Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 12(1): 447, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33469018

RESUMEN

Cerebrospinal fluid (CSF) provides vital support for the brain. Abnormal CSF accumulation, such as hydrocephalus, can negatively affect perinatal neurodevelopment. The mechanisms regulating CSF clearance during the postnatal critical period are unclear. Here, we show that CSF K+, accompanied by water, is cleared through the choroid plexus (ChP) during mouse early postnatal development. We report that, at this developmental stage, the ChP showed increased ATP production and increased expression of ATP-dependent K+ transporters, particularly the Na+, K+, Cl-, and water cotransporter NKCC1. Overexpression of NKCC1 in the ChP resulted in increased CSF K+ clearance, increased cerebral compliance, and reduced circulating CSF in the brain without changes in intracranial pressure in mice. Moreover, ChP-specific NKCC1 overexpression in an obstructive hydrocephalus mouse model resulted in reduced ventriculomegaly. Collectively, our results implicate NKCC1 in regulating CSF K+ clearance through the ChP in the critical period during postnatal neurodevelopment in mice.


Asunto(s)
Líquido Cefalorraquídeo/metabolismo , Plexo Coroideo/patología , Hidrocefalia/patología , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Animales Recién Nacidos , Plexo Coroideo/diagnóstico por imagen , Plexo Coroideo/crecimiento & desarrollo , Plexo Coroideo/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Hidrocefalia/congénito , Hidrocefalia/diagnóstico , Hidrocefalia/fisiopatología , Inyecciones Intraventriculares , Presión Intracraneal/fisiología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Transgénicos , Miembro 2 de la Familia de Transportadores de Soluto 12/genética
2.
Mol Neurodegener ; 15(1): 29, 2020 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-32448329

RESUMEN

Huntington's disease (HD) is an incurable neurodegenerative disorder caused by CAG trinucleotide expansions in the huntingtin gene. Markers of both systemic and CNS immune activation and inflammation have been widely noted in HD and mouse models of HD. In particular, elevation of the pro-inflammatory cytokine interleukin-6 (IL-6) is the earliest reported marker of immune activation in HD, and this elevation has been suggested to contribute to HD pathogenesis. To test the hypothesis that IL-6 deficiency would be protective against the effects of mutant huntingtin, we generated R6/2 HD model mice that lacked IL-6. Contrary to our prediction, IL-6 deficiency exacerbated HD-model associated behavioral phenotypes. Single nuclear RNA Sequencing (snRNA-seq) analysis of striatal cell types revealed that IL-6 deficiency led to the dysregulation of various genes associated with synaptic function, as well as the BDNF receptor Ntrk2. These data suggest that IL-6 deficiency exacerbates the effects of mutant huntingtin through dysregulation of genes of known relevance to HD pathobiology in striatal neurons, and further suggest that modulation of IL-6 to a level that promotes proper regulation of genes associated with synaptic function may hold promise as an HD therapeutic target.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Huntington/metabolismo , Interleucina-6/deficiencia , Fenotipo , Animales , Encéfalo/fisiopatología , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Huntington/genética , Interleucina-6/metabolismo , Ratones Transgénicos
3.
Neuron ; 106(1): 76-89.e8, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32004439

RESUMEN

Unbiased in vivo genome-wide genetic screening is a powerful approach to elucidate new molecular mechanisms, but such screening has not been possible to perform in the mammalian central nervous system (CNS). Here, we report the results of the first genome-wide genetic screens in the CNS using both short hairpin RNA (shRNA) and CRISPR libraries. Our screens identify many classes of CNS neuronal essential genes and demonstrate that CNS neurons are particularly sensitive not only to perturbations to synaptic processes but also autophagy, proteostasis, mRNA processing, and mitochondrial function. These results reveal a molecular logic for the common implication of these pathways across multiple neurodegenerative diseases. To further identify disease-relevant genetic modifiers, we applied our screening approach to two mouse models of Huntington's disease (HD). Top mutant huntingtin toxicity modifier genes included several Nme genes and several genes involved in methylation-dependent chromatin silencing and dopamine signaling, results that reveal new HD therapeutic target pathways.


Asunto(s)
Supervivencia Celular/genética , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Neostriado/metabolismo , Neuronas/metabolismo , Animales , Conducta Animal , Sistemas CRISPR-Cas , Técnicas de Silenciamiento del Gen , Biblioteca de Genes , Genes Esenciales/genética , Ratones , Ratones Transgénicos , Nucleósido Difosfato Quinasas NM23/genética , Nucleósido Difosfato Quinasa D/genética , Agregado de Proteínas , Interferencia de ARN , ARN Guía de Kinetoplastida , ARN Interferente Pequeño , Receptores de Dopamina D2/genética , Análisis de Secuencia de ARN
4.
Cell ; 178(3): 521-535.e23, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31348885

RESUMEN

Intracellular accumulation of misfolded proteins causes toxic proteinopathies, diseases without targeted therapies. Mucin 1 kidney disease (MKD) results from a frameshift mutation in the MUC1 gene (MUC1-fs). Here, we show that MKD is a toxic proteinopathy. Intracellular MUC1-fs accumulation activated the ATF6 unfolded protein response (UPR) branch. We identified BRD4780, a small molecule that clears MUC1-fs from patient cells, from kidneys of knockin mice and from patient kidney organoids. MUC1-fs is trapped in TMED9 cargo receptor-containing vesicles of the early secretory pathway. BRD4780 binds TMED9, releases MUC1-fs, and re-routes it for lysosomal degradation, an effect phenocopied by TMED9 deletion. Our findings reveal BRD4780 as a promising lead for the treatment of MKD and other toxic proteinopathies. Generally, we elucidate a novel mechanism for the entrapment of misfolded proteins by cargo receptors and a strategy for their release and anterograde trafficking to the lysosome.


Asunto(s)
Benzamidas/metabolismo , Compuestos Bicíclicos con Puentes/farmacología , Heptanos/farmacología , Lisosomas/efectos de los fármacos , Proteínas de Transporte Vesicular/metabolismo , Factor de Transcripción Activador 6/metabolismo , Animales , Benzamidas/química , Benzamidas/farmacología , Compuestos Bicíclicos con Puentes/uso terapéutico , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Mutación del Sistema de Lectura , Heptanos/uso terapéutico , Humanos , Receptores de Imidazolina/antagonistas & inhibidores , Receptores de Imidazolina/genética , Receptores de Imidazolina/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/citología , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Lisosomas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mucina-1/química , Mucina-1/genética , Mucina-1/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteínas de Transporte Vesicular/química
5.
Proc Natl Acad Sci U S A ; 115(8): E1896-E1905, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29437957

RESUMEN

The decline of cognitive function occurs with aging, but the mechanisms responsible are unknown. Astrocytes instruct the formation, maturation, and elimination of synapses, and impairment of these functions has been implicated in many diseases. These findings raise the question of whether astrocyte dysfunction could contribute to cognitive decline in aging. We used the Bac-Trap method to perform RNA sequencing of astrocytes from different brain regions across the lifespan of the mouse. We found that astrocytes have region-specific transcriptional identities that change with age in a region-dependent manner. We validated our findings using fluorescence in situ hybridization and quantitative PCR. Detailed analysis of the differentially expressed genes in aging revealed that aged astrocytes take on a reactive phenotype of neuroinflammatory A1-like reactive astrocytes. Hippocampal and striatal astrocytes up-regulated a greater number of reactive astrocyte genes compared with cortical astrocytes. Moreover, aged brains formed many more A1 reactive astrocytes in response to the neuroinflammation inducer lipopolysaccharide. We found that the aging-induced up-regulation of reactive astrocyte genes was significantly reduced in mice lacking the microglial-secreted cytokines (IL-1α, TNF, and C1q) known to induce A1 reactive astrocyte formation, indicating that microglia promote astrocyte activation in aging. Since A1 reactive astrocytes lose the ability to carry out their normal functions, produce complement components, and release a toxic factor which kills neurons and oligodendrocytes, the aging-induced up-regulation of reactive genes by astrocytes could contribute to the cognitive decline in vulnerable brain regions in normal aging and contribute to the greater vulnerability of the aged brain to injury.


Asunto(s)
Envejecimiento/metabolismo , Astrocitos/metabolismo , Envejecimiento/genética , Envejecimiento/psicología , Animales , Cognición , Femenino , Perfilación de la Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Neuronas/metabolismo , ARN/genética , ARN/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
Cell Metab ; 22(6): 1059-67, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26655697

RESUMEN

In this report we evaluated the functions of hypothalamic amylin in vivo and in vitro. Profiling of hypothalamic neurons revealed that islet amyloid polypeptide (Iapp, precursor to amylin) is expressed in neurons in the lateral hypothalamus, arcuate nucleus, medial preoptic area, and elsewhere. Hypothalamic expression of lapp is markedly decreased in ob/ob mice and normalized by exogenous leptin. In slices, amylin and leptin had similar electrophysiologic effects on lateral hypothalamic leptin receptor ObRb-expressing neurons, while the amylin antagonist AC187 inhibited their activity and blunted the effect of leptin. Finally, i.c.v. infusion of AC187 acutely reduced the anorectic effects of leptin. These data show that hypothalamic amylin is transcriptionally regulated by leptin, that it can act directly on ObRb neurons in concert with leptin, and that it regulates feeding. These findings provide a potential mechanism for the increased efficacy of a metreleptin/pramlintide combination therapy for obesity.


Asunto(s)
Hipotálamo/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Leptina/metabolismo , Animales , Ingestión de Alimentos/efectos de los fármacos , Encefalinas/genética , Encefalinas/metabolismo , Femenino , Hipotálamo/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Leptina/sangre , Leptina/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía Confocal , Neuronas/metabolismo , Técnicas de Placa-Clamp , Fragmentos de Péptidos/farmacología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
7.
J Neurosci ; 35(30): 10762-72, 2015 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-26224859

RESUMEN

Hyperactivation of the mechanistic target of rapamycin (mTOR) kinase, as a result of loss-of-function mutations in tuberous sclerosis complex 1 (TSC1) or TSC2 genes, causes protein synthesis dysregulation, increased cell size, and aberrant neuronal connectivity. Dysregulated synthesis of synaptic proteins has been implicated in the pathophysiology of autism spectrum disorder (ASD) associated with TSC and fragile X syndrome. However, cell type-specific translational profiles in these disease models remain to be investigated. Here, we used high-fidelity and unbiased Translating Ribosome Affinity Purification (TRAP) methodology to purify ribosome-associated mRNAs and identified translational alterations in a rat neuronal culture model of TSC. We find that expression of many stress and/or activity-dependent proteins is highly induced while some synaptic proteins are repressed. Importantly, transcripts for the activating transcription factor-3 (Atf3) and mitochondrial uncoupling protein-2 (Ucp2) are highly induced in Tsc2-deficient neurons, as well as in a neuron-specific Tsc1 conditional knock-out mouse model, and show differential responses to the mTOR inhibitor rapamycin. Gelsolin, a known target of Atf3 transcriptional activity, is also upregulated. shRNA-mediated block of Atf3 induction suppresses expression of gelsolin, an actin-severing protein, and rescues spine deficits found in Tsc2-deficient neurons. Together, our data demonstrate that a cell-autonomous program consisting of a stress-induced Atf3-gelsolin cascade affects the change in dendritic spine morphology following mTOR hyperactivation. This previously unidentified molecular cascade could be a therapeutic target for treating mTORopathies. SIGNIFICANCE STATEMENT: Tuberous sclerosis complex (TSC) is a genetic disease associated with epilepsy and autism. Dysregulated protein synthesis has been implicated as a cause of this disease. However, cell type-specific translational profiles that are aberrant in this disease are unknown. Here we show that expression of many stress and/or activity-dependent proteins is highly induced while some synaptic proteins are repressed in neurons missing the Tsc2 gene expression. Identification of genes whose translation is abnormal in TSC may provide insights to previously unidentified therapeutic targets.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Gelsolina/metabolismo , Esclerosis Tuberosa/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Inmunohistoquímica , Masculino , Ratones , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Transcriptoma , Transfección , Esclerosis Tuberosa/patología
8.
J Neurochem ; 132(6): 677-86, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25639954

RESUMEN

Metabotropic glutamate receptor 5 (mGluR5) regulates excitatory post-synaptic signaling in the central nervous system (CNS) and is implicated in various CNS disorders. Protein kinase A (PKA) signaling is known to play a critical role in neuropsychiatric disorders such as Parkinson's disease, schizophrenia, and addiction. Dopamine signaling is known to modulate the properties of mGluR5 in a cAMP- and PKA-dependent manner, suggesting that mGluR5 may be a direct target for PKA. Our study identifies mGluR5 at Ser870 as a direct substrate for PKA phosphorylation and demonstrates that this phosphorylation plays a critical role in the PKA-mediated modulation of mGluR5 functions such as extracellular signal-regulated kinase phosphorylation and intracellular Ca(2+) oscillations. The identification of the molecular mechanism by which PKA signaling modulates mGluR5-mediated cellular responses contributes to the understanding of the interaction between dopaminergic and glutamatergic neuronal signaling. We identified serine residue 870 (S870) in metabotropic glutamate receptor 5 (mGluR5) as a direct substrate for protein kinase A (PKA). The phosphorylation of this site regulates the ability of mGluR5 to induce extracellular signal-regulated kinase (ERK) phosphorylation and intracellular Ca(2+) oscillations. This study provides a direct molecular mechanism by which PKA signaling interacts with glutamate neurotransmission.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptor del Glutamato Metabotropico 5/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Fosforilación/fisiología
9.
Proc Natl Acad Sci U S A ; 111(49): 17636-41, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25413364

RESUMEN

Recurrent axon collaterals are a major means of communication between spiny projection neurons (SPNs) in the striatum and profoundly affect the function of the basal ganglia. However, little is known about the molecular and cellular mechanisms that underlie this communication. We show that intrastriatal nitric oxide (NO) signaling elevates the expression of the vesicular GABA transporter (VGAT) within recurrent collaterals of SPNs. Down-regulation of striatal NO signaling resulted in an attenuation of GABAergic signaling in SPN local collaterals, down-regulation of VGAT expression in local processes of SPNs, and impaired motor behavior. PKG1 and cAMP response element-binding protein are involved in the signal transduction that transcriptionally regulates VGAT by NO. These data suggest that transcriptional control of the vesicular GABA transporter by NO regulates GABA transmission and action selection.


Asunto(s)
Ganglios Basales/metabolismo , Guanilato Ciclasa/química , Neuronas/metabolismo , Óxido Nítrico/química , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/química , Animales , Axones/metabolismo , AMP Cíclico/metabolismo , Dopamina/metabolismo , Electrofisiología , Retroalimentación Fisiológica , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Levodopa/química , Masculino , Ratones , Plasticidad Neuronal , Oxidopamina/química , Transducción de Señal , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
10.
Proc Natl Acad Sci U S A ; 104(10): 4159-64, 2007 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-17360493

RESUMEN

Alzheimer's disease (AD) is associated with accumulation of the neurotoxic peptide amyloid-beta (Abeta), which is produced by sequential cleavage of amyloid precursor protein (APP) by the aspartyl protease beta-secretase and the presenilin-dependent protease gamma-secretase. An increase of casein kinase 1 (CK1) expression has been described in the human AD brain. We show, by using in silico analysis, that APP, beta-secretase, and gamma-secretase subunits contain, in their intracellular regions, multiple CK1 consensus phosphorylation sites, many of which are conserved among human, rat, and mouse species. Overexpression of constitutively active CK1epsilon, one of the CK1 isoforms expressed in brain, leads to an increase in Abeta peptide production. Conversely, three structurally dissimilar CK1-specific inhibitors significantly reduced endogenous Abeta peptide production. By using mammalian cells expressing the beta C-terminal fragment of APP, it was possible to demonstrate that CK1 inhibitors act at the level of gamma-secretase cleavage. Importantly, Notch cleavage was not affected. Our results indicate that CK1 represents a therapeutic target for prevention of Abeta formation in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Quinasa de la Caseína I/fisiología , Regulación de la Expresión Génica , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Quinasa de la Caseína I/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Enfermedades Neurodegenerativas/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA