Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Intervalo de año de publicación
1.
Neuron ; 111(6): 824-838.e7, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36610398

RESUMEN

Tauopathies are neurodegenerative diseases that involve the pathological accumulation of tau proteins; in this family are Alzheimer disease, corticobasal degeneration, and chronic traumatic encephalopathy, among others. Hypothesizing that reducing this accumulation could mitigate pathogenesis, we performed a cross-species genetic screen targeting 6,600 potentially druggable genes in human cells and Drosophila. We found and validated 83 hits in cells and further validated 11 hits in the mouse brain. Three of these hits (USP7, RNF130, and RNF149) converge on the C terminus of Hsc70-interacting protein (CHIP) to regulate tau levels, highlighting the role of CHIP in maintaining tau proteostasis in the brain. Knockdown of each of these three genes in adult tauopathy mice reduced tau levels and rescued the disease phenotypes. This study thus identifies several points of intervention to reduce tau levels and demonstrates that reduction of tau levels via regulation of this pathway is a viable therapeutic strategy for Alzheimer disease and other tauopathies.


Asunto(s)
Tauopatías , Proteínas tau , Adulto , Animales , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Drosophila/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Tauopatías/tratamiento farmacológico , Tauopatías/genética , Tauopatías/metabolismo , Peptidasa Específica de Ubiquitina 7/metabolismo
2.
Blood Adv ; 6(12): 3803-3811, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35500223

RESUMEN

Fanconi anemia (FA), a genetic DNA repair disorder characterized by marrow failure and cancer susceptibility. In FA mice, metformin improves blood counts and delays tumor development. We conducted a single institution study of metformin in nondiabetic patients with FA to determine feasibility and tolerability of metformin treatment and to assess for improvement in blood counts. Fourteen of 15 patients with at least 1 cytopenia (hemoglobin < 10 g/dL; platelet count < 100 000 cells/µL; or an absolute neutrophil count < 1000 cells/µL) were eligible to receive metformin for 6 months. Median patient age was 9.4 years (range 6.0-26.5 ). Thirteen of 14 subjects (93%) tolerated maximal dosing for age; 1 subject had dose reduction for grade 2 gastrointestinal symptoms. No subjects developed hypoglycemia or metabolic acidosis. No subjects had dose interruptions caused by toxicity, and no grade 3 or higher adverse events attributed to metformin were observed. Hematologic response based on modified Myelodysplastic Syndrome International Working Group criteria was observed in 4 of 13 evaluable patients (30.8%; 90% confidence interval, 11.3-57.3). Median time to response was 84.5 days (range 71-128 days). Responses were noted in neutrophils (n = 3), platelets (n = 1), and red blood cells (n = 1). No subjects met criteria for disease progression or relapse during treatment. Correlative studies explored potential mechanisms of metformin activity in FA. Plasma proteomics showed reduction in inflammatory pathways with metformin. Metformin is safe and tolerable in nondiabetic patients with FA and may provide therapeutic benefit. This trial was registered at as #NCT03398824.


Asunto(s)
Anemia de Fanconi , Metformina , Niño , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/genética , Humanos , Metformina/uso terapéutico , Adulto Joven
3.
Nanomaterials (Basel) ; 11(9)2021 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-34578642

RESUMEN

Formic acid is an advantageous liquid organic hydrogen carrier. It is relatively nontoxic and can be synthesized by the reaction of CO2 with sustainable hydrogen or by biomass decomposition. As an alternative to more widely studied powdery catalysts, supported Pd-C catalytic thin films with controlled nanostructure and compositions were newly prepared in this work by magnetron sputtering on structured supports and tested for the formic acid decomposition reaction. A two-magnetron configuration (carbon and tailored Pd-C targets) was used to achieve a reduction in Pd consumption and high catalyst surface roughness and dispersion by increasing the carbon content. Activity and durability tests were carried out for the gas phase formic acid decomposition reaction on SiC foam monoliths coated with the Pd-C films and the effects of column width, surface roughness and thermal pre-reduction time were investigated. Activity of 5.04 molH2·gPd-1·h-1 and 92% selectivity to the dehydrogenation reaction were achieved at 300 °C for the catalyst with a lower column width and higher carbon content and surface roughness. It was also found that deactivation occurs when Pd is sintered due to the elimination of carbon and/or the segregation and agglomeration of Pd upon cycling. Magnetron sputtering deposition appears as a promising and scalable route for the one-step preparation of Pd-C catalytic films by overcoming the different deposition characteristics of Pd and C with an appropriate experimental design.

4.
Elife ; 102021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33871358

RESUMEN

Most research on neurodegenerative diseases has focused on neurons, yet glia help form and maintain the synapses whose loss is so prominent in these conditions. To investigate the contributions of glia to Huntington's disease (HD), we profiled the gene expression alterations of Drosophila expressing human mutant Huntingtin (mHTT) in either glia or neurons and compared these changes to what is observed in HD human and HD mice striata. A large portion of conserved genes are concordantly dysregulated across the three species; we tested these genes in a high-throughput behavioral assay and found that downregulation of genes involved in synapse assembly mitigated pathogenesis and behavioral deficits. To our surprise, reducing dNRXN3 function in glia was sufficient to improve the phenotype of flies expressing mHTT in neurons, suggesting that mHTT's toxic effects in glia ramify throughout the brain. This supports a model in which dampening synaptic function is protective because it attenuates the excitotoxicity that characterizes HD.


When a neuron dies, through injury or disease, the body loses all communication that passes through it. The brain compensates by rerouting the flow of information through other neurons in the network. Eventually, if the loss of neurons becomes too great, compensation becomes impossible. This process happens in Alzheimer's, Parkinson's, and Huntington's disease. In the case of Huntington's disease, the cause is mutation to a single gene known as huntingtin. The mutation is present in every cell in the body but causes particular damage to parts of the brain involved in mood, thinking and movement. Neurons and other cells respond to mutations in the huntingtin gene by turning the activities of other genes up or down, but it is not clear whether all of these changes contribute to the damage seen in Huntington's disease. In fact, it is possible that some of the changes are a result of the brain trying to protect itself. So far, most research on this subject has focused on neurons because the huntingtin gene plays a role in maintaining healthy neuronal connections. But, given that all cells carry the mutated gene, it is likely that other cells are also involved. The glia are a diverse group of cells that support the brain, providing care and sustenance to neurons. These cells have a known role in maintaining the connections between neurons and may also have play a role in either causing or correcting the damage seen in Huntington's disease. The aim of Onur et al. was to find out which genes are affected by having a mutant huntingtin gene in neurons or glia, and whether severity of Huntington's disease improved or worsened when the activity of these genes changed. First, Onur et al. identified genes affected by mutant huntingtin by comparing healthy human brains to the brains of people with Huntington's disease. Repeating the same comparison in mice and fruit flies identified genes affected in the same way across all three species, revealing that, in Huntington's disease, the brain dials down glial cell genes involved in maintaining neuronal connections. To find out how these changes in gene activity affect disease severity and progression, Onur et al. manipulated the activity of each of the genes they had identified in fruit flies that carried mutant versions of huntingtin either in neurons, in glial cells or in both cell types. They then filmed the flies to see the effects of the manipulation on movement behaviors, which are affected by Huntington's disease. This revealed that purposely lowering the activity of the glial genes involved in maintaining connections between neurons improved the symptoms of the disease, but only in flies who had mutant huntingtin in their glial cells. This indicates that the drop in activity of these genes observed in Huntington's disease is the brain trying to protect itself. This work suggests that it is important to include glial cells in studies of neurological disorders. It also highlights the fact that changes in gene expression as a result of a disease are not always bad. Many alterations are compensatory, and try to either make up for or protect cells affected by the disease. Therefore, it may be important to consider whether drugs designed to treat a condition by changing levels of gene activity might undo some of the body's natural protection. Working out which changes drive disease and which changes are protective will be essential for designing effective treatments.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Sinapsis Eléctricas/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Neuroglía/metabolismo , Transmisión Sináptica , Animales , Conducta Animal , Encéfalo/patología , Encéfalo/fisiopatología , Estudios de Casos y Controles , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster , Sinapsis Eléctricas/patología , Femenino , Redes Reguladoras de Genes , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Locomoción , Masculino , Ratones Transgénicos , Mutación , Neuroglía/patología , Transcriptoma , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
5.
Hum Mol Genet ; 28(12): 2014-2029, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30753434

RESUMEN

An early hallmark of Alzheimer's disease is the accumulation of amyloid-ß (Aß), inspiring numerous therapeutic strategies targeting this peptide. An alternative approach is to destabilize the amyloid beta precursor protein (APP) from which Aß is derived. We interrogated innate pathways governing APP stability using a siRNA screen for modifiers whose own reduction diminished APP in human cell lines and transgenic Drosophila. As proof of principle, we validated PKCß-a known modifier identified by the screen-in an APP transgenic mouse model. PKCß was genetically targeted using a novel adeno-associated virus shuttle vector to deliver microRNA-adapted shRNA via intracranial injection. In vivo reduction of PKCß initially diminished APP and delayed plaque formation. Despite persistent PKCß suppression, the effect on APP and amyloid diminished over time. Our study advances this approach for mining druggable modifiers of disease-associated proteins, while cautioning that prolonged in vivo validation may be needed to reveal emergent limitations on efficacy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/metabolismo , Proteína Quinasa C beta/antagonistas & inhibidores , Enfermedad de Alzheimer/genética , Amiloidosis/terapia , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Drosophila , Pruebas Genéticas , Terapia Genética , Humanos , Ratones , Ratones Transgénicos , Células 3T3 NIH , Fosforilación , Placa Amiloide/patología , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
6.
Cell Syst ; 7(1): 28-40.e4, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-29936182

RESUMEN

Discriminating transcriptional changes that drive disease pathogenesis from nonpathogenic and compensatory responses is a daunting challenge. This is particularly true for neurodegenerative diseases, which affect the expression of thousands of genes in different brain regions at different disease stages. Here we integrate functional testing and network approaches to analyze previously reported transcriptional alterations in the brains of Huntington disease (HD) patients. We selected 312 genes whose expression is dysregulated both in HD patients and in HD mice and then replicated and/or antagonized each alteration in a Drosophila HD model. High-throughput behavioral testing in this model and controls revealed that transcriptional changes in synaptic biology and calcium signaling are compensatory, whereas alterations involving the actin cytoskeleton and inflammation drive disease. Knockdown of disease-driving genes in HD patient-derived cells lowered mutant Huntingtin levels and activated macroautophagy, suggesting a mechanism for mitigating pathogenesis. Our multilayered approach can thus untangle the wealth of information generated by transcriptomics and identify early therapeutic intervention points.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Enfermedad de Huntington/genética , Animales , Encéfalo/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Enfermedad de Huntington/fisiopatología , Células Madre Pluripotentes Inducidas , Masculino , Transcriptoma/genética
7.
Elife ; 62017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29256861

RESUMEN

The discovery of the causative gene for Huntington's disease (HD) has promoted numerous efforts to uncover cellular pathways that lower levels of mutant huntingtin protein (mHtt) and potentially forestall the appearance of HD-related neurological defects. Using a cell-based model of pathogenic huntingtin expression, we identified a class of compounds that protect cells through selective inhibition of a lipid kinase, PIP4Kγ. Pharmacological inhibition or knock-down of PIP4Kγ modulates the equilibrium between phosphatidylinositide (PI) species within the cell and increases basal autophagy, reducing the total amount of mHtt protein in human patient fibroblasts and aggregates in neurons. In two Drosophila models of Huntington's disease, genetic knockdown of PIP4K ameliorated neuronal dysfunction and degeneration as assessed using motor performance and retinal degeneration assays respectively. Together, these results suggest that PIP4Kγ is a druggable target whose inhibition enhances productive autophagy and mHtt proteolysis, revealing a useful pharmacological point of intervention for the treatment of Huntington's disease, and potentially for other neurodegenerative disorders.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/prevención & control , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Animales , Autofagia , Células Cultivadas , Modelos Animales de Enfermedad , Drosophila , Fibroblastos/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Modelos Biológicos , Neuronas/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Agregación Patológica de Proteínas , Proteolisis
8.
Elife ; 52016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27779468

RESUMEN

Several neurodegenerative diseases are driven by the toxic gain-of-function of specific proteins within the brain. Elevated levels of alpha-synuclein (α-Syn) appear to drive neurotoxicity in Parkinson's disease (PD); neuronal accumulation of tau is a hallmark of Alzheimer's disease (AD); and their increased levels cause neurodegeneration in humans and model organisms. Despite the clinical differences between AD and PD, several lines of evidence suggest that α-Syn and tau overlap pathologically. The connections between α-Syn and tau led us to ask whether these proteins might be regulated through a shared pathway. We therefore screened for genes that affect post-translational levels of α-Syn and tau. We found that TRIM28 regulates α-Syn and tau levels and that its reduction rescues toxicity in animal models of tau- and α-Syn-mediated degeneration. TRIM28 stabilizes and promotes the nuclear accumulation and toxicity of both proteins. Intersecting screens across comorbid proteinopathies thus reveal shared mechanisms and therapeutic entry points.


Asunto(s)
Núcleo Celular/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Enfermedad de Parkinson/fisiopatología
9.
Neuron ; 92(2): 407-418, 2016 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-27720485

RESUMEN

Many neurodegenerative proteinopathies share a common pathogenic mechanism: the abnormal accumulation of disease-related proteins. As growing evidence indicates that reducing the steady-state levels of disease-causing proteins mitigates neurodegeneration in animal models, we developed a strategy to screen for genes that decrease the levels of tau, whose accumulation contributes to the pathology of both Alzheimer disease (AD) and progressive supranuclear palsy (PSP). Integrating parallel cell-based and Drosophila genetic screens, we discovered that tau levels are regulated by Nuak1, an AMPK-related kinase. Nuak1 stabilizes tau by phosphorylation specifically at Ser356. Inhibition of Nuak1 in fruit flies suppressed neurodegeneration in tau-expressing Drosophila, and Nuak1 haploinsufficiency rescued the phenotypes of a tauopathy mouse model. These results demonstrate that decreasing total tau levels is a valid strategy for mitigating tau-related neurodegeneration and reveal Nuak1 to be a novel therapeutic entry point for tauopathies.


Asunto(s)
Conducta Animal , Proteínas Quinasas/genética , Proteínas Represoras/genética , Tauopatías/genética , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Animales , Línea Celular Tumoral , Condicionamiento Psicológico , Modelos Animales de Enfermedad , Drosophila , Miedo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Ratones , Fosforilación/genética , Parálisis Supranuclear Progresiva/genética
10.
ACS Appl Mater Interfaces ; 7(25): 13889-97, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26046812

RESUMEN

The fabrication of single-material photonic-multilayer devices is explored using a new methodology to produce porous silicon layers by magnetron sputtering. Our bottom-up methodology produces highly stable amorphous porous silicon films with a controlled refractive index using magnetron sputtering and incorporating a large amount of deposition gas inside the closed pores. The influence of the substrate bias on the formation of the closed porosity was explored here for the first time when He was used as the deposition gas. We successfully simulated, designed, and characterized Bragg reflectors and an optical microcavity that integrates these porous layers. The sharp interfaces between the dense and porous layers combined with the adequate control of the refractive index and thickness allowed for excellent agreement between the simulation and the experiments. The versatility of the magnetron sputtering technique allowed for the preparation of these structures for a wide range of substrates such as polymers while also taking advantage of the oblique angle deposition to prepare Bragg reflectors with a controlled lateral gradient in the stop band wavelengths.

11.
Insuf. card ; 10(2): 57-65, abr. 2015. ilus, tab
Artículo en Español | LILACS | ID: lil-757080

RESUMEN

Introducción. El tamaño y la función de la aurícula izquierda (AI) son predictores de mal pronóstico. Nuevas técnicas ecocardiográficas permiten evaluar la función global y regional de la AI. Objetivo. Comparar la función de la AI utilizando la deformidad miocárdica auricular (DMA) en pacientes con dilatación de la AI y sujetos controles. Métodos. Estudio prospectivo, en mayores de 18 años, estudiados entre Julio y Diciembre de 2013. Se registraron datos epidemiológicos y se tomaron medidas ecocardiográficas en modo M y Doppler y speckle tracking. Resultados. Se estudiaron 50 pacientes divididos en 2 grupos: AI dilatada (A) y AI normal (B). Hubo una correlación lineal significativa entre el área y el volumen de la AI en el grupo A y diferencias significativas en el strain longitudinal global entre ambos grupos. El strain de la AI fue mayor en el grupo B. Hubo una correlación lineal inversa significativa en el grupo A entre la velocidad, strain y strain rate globales con el volumen de la AI. Conclusiones. El aumento del volumen de la AI se asoció a una disminución de la función de reservorio de la AI. La DMA nos permite una evaluación regional y global no invasiva y confiable de la función auricular izquierda.


Introduction. The size and function of the left atrium (LA) are predictors of poor prognosis. New echocardiographic techniques allow the assessment of global and regional function of the LA. Objective. To compare the role of LA using atrial myocardial deformation (AMD) in patients with dilatation of LA and in control subjects. Methods. This is a prospective study in 50 patients over 18 years, studied between July and December 2013. Epidemiological data were recorded and echocardiographic measurements were taken in M mode, Doppler and speckle tracking. Results. The 50 patients were divided into two groups: dilated LA (A) and normal LA (B) were studied. There was a significant linear correlation between the area and volume of the LA in A group and significant differences in the global longitudinal strain between the two groups. The percentage of AMD was higher in B group. There was a significant inverse linear correlation in A group, between velocity, global strain and global strain with the LA volume. Conclusions. The increase in LA volume was associated with a decreased function of reservoir of LA. The AMD allows us to assess the regional and global non-invasive of LA early dysfunction.


Introdução. O tamanho e a função do átrio esquerdo (AE) são preditores de pior prognóstico. Novas técnicas de ecocardiografia permitem avaliação da função global e regional do AE. Objetivo. Comparar o papel do AE usando deformação do miocárdio atrial (DMA) em pacientes com AE dilatados e indivíduos controle. Métodos. Estudo prospectivo em 50 pacientes com mais de 18 anos, estudados entre Julho e Dezembro de 2013. Os dados epidemiológicos foram registrados e as medidas ecocardiográficas foram tomadas em modo M com Doppler e specke tracking. Resultados. Foram estudados 50 pacientes divididos em dois grupos: AE dilatado (A) y AE normal (B). Houve uma correlação linear significativa entre a área e o volume do AE no grupo A e diferenças significativas na deformação longitudinal global entre os dois grupos. O strain de AE foi maior no grupo B. Houve uma correlação linear inversa significativa no grupo A entre a velocidade, strain e strain rate globais com o volume do AE. Conclusões. O aumento no volume do AE foi associado com uma diminuição da função de reservatório do AE. A DMA permite uma avaliação regional e global não-invasivo e confiável da função atrial esquerda.


Asunto(s)
Humanos , Función del Atrio Izquierdo , Técnicas de Imagen Cardíaca
12.
PLoS Genet ; 9(4): e1003445, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23637619

RESUMEN

We report the identification and characterization of a previously unknown suppressor of myopathy caused by expansion of CUG repeats, the mutation that triggers Myotonic Dystrophy Type 1 (DM1). We screened a collection of genes encoding RNA-binding proteins as candidates to modify DM1 pathogenesis using a well established Drosophila model of the disease. The screen revealed smaug as a powerful modulator of CUG-induced toxicity. Increasing smaug levels prevents muscle wasting and restores muscle function, while reducing its function exacerbates CUG-induced phenotypes. Using human myoblasts, we show physical interactions between human Smaug (SMAUG1/SMAD4A) and CUGBP1. Increased levels of SMAUG1 correct the abnormally high nuclear accumulation of CUGBP1 in myoblasts from DM1 patients. In addition, augmenting SMAUG1 levels leads to a reduction of inactive CUGBP1-eIF2α translational complexes and to a correction of translation of MRG15, a downstream target of CUGBP1. Therefore, Smaug suppresses CUG-mediated muscle wasting at least in part via restoration of translational activity of CUGBP1.


Asunto(s)
Distrofia Miotónica , Proteínas de Unión al ARN , Regulación de la Expresión Génica , Humanos , Mioblastos/metabolismo , Distrofia Miotónica/genética , Proteínas de Unión al ARN/genética
13.
PLoS Genet ; 8(11): e1003042, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209424

RESUMEN

A genome-scale RNAi screen was performed in a mammalian cell-based assay to identify modifiers of mutant huntingtin toxicity. Ontology analysis of suppressor data identified processes previously implicated in Huntington's disease, including proteolysis, glutamate excitotoxicity, and mitochondrial dysfunction. In addition to established mechanisms, the screen identified multiple components of the RRAS signaling pathway as loss-of-function suppressors of mutant huntingtin toxicity in human and mouse cell models. Loss-of-function in orthologous RRAS pathway members also suppressed motor dysfunction in a Drosophila model of Huntington's disease. Abnormal activation of RRAS and a down-stream effector, RAF1, was observed in cellular models and a mouse model of Huntington's disease. We also observe co-localization of RRAS and mutant huntingtin in cells and in mouse striatum, suggesting that activation of R-Ras may occur through protein interaction. These data indicate that mutant huntingtin exerts a pathogenic effect on this pathway that can be corrected at multiple intervention points including RRAS, FNTA/B, PIN1, and PLK1. Consistent with these results, chemical inhibition of farnesyltransferase can also suppress mutant huntingtin toxicity. These data suggest that pharmacological inhibition of RRAS signaling may confer therapeutic benefit in Huntington's disease.


Asunto(s)
Enfermedad de Huntington , Proteínas del Tejido Nervioso , Interferencia de ARN , Proteínas ras , Animales , Cuerpo Estriado/ultraestructura , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Farnesiltransferasa/antagonistas & inhibidores , Farnesiltransferasa/metabolismo , Genoma Humano , Células HEK293 , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Redes y Vías Metabólicas , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/toxicidad , Proteínas del Tejido Nervioso/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología , Proteínas ras/antagonistas & inhibidores , Proteínas ras/genética , Proteínas ras/metabolismo
14.
Neuron ; 67(2): 199-212, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20670829

RESUMEN

Proteolytic cleavage of huntingtin (Htt) is known to be a key event in the pathogenesis of Huntington's disease (HD). Our understanding of proteolytic processing of Htt has thus far focused on the protease families-caspases and calpains. Identifying critical proteases involved in Htt proteolysis and toxicity using an unbiased approach has not been reported. To accomplish this, we designed a high-throughput western blot-based screen to examine the generation of the smallest N-terminal polyglutamine-containing Htt fragment. We screened 514 siRNAs targeting the repertoire of human protease genes. This screen identified 11 proteases that, when inhibited, reduced Htt fragment accumulation. Three of these belonged to the matrix metalloproteinase (MMP) family. One family member, MMP-10, directly cleaves Htt and prevents cell death when knocked down in striatal Hdh(111Q/111Q) cells. Correspondingly, MMPs are activated in HD mouse models, and loss of function of Drosophila homologs of MMPs suppresses Htt-induced neuronal dysfunction in vivo.


Asunto(s)
Enfermedad de Huntington/genética , Metaloproteinasas de la Matriz/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/toxicidad , Proteínas Nucleares/metabolismo , Proteínas Nucleares/toxicidad , Animales , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Transformada , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Drosophila , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Proteína Huntingtina , Metaloproteinasas de la Matriz/clasificación , Metaloproteinasas de la Matriz/genética , Ratones , Ratones Mutantes Neurológicos , Mutación/genética , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/genética , Péptidos/genética , Péptidos/metabolismo , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Transfección/métodos
15.
Cell Tissue Res ; 339(2): 321-36, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19941006

RESUMEN

The distribution of leucokinin (LK) neurons in the central nervous system (CNS) of Drosophila melanogaster was described by immunolabelling many years ago. However, no detailed underlying information of the input or output connections of their neurites was then available. Here, we provide a more accurate morphological description by employing a novel LK-specific GAL4 line that recapitulates LK expression. In order to analyse the possible afferent and efferent neural candidates of LK neurons, we used this lk-GAL4 line together with other CNS-Gal4 lines, combined with antisera against various neuropeptides or neurotransmitters. We found four kinds of LK neurons in the brain. (1) The lateral horn neurons connect the antennal glomerula to the mushroom bodies. (2) The suboesophageal neurons connect the gustatory receptors to the suboesophageal ganglia and ventral nerve cord. (3) The anterior neurons innervate the corpus cardiacum of the ring gland but LK expression is surprisingly not detectable from the third instar onwards in these neurons. (4) A set of abdominal ganglion neurons connect to the dorsal median tract in larvae and send their axons to a segmental muscle 8. Thus, the methods employed in our study can be used to identify individual neuropeptidergic neurons and thereby characterize functional cues or developmental transformations in their differentiation.


Asunto(s)
Proteínas de Drosophila/biosíntesis , Drosophila melanogaster/anatomía & histología , Neuronas/metabolismo , Neuropéptidos/biosíntesis , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ganglios/metabolismo , Larva/anatomía & histología , Larva/genética , Larva/metabolismo , Músculos/metabolismo , Cuerpos Pedunculados/metabolismo , Vías Nerviosas , Neuritas/metabolismo , Neuropéptidos/genética , Sistemas Neurosecretores/metabolismo , Neurotransmisores/metabolismo
16.
Neuron ; 55(4): 565-71, 2007 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-17698010

RESUMEN

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a recently described neurodegenerative disorder of older adult carriers of premutation alleles (60-200 CGG repeats) in the fragile X mental retardation gene (FMR1). It has been proposed that FXTAS is an RNA-mediated neurodegenerative disease caused by the titration of RNA-binding proteins by the CGG repeats. To test this hypothesis, we utilize a transgenic Drosophila model of FXTAS that expresses a premutation-length repeat (90 CGG repeats) from the 5' UTR of the human FMR1 gene and displays neuronal degeneration. Here, we show that overexpression of RNA-binding proteins hnRNP A2/B1 and CUGBP1 suppresses the phenotype of the CGG transgenic fly. Furthermore, we show that hnRNP A2/B1 directly interacts with riboCGG repeats and that the CUGBP1 protein interacts with the riboCGG repeats via hnRNP A2/B1.


Asunto(s)
Proteínas de Drosophila/metabolismo , Síndrome del Cromosoma X Frágil/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas de Unión al ARN/metabolismo , Expansión de Repetición de Trinucleótido/genética , Animales , Animales Modificados Genéticamente , Proteínas CELF1 , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Ojo/patología , Ojo/ultraestructura , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/complicaciones , Síndrome del Cromosoma X Frágil/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Inmunoprecipitación/métodos , Microscopía Electrónica de Rastreo/métodos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Proteínas de Unión al ARN/genética
17.
PLoS Genet ; 3(12): e234, 2007 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-18166084

RESUMEN

Spinocerebellar ataxias (SCAs) are a genetically heterogeneous group of neurodegenerative disorders sharing atrophy of the cerebellum as a common feature. SCA1 and SCA2 are two ataxias caused by expansion of polyglutamine tracts in Ataxin-1 (ATXN1) and Ataxin-2 (ATXN2), respectively, two proteins that are otherwise unrelated. Here, we use a Drosophila model of SCA1 to unveil molecular mechanisms linking Ataxin-1 with Ataxin-2 during SCA1 pathogenesis. We show that wild-type Drosophila Ataxin-2 (dAtx2) is a major genetic modifier of human expanded Ataxin-1 (Ataxin-1[82Q]) toxicity. Increased dAtx2 levels enhance, and more importantly, decreased dAtx2 levels suppress Ataxin-1[82Q]-induced neurodegeneration, thereby ruling out a pathogenic mechanism by depletion of dAtx2. Although Ataxin-2 is normally cytoplasmic and Ataxin-1 nuclear, we show that both dAtx2 and hAtaxin-2 physically interact with Ataxin-1. Furthermore, we show that expanded Ataxin-1 induces intranuclear accumulation of dAtx2/hAtaxin-2 in both Drosophila and SCA1 postmortem neurons. These observations suggest that nuclear accumulation of Ataxin-2 contributes to expanded Ataxin-1-induced toxicity. We tested this hypothesis engineering dAtx2 transgenes with nuclear localization signal (NLS) and nuclear export signal (NES). We find that NLS-dAtx2, but not NES-dAtx2, mimics the neurodegenerative phenotypes caused by Ataxin-1[82Q], including repression of the proneural factor Senseless. Altogether, these findings reveal a previously unknown functional link between neurodegenerative disorders with common clinical features but different etiology.


Asunto(s)
Proteínas del Tejido Nervioso/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Proteínas Nucleares/fisiología , Animales , Ataxina-1 , Ataxinas , Drosophila , Modelos Biológicos , Proteínas del Tejido Nervioso/genética
18.
J Biol Chem ; 281(36): 26714-24, 2006 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16831871

RESUMEN

CHIP (C terminus of Hsc-70 interacting protein) is an E3 ligase that links the protein folding machinery with the ubiquitin-proteasome system and has been implicated in disorders characterized by protein misfolding and aggregation. Here we investigate the role of CHIP in protecting from ataxin-1-induced neurodegeneration. Ataxin-1 is a polyglutamine protein whose expansion causes spinocerebellar ataxia type-1 (SCA1) and triggers the formation of nuclear inclusions (NIs). We find that CHIP and ataxin-1 proteins directly interact and co-localize in NIs both in cell culture and SCA1 postmortem neurons. CHIP promotes ubiquitination of expanded ataxin-1 both in vitro and in cell culture. The Hsp70 chaperone increases CHIP-mediated ubiquitination of ataxin-1 in vitro, and the tetratricopeptide repeat domain, which mediates CHIP interactions with chaperones, is required for ataxin-1 ubitiquination in cell culture. Interestingly, CHIP also interacts with and ubiquitinates unexpanded ataxin-1. Overexpression of CHIP in a Drosophila model of SCA1 decreases the protein steady-state levels of both expanded and unexpanded ataxin-1 and suppresses their toxicity. Finally we investigate the ability of CHIP to protect against toxicity caused by expanded polyglutamine tracts in different protein contexts. We find that CHIP is not effective in suppressing the toxicity caused by a bare 127Q tract with only a short hemagglutinin tag, but it is very efficient in suppressing toxicity caused by a 128Q tract in the context of an N-terminal huntingtin backbone. These data underscore the importance of the protein framework for modulating the effects of polyglutamine-induced neurodegeneration.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/toxicidad , Proteínas Nucleares/metabolismo , Proteínas Nucleares/toxicidad , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Animales , Ataxina-1 , Ataxinas , Células Cultivadas , Drosophila melanogaster/anatomía & histología , Humanos , Cuerpos de Inclusión Intranucleares/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Proteínas Nucleares/genética , Péptidos/toxicidad , Células Fotorreceptoras de Invertebrados/citología , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Conformación Proteica , Pliegue de Proteína , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología , Transgenes , Ubiquitina-Proteína Ligasas/genética
19.
Hum Mol Genet ; 15(13): 2138-45, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16723374

RESUMEN

Myotonic dystrophy type 1 (DM1) is a neuromuscular disorder caused by a CTG expansion in the 3' UTR of the dystrophia myotonica protein kinase (DMPK) gene. It has been hypothesized that the pathogenesis in DM1 is triggered by a toxic gain of function of the expanded DMPK RNA. This expanded RNA is retained in nuclear foci where it sequesters and induces alterations in the levels of RNA-binding proteins (RNA-BP). To model DM1 and study the implication of RNA-BP in CUG-induced toxicity, we have generated a Drosophila DM1 model expressing a non-coding mRNA containing 480 interrupted CUG repeats; i.e. [(CUG)20CUCGA]24. This (iCUG)480 transcript accumulates in nuclear foci and its expression leads to muscle wasting and degeneration in Drosophila. We also report that altering the levels of two RNA-BP known to be involved in DM1 pathogenesis, MBNL1 and CUGBP1, modify the (iCUG)480 degenerative phenotypes. Expanded CUG-induced toxicity in Drosophila is suppressed when MBNL1 expression levels are increased, and enhanced when MBNL1 levels are reduced. In addition, (iCUG)480 also causes a decrease in the levels of soluble MBNL1 that is sequestered in the CUG-containing nuclear foci. In contrast, increasing the levels of CUGBP1 worsens (iCUG)480-induced degeneration even though CUGBP1 distribution is not altered by the expression of the expanded triplet repeat. Our data supports a mechanism for DM1 pathogenesis in which decreased levels of MBNL and increased levels of CUGBP mediate the RNA-induced toxicity observed in DM1. Perhaps more importantly, they also provide proof of the principle that CUG-induced muscle toxicity can be suppressed.


Asunto(s)
Drosophila melanogaster/genética , Distrofia Miotónica/genética , Proteínas de Unión al ARN/fisiología , Expansión de Repetición de Trinucleótido/genética , Animales , Animales Modificados Genéticamente , Northern Blotting , Proteínas CELF1 , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestructura , Ojo/metabolismo , Ojo/ultraestructura , Humanos , Hibridación in Situ , Microscopía Electrónica de Rastreo , Músculos/metabolismo , Músculos/ultraestructura , Distrofia Miotónica/metabolismo , Distrofia Miotónica/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...