fos protein levels in the contralateral compared to the ipsilateral hemisphere; however, the increase in CGG KI mice was less than wildtype mice. Increases in Fmrp and Arc proteins scaled with learning, whereas this relationship was not observed with the c-Fos levels. These data suggest the possibility that reduced levels of activity-dependent proteins associated with synaptic plasticity such as Fmrp and Arc may contribute to the neurocognitive phenotype reported in the CGG KI mice and the fragile X premutation.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Actividad Motora/fisiología , Corteza Motora/metabolismo , Animales , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Técnicas de Sustitución del Gen , Masculino , Ratones , Mutación
20.
Circ Res ; 110(12): 1564-74, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22550138

RESUMEN

RATIONALE: Congenital heart malformations are a major cause of morbidity and mortality, especially in young children. Failure to establish normal left-right (L-R) asymmetry often results in cardiovascular malformations and other laterality defects of visceral organs. OBJECTIVE: To identify genetic mutations causing cardiac laterality defects. METHODS AND RESULTS: We performed a genome-wide linkage analysis in patients with cardiac laterality defects from a consanguineous family. The patients had combinations of defects that included dextrocardia, transposition of great arteries, double-outlet right ventricle, atrioventricular septal defects, and caval vein abnormalities. Sequencing of positional candidate genes identified mutations in NPHP4. We performed mutation analysis of NPHP4 in 146 unrelated patients with similar cardiac laterality defects. Forty-one percent of these patients also had laterality defects of the abdominal organs. We identified 8 additional missense variants that were absent or very rare in control subjects. To study the role of nphp4 in establishing L-R asymmetry, we used antisense morpholinos to knockdown nphp4 expression in zebrafish. Depletion of nphp4 disrupted L-R patterning as well as cardiac and gut laterality. Cardiac laterality defects were partially rescued by human NPHP4 mRNA, whereas mutant NPHP4 containing genetic variants found in patients failed to rescue. We show that nphp4 is involved in the formation of motile cilia in Kupffer's vesicle, which generate asymmetrical fluid flow necessary for normal L-R asymmetry. CONCLUSIONS: NPHP4 mutations are associated with cardiac laterality defects and heterotaxy. In zebrafish, nphp4 is essential for the development and function of Kupffer's vesicle cilia and is required for global L-R patterning.


Asunto(s)
Pleiotropía Genética/genética , Variación Genética/genética , Estudio de Asociación del Genoma Completo/métodos , Cardiopatías Congénitas/genética , Proteínas/genética , Secuencia de Aminoácidos , Animales , Estudios de Cohortes , Femenino , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/patología , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA