Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Biol Chem ; 296: 100760, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33965374

RESUMEN

One of the defining pathological features of Alzheimer's disease (AD) is the deposition of neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau in the brain. Aberrant activation of kinases in AD has been suggested to enhance phosphorylation and toxicity of tau, making the responsible tau kinases attractive therapeutic targets. The full complement of tau-interacting kinases in AD brain and their activity in disease remains incompletely defined. Here, immunoaffinity enrichment coupled with mass spectrometry (MS) identified TANK-binding kinase 1 (TBK1) as a tau-interacting partner in human AD cortical brain tissues. We validated this interaction in human AD, familial frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) caused by mutations in MAPT (R406W & P301L) and corticobasal degeneration (CBD) postmortem brain tissues as well as human cell lines. Further, we document increased TBK1 activation in both AD and FTDP-17 and map TBK1 phosphorylation sites on tau based on in vitro kinase assays coupled to MS. Lastly, in a Drosophila tauopathy model, activating expression of a conserved TBK1 ortholog triggers tau hyperphosphorylation and enhanced neurodegeneration, whereas knockdown had the reciprocal effect, suppressing tau toxicity. Collectively, our findings suggest that increased TBK1 activation may promote tau hyperphosphorylation and neuronal loss in AD and related tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Animales , Drosophila , Femenino , Células HEK293 , Humanos , Masculino , Tauopatías/patología
2.
J Neurosci ; 39(45): 8877-8884, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31558618

RESUMEN

Alcohol use is highly prevalent in the United States and across the world, and every year millions of people suffer from alcohol use disorders (AUDs). Although the genetic contribution to developing AUDs is estimated to be 50-60%, many of the underlying molecular mechanisms remain unclear. Previous studies from our laboratory revealed that Drosophila melanogaster lacking RhoGAP18B and Ras Suppressor 1 (Rsu1) display reduced sensitivity to ethanol-induced sedation. Both Rsu1 and RhoGAP18B are negative regulators of the small Rho-family GTPase, Rac1, a modulator of actin dynamics. Here we investigate the role of Rac1 and its downstream target, the actin-severing protein cofilin, in alcohol consumption preference. We show that these two regulators of actin dynamics can alter male experience-dependent alcohol preference in a bidirectional manner: expressing either activated Rac1 or dominant-negative cofilin in the mushroom bodies (MBs) abolishes experience-dependent alcohol preference. Conversely, dominant-negative Rac1 or activated cofilin MB expression lead to faster acquisition of alcohol preference. Our data show that Rac1 and cofilin activity are key to determining the rate of acquisition of alcohol preference, revealing a critical role of actin dynamics regulation in the development of voluntary self-administration in DrosophilaSIGNIFICANCE STATEMENT The risks for developing an alcohol use disorder (AUD) are strongly determined by genetic factors. Understanding the genes and molecular mechanisms that contribute to that risk is therefore a necessary first step for the development of targeted therapeutic intervention. Here we show that regulators of actin cytoskeleton dynamics can bidirectionally determine the acquisition rate of alcohol self-administration, highlighting this process as a key mechanism contributing to the risk of AUD development.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Alcoholismo/genética , Proteínas de Drosophila/genética , Proteínas de Microfilamentos/genética , Cuerpos Pedunculados/metabolismo , Proteínas de Unión al GTP rac/genética , Factores Despolimerizantes de la Actina/metabolismo , Alcoholismo/metabolismo , Animales , Condicionamiento Clásico , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Masculino , Proteínas de Microfilamentos/metabolismo , Cuerpos Pedunculados/fisiología , Proteínas de Unión al GTP rac/metabolismo
3.
Proc Natl Acad Sci U S A ; 112(30): E4085-93, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26170296

RESUMEN

Alcohol abuse is highly prevalent, but little is understood about the molecular causes. Here, we report that Ras suppressor 1 (Rsu1) affects ethanol consumption in flies and humans. Drosophila lacking Rsu1 show reduced sensitivity to ethanol-induced sedation. We show that Rsu1 is required in the adult nervous system for normal sensitivity and that it acts downstream of the integrin cell adhesion molecule and upstream of the Ras-related C3 botulinum toxin substrate 1 (Rac1) GTPase to regulate the actin cytoskeleton. In an ethanol preference assay, global loss of Rsu1 causes high naïve preference. In contrast, flies lacking Rsu1 only in the mushroom bodies of the brain show normal naïve preference but then fail to acquire ethanol preference like normal flies. Rsu1 is, thus, required in distinct neurons to modulate naïve and acquired ethanol preference. In humans, we find that polymorphisms in RSU1 are associated with brain activation in the ventral striatum during reward anticipation in adolescents and alcohol consumption in both adolescents and adults. Together, these data suggest a conserved role for integrin/Rsu1/Rac1/actin signaling in modulating reward-related phenotypes, including ethanol consumption, across phyla.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Proteínas de Drosophila/fisiología , Factores de Transcripción/fisiología , Actinas/metabolismo , Adolescente , Adulto , Animales , Encéfalo/metabolismo , Moléculas de Adhesión Celular/metabolismo , Niño , Estudios de Cohortes , Citoesqueleto/metabolismo , Proteínas de Drosophila/genética , Etanol/química , Femenino , GTP Fosfohidrolasas/metabolismo , Genes Dominantes , Humanos , Integrinas/metabolismo , Masculino , Mutación , Neuronas/metabolismo , Polimorfismo Genético , Encuestas y Cuestionarios , Factores de Transcripción/genética
4.
Addict Biol ; 19(3): 392-401, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24164972

RESUMEN

To understand the molecular and neural mechanisms underlying alcohol addiction, many models ranging from vertebrates to invertebrates have been developed. In Drosophila melanogaster, behavioral paradigms from assaying acute responses to alcohol and to behaviors more closely modeling addiction have emerged in recent years. However, both the CAFÉ assay, similar to a two-bottle choice consumption assay, as well as conditioned odor preference, where ethanol is used as the reinforcer, are labor intensive and have low throughput. To address this limitation, we have established a novel ethanol consumption preference assay, called FRAPPÉ, which allows for fast, high throughput measurement of consumption in individual flies, using a fluorescence plate reader. We show that naïve flies do not prefer to consume ethanol, but various pre-exposures, such as ethanol vapor or voluntary ethanol consumption, induce ethanol preference. This ethanol-primed preference is long lasting and is not driven by calories contained in ethanol during the consumption choice. Our novel experience-dependent model of ethanol preference in Drosophila-a highly genetically tractable organism-therefore recapitulates salient features of human alcohol abuse and will facilitate the molecular understanding of the development of alcohol preference.


Asunto(s)
Alcoholismo/fisiopatología , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Animales , Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Condicionamiento Psicológico/efectos de los fármacos , Drosophila melanogaster , Ingestión de Energía/fisiología , Etanol/administración & dosificación , Conducta Alimentaria/efectos de los fármacos , Masculino , Factores de Tiempo
5.
Rev Neurosci ; 24(5): 471-84, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24077615

RESUMEN

Over the past decade, the function of the cytoskeleton has been studied extensively in developing and mature neurons. Actin, a major cytoskeletal protein, is indispensable for the structural integrity and plasticity of neurons and their synapses. Disruption of actin dynamics has significant consequence for neurons, neuronal circuits, and the functions they govern. In particular, cell adhesion molecules, members of the Rho family of GTPases, and actin-binding proteins are important modulators of actin dynamics and neuronal as well as behavioral plasticity. In this review, we discuss recent advances in Drosophila that highlight the importance of actin regulatory proteins in mediating fly behaviors such as circadian rhythm, courtship behavior, learning and memory, and the development of drug addiction.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Conducta Animal/fisiología , Animales , Moléculas de Adhesión Celular/metabolismo , Drosophila , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Sinapsis/metabolismo , Proteínas de Unión al GTP rho/metabolismo
6.
Elife ; 92020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286230

RESUMEN

Retromer, including Vps35, Vps26, and Vps29, is a protein complex responsible for recycling proteins within the endolysosomal pathway. Although implicated in both Parkinson's and Alzheimer's disease, our understanding of retromer function in the adult brain remains limited, in part because Vps35 and Vps26 are essential for development. In Drosophila, we find that Vps29 is dispensable for embryogenesis but required for retromer function in aging adults, including for synaptic transmission, survival, and locomotion. Unexpectedly, in Vps29 mutants, Vps35 and Vps26 proteins are normally expressed and associated, but retromer is mislocalized from neuropil to soma with the Rab7 GTPase. Further, Vps29 phenotypes are suppressed by reducing Rab7 or overexpressing the GTPase activating protein, TBC1D5. With aging, retromer insufficiency triggers progressive endolysosomal dysfunction, with ultrastructural evidence of impaired substrate clearance and lysosomal stress. Our results reveal the role of Vps29 in retromer localization and function, highlighting requirements for brain homeostasis in aging.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Transmisión Sináptica/fisiología , Proteínas de Transporte Vesicular/metabolismo , Animales , Drosophila , Proteínas de Drosophila/metabolismo
7.
Cell Rep ; 28(7): 1799-1813.e5, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31412248

RESUMEN

The Alzheimer's disease (AD) susceptibility gene, CD2-associated protein (CD2AP), encodes an actin binding adaptor protein, but its function in the nervous system is largely unknown. Loss of the Drosophila ortholog cindr enhances neurotoxicity of human Tau, which forms neurofibrillary tangle pathology in AD. We show that Cindr is expressed in neurons and present at synaptic terminals. cindr mutants show impairments in synapse maturation and both synaptic vesicle recycling and release. Cindr associates and genetically interacts with 14-3-3ζ, regulates the ubiquitin-proteasome system, and affects turnover of Synapsin and the plasma membrane calcium ATPase (PMCA). Loss of cindr elevates PMCA levels and reduces cytosolic calcium. Studies of Cd2ap null mice support a conserved role in synaptic proteostasis, and CD2AP protein levels are inversely related to Synapsin abundance in human postmortem brains. Our results reveal CD2AP neuronal requirements with relevance to AD susceptibility, including for proteostasis, calcium handling, and synaptic structure and function.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer , Proteínas del Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Microfilamentos/metabolismo , Neuronas/metabolismo , Proteostasis , Proteínas 14-3-3/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas del Citoesqueleto/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Femenino , Humanos , Masculino , Ratones , Proteínas de Microfilamentos/genética , Neuronas/citología , Proteoma/análisis , Proteoma/metabolismo , Transmisión Sináptica
8.
PLoS One ; 10(9): e0137465, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26366560

RESUMEN

Responses to the effects of ethanol are highly conserved across organisms, with reduced responses to the sedating effects of ethanol being predictive of increased risk for human alcohol dependence. Previously, we described that regulators of actin dynamics, such as the Rho-family GTPases Rac1, Rho1, and Cdc42, alter Drosophila's sensitivity to ethanol-induced sedation. The GTPase activating protein RhoGAP18B also affects sensitivity to ethanol. To better understand how different RhoGAP18B isoforms affect ethanol sedation, we examined them for their effects on cell shape, GTP-loading of Rho-family GTPase, activation of the actin-severing cofilin, and actin filamentation. Our results suggest that the RhoGAP18B-PA isoform acts on Cdc42, while PC and PD act via Rac1 and Rho1 to activate cofilin. In vivo, a loss-of-function mutation in the cofilin-encoding gene twinstar leads to reduced ethanol-sensitivity and acts in concert with RhoGAP18B. Different RhoGAP18B isoforms, therefore, act on distinct subsets of Rho-family GTPases to modulate cofilin activity, actin dynamics, and ethanol-induced behaviors.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Alcoholes/farmacología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Proteínas Activadoras de GTPasa/metabolismo , Hipnóticos y Sedantes/farmacología , Animales , Conducta Animal/efectos de los fármacos , Línea Celular , Forma de la Célula/efectos de los fármacos , Drosophila melanogaster/citología , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Animales , Isoformas de Proteínas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA