Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Sci Rep ; 5: 10256, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26194093

RESUMEN

ß-amyloid (Aß) oligomers have been closely implicated in the pathogenesis of Alzheimer's disease (AD). We found, for the first time, that bis(heptyl)-cognitin, a novel dimeric acetylcholinesterase (AChE) inhibitor derived from tacrine, prevented Aß oligomers-induced inhibition of long-term potentiation (LTP) at concentrations that did not interfere with normal LTP. Bis(heptyl)-cognitin also prevented Aß oligomers-induced synaptotoxicity in primary hippocampal neurons. In contrast, tacrine and donepezil, typical AChE inhibitors, could not prevent synaptic impairments in these models, indicating that the modification of Aß oligomers toxicity by bis(heptyl)-cognitin might be attributed to a mechanism other than AChE inhibition. Studies by using dot blotting, immunoblotting, circular dichroism spectroscopy, and transmission electron microscopy have shown that bis(heptyl)-cognitin altered Aß assembly via directly inhibiting Aß oligomers formation and reducing the amount of preformed Aß oligomers. Molecular docking analysis further suggested that bis(heptyl)-cognitin presumably interacted with the hydrophobic pockets of Aß, which confers stabilizing powers and assembly alteration effects on Aß. Most importantly, bis(heptyl)-cognitin significantly reduced cognitive impairments induced by intra-hippocampal infusion of Aß oligomers in mice. These results clearly demonstrated how dimeric agents prevent Aß oligomers-induced synaptic and memory impairments, and offered a strong support for the beneficial therapeutic effects of bis(heptyl)-cognitin in the treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Sinapsis/patología , Tacrina/análogos & derivados , Tacrina/uso terapéutico , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/ultraestructura , Animales , Células Cultivadas , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Estimulación Eléctrica , Hipocampo/patología , Interacciones Hidrofóbicas e Hidrofílicas , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones Endogámicos ICR , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Estructura Secundaria de Proteína , Ratas Sprague-Dawley , Ratas Wistar , Sinapsis/efectos de los fármacos , Tacrina/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
2.
J Alzheimers Dis ; 39(2): 239-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24150104

RESUMEN

Rosiglitazone has been known to attenuate neurodegeneration in Alzheimer's disease (AD), but the underlying mechanisms remain to be fully elucidated. In this study, living-cell image, immunocytochemistry, and electrophysiology were used to examine the effects of soluble amyloid-ß protein (Aß) oligomers and rosiglitazone on the synapse formation, plasticity, and mitochondrial distribution in cultured neurons. Incubation of hippocampal cultures with amyloid-ß (Aß)42 oligomers (0.5 µM) for 3 h significantly decreased dendritic filopodium and synapse density. Pretreatment with rosiglitazone (0.5-5 µM) for 24 h prevented the Aß42-induced loss of dendritic filopodium and synapse in a dose-dependent manner. However, neither Aß42 oligomer nor rosiglitazone has a significant effect on the velocity and length of dendritic filopodia. Electrophysiological recording showed that acute exposure of slices with 0.5 µM Aß42 oligomers impaired hippocampal long-term potentiation (LTP). Pre-incubation of hippocampal slices with rosiglitazone significantly attenuated the Aß42-induced LTP deficit, which depended on rosiglitazone concentrations (1-5 µM) and pretreatment period (1-5 h). The beneficial effects of rosiglitazone were abolished by the peroxisome proliferator-activated receptor gamma (PPARγ) specific antagonist, GW9662. Moreover, the mitochondrial numbers in the dendrite and spine were decreased by Aß42 oligomers, which can be prevented by rosiglitazone. In conclusion, our data suggested that rosiglitazone prevents Aß42 oligomers-induced impairment via increasing mitochondrial numbers in the dendrite and spine, improving synapse formation and plasticity. This process is most likely through the PPARγ-dependent pathway and in concentration and time dependent manners. The study provides novel insights into the mechanisms for the protective effects of rosiglitzone on AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Mitocondrias/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Nootrópicos/farmacología , Fragmentos de Péptidos/toxicidad , Tiazolidinedionas/farmacología , Anilidas/farmacología , Animales , Células Cultivadas , Fármacos del Sistema Nervioso Central/farmacología , Dendritas/efectos de los fármacos , Dendritas/patología , Dendritas/fisiología , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/patología , Espinas Dendríticas/fisiología , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/fisiopatología , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Mitocondrias/patología , Mitocondrias/fisiología , Plasticidad Neuronal/fisiología , Neuronas/patología , Neuronas/fisiología , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , Ratas , Ratas Wistar , Rosiglitazona , Sinapsis/efectos de los fármacos , Sinapsis/patología , Sinapsis/fisiología , Factores de Tiempo
3.
Neural Regen Res ; 8(1): 49-55, 2013 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25206371

RESUMEN

Previous studies indicate that memantine, a low-affinity N-methyl-D-aspartate receptor antagonist, exerted acute protective effects against amyloid-ß protein-induced neurotoxicity. In the present study, the chronic effects and mechanisms of memantine were investigated further using electrophysiological methods. The results showed that 7-day intraperitoneal application of memantine, at doses of 5 mg/kg or 20 mg/kg, did not alter hippocampal long-term potentiation induction in rats, while 40 mg/kg memantine presented potent long-term potentiation inhibition. Then further in vitro studys were carried out in 5 mg/kg and 20 mg/kg memantine treated rats. We found that 20 mg/kg memantine attenuated the potent long-term potentiation inhibition caused by exposure to amyloid-ß protein in the dentate gyrus in vitro. These findings are the first to demonstrate the antagonizing effect of long-term systematic treatment of memantine against amyloid-ß protein triggered long-term potentiation inhibition to improve synaptic plasticity.

4.
J Physiol ; 590(16): 3771-86, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22615437

RESUMEN

Synaptic plasticity of NMDA receptors (NMDARs) has been recently described in a number of brain regions and we have previously characterised LTP and LTD of glutamatergic NMDA receptor-mediated EPSCs (NMDAR-EPSCs) in granule cells of dentate gyrus. The functional significance of NMDAR plasticity at perforant path synapses on hippocampal network activity depends on whether this is a common feature of perforant path synapses on all postsynaptic target cells or if this plasticity occurs only at synapses on principal cells. We recorded NMDAR-EPSCs at medial perforant path synapses on interneurons in dentate gyrus which had significantly slower decay kinetics compared to those recorded in granule cells. NMDAR pharmacology in interneurons was consistent with expression of both GluN2B- and GluN2D-containing receptors. In contrast to previously described high frequency stimulation-induced bidirectional plasticity of NMDAR-EPSCs in granule cells, only LTD of NMDAR-EPSCs was induced in interneurons in our standard experimental conditions. In interneurons, LTD of NMDAR-EPSCs was associated with a loss of sensitivity to a GluN2D-selective antagonist and was inhibited by the actin stabilising agent, jasplakinolide. While LTP of NMDAR-EPSCs can be readily induced in granule cells, this form of plasticity was only observed in interneurons when extracellular calcium was increased above physiological concentrations during HFS or when PKC was directly activated by phorbol ester, suggesting that opposing forms of plasticity at inputs to interneurons and principal cells may act to regulate granule cell dendritic integration and processing.


Asunto(s)
Dendritas/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Interneuronas/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Actinas , Potenciales de Acción , Animales , Giro Dentado/fisiología , Regulación de la Expresión Génica , Masculino , Plasticidad Neuronal , Ratas , Ratas Wistar , Sinapsis
5.
J Neurophysiol ; 107(1): 205-15, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21975445

RESUMEN

The induction of long-term potentiation (LTP) and long-term depression (LTD) of excitatory postsynaptic currents was investigated in proximal synapses of layer 2/3 pyramidal cells of the rat medial prefrontal cortex. The spike timing-dependent plasticity (STDP) induction protocol of negative timing, with postsynaptic leading presynaptic stimulation of action potentials (APs), induced LTD as expected from the classical STDP rule. However, the positive STDP protocol of presynaptic leading postsynaptic stimulation of APs predominantly induced a presynaptically expressed LTD rather than the expected postsynaptically expressed LTP. Thus the induction of plasticity in layer 2/3 pyramidal cells does not obey the classical STDP rule for positive timing. This unusual STDP switched to a classical timing rule if the slow Ca(2+)-dependent, K(+)-mediated afterhyperpolarization (sAHP) was inhibited by the selective blocker N-trityl-3-pyridinemethanamine (UCL2077), by the ß-adrenergic receptor agonist isoproterenol, or by the cholinergic agonist carbachol. Thus we demonstrate that neuromodulators can affect synaptic plasticity by inhibition of the sAHP. These findings shed light on a fundamental question in the field of memory research regarding how environmental and behavioral stimuli influence LTP, thereby contributing to the modulation of memory.


Asunto(s)
Potenciales de Acción/fisiología , Modelos Neurológicos , Red Nerviosa/fisiología , Inhibición Neural/fisiología , Plasticidad Neuronal/fisiología , Corteza Prefrontal/fisiología , Tractos Piramidales/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Ratas , Ratas Wistar
6.
Eur J Pharmacol ; 677(1-3): 63-70, 2012 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-22200627

RESUMEN

Nicotinic acetylcholine receptors mediate fast cholinergic modulation of glutamatergic transmission and synaptic plasticity. Here we investigated the effects of subtype selective activation of the α7 nicotinic acetylcholine receptors on hippocampal transmission and the inhibition of synaptic long-term potentiation by the Alzheimer's disease associated amyloid ß-protein (Aß). The α7 nicotinic acetylcholine receptor agonist "compound A" ((R)-N-(1-azabicyclo[2.2.2]oct-3-yl)(5-(2-pyridyl))thiophene-2-carboxamide) induced a rapid-onset persistent enhancement of synaptic transmission in the dentate gyrus in vitro. Consistent with a requirement for activation of α7 nicotinic acetylcholine receptors, the type II α7-selective positive allosteric modulator PheTQS ((3aR, 4S, 9bS)-4-(4-methylphenyl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinoline-8-sulfonamide) potentiated, and the antagonist methyllycaconitine (MLA) prevented the persistent enhancement. Systemic injection of the agonist also induced a similar MLA-sensitive persistent enhancement of synaptic transmission in the CA1 area in vivo. Remarkably, although compound A did not affect control long-term potentiation (LTP) in vitro, it prevented the inhibition of LTP by Aß1-42 and this effect was inhibited by MLA. These findings strongly indicate that activation of α7 nicotinic acetylcholine receptors is sufficient to persistently enhance hippocampal synaptic transmission and to overcome the inhibition of LTP by Aß.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Hipocampo/citología , Hipocampo/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores Nicotínicos/metabolismo , Transmisión Sináptica/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Animales , Interacciones Farmacológicas , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Masculino , Agonistas Nicotínicos/farmacología , Ratas , Ratas Wistar , Sulfonamidas/química , Sulfonamidas/farmacología , Tiofenos/química , Tiofenos/farmacología , Receptor Nicotínico de Acetilcolina alfa 7
7.
Nat Chem Biol ; 8(1): 93-101, 2011 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-22101602

RESUMEN

Several lines of evidence indicate that prefibrillar assemblies of amyloid-ß (Aß) polypeptides, such as soluble oligomers or protofibrils, rather than mature, end-stage amyloid fibrils cause neuronal dysfunction and memory impairment in Alzheimer's disease. These findings suggest that reducing the prevalence of transient intermediates by small molecule-mediated stimulation of amyloid polymerization might decrease toxicity. Here we demonstrate the acceleration of Aß fibrillogenesis through the action of the orcein-related small molecule O4, which directly binds to hydrophobic amino acid residues in Aß peptides and stabilizes the self-assembly of seeding-competent, ß-sheet-rich protofibrils and fibrils. Notably, the O4-mediated acceleration of amyloid fibril formation efficiently decreases the concentration of small, toxic Aß oligomers in complex, heterogeneous aggregation reactions. In addition, O4 treatment suppresses inhibition of long-term potentiation by Aß oligomers in hippocampal brain slices. These results support the hypothesis that small, diffusible prefibrillar amyloid species rather than mature fibrillar aggregates are toxic for mammalian cells.


Asunto(s)
Amiloide/química , Oxazinas/química , Fragmentos de Péptidos/química , Secuencia de Aminoácidos , Amiloide/toxicidad , Amiloide/ultraestructura , Línea Celular Tumoral , Hipocampo/química , Hipocampo/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Microscopía Electrónica de Transmisión , Modelos Moleculares , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/ultraestructura , Estructura Secundaria de Proteína , Transmisión Sináptica
8.
Neurobiol Aging ; 32(4): 614-23, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19446369

RESUMEN

Soluble amyloid-ß protein (Aß) may cause cognitive impairment in Alzheimer's disease in the absence of significant neurodegeneration. Here, the ability of the NMDA receptor (NMDAR) antagonist memantine to prevent synthetic Aß-mediated rapid functional deficits in learned behavior and synaptic plasticity was assessed in the rat. In vitro, pretreatment with a clinically relevant, NMDAR blocking concentration of memantine partially inhibited the induction of long-term potentiation (LTP) in the dentate gyrus and prevented further inhibition caused by exposure to Aß(1-42). Whereas systemic injection with memantine alone inhibited LTP in the CA1 area in vivo, a subthreshold dose partially abrogated the inhibition of LTP by intracerebroventricular soluble Aß(1-42). Similarly, systemic treatment with memantine alone impaired performance of an operant learning task and a subthreshold dose prevented the Aß(1-42)-mediated increase in perseveration errors. The acute protection afforded by memantine, albeit in a narrow dose range, against the rapid disruptive effects of soluble Aß(1-42) on synaptic plasticity and learned behavior strongly implicate NMDAR-dependent reversible dysfunction of synaptic mechanisms in Aß-mediated cognitive impairment.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Memantina/farmacología , Fármacos Neuroprotectores/farmacología , Sinapsis/efectos de los fármacos , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Electrofisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Ratas Wistar
9.
Proc Natl Acad Sci U S A ; 106(48): 20504-9, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19918059

RESUMEN

Currently, treatment with the relatively low-affinity NMDA receptor antagonist memantine provides limited benefit in Alzheimer's disease (AD). One probable dose-limiting factor in the use of memantine is the inhibition of NMDA receptor-dependent synaptic plasticity mechanisms believed to underlie certain forms of memory. Moreover, amyloid-beta protein (Abeta) oligomers that are implicated in causing the cognitive deficits of AD potently inhibit this form of plasticity. Here we examined if subtype-preferring NMDA receptor antagonists could preferentially protect against the inhibition of NMDA receptor-dependent plasticity of excitatory synaptic transmission by Abeta in the hippocampus in vivo. Using doses that did not affect control plasticity, antagonists selective for NMDA receptors containing GluN2B but not other GluN2 subunits prevented Abeta(1-42) -mediated inhibition of plasticity. Evidence that the proinflammatory cytokine TNFalpha mediates this deleterious action of Ass was provided by the ability of TNFalpha antagonists to prevent Abeta(1-42) inhibition of plasticity and the abrogation of a similar disruptive effect of TNFalpha using a GluN2B-selective antagonist. Moreover, at nearby synapses that were resistant to the inhibitory effect of TNFalpha, Abeta(1-42) did not significantly affect plasticity. These findings suggest that preferentially targeting GluN2B subunit-containing NMDARs may provide an effective means of preventing cognitive deficits in early Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Ácido Glutámico/metabolismo , Memantina/farmacología , Plasticidad Neuronal/fisiología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sinapsis/fisiología , Enfermedad de Alzheimer/prevención & control , Análisis de Varianza , Animales , Relación Dosis-Respuesta a Droga , Electrofisiología , Hipocampo/fisiología , Masculino , Memantina/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
10.
Int J Neuropsychopharmacol ; 12(4): 553-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18976544

RESUMEN

An animal model of depression combining genetic vulnerability and early-life stress (ELS) was prepared by submitting the Flinders Sensitive Line (FSL) rats to a standard paradigm of maternal separation. We analysed hippocampal synaptic transmission and plasticity in vivo and ionotropic receptors for glutamate in FSL rats, in their controls Flinders Resistant Line (FRL) rats, and in both lines subjected to ELS. A strong inhibition of long-term potentiation (LTP) and lower synaptic expression of NR1 subunit of the NMDA receptor were found in FSL rats. Remarkably, ELS induced a remodelling of synaptic plasticity only in FSL rats, reducing inhibition of LTP; this was accompanied by marked increase of synaptic NR1 subunit and GluR2/3 subunits of AMPA receptors. Chronic treatment with escitalopram inhibited LTP in FRL rats, but this effect was attenuated by prior ELS. The present results suggest that early gene-environment interactions cause lifelong synaptic changes affecting functional and molecular aspects of plasticity, partly reversed by antidepressant treatments.


Asunto(s)
Depresión/genética , Depresión/patología , Plasticidad Neuronal/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Estrés Psicológico/patología , Sinapsis/fisiología , Animales , Western Blotting , Depresión/psicología , Estimulación Eléctrica , Electrofisiología , Ambiente , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Ratas , Receptores de Glutamato/efectos de los fármacos , Receptores de Glutamato/genética , Estrés Psicológico/psicología , Sinaptosomas/fisiología
11.
Neurobiol Aging ; 30(10): 1608-13, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18272254

RESUMEN

Beta-amyloid (Abeta) is the main component of the extracellular plaques present in patients with Alzheimer's disease (AD) and studies have shown that exogenous application of Abeta results in neurodegeneration. As a model of the neurodegenerative action of Abeta, we have previously shown that acutely applied Abeta inhibits the induction of LTP in the hippocampus in vitro. In the present studies, we have studied the effect of beta-adrenoceptor activation on the Abeta inhibition of LTP. Pharmacological activation of beta2 adrenoceptors, but not of beta1 adrenoceptors, was found to prevent the Abeta evoked inhibition of LTP in the dentate gyrus of adult animals. The prevention of the effect of Abeta was shown to occur via the cAMP/PKA signaling pathway as the adenylate cyclase-stimulating agent forskolin prevented the Abeta inhibition of LTP, an action prevented by the PKA inhibitor, Rp-8-Br-cAMPs. We suggest microglia as a likely site of action of the neuroprotective effect of beta2 adrenoceptor activation. Therapeutic treatment for AD may include agents that activate beta2 receptors and elevate cAMP.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Giro Dentado/fisiología , Potenciación a Largo Plazo/fisiología , Receptores Adrenérgicos beta 2/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Adenilil Ciclasas/metabolismo , Adrenérgicos/farmacología , Agonistas de Receptores Adrenérgicos beta 1 , Agonistas de Receptores Adrenérgicos beta 2 , Antagonistas de Receptores Adrenérgicos beta 2 , Animales , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Giro Dentado/efectos de los fármacos , Dobutamina/farmacología , Inhibidores Enzimáticos/farmacología , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Propanolaminas/farmacología , Ratas , Receptores Adrenérgicos beta 1/metabolismo , Transducción de Señal/efectos de los fármacos , Terbutalina/farmacología , Tionucleótidos/farmacología
12.
Eur J Neurosci ; 29(1): 65-75, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19077124

RESUMEN

We have previously shown that activation of nicotinic acetylcholine receptors (nAChRs) enhanced long-term potentiation (LTP) in the rat dentate gyrus in vitro via activation of alpha7 nAChR. In the present studies, mechanisms underlying the acute and chronic nicotinic enhancement of LTP were examined. In particular, the involvement of activation of intracellular kinases was examined using selective kinase antagonists, and the effects of enhancing cholinergic function with positive allosteric modulators of the alpha7 nAChR and with acetylcholinesterase (AChE) inhibitors were also investigated. Activation of extracellular signal-regulated kinase (ERK) and cAMP-dependent protein kinase (PKA) was found to be involved in the induction of the acute nicotinic enhancement of LTP, although not control LTP. In contrast, activation of the tyrosine kinase Src, Ca(2+)-calmodulin-dependent protein kinase II, Janus kinase 2 and p38 mitogen-activated protein kinase was not involved in the acute nicotinic enhancement of LTP, although Src activation was necessary for control LTP. Moreover, activation of phosphoinositide 3-kinase was involved in the acute nicotinic enhancement of LTP to a much lesser extent than in control LTP. Chronic nicotine enhancement of LTP was found to be dependent on PKA, ERK and Src kinases. Acute nicotinic enhancement of LTP was occluded by chronic nicotine treatment. The positive allosteric modulator PNU-120596 was found to strongly reduce the threshold for nicotinic enhancement of LTP, an affect mediated via the alpha7 nAChR as it was blocked by the selective antagonist methyllycaconitine. The AChE inhibitors tacrine and physostigmine enhanced control LTP.


Asunto(s)
Acetilcolina/metabolismo , Giro Dentado/metabolismo , Potenciación a Largo Plazo/fisiología , Fosfotransferasas/metabolismo , Receptores Nicotínicos/metabolismo , Transmisión Sináptica/fisiología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Animales , Inhibidores de la Colinesterasa/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Giro Dentado/efectos de los fármacos , Isoxazoles/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Técnicas de Cultivo de Órganos , Compuestos de Fenilurea/farmacología , Ratas , Ratas Wistar , Receptores Nicotínicos/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Receptor Nicotínico de Acetilcolina alfa 7 , Familia-src Quinasas/efectos de los fármacos , Familia-src Quinasas/metabolismo
13.
J Neurosci ; 28(45): 11685-94, 2008 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-18987204

RESUMEN

Long-term potentiation of NMDA-receptor-mediated synaptic transmission (NMDAR-LTP) is a little-understood form of plasticity. In the present study, we investigated whether NMDAR-LTP in the dentate gyrus involves recruitment of extrasynaptic NMDARs, because NMDARs are expressed both synaptically and extrasynaptically with evidence for subtype differences at different locations. We show that before induction of NMDAR-LTP, pharmacological inhibition of glutamate transporters resulted in glutamate spillover from the synapse and activation of extrasynaptic NMDARs. After the induction of NMDAR-LTP, such activation of extrasynaptic NMDARs was absent. Activation of extrasynaptic NMDARs after glutamate uptake inhibition also occurred when synaptic NMDARs were inhibited with MK801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d] cyclohepten-5,10-imine maleate], and this extrasynaptically mediated NMDAR-EPSC was strongly reduced by prior induction of NMDAR-LTP. The extrasynaptic NMDARs were shown to be NR2D-containing, because the activation of extrasynaptic NMDARs by glutamate spillover was prevented by the NR2D-selective antagonists PPDA [(2R*,3S*)-1-(phenanthrenyl-2-carbonyl)piperazine-2,3-dicarboxylic acid] and UBP141. Further studies using selective antagonists for NR2A- and NR2B-containing NMDARs demonstrated that synaptic NMDARs are predominantly NR2A-containing and NR2B-containing receptors, whereas the extrasynaptic NMDARs are complex multimeric receptors with NR2A, NR2B, or NR2D subunits. Our results show that LTP of NMDAR-EPSCs involves movement of NMDARs from an extrasynaptic to a synaptic location and suggest a novel physiological role for extrasynaptic NMDARs.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Potenciación a Largo Plazo/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Ácido Aspártico/farmacología , Compuestos de Diazonio/farmacología , Maleato de Dizocilpina/farmacología , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Glutámico/metabolismo , Hipocampo/citología , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Técnicas de Placa-Clamp , Piridinas/farmacología , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Factores de Tiempo
14.
Neuropharmacology ; 55(2): 175-82, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18538800

RESUMEN

The serotonergic system is known to modulate and mediate many of the central nervous system effects of stress. Here we investigated the ability of serotonergic agents to reverse the inhibition of the induction of hippocampal long-term potentiation (LTP) caused by prior exposure to inescapable stress. Elevated platform stress prevented the induction of LTP in the CA1 area of anaesthetized rats. An agent that increases extracellular 5-HT concentration, fenfluramine (5 mg/kg, i.p.) enabled the induction of LTP in previously stressed animals. Consistent with a role for enhanced activation of 5-HT(2) receptors, the facilitatory effect of fenfluramine was prevented by the 5-HT(2) receptor antagonist cinanserin (30 mg/kg). Agents that directly activate 5-HT(2) receptors, including the 5-HT(2B) receptor agonist BW 723C86 (30 mg/kg) and the 5-HT(2C) receptor agonist MK-212 (3 mg/kg), mimicked the restorative effect of fenfluramine. Fenfluramine also opposed inhibition of LTP caused by the NMDA-receptor antagonist D-AP5 (100 nmol, i.c.v.) which suggests that the facilitatory action of serotonergic agents is not restricted to stress-mediated inhibition of LTP. These findings support an important role for activation of 5-HT(2) receptors by systemically applied agents to enable recovery from the inhibition of LTP by stress.


Asunto(s)
Hipocampo/fisiopatología , Potenciación a Largo Plazo/fisiología , Inhibición Neural/fisiología , Receptores de Serotonina 5-HT2/fisiología , Estrés Psicológico/patología , 2-Amino-5-fosfonovalerato/farmacología , Animales , Cinanserina/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Vías de Administración de Medicamentos , Interacciones Farmacológicas , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Fenfluramina/farmacología , Indoles/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de la radiación , Masculino , Inhibición Neural/efectos de los fármacos , Ratas , Ratas Wistar , Restricción Física/métodos , Serotoninérgicos/farmacología , Estrés Psicológico/etiología , Tiofenos/farmacología
15.
Neurosci Lett ; 436(2): 235-8, 2008 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-18403113

RESUMEN

The involvement of group I metabotropic glutamate receptors (mGluRs) and ryanodine receptors was investigated in the induction of LTP induced either by application of one standard high frequency stimulation (HFS) or by strong multiple HFS in the medial perforant path to granule cell synapse of the rat dentate gyrus. Whilst a standard brief HFS induced LTP close to 50%, strong stimulation consisting of multiple HFS induced a much larger LTP. mGluR5 was found to be partially involved in the induction of the enhanced LTP induced by the strong HFS but not in the standard LTP induced by the brief HFS. Thus the mGluR5 antagonists LY341495 and MPEP partially inhibited the induction of LTP induced by strong HFS but did not inhibit LTP induced by a standard HFS. Ryanodine was found to partially inhibit LTP induced by the strong HFS but not to inhibit the standard LTP induced by the brief HFS, demonstrating the involvement of Ca-induced Ca release from ryanodine-sensitive Ca stores in the former. These studies demonstrate that the large amplitude LTP induced by strong stimulation involves additional mechanisms to the LTP induced by brief HFS, in particular involving activation of mGluR5 and RyR-sensitive Ca stores.


Asunto(s)
Giro Dentado/efectos de la radiación , Estimulación Eléctrica , Potenciación a Largo Plazo/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Aminoácidos/farmacología , Animales , Giro Dentado/fisiología , Relación Dosis-Respuesta en la Radiación , Antagonistas de Aminoácidos Excitadores/farmacología , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de la radiación , Técnicas de Placa-Clamp , Piridinas/farmacología , Ratas , Rianodina/farmacología , Xantenos/farmacología
16.
J Neurosci ; 28(16): 4231-7, 2008 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-18417702

RESUMEN

The current development of immunotherapy for Alzheimer's disease is based on the assumption that human-derived amyloid beta protein (Abeta) can be targeted in a similar manner to animal cell-derived or synthetic Abeta. Because the structure of Abeta depends on its source and the presence of cofactors, it is of great interest to determine whether human-derived oligomeric Abeta species impair brain function and, if so, whether or not their disruptive effects can be prevented using antibodies. We report that untreated ex vivo human CSF that contains Abeta dimers rapidly inhibits hippocampal long-term potentiation in vivo and that acute systemic infusion of an anti-Abeta monoclonal antibody can prevent this disruption of synaptic plasticity. Abeta monomer isolated from human CSF did not affect long-term potentiation. These results strongly support a strategy of passive immunization against soluble Abeta oligomers in early Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/líquido cefalorraquídeo , Péptidos beta-Amiloides/inmunología , Inmunización Pasiva/métodos , Plasticidad Neuronal/inmunología , Sinapsis/inmunología , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/administración & dosificación , Animales , Anticuerpos Monoclonales/administración & dosificación , Células CHO , Cricetinae , Cricetulus , Dimerización , Humanos , Potenciación a Largo Plazo/inmunología , Masculino , Ratas , Ratas Wistar
17.
Brain ; 131(Pt 3): 651-64, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18292081

RESUMEN

Non-steroidal anti-inflammatory agents (NSAIDs) are associated with a marked reduction in the risk of developing Alzheimer's disease, a form of dementia characterized by the accumulation of amyloid plaques containing the amyloid-beta protein (Abeta). Studies of the effects of NSAIDs upon the inflammatory response surrounding amyloid plaques and upon the generation of Abeta from the amyloid precursor protein (APP) have led to two proposed mechanisms by which NSAIDs may protect against Alzheimer's disease: one, the selective lowering of Abeta42 by a subset of NSAIDs; and two, the reduction of inflammation. Although Alzheimer's disease is a disorder of brain and synaptic function, the effects of NSAIDs on Abeta-mediated suppression of synaptic plasticity and memory function have never been reported. We therefore investigated how three different NSAIDs, chosen for their distinct effects on Abeta42 production and the inhibition of the cyclooxygenase (COX) isoenzymes, COX-1 and COX-2, affect memory function and synaptic plasticity. By focusing upon brain and synapse function, we made novel observations about the effects of NSAIDs on Abeta-mediated neural processes. Here we report that the selective inhibition of COX-2, but not COX-1, acutely prevented the suppression of hippocampal long-term plasticity (LTP) by Abeta. The non-selective NSAIDs, ibuprofen and naproxen, and a selective COX-2 inhibitor, MF-tricyclic, each restored memory function in Tg2576 mice over-expressing APP, and also blocked Abeta-mediated inhibition of LTP. There was no advantage of ibuprofen, a selective Abeta42-lowering agent (SALA), over the non-SALAs, naproxen and MF-tricyclic. The beneficial effects on memory did not depend upon lowered levels of Abeta42 or the inflammatory cytokines, tumour necrosis factor alpha (TNF-alpha) and interleukin 1beta (IL-1beta). Intriguingly, improved memory function was inversely related to prostaglandin E2 (PGE2) levels. Conversely, exogenous PGE2 prevented the restorative effects of COX-2 inhibitors on LTP. The data indicate that the inhibition of COX-2 blocks Abeta-mediated suppression of LTP and memory function, and that this block occurs independently of reductions in Abeta42 or decreases in inflammation. The results lead us to propose a third possible mechanism by which NSAIDs may protect against Alzheimer's disease, involving the blockade of a COX-2-mediated PGE2 response at synapses.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Inhibidores de la Ciclooxigenasa 2/farmacología , Memoria/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Péptidos beta-Amiloides/farmacología , Animales , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Inhibidores de la Ciclooxigenasa/farmacología , Inhibidores de la Ciclooxigenasa/uso terapéutico , Dinoprostona/fisiología , Furanos/farmacología , Furanos/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ibuprofeno/farmacología , Ibuprofeno/uso terapéutico , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/prevención & control , Ratones , Ratones Endogámicos C57BL , Naproxeno/farmacología , Naproxeno/uso terapéutico , Fragmentos de Péptidos/farmacología , Ratas , Sinapsis/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
18.
Neurobiol Aging ; 29(10): 1485-93, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17442458

RESUMEN

Beta-amyloid (Abeta) is the principal component of the extracellular plaques present in patients with Alzheimer's disease. Several studies have recently shown that acutely applied Abeta inhibits the induction of LTP in the hippocampus. In the present studies, we have investigated the role of integrins in such Abeta-mediated block of LTP in the dentate gyrus in vitro and in the CA1 in vivo. Selective antibodies to the alpha v integrin subunit were found to prevent the Abeta inhibition of LTP, both in the dentate gyrus in vitro and in the CA1 in vivo. In contrast, two control antibodies did not prevent such action of Abeta. In addition, a small molecule nonpeptide antagonist of alpha v-containing integrins and two other antagonistic ligands of integrins, superfibronectin and the disintegrin echistatin, also prevented the Abeta inhibition of LTP. These studies indicate that alpha v integrins may be important mediators of synaptic dysfunction prior to neurodegeneration in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Integrina alfaV/metabolismo , Potenciación a Largo Plazo/fisiología , Inhibición Neural/fisiología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/toxicidad , Animales , Anticuerpos/farmacología , Giro Dentado/metabolismo , Giro Dentado/fisiopatología , Hipocampo/fisiopatología , Péptidos y Proteínas de Señalización Intercelular , Ligandos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Inhibición Neural/efectos de los fármacos , Técnicas de Cultivo de Órganos , Péptidos/farmacología , Ratas , Ratas Wistar , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
19.
Eur J Pharmacol ; 577(1-3): 71-7, 2007 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-17900561

RESUMEN

Dopamine D(1)/D(5) receptor agonists may enhance cognition by mimicking dopamine's neurophysiological actions on the processes underlying learning and memory. The present study examined the task- and performance- dependence of the cognitive effects of a partial agonist at dopamine D(1)/D(5) receptors, SKF 38393 [(+/-)-1-phenyl-2,3,4,5-tetrahydro-(1H)-3-benzazepine-7,8-diol hydrobromide], in rats. Spatial working memory was assessed in a T-maze, spatial reference memory in a water maze and habituation learning in a novel environment, a hole board. The muscarinic acetylcholine receptor antagonist scopolamine (1.5 mg/kg, i.p.) was used to cause an impairment of performance of these learning tasks. Administration of SKF 38393 (6 mg/kg, i.p.) alone had no significant effect on spontaneous alternation in the T-maze, latency to escape to a hidden platform in the water maze or the habituation of spontaneous behaviour in the hole board. In contrast, in scopolamine-treated rats, whereas SKF 38393 prevented the scopolamine-induced deficit in the T-maze, it exacerbated the impairment in the water maze and did not significantly alter the disruption of habituation. These results suggest that dopamine D(1)/D(5) receptor activation has performance- and task-dependent effects on cognitive function.


Asunto(s)
2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Agonistas de Dopamina/farmacología , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Receptores de Dopamina D5/agonistas , Animales , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Ratas , Ratas Wistar , Escopolamina/farmacología
20.
Neuropharmacology ; 53(1): 188-95, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17610911

RESUMEN

Nicotine has been postulated to be a possible neuroprotective agent in Alzheimer's Disease (AD). In the present studies, the effect of beta-amyloid (Abeta) was investigated on the nicotine enhancement of high-frequency-induced LTP. Perfusion of nicotine substantially enhanced HFS-induced LTP in both rat and mouse dentate gyrus. The enhancing action of nicotine was mediated via alpha7 nAChRs as it was absent in mice null for alpha7 nAChR. Abeta strongly inhibited the induction of LTP in control animals, with LTP being completely inhibited at 1h post-HFS. Although Abeta also inhibited LTP in the presence of nicotine, the extent of the inhibition of LTP in nicotine perfused slices was similar to that in control, resulting in substantial LTP remaining in the presence of Abeta in the nicotine perfused slices. The nicotine enhanced LTP and the LTP remaining in the presence of Abeta and nicotine, although not the control LTP was dependent on activation of PKA. Chronic nicotine treatment also enhanced HFS-LTP recorded in acute slices taken from the nicotine-treated animals, and such LTP was only partially inhibited by Abeta. We postulate that nicotine-enhanced LTP has certain different mechanisms to that of control LTP which results in a resistance to inhibition by Abeta.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Hipocampo/citología , Potenciación a Largo Plazo/efectos de los fármacos , Neuronas/efectos de los fármacos , Nicotina/farmacología , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta en la Radiación , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de la radiación , Isoquinolinas/farmacología , Potenciación a Largo Plazo/fisiología , Potenciación a Largo Plazo/efectos de la radiación , Masculino , Ratones , Ratones Noqueados , Neuronas/efectos de la radiación , Ratas , Ratas Wistar , Receptores Nicotínicos/deficiencia , Receptores Nicotínicos/fisiología , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA