Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(45): e2301534120, 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37903257

RESUMEN

L-type voltage-gated calcium (Ca2+) channels (L-VGCC) dysfunction is implicated in several neurological and psychiatric diseases. While a popular therapeutic target, it is unknown whether molecular mechanisms leading to disrupted L-VGCC across neurodegenerative disorders are conserved. Importantly, L-VGCC integrate synaptic signals to facilitate a plethora of cellular mechanisms; however, mechanisms that regulate L-VGCC channel density and subcellular compartmentalization are understudied. Herein, we report that in disease models with overactive mammalian target of rapamycin complex 1 (mTORC1) signaling (or mTORopathies), deficits in dendritic L-VGCC activity are associated with increased expression of the RNA-binding protein (RBP) Parkinsonism-associated deglycase (DJ-1). DJ-1 binds the mRNA coding for the alpha and auxiliary Ca2+ channel subunits CaV1.2 and α2δ2, and represses their mRNA translation, only in the disease states, specifically preclinical models of tuberous sclerosis complex (TSC) and Alzheimer's disease (AD). In agreement, DJ-1-mediated repression of CaV1.2/α2δ2 protein synthesis in dendrites is exaggerated in mouse models of AD and TSC, resulting in deficits in dendritic L-VGCC calcium activity. Finding of DJ-1-regulated L-VGCC activity in dendrites in TSC and AD provides a unique signaling pathway that can be targeted in clinical mTORopathies.


Asunto(s)
Enfermedad de Alzheimer , Esclerosis Tuberosa , Animales , Ratones , Enfermedad de Alzheimer/genética , Calcio/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Dendritas/metabolismo , Mamíferos/metabolismo , Esclerosis Tuberosa/genética
2.
J Neurochem ; 152(5): 523-541, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31376158

RESUMEN

N-methyl-d-aspartate receptors (NMDARs) mediate both physiological and pathophysiological processes, although selective ligands lack broad clinical utility. NMDARs are composed of multiple subunits, but N-methyl-d-aspartate receptor subunit 2 (GluN2) is predominately responsible for functional heterogeneity. Specifically, the GluN2A- and GluN2B-containing subtypes are enriched in adult hippocampus and cortex and impact neuronal communication via dynamic trafficking into and out of the synapse. We sought to understand if ((2S, 3R)-3-hydroxy-2-((R)-5-isobutyryl-1-oxo-2,5-diazaspiro[3,4]octan-2-yl) butanamide (NYX-2925), a novel NMDAR modulator, alters synaptic levels of GluN2A- or GluN2B-containing NMDARs. Low-picomolar NYX-2925 increased GluN2B colocalization with the excitatory post-synaptic marker post-synaptic density protein 95 (PSD-95) in rat primary hippocampal neurons within 30 min. Twenty-four hours following oral administration, 1 mg/kg NYX-2925 increased GluN2B in PSD-95-associated complexes ex vivo, and low-picomolar NYX-2925 regulated numerous trafficking pathways in vitro. Because the NYX-2925 concentration that increases synaptic GluN2B was markedly below that which enhances long-term potentiation (mid-nanomolar), we sought to elucidate the basis of this effect. Although NMDAR-dependent, NYX-2925-mediated colocalization of GluN2B with PSD-95 occurred independent of ion flux, as colocalization increased in the presence of either the NMDAR channel blocker (5R,10S)-(-)-5-Methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate or glycine site antagonist 7-chlorokynurenic acid. Moreover, while mid-nanomolar NYX-2925 concentrations, which do not increase synaptic GluN2B, enhanced calcium transients, functional plasticity was only enhanced by picomolar NYX-2925. Thus, NYX-2925 concentrations that increase synaptic GluN2B facilitated the chemical long-term potentiation induced insertion of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluA1 subunit levels. Basal (unstimulated by chemical long-term potentiation) levels of synaptic GluA1 were only increased by mid-nanomolar NYX-2925. These data suggest that NYX-2925 facilitates homeostatic plasticity by initially increasing synaptic GluN2B via metabotropic-like NMDAR signaling. Cover Image for this issue: doi: 10.1111/jnc.14735.


Asunto(s)
Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Compuestos de Espiro/farmacología , Sinapsis/metabolismo , Animales , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Receptores AMPA/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos
3.
Nat Commun ; 7: 12867, 2016 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-27666021

RESUMEN

Alcohol promotes lasting neuroadaptive changes that may provide relief from depressive symptoms, often referred to as the self-medication hypothesis. However, the molecular/synaptic pathways that are shared by alcohol and antidepressants are unknown. In the current study, acute exposure to ethanol produced lasting antidepressant and anxiolytic behaviours. To understand the functional basis of these behaviours, we examined a molecular pathway that is activated by rapid antidepressants. Ethanol, like rapid antidepressants, alters γ-aminobutyric acid type B receptor (GABABR) expression and signalling, to increase dendritic calcium. Furthermore, new GABABRs are synthesized in response to ethanol treatment, requiring fragile-X mental retardation protein (FMRP). Ethanol-dependent changes in GABABR expression, dendritic signalling, and antidepressant efficacy are absent in Fmr1-knockout (KO) mice. These findings indicate that FMRP is an important regulator of protein synthesis following alcohol exposure, providing a molecular basis for the antidepressant efficacy of acute ethanol exposure.

4.
J Biol Chem ; 290(26): 16357-71, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25944900

RESUMEN

The fate of a memory, whether stored or forgotten, is determined by the ability of an active or tagged synapse to undergo changes in synaptic efficacy requiring protein synthesis of plasticity-related proteins. A synapse can be tagged, but without the "capture" of plasticity-related proteins, it will not undergo long lasting forms of plasticity (synaptic tagging and capture hypothesis). What the "tag" is and how plasticity-related proteins are captured at tagged synapses are unknown. Ca(2+)/calmodulin-dependent protein kinase II α (CaMKIIα) is critical in learning and memory and is synthesized locally in neuronal dendrites. The mechanistic (mammalian) target of rapamycin (mTOR) is a protein kinase that increases CaMKIIα protein expression; however, the mechanism and site of dendritic expression are unknown. Herein, we show that mTOR activity mediates the branch-specific expression of CaMKIIα, favoring one secondary, daughter branch over the other in a single neuron. mTOR inhibition decreased the dendritic levels of CaMKIIα protein and mRNA by shortening its poly(A) tail. Overexpression of the RNA-stabilizing protein HuD increased CaMKIIα protein levels and preserved its selective expression in one daughter branch over the other when mTOR was inhibited. Unexpectedly, deleting the third RNA recognition motif of HuD, the domain that binds the poly(A) tail, eliminated the branch-specific expression of CaMKIIα when mTOR was active. These results provide a model for one molecular mechanism that may underlie the synaptic tagging and capture hypothesis where mTOR is the tag, preventing deadenylation of CaMKIIα mRNA, whereas HuD captures and promotes its expression in a branch-specific manner.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Dendritas/metabolismo , Proteínas ELAV/metabolismo , ARN Mensajero/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Dendritas/enzimología , Dendritas/genética , Proteínas ELAV/genética , Proteína 4 Similar a ELAV , Hipocampo/citología , Hipocampo/enzimología , Hipocampo/metabolismo , Neuronas/metabolismo , Unión Proteica , ARN Mensajero/genética , Ratas , Sinapsis/enzimología , Sinapsis/genética , Sinapsis/metabolismo , Serina-Treonina Quinasas TOR/genética
5.
Ann Neurol ; 75(6): 864-75, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24659129

RESUMEN

OBJECTIVE: Acquired epilepsy is frequently associated with structural lesions after trauma, stroke, and infections. Although seizures are often difficult to treat, there is no clinically applicable strategy to prevent the development of epilepsy in patients at risk. We have recently shown that vascular injury is associated with activation of albumin-mediated transforming growth factor ß (TGF-ß) signaling, and followed by local inflammatory response and epileptiform activity ex vivo. Here we investigated albumin-mediated TGF-ß signaling and tested the efficacy of blocking the TGF-ß pathway in preventing epilepsy. METHODS: We addressed the role of TGF-ß signaling in epileptogenesis in 2 different rat models of vascular injury, combining in vitro and in vivo biochemical assays, gene expression, and magnetic resonance and direct optical imaging for blood-brain barrier permeability and vascular reactivity. Long-term electrocorticographic recordings were acquired in freely behaving animals. RESULTS: We demonstrate that serum-derived albumin preferentially induces activation of the activin receptor-like kinase 5 pathway of TGF-ß receptor I in astrocytes. We further show that the angiotensin II type 1 receptor antagonist, losartan, previously identified as a blocker of peripheral TGF-ß signaling, effectively blocks albumin-induced TGF-ß activation in the brain. Most importantly, losartan prevents the development of delayed recurrent spontaneous seizures, an effect that persists weeks after drug withdrawal. INTERPRETATION: TGF-ß signaling, activated in astrocytes by serum-derived albumin, is involved in epileptogenesis. We propose losartan, a drug approved by the US Food and Drug Administration, as an efficient antiepileptogenic therapy for epilepsy associated with vascular injury.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/prevención & control , Losartán/uso terapéutico , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Recién Nacidos , Anticonvulsivantes/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Benzamidas/farmacología , Barrera Hematoencefálica/fisiología , Células Cultivadas , Corteza Cerebral/citología , Dioxoles/farmacología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Endocitosis/efectos de los fármacos , Epilepsia/inducido químicamente , Epilepsia/patología , Epilepsia/fisiopatología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética
6.
J Cell Biol ; 202(1): 53-69, 2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-23836929

RESUMEN

Little is known about how a neuron undergoes site-specific changes in intrinsic excitability during neuronal activity. We provide evidence for a novel mechanism for mTORC1 kinase-dependent translational regulation of the voltage-gated potassium channel Kv1.1 messenger RNA (mRNA). We identified a microRNA, miR-129, that repressed Kv1.1 mRNA translation when mTORC1 was active. When mTORC1 was inactive, we found that the RNA-binding protein, HuD, bound to Kv1.1 mRNA and promoted its translation. Unexpectedly, inhibition of mTORC1 activity did not alter levels of miR-129 and HuD to favor binding to Kv1.1 mRNA. However, reduced mTORC1 signaling caused the degradation of high affinity HuD target mRNAs, freeing HuD to bind Kv1.1 mRNA. Hence, mTORC1 activity regulation of mRNA stability and high affinity HuD-target mRNA degradation mediates the bidirectional expression of dendritic Kv1.1 ion channels.


Asunto(s)
Proteínas ELAV/metabolismo , Canal de Potasio Kv.1.1/metabolismo , MicroARNs/metabolismo , Proteínas/metabolismo , Proteolisis , ARN Mensajero/metabolismo , Animales , Sitios de Unión , Proteínas ELAV/genética , Proteína 4 Similar a ELAV , Activación Enzimática , Células HEK293 , Humanos , Canal de Potasio Kv.1.1/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , MicroARNs/genética , Complejos Multiproteicos , Plasticidad Neuronal , Neuronas/metabolismo , Biosíntesis de Proteínas , Proteínas/genética , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , ARN Mensajero/genética , Ratas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR , Transfección
7.
Cardiovasc Psychiatry Neurol ; 2011: 461263, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21547249

RESUMEN

Clinical and experimental data suggest that stress contributes to the pathology of epilepsy. We review mechanisms by which stress, primarily via stress hormones, may exacerbate epilepsy, focusing on the intersection between stress-induced pathways and the progression of pathological events that occur before, during, and after the onset of epileptogenesis. In addition to this temporal nuance, we discuss other complexities in stress-epilepsy interactions, including the role of blood-brain barrier dysfunction, neuron-glia interactions, and inflammatory/cytokine pathways that may be protective or damaging depending on context. We advocate the use of global analytical tools, such as microarray, in support of a shift away from a narrow focus on seizures and towards profiling the complex, early process of epileptogenesis, in which multiple pathways may interact to dictate the ultimate onset of chronic, recurring seizures.

8.
J Neurosci ; 29(34): 10588-99, 2009 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-19710312

RESUMEN

Focal epilepsy often develops following traumatic, ischemic, or infectious brain injury. While the electrical activity of the epileptic brain is well characterized, the mechanisms underlying epileptogenesis are poorly understood. We have recently shown that in the rat neocortex, long-lasting breakdown of the blood-brain barrier (BBB) or direct exposure of the neocortex to serum-derived albumin leads to rapid upregulation of the astrocytic marker GFAP (glial fibrillary acidic protein), followed by delayed (within 4-7 d) development of an epileptic focus. We investigated the role of astrocytes in epileptogenesis in the BBB-breakdown and albumin models of epileptogenesis. We found similar, robust changes in astrocytic gene expression in the neocortex within hours following treatment with deoxycholic acid (BBB breakdown) or albumin. These changes predict reduced clearance capacity for both extracellular glutamate and potassium. Electrophysiological recordings in vitro confirmed the reduced clearance of activity-dependent accumulation of both potassium and glutamate 24 h following exposure to albumin. We used a NEURON model to simulate the consequences of reduced astrocytic uptake of potassium and glutamate on EPSPs. The model predicted that the accumulation of glutamate is associated with frequency-dependent (>100 Hz) decreased facilitation of EPSPs, while potassium accumulation leads to frequency-dependent (10-50 Hz) and NMDA-dependent synaptic facilitation. In vitro electrophysiological recordings during epileptogenesis confirmed frequency-dependent synaptic facilitation leading to seizure-like activity. Our data indicate a transcription-mediated astrocytic transformation early during epileptogenesis. We suggest that the resulting reduction in the clearance of extracellular potassium underlies frequency-dependent neuronal hyperexcitability and network synchronization.


Asunto(s)
Astrocitos , Epilepsia/patología , Epilepsia/fisiopatología , Regulación de la Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácido Glutámico/metabolismo , Potasio/metabolismo , Albúminas , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Simulación por Computador , Ácido Desoxicólico/toxicidad , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Epilepsia/complicaciones , Agonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Masculino , Modelos Neurológicos , Neocórtex/patología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Técnicas de Placa-Clamp , Ratas , Ratas Wistar
9.
J Neurosci ; 29(28): 8927-35, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19605630

RESUMEN

Brain injury may result in the development of epilepsy, one of the most common neurological disorders. We previously demonstrated that albumin is critical in the generation of epilepsy after blood-brain barrier (BBB) compromise. Here, we identify TGF-beta pathway activation as the underlying mechanism. We demonstrate that direct activation of the TGF-beta pathway by TGF-beta1 results in epileptiform activity similar to that after exposure to albumin. Coimmunoprecipitation revealed binding of albumin to TGF-beta receptor II, and Smad2 phosphorylation confirmed downstream activation of this pathway. Transcriptome profiling demonstrated similar expression patterns after BBB breakdown, albumin, and TGF-beta1 exposure, including modulation of genes associated with the TGF-beta pathway, early astrocytic activation, inflammation, and reduced inhibitory transmission. Importantly, TGF-beta pathway blockers suppressed most albumin-induced transcriptional changes and prevented the generation of epileptiform activity. Our present data identifies the TGF-beta pathway as a novel putative epileptogenic signaling cascade and therapeutic target for the prevention of injury-induced epilepsy.


Asunto(s)
Encéfalo/metabolismo , Epilepsia/etiología , Perfilación de la Expresión Génica/métodos , Expresión Génica/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Potenciales de Acción/fisiología , Albúminas/metabolismo , Animales , Anticuerpos/farmacología , Astrocitos/metabolismo , Benzamidas/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/fisiopatología , Análisis por Conglomerados , Dioxoles/farmacología , Modelos Animales de Enfermedad , Estimulación Eléctrica/efectos adversos , Epilepsia/complicaciones , Epilepsia/patología , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo/métodos , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Inmunoprecipitación/métodos , Técnicas In Vitro , Inflamación/etiología , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Análisis por Micromatrices/métodos , Ratas , Ratas Wistar , Proteína Smad2/metabolismo , Estadísticas no Paramétricas , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta2/antagonistas & inhibidores , Factor de Crecimiento Transformador beta2/inmunología , Factor de Crecimiento Transformador beta2/metabolismo , Ácido gamma-Aminobutírico/genética , Ácido gamma-Aminobutírico/metabolismo
10.
J Pharmacol Exp Ther ; 315(2): 517-25, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16033909

RESUMEN

Propofol (2,6-diisopropylphenol) is a widely used intravenous general anesthetic, which has been reported to produce bradycardia in patients at concentrations associated with profound sedation and loss of consciousness. Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels conduct a monovalent cationic current I(h) (also known as I(q) or I(f)) that contributes to autorhythmicity in both the brain and heart. Here we studied the effects of propofol on recombinant HCN1, HCN2, and HCN4 channels and found that the drug inhibits and slows activation of all three channels at clinically relevant concentrations. In oocyte expression studies, HCN1 channel activation was most sensitive to slowing by propofol (EC(50) values of 5.6 +/- 1.0 microM for fast component and 31.5 +/- 7.5 microM for slow component). HCN1 channels also showed a marked propofol-induced hyperpolarizing shift in the voltage dependence of activation (EC(50) of 6.7 +/- 1.0 microM) and accelerated deactivation (EC(50) of 4.5 +/- 0.9 microM). Furthermore, propofol reduced heart rate in an isolated guinea pig heart preparation over the same range of concentrations. These data suggest that propofol modulation of HCN channel gating is an important molecular mechanism that can contribute to the depression of central nervous system function and also lead to bradyarrhythmias in patients receiving propofol during surgical anesthesia.


Asunto(s)
Anestésicos Intravenosos/farmacología , Activación del Canal Iónico/efectos de los fármacos , Canales Iónicos/efectos de los fármacos , Proteínas Musculares/efectos de los fármacos , Proteínas del Tejido Nervioso/efectos de los fármacos , Propofol/farmacología , Animales , Relojes Biológicos/efectos de los fármacos , Canales Catiónicos Regulados por Nucleótidos Cíclicos , ADN Complementario/biosíntesis , ADN Complementario/genética , Electrocardiografía/efectos de los fármacos , Electrofisiología , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Cinética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Oocitos/efectos de los fármacos , Técnicas de Placa-Clamp , Canales de Potasio , Nodo Sinoatrial/citología , Nodo Sinoatrial/efectos de los fármacos , Xenopus laevis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA