Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Psychopharmacol ; 37(8): 822-835, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37165655

RESUMEN

BACKGROUND: Understanding the effects of the N-methyl-D-aspartate receptor (NMDA-R) antagonist ketamine on brain function is of considerable interest due to the discovery of its fast-acting antidepressant properties. It is well known that gamma oscillations are increased when ketamine is administered to rodents and humans, and increases in the auditory steady-state response (ASSR) have also been observed. AIMS: To elucidate the cellular substrate of the increase in network activity and synchrony observed by sub-anesthetic doses of ketamine, the aim was to investigate spike timing and regularity and determine how this is affected by the animal's motor state. METHODS: Single unit activity and local field potentials from the auditory cortex of awake, freely moving rats were recorded with microelectrode arrays during an ASSR paradigm. RESULTS: Ketamine administration yielded a significant increase in ASSR power and phase locking, both significantly modulated by motor activity. Before drug administration, putative fast-spiking interneurons (FSIs) were significantly more entrained to the stimulus than putative pyramidal neurons (PYRs). The degree of entrainment significantly increased at lower doses of ketamine (3 and 10 mg/kg for FSIs, 10 mg/kg for PYRs). At the highest dose (30 mg/kg), a strong increase in tonic firing of PYRs was observed. CONCLUSIONS: These findings suggest an involvement of FSIs in the increased network synchrony and provide a possible cellular explanation for the well-documented effects of ketamine-induced increase in power and synchronicity during ASSR. The results support the importance to evaluate different motor states separately for more translational preclinical research.


Asunto(s)
Anestésicos , Corteza Auditiva , Ketamina , Humanos , Ratas , Animales , Ketamina/farmacología , Corteza Auditiva/fisiología , Potenciales Evocados Auditivos , Estimulación Acústica/métodos , Anestésicos/farmacología
2.
J Alzheimers Dis ; 78(1): 291-308, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32955458

RESUMEN

BACKGROUND: The European PharmaCog study (http://www.pharmacog.org) has reported a reduction in delta (1-6 Hz) electroencephalographic (EEG) power (density) during cage exploration (active condition) compared with quiet wakefulness (passive condition) in PDAPP mice (hAPP Indiana V717F mutation) modeling Alzheimer's disease (AD) amyloidosis and cognitive deficits. OBJECTIVE: Here, we tested the reproducibility of that evidence in TASTPM mice (double mutation in APP KM670/671NL and PSEN1 M146V), which develop brain amyloidosis and cognitive deficits over aging. The reliability of that evidence was examined in four research centers of the PharmaCog study. METHODS: Ongoing EEG rhythms were recorded from a frontoparietal bipolar channel in 29 TASTPM and 58 matched "wild type" C57 mice (range of age: 12-24 months). Normalized EEG power was calculated. Frequency and amplitude of individual delta and theta frequency (IDF and ITF) peaks were considered during the passive and active conditions. RESULTS: Compared with the "wild type" group, the TASTPM group showed a significantly lower reduction in IDF power during the active over the passive condition (p < 0.05). This effect was observed in 3 out of 4 EEG recording units. CONCLUSION: TASTPM mice were characterized by "poor reactivity" of delta EEG rhythms during the cage exploration in line with previous evidence in PDAPP mice. The reliability of that result across the centers was moderate, thus unveiling pros and cons of multicenter preclinical EEG trials in TASTPM mice useful for planning future studies.


Asunto(s)
Enfermedad de Alzheimer/genética , Electroencefalografía/métodos , Precursor de Proteína beta-Amiloide/genética , Amiloidosis , Animales , Encéfalo/metabolismo , Disfunción Cognitiva , Ratones , Ratones Transgénicos , Movimiento , Reproducibilidad de los Resultados , Vigilia
3.
Eur J Pharmacol ; 887: 173440, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32745603

RESUMEN

Activation of the voltage-gated Kv7 channels holds therapeutic promise in several neurological and psychiatric disorders, including epilepsy, schizophrenia, and depression. Here, we present a pharmacological characterization of Lu AA41178, a novel, pan-selective Kv7.2-7.5 opener, using both in vitro assays and a broad range of in vivo assays with relevance to epilepsy, schizophrenia, and depression. Electrophysiological characterization in Xenopus oocytes expressing human Kv7.2-Kv7.5 confirmed Lu AA41178 as a pan-selective opener of Kv7 channels by significantly left-shifting the activation threshold. Additionally, Lu AA41178 was tested in vitro for off-target effects, demonstrating a clean Kv7-selective profile, with no impact on common cardiac ion channels, and no potentiating activity on GABAA channels. Lu AA41178 was evaluated across preclinical in vivo assays with relevance to neurological and psychiatric disorders. In the maximum electroshock seizure threshold test and PTZ seizure threshold test, Lu AA41178 significantly increased the seizure thresholds in mice, demonstrating anticonvulsant efficacy. Lu AA41178 demonstrated antipsychotic-like activity by reducing amphetamine-induced hyperlocomotion in mice as well as lowering conditioned avoidance responses in rats. In the mouse forced swim test, a model with antidepressant predictivity, Lu AA41178 significantly reduced immobility. Additionally, behavioral effects typically observed with Kv7 openers was also characterized. In vivo assays were accompanied by plasma and brain exposures, revealing minimum effective plasma levels <1000 ng/ml. Lu AA41178, a potent opener of neuronal Kv7 channels demonstrate efficacy in assays of epilepsy, schizophrenia and depression and might serve as a valuable tool for exploring the role of Kv7 channels in both neurological and psychiatric disorders.


Asunto(s)
Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Canal de Potasio KCNQ2/agonistas , Trastornos Mentales/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Canal de Potasio KCNQ2/metabolismo , Masculino , Trastornos Mentales/metabolismo , Trastornos Mentales/psicología , Ratones , Ratones Endogámicos C57BL , Psicotrópicos/farmacología , Psicotrópicos/uso terapéutico , Ratas , Ratas Wistar , Convulsiones/metabolismo , Convulsiones/psicología , Resultado del Tratamiento , Xenopus laevis
4.
Neuroscience ; 424: 172-181, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31678343

RESUMEN

The antidepressant drug vortioxetine has a multimodal action modulating neurotransmission through inhibition of the serotonin transporter and modulation of serotonin receptors. Vortioxetine has also been shown to alleviate cognitive symptoms in preclinical studies and in patients with depression. However, it is largely unclear how vortioxetine affects the brain processing in humans. The present study was conducted in 32 healthy males in a randomized, double-blinded, placebo-controlled, active comparator, four-way crossover design. Treatments were 10 and 20 mg/day vortioxetine, 15 mg/day escitalopram, and placebo, administered orally once daily for three days. Results were compared to placebo. Treatment effect was assessed by recording spontaneous electroencephalography (EEG) and 40 Hz auditory steady state responses. For the spontaneous EEG, both vortioxetine and escitalopram decreased the frequency content in the theta band (4-8 Hz) and increased power in the beta (12-32 Hz) and gamma (32-45 Hz) bands. Vortioxetine and escitalopram decreased connectivity during rest in the theta band and increased connectivity in the gamma bands. Finally, both treatments caused decreased power in the evoked gamma band in response to 40 Hz auditory stimulation. Although the global EEG changes were comparable between vortioxetine and escitalopram, subtle differences between treatment effects on the EEG in terms of effect size and regional distribution of the EEG changes were apparent. To our knowledge, the current results are the first data on how vortioxetine affects EEG in humans. The present study calls for further investigations addressing the possible electrophysiological and cognitive effects of vortioxetine.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Citalopram/farmacología , Electroencefalografía/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Vortioxetina/farmacología , Adulto , Citalopram/sangre , Estudios Cruzados , Método Doble Ciego , Electroencefalografía/métodos , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Inhibidores Selectivos de la Recaptación de Serotonina/sangre , Vortioxetina/sangre , Adulto Joven
5.
Eur J Neurosci ; 46(3): 1887-1896, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28635024

RESUMEN

Nav 1.1 (SCN1A) channels primarily located in gamma-aminobutyric acid (GABA)ergic fast-spiking interneurons are pivotal for action potential generation and propagation in these neurons. Inappropriate function of fast-spiking interneurons, leading to disinhibition of pyramidal cells and network desynchronization, correlates with decreased cognitive capability. Further, reduced functionality of Nav 1.1 channels is linked to various diseases in the central nervous system. There is, at present, however no subtype selective pharmacological activators of Nav 1.1 channels available for studying pharmacological modulation of interneuron function. In the current study, we identified a small molecule Nav 1.1 activator, 3-amino-5-(4-methoxyphenyl)thiophene-2-carboxamide, named AA43279, and provided an in vitro to in vivo characterization of the compound. In HEK-293 cells expressing human Nav 1.1 channels, AA43279 increased the Nav 1.1-mediated current in a concentration-dependent manner mainly by impairing the fast inactivation kinetics of the channels. In rat hippocampal brain slices, AA43279 increased the firing activity of parvalbumin-expressing, fast-spiking GABAergic interneurons and increased the spontaneous inhibitory post-synaptic currents (sIPSCs) recorded from pyramidal neurons. When tested in vivo, AA43279 had anti-convulsive properties in the maximal electroshock seizure threshold test. AA43279 was tested for off-target effects on 72 different proteins, including Nav 1.2, Nav 1.4, Nav 1.5, Nav 1.6 and Nav 1.7 and exhibited reasonable selectivity. Taken together, AA43279 might constitute a valuable tool compound for revealing biological functions of Nav 1.1 channels.


Asunto(s)
Anticonvulsivantes/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Interneuronas/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Convulsiones/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/farmacología , Tiofenos/farmacología , Potenciales de Acción , Animales , Anticonvulsivantes/síntesis química , Anticonvulsivantes/uso terapéutico , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Potenciales Postsinápticos Excitadores , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Células HEK293 , Humanos , Interneuronas/metabolismo , Interneuronas/fisiología , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/uso terapéutico
6.
Neurobiol Aging ; 49: 20-30, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27728831

RESUMEN

Resting state electroencephalographic (EEG) rhythms reflect the fluctuation of cortical arousal and vigilance in a typical clinical setting, namely the EEG recording for few minutes with eyes closed (i.e., passive condition) and eyes open (i.e., active condition). Can this procedure be back-translated to C57 (wild type) mice for aging studies? On-going EEG rhythms were recorded from a frontoparietal bipolar channel in 85 (19 females) C57 mice. Male mice were subdivided into 3 groups: 25 young (4.5-6 months), 18 middle-aged (12-15 months), and 23 old (20-24 months) mice to test the effect of aging. EEG power density was compared between short periods (about 5 minutes) of awake quiet behavior (passive) and dynamic exploration of the cage (active). Compared with the passive condition, the active condition induced decreased EEG power at 1-2 Hz and increased EEG power at 6-10 Hz in the group of 85 mice. Concerning the aging effects, the passive condition showed higher EEG power at 1-2 Hz in the old group than that in the others. Furthermore, the active condition exhibited a maximum EEG power at 6-8 Hz in the former group and 8-10 Hz in the latter. In the present conditions, delta and theta EEG rhythms reflected changes in cortical arousal and vigilance in freely behaving C57 mice across aging. These changes resemble the so-called slowing of resting state EEG rhythms observed in humans across physiological and pathological aging. The present EEG procedures may be used to enhance preclinical phases of drug discovery in mice for understanding the neurophysiological effects of new compounds against brain aging.


Asunto(s)
Envejecimiento/fisiología , Nivel de Alerta/fisiología , Corteza Cerebral/fisiología , Electroencefalografía , Animales , Descubrimiento de Drogas , Femenino , Masculino , Ratones Endogámicos C57BL , Descanso/fisiología , Vigilia/fisiología
7.
Eur Neuropsychopharmacol ; 26(7): 1150-60, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27061851

RESUMEN

The 15q13.3 microdeletion copy number variation is strongly associated with schizophrenia and epilepsy. The CHRNA7 gene, encoding nicotinic acetylcholine alpha 7 receptors (nAChA7Rs), is hypothesized to be one of the main genes in this deletion causing the neuropsychiatric phenotype. Here we used a recently developed 15q13.3 microdeletion mouse model to explore whether an established schizophrenia-associated connectivity phenotype is replicated in a murine model, and whether positive modulation of nAChA7 receptor might pharmacologically normalize the connectivity patterns. Resting-state fMRI data were acquired from male mice carrying a hemizygous 15q13.3 microdeletion (N=9) and from wild-type mice (N=9). To study the connectivity profile of 15q13.3 mice and test the effect of nAChA7 positive allosteric modulation, the 15q13.3 mice underwent two imaging sessions, one week apart, receiving a single intraperitoneal injection of either 15mg/kg Lu AF58801 or saline. The control group comprised wild-type mice treated with saline. We performed seed-based functional connectivity analysis to delineate aberrant connectivity patterns associated with the deletion (15q13.3 mice (saline treatment) versus wild-type mice (saline treatment)) and their modulation by Lu AF58801 (15q13.3 mice (Lu AF58801 treatment) versus 15q13.3 mice (saline treatment)). Compared to wild-type mice, 15q13.3 mice evidenced a predominant hyperconnectivity pattern. The main effect of Lu AF58801 was a normalization of elevated functional connectivity between prefrontal and frontal, hippocampal, striatal, thalamic and auditory regions. The strongest effects were observed in brain regions expressing nAChA7Rs, namely hippocampus, cerebral cortex and thalamus. These effects may underlie the antiepileptic, pro-cognitive and auditory gating deficit-reversal effects of nAChA7R stimulation.


Asunto(s)
Encéfalo/efectos de los fármacos , Colinérgicos/farmacología , Trastornos de los Cromosomas/tratamiento farmacológico , Discapacidad Intelectual/tratamiento farmacológico , Esquizofrenia/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Mapeo Encefálico , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/metabolismo , Cromosomas Humanos Par 15/genética , Cromosomas Humanos Par 15/metabolismo , Ciclopropanos/farmacología , Modelos Animales de Enfermedad , Endofenotipos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones Transgénicos , Vías Nerviosas/diagnóstico por imagen , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/farmacología , Descanso , Esquizofrenia/metabolismo , Convulsiones/genética , Convulsiones/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/genética
8.
Neuropsychopharmacology ; 41(2): 638-45, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26129678

RESUMEN

Sensory gating is the brain's ability to filter out irrelevant information before it reaches high levels of conscious processing. In the current study we aimed to investigate the involvement of the noradrenergic and dopaminergic neurotransmitter systems in sensory gating. Furthermore, we investigated cross-species reliability by comparing effects in both healthy humans and rats, while keeping all experimental conditions as similar as possible between the species. The design of the human experiment (n=21) was a double-blind, placebo-controlled, cross-over study where sensory gating was assessed following a dose of either reboxetine (8 mg), haloperidol (2 mg), their combination or placebo at four separate visits. Similarly in the animal experiment sensory gating was assessed in rats, (n=22) following a dose of reboxetine (2 mg/kg), haloperidol (0.08 mg/kg), their combination or placebo. The sensory gating paradigms in both experiments were identical. In humans, we found significantly reduced P50 suppression following separate administration of reboxetine or haloperidol, while their combined administration did not reach statistical significance compared with placebo. In the rats, we found a similar significant reduction of sensory gating (N40) following treatment with haloperidol and the combination of haloperidol and reboxetine, but not with separate reboxetine treatment, compared with placebo. Our study indicates that even when experimental conditions are kept as similar as possible, direct human to rat cross-species translation of pharmacological effects on sensory gating is challenging, which calls for more focussed research in this important translational area.


Asunto(s)
Inhibidores de Captación Adrenérgica/administración & dosificación , Antagonistas de Dopamina/administración & dosificación , Haloperidol/administración & dosificación , Morfolinas/administración & dosificación , Filtrado Sensorial/efectos de los fármacos , Filtrado Sensorial/fisiología , Administración Oral , Adolescente , Adulto , Animales , Percepción Auditiva/efectos de los fármacos , Percepción Auditiva/fisiología , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/fisiología , Estudios Cruzados , Método Doble Ciego , Potenciales Evocados Auditivos/efectos de los fármacos , Potenciales Evocados Auditivos/fisiología , Humanos , Masculino , Ratas , Reboxetina , Especificidad de la Especie , Adulto Joven
9.
Eur J Pharmacol ; 758: 164-70, 2015 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-25845309

RESUMEN

The effect of two positive modulators, RE1 and EX15, on the voltage-gated K(+) channel Kv3.1 was investigated using the whole-cell patch-clamp technique on HEK293 cells expressing Kv3.1a. RE1 and EX15 increased the Kv3.1 currents in a concentration-dependent manner with an EC50 value of 4.5 and 1.3µM, respectively. However, high compound concentrations caused an inhibition of the Kv3.1 current. The compound-induced activation of Kv3.1 channels showed a profound hyperpolarized shift in activation kinetics. 30µM RE1 shifted V1/2 from 5.63±0.31mV to -9.71±1.00mV and 10µM EX15 induced a shift from 10.77±0.32mV to -15.11±1.57mV. The activation time constant (Tauact) was reduced for both RE1 and EX15, with RE1 being the fastest activator. The deactivation time constant (Taudeact) was also markedly reduced for both RE1 and EX15, with EX15 inducing the most prominent effect. Furthermore, subjected to depolarizing pulses at 30Hz, both compounds were showing a use-dependent effect resulting in a reduction of the compound-mediated effect. However, during these conditions, RE1- and EX15-modified current amplitudes still exceeded the control condition amplitudes by up to 200%. In summary, the present study introduces the first detailed biophysical characterization of two new Kv3.1 channel modifying compounds with different modulating properties.


Asunto(s)
Canales de Potasio Shaw/agonistas , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Hidantoínas/farmacología , Cinética , Piridinas/farmacología , Canales de Potasio Shaw/fisiología
10.
Basic Clin Pharmacol Toxicol ; 116(3): 187-200, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25441336

RESUMEN

The nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels broadly involved in regulating neurotransmission in the central nervous system (CNS) by conducting cation currents through the membrane of neurons. Many different nAChR subtypes exist with each their functional characteristics, expression pattern and pharmacological profile. The focus of the present MiniReview is on the heteromeric α4ß2 nAChR, as activity at this subtype contributes to cognitive functioning through interactions with multiple neurotransmitter systems and is implicated in various CNS disorders, for example schizophrenia and Alzheimer's disease. Additionally, the α4ß2 nAChR provides an extra layer of molecular complexity by existing in two different stoichiometries determined by the subunit composition. Such findings have founded the rationale that pharmacological modulation of the α4ß2 nAChR may be used as a treatment approach in various CNS disorders. As subtype-selective agonists and other cholinergic ligands have only shown limited therapeutic success, the focus of recent drug development endeavours has largely shifted to positive allosteric modulators (PAMs). By potentiating the action of an agonist through binding to an allosteric site, a PAM can enhance cholinergic neurotransmission, thus compensating for compromised neuronal communication in a pathophysiological setting. The pharmacological advantages of PAMs compared to other types of ligands include minimal interference with spatial and temporal aspects of neurotransmission as well as higher subtype selectivity, hypothetically resulting in high clinical efficacy with minimal adverse effects. In this MiniReview, we describe the currently identified compounds, which potentiate the effects of agonists at the α4ß2 nAChR. The potential clinical advantages and concerns of PAMs are discussed in the light of the role of α4ß2 nAChRs as key regulators of fast synaptic transmission.


Asunto(s)
Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Agonistas Nicotínicos/uso terapéutico , Receptores Nicotínicos/efectos de los fármacos , Regulación Alostérica , Sitio Alostérico , Animales , Enfermedades del Sistema Nervioso Central/fisiopatología , Diseño de Fármacos , Humanos , Ligandos , Agonistas Nicotínicos/efectos adversos , Agonistas Nicotínicos/farmacología , Receptores Nicotínicos/metabolismo , Transmisión Sináptica/efectos de los fármacos
11.
J Alzheimers Dis ; 42(4): 1179-89, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25024305

RESUMEN

P300 (P3) event-related potentials (ERPs) have been suggested to be an endogenous marker of cognitive function and auditory oddball paradigms are frequently used to evaluate P3 ERPs in clinical settings. Deficits in P3 amplitude and latency reflect some of the neurological dysfunctions related to several psychiatric and neurological diseases, e.g., Alzheimer's disease (AD). However, only a very limited number of rodent studies have addressed the back-translational validity of the P3-like ERPs as suitable markers of cognition. Thus, the potential of rodent P3-like ERPs to predict pro-cognitive effects in humans remains to be fully validated. The current study characterizes P3-like ERPs in the 192-IgG-SAP (SAP) rat model of the cholinergic degeneration associated with AD. Following training in a combined auditory oddball and lever-press setup, rats were subjected to bilateral intracerebroventricular infusion of 1.25 µg SAP or PBS (sham lesion) and recording electrodes were implanted in hippocampal CA1. Relative to sham-lesioned rats, SAP-lesioned rats had significantly reduced amplitude of P3-like ERPs. P3 amplitude was significantly increased in SAP-treated rats following pre-treatment with 1 mg/kg donepezil. Infusion of SAP reduced the hippocampal choline acetyltransferase activity by 75%. Behaviorally defined cognitive performance was comparable between treatment groups. The present study suggests that AD-like deficits in P3-like ERPs may be mimicked by the basal forebrain cholinergic degeneration induced by SAP. SAP-lesioned rats may constitute a suitable model to test the efficacy of pro-cognitive substances in an applied experimental setup.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Región CA1 Hipocampal/fisiopatología , Neuronas Colinérgicas/fisiología , Potenciales Evocados Auditivos/fisiología , Degeneración Nerviosa/fisiopatología , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Anticuerpos Monoclonales , Percepción Auditiva/efectos de los fármacos , Percepción Auditiva/fisiología , Región CA1 Hipocampal/efectos de los fármacos , Colina O-Acetiltransferasa/metabolismo , Neuronas Colinérgicas/efectos de los fármacos , Modelos Animales de Enfermedad , Donepezilo , Electrodos Implantados , Potenciales Evocados Auditivos/efectos de los fármacos , Indanos/farmacología , Masculino , Degeneración Nerviosa/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Pruebas Neuropsicológicas , Piperidinas/farmacología , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Ratas Sprague-Dawley , Proteínas Inactivadoras de Ribosomas Tipo 1 , Saporinas
12.
Behav Brain Res ; 266: 85-93, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24613239

RESUMEN

Patients with schizophrenia exhibit disturbances in information processing. These disturbances can be investigated with different paradigms of auditory event related potentials (ERP), such as sensory gating in a double click paradigm (P50 suppression) and the mismatch negativity (MMN) component in an auditory oddball paradigm. The aim of the current study was to test if rats subjected to social isolation, which is believed to induce some changes that mimic features of schizophrenia, displays alterations in sensory gating and MMN-like response. Male Lister-Hooded rats were separated into two groups; one group socially isolated (SI) for 8 weeks and one group housed (GH). Both groups were then tested in a double click sensory gating paradigm and an auditory oddball paradigm (MMN-like) paradigm. It was observed that the SI animals showed reduced sensory gating of the cortical N1 amplitude. Furthermore, the SI animals showed significant reduction in cortical MMN-like response compared with the GH animals. No deficits in sensory gating or MMN-like response were observed in the hippocampus (CA3) of the SI animals compared with GH animals. In conclusion, the change in sensory gating of the N1 amplitude supports previous findings in SI rats and the reduced MMN-like response is similar to the deficits of MMN seen in patients with schizophrenia. Since reduced auditory MMN amplitude is believed to be more selectively associated with schizophrenia than other measures of sensory gating deficits, the current study supports the face validity of the SI reared rat model for schizophrenia.


Asunto(s)
Variación Contingente Negativa/fisiología , Potenciales Evocados Auditivos/fisiología , Esquizofrenia/fisiopatología , Filtrado Sensorial/fisiología , Aislamiento Social/psicología , Estimulación Acústica , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Masculino , Actividad Motora , Ratas , Psicología del Esquizofrénico
13.
Brain Res ; 1543: 253-62, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24231553

RESUMEN

The Alzheimer's disease (AD) mouse model Tg2576 overexpresses an AD associated mutant variant of human APP and accumulates amyloid beta (Aß) in an age-dependent manner. Using the selective cholinergic immunotoxin mu p75-saporin (SAP), we induced a partial basal forebrain cholinergic degeneration (BFCD) in 3 months old male Tg2576 mice to co-express cholinergic degeneration with Aß overexpression as these characteristics constitutes key hallmarks of AD. At 9 months, SAP lesioned Tg2576 mice were cognitively impaired in two spatial paradigms addressing working memory and mid to long-term memory. Conversely, there was no deterioration of cognitive functioning in sham lesioned Tg2576 mice or wild type littermates (wt) receiving the immunotoxin. At 10 months of age, release of acetylcholine (ACh) was addressed by microdialysis in conscious mice. Scopolamine-induced increases in hippocampal ACh efflux was significantly reduced in SAP lesioned Tg2576 mice compared to sham lesioned Tg2576 mice. Intriguingly, there was no significant difference in ACh efflux between wt treatment groups. Following SAP treatment, choline acetyltransferase activity was reduced in the hippocampus and frontal cortex and the reduction was comparable between groups. Our results suggest that partial BFCD acts collectively with increased levels of Aß to induce cognitive decline and to compromise cholinergic release. Tg2576 mice with BFCD may constitute a new and suitable AD mouse model to study the interrelations between cholinergic deficits and amsyloid deposition.


Asunto(s)
Acetilcolina/metabolismo , Neuronas Colinérgicas/patología , Hipocampo/metabolismo , Trastornos de la Memoria/patología , Degeneración Nerviosa/etiología , Prosencéfalo/patología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Colina O-Acetiltransferasa/metabolismo , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Humanos , Inmunotoxinas/farmacología , Aprendizaje por Laberinto , Trastornos de la Memoria/etiología , Ratones , Ratones Transgénicos , Presenilina-1/genética , Reconocimiento en Psicología/fisiología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Saporinas
14.
Neuropharmacology ; 79: 444-55, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24361451

RESUMEN

The P300 (P3) event-related potential (ERP) is a neurophysiological signal believed to reflect cognitive processing of salient cues, and is thus used as a measure of attention and working memory. Additionally, P3 amplitude and latency is altered in neurological diseases and can be pharmacologically modulated. As P3-like ERPs can be recorded in rodents, it may serve as a potential translational biomarker of value for drug discovery. Here we investigated whether a positive allosteric modulator of α4ß2 nicotinic acetylcholine receptors, NS9283, and the psychostimulant modafinil could modulate P3-like ERPs in healthy adult rats performing an auditory oddball discrimination task. ERPs were recorded with electroencephalography electrodes implanted into mediodorsal (MD) thalamus, medial prefrontal cortex, hippocampus and auditory cortex (AC). P3-like ERPs were detected in all brain regions, displaying larger amplitudes in target trials compared to non-target trials. Administration of modafinil (64 mg/kg) decreased P3-like ERP latency in MD thalamus and AC, whereas NS9283 augmented P3-like ERP amplitude in MD thalamus at 0.3 mg/kg and in AC at 3.0 mg/kg. Additionally, N1 pre-attention peak amplitude in MD thalamus was increased with 0.3 mg/kg NS9283. Neither of the compounds enhanced task performance. Rather, modafinil lowered correct rejections in non-target trials. In summary, our findings reveal pharmacological modulation of the rat P3-like ERP in cortical and subcortical regions by modafinil and NS9283. These findings encourage further exploration of the rat P3-like ERP in order to promote the understanding of its meaning within cognition, as well as its applicability as a translatable biomarker in drug development.


Asunto(s)
Percepción Auditiva/efectos de los fármacos , Compuestos de Bencidrilo/farmacología , Encéfalo/efectos de los fármacos , Agonistas Nicotínicos/farmacología , Oxadiazoles/farmacología , Piridinas/farmacología , Promotores de la Vigilia/farmacología , Estimulación Acústica , Animales , Percepción Auditiva/fisiología , Encéfalo/fisiología , Discriminación en Psicología/efectos de los fármacos , Discriminación en Psicología/fisiología , Electrodos Implantados , Electroencefalografía , Potenciales Evocados Auditivos/efectos de los fármacos , Potenciales Evocados Auditivos/fisiología , Modafinilo , Pruebas Neuropsicológicas , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/metabolismo , Análisis y Desempeño de Tareas , Factores de Tiempo
15.
Clin Neurophysiol ; 124(3): 437-51, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23040292

RESUMEN

Different kinds of challenge can alter spontaneous ongoing electroencephalographic (EEG) rhythms in animal models, thus providing paradigms to evaluate treatment effects in drug discovery. The effects of challenges represented by pharmacological agents, hypoxia, sleep deprivation and transcranial magnetic stimulation (TMS) on EEG rhythms are here reviewed to build a knowledge platform for innovative translational models for drug discovery in Alzheimer's disease (AD). It has been reported that antagonists of cholinergic neurotransmission cause synchronisation of spontaneous ongoing EEG rhythms in terms of enhanced power of EEG low frequencies and decreased power of EEG high frequencies. Acetylcholinesterase inhibitors and serotonergic drugs may restore a normal pattern of EEG desynchronisation. Sleep deprivation and hypoxia challenges have also been reported to elicit abnormal synchronisation of spontaneous ongoing EEG rhythms in rodents. The feasibility and reproducibility of TMS have been demonstrated in rodents but information on a consistent modulation of EEG after TMS manipulation is very limited. Transgenic mice over-expressing human amyloid precursor protein complementary DNAs (cDNAs) harbouring the 'Swedish' mutation and PS-1 cDNAs harbouring the A264E mutation, which recapitulate some of the pathological features of AD, exhibit alterations of spontaneous ongoing EEG rhythms at several low and high frequencies. This does not appear, however, to be a consequence of beta-amyloid deposition in the brain. The present review provides a critical evaluation of changes of spontaneous ongoing EEG rhythms due to the experimental manipulations described above, in order to stimulate the promote more adherent models fitting dynamics in humans.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Hipoxia/fisiopatología , Privación de Sueño/fisiopatología , Estimulación Magnética Transcraneal , Enfermedad de Alzheimer/fisiopatología , Animales , Electroencefalografía , Ratones , Ratas , Investigación Biomédica Traslacional
16.
Behav Brain Res ; 240: 146-52, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23178660

RESUMEN

Cholinergic dysfunction and deposition of plaques containing amyloid ß-peptides (Aß) are two of the characteristics of Alzheimer's disease. Here, we combine APPswe/PS1dE9 (APP/PS1) mice with the cholinergic immunotoxin mu p75-saporin (SAP) to integrate partial basal forebrain cholinergic degeneration and the neuropathology of APP/PS1 mice. By 6 months of age, APP/PS1 mice and wild type littermates (Wt) received intracerebroventricular injection of 0.6 µg SAP (lesion) or PBS (sham). Two months following surgery, APP/PS1 mice treated with SAP were significantly impaired compared to sham treated APP/PS1 mice in a behavioural paradigm addressing working memory. Conversely, the performance of Wt mice was unaffected by SAP treatment. Choline acetyltransferase activity was reduced in the hippocampus and frontal cortex following SAP treatment. The selective effect of a mild SAP lesion in APP/PS1 mice was not due to a more extensive cholinergic degeneration since the reduction in choline acetyltransferase activity was similar following SAP treatment in APP/PS1 mice and Wt. Interestingly, plaque load was significantly increased in SAP treated APP/PS1 mice relative to sham lesioned APP/PS1 mice. Additionally, APP/PS1 mice treated with SAP showed a tendency towards an increased level of soluble and insoluble Aß1-40 and Aß1-42 measured in brain tissue homogenate. Our results suggest that the combination of cholinergic degeneration and Aß overexpression in the APP/PS1 mouse model results in cognitive decline and accelerated plaque burden. SAP treated APP/PS1 mice might thus constitute an improved model of Alzheimer's disease-like neuropathology and cognitive deficits compared to the conventional APP/PS1 model without selective removal of basal forebrain cholinergic neurons.


Asunto(s)
Enfermedad de Alzheimer/etiología , Neuronas Colinérgicas/efectos de los fármacos , Modelos Animales de Enfermedad , Inmunotoxinas/farmacología , Placa Amiloide/patología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Colina O-Acetiltransferasa/efectos de los fármacos , Colina O-Acetiltransferasa/metabolismo , Neuronas Colinérgicas/patología , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inyecciones Intraventriculares , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Transgénicos , Placa Amiloide/fisiopatología , Presenilina-1/genética , Saporinas
17.
Behav Brain Res ; 213(2): 142-7, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20417666

RESUMEN

The use of translational approaches to validate animal models is needed for the development of treatments that can effectively alleviate cognitive impairments associated with schizophrenia, which are unsuccessfully treated by the current available therapies. Deficits in pre-attentive stages of sensory information processing seen in schizophrenia patients, can be assessed by highly homologues methods in both humans and rodents, evident by the prepulse inhibition (PPI) of the auditory startle response and the P50 (termed P1 here) suppression paradigms. Treatment with the NMDA receptor antagonist PCP on postnatal days 7, 9, and 11 reliably induce cognitive impairments resembling those presented by schizophrenia patients. Here we evaluate the potential of early postnatal PCP (20mg/kg) treatment in Lister Hooded rats to induce post-pubertal deficits in PPI and changes, such as reduced gating, in the P1 suppression paradigm in the EEG. The results indicate that early postnatal PCP treatment to rats leads to a reduction in PPI of the acoustic startle response. Furthermore, treated animals were assessed in the P1 suppression paradigm and produced significant changes in auditory-evoked potentials (AEP), specifically by an increased P1 amplitude and reduced P2 (P200 in humans) gating. However, the treatment neither disrupted normal P1 gating nor reduced N1 (N100 in humans) amplitude, representing two phenomena that are usually found to be disturbed in schizophrenia. In conclusion, the current findings confirm measures of early information processing to show high resemblance between rodents and humans, and indicate that early postnatal PCP-treated rats show deficits in pre-attentional processing, which are distinct from those observed in schizophrenia patients.


Asunto(s)
Modelos Animales de Enfermedad , Potenciales Evocados Auditivos/fisiología , Esquizofrenia/fisiopatología , Filtrado Sensorial/fisiología , Animales , Animales Recién Nacidos , Fenciclidina , Ratas , Ratas Endogámicas
18.
J Neurochem ; 110(5): 1377-87, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19519664

RESUMEN

Alzheimer's disease (AD) is hypothesized to result from elevated brain levels of beta-amyloid peptide (Abeta) which is the main component of plaques found in AD brains and which cause memory impairment in mice. Therefore, there has been a major focus on the development of inhibitors of the Abeta producing enzymes gamma-secretase and beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1). In this study, we investigated the Abeta-lowering effects of the BACE1 inhibitor LY2434074 in vitro and in vivo, comparing it to the well characterized gamma-secretase inhibitor LY450139. We sampled interstitial fluid Abeta from awake APPswe/PS1dE9 AD mice by in vivo Abeta microdialysis. In addition, we measured levels of endogenous brain Abeta extracted from wildtype C57BL/6 mice. In our in vitro assays both compounds showed similar Abeta-lowering effects. However, while systemic administration of LY450139 resulted in transient reduction of Abeta in both in vivo models, we were unable to show any Abeta-lowering effect by systemic administration of the BACE1 inhibitor LY2434074 despite brain exposure exceeding the in vitro IC(50) value several fold. In contrast, significant reduction of 40-50% of interstitial fluid Abeta and wildtype cortical Abeta was observed when infusing LY2434074 directly into the brain by means of reverse microdialysis or by dosing the BACE1 inhibitor to p-glycoprotein (p-gp) mutant mice. The effects seen in p-gp mutant mice and subsequent data from our cell-based p-gp transport assay suggested that LY2434074 is a p-gp substrate. This may partly explain why BACE1 inhibition by LY2434074 has lower in vivo efficacy, with respect to decreased Abeta40 levels, compared with gamma-secretase inhibition by LY450139.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Encéfalo/enzimología , Alanina/análogos & derivados , Alanina/farmacología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Azepinas/farmacología , Encéfalo/efectos de los fármacos , Línea Celular , Células Cultivadas , Perros , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de Proteasas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...