Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 13187, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162968

RESUMEN

Electrophysiological studies show that nicotine enhances neural responses to characteristic frequency stimuli. Previous behavioral studies partially corroborate these findings in young adults, showing that nicotine selectively enhances auditory processing in difficult listening conditions. The present work extended previous work to include both young and older adults and assessed the nicotine effect on sound frequency and intensity discrimination. Hypotheses were that nicotine improves auditory performance and that the degree of improvement is inversely proportional to baseline performance. Young (19-23 years old) normal-hearing nonsmokers and elderly (61-80) nonsmokers with normal hearing between 500 and 2000 Hz received nicotine gum (6 mg) or placebo gum in a single-blind, randomized crossover design. Participants performed three experiments (frequency discrimination, frequency modulation identification, and intensity discrimination) before and after treatment. The perceptual differences were analyzed between pre- and post-treatment, as well as between post-treatment nicotine and placebo conditions as a function of pre-treatment baseline performance. Compared to pre-treatment performance, nicotine significantly improved frequency discrimination. Compared to placebo, nicotine significantly improved performance for intensity discrimination, and the improvement was more pronounced in the elderly with lower baseline performance. Nicotine had no effect on frequency modulation identification. Nicotine effects are task-dependent, reflecting possible interplays of subjects, tasks and neural mechanisms.


Asunto(s)
Envejecimiento/fisiología , Percepción Auditiva/efectos de los fármacos , Nicotina/farmacología , No Fumadores , Afecto/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Estudios Cruzados , Discriminación en Psicología/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nicotina/administración & dosificación , Chicles de Nicotina , No Fumadores/psicología , Oxígeno/sangre , Percepción de la Altura Tonal/efectos de los fármacos , Desempeño Psicomotor , Receptores Nicotínicos/efectos de los fármacos , Receptores Nicotínicos/fisiología , Proyectos de Investigación , Relación Señal-Ruido , Método Simple Ciego , Adulto Joven
2.
Front Neural Circuits ; 15: 597401, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679335

RESUMEN

Systemic nicotine enhances neural processing in primary auditory cortex (A1) as determined using tone-evoked, current-source density (CSD) measurements. For example, nicotine enhances the characteristic frequency (CF)-evoked current sink in layer 4 of A1, increasing amplitude and decreasing latency. However, since presenting auditory stimuli within a stream of stimuli increases the complexity of response dynamics, we sought to determine the effects of nicotine on CSD responses to trains of CF stimuli (one-second trains at 2-40 Hz; each train repeated 25 times). CSD recordings were obtained using a 16-channel multiprobe inserted in A1 of urethane/xylazine-anesthetized mice, and analysis focused on two current sinks in the middle (layer 4) and deep (layers 5/6) layers. CF trains produced adaptation of the layer 4 response that was weak at 2 Hz, stronger at 5-10 Hz and complete at 20-40 Hz. In contrast, the layer 5/6 current sink exhibited less adaptation at 2-10 Hz, and simultaneously recorded auditory brainstem responses (ABRs) showed no adaptation even at 40 Hz. Systemic nicotine (2.1 mg/kg) enhanced layer 4 responses throughout the one-second stimulus train at rates ≤10 Hz. Nicotine enhanced both response amplitude within each train and the consistency of response timing across 25 trials. Nicotine did not alter the degree of adaptation over one-second trials, but its effect to increase amplitudes revealed a novel, slower form of adaptation that developed over multiple trials. Nicotine did not affect responses that were fully adapted (20-40 Hz trains), nor did nicotine affect any aspect of the layer 5/6 current sink or ABRs. The overall effect of nicotine in layer 4 was to enhance all responses within each train, to emphasize earlier trials across multiple trials, and to improve the consistency of timing across all trials. These effects may improve processing of complex acoustic streams, including speech, that contain information in the 2-10 Hz range.


Asunto(s)
Corteza Auditiva/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Potenciales Evocados Auditivos/efectos de los fármacos , Nicotina/farmacología , Estimulación Acústica/métodos , Acústica , Animales , Corteza Auditiva/fisiología , Potenciales Evocados Auditivos/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Masculino , Ratones , Factores de Tiempo
3.
Psychopharmacology (Berl) ; 237(3): 833-840, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31832719

RESUMEN

RATIONALE: Electrophysiological studies show that systemic nicotine narrows frequency receptive fields and increases gain in neural responses to characteristic frequency stimuli. We postulated that nicotine enhances related auditory processing in humans. OBJECTIVES: The main hypothesis was that nicotine improves auditory performance. A secondary hypothesis was that the degree of nicotine-induced improvement depends on the individual's baseline performance. METHODS: Young (18-27 years old), normal-hearing nonsmokers received nicotine (Nicorette gum, 6mg) or placebo gum in a single-blind, randomized, crossover design. Subjects performed four experiments involving tone-in-noise detection, temporal gap detection, spectral ripple discrimination, and selective auditory attention before and after treatment. The perceptual differences between posttreatment nicotine and placebo conditions were measured and analyzed as a function of the pre-treatment baseline performance. RESULTS: Nicotine significantly improved performance in the more difficult tasks of tone-in-noise detection and selective attention (effect size = - 0.3) but had no effect on relatively easier tasks of temporal gap detection and spectral ripple discrimination. The two tasks showing significant nicotine effects further showed no baseline-dependent improvement. CONCLUSIONS: Nicotine improves auditory performance in difficult listening situations. The present results support future investigation of nicotine effects in clinical populations with auditory processing deficits or reduced cholinergic activation.


Asunto(s)
Percepción Auditiva/efectos de los fármacos , Audición/efectos de los fármacos , Chicles de Nicotina , Nicotina/administración & dosificación , No Fumadores/psicología , Estimulación Acústica/métodos , Estimulación Acústica/psicología , Adolescente , Adulto , Atención/efectos de los fármacos , Atención/fisiología , Percepción Auditiva/fisiología , Estudios Cruzados , Femenino , Voluntarios Sanos , Audición/fisiología , Humanos , Masculino , Oximetría/métodos , Método Simple Ciego , Adulto Joven
4.
Synapse ; 73(9): e22116, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31081950

RESUMEN

Nicotine activates nicotinic acetylcholine receptors and improves cognitive and sensory function, in part by its actions in cortical regions. Physiological studies show that nicotine amplifies stimulus-evoked responses in sensory cortex, potentially contributing to enhancement of sensory processing. However, the role of specific cell types and circuits in the nicotinic modulation of sensory cortex remains unclear. Here, we performed whole-cell recordings from pyramidal (Pyr) neurons and inhibitory interneurons expressing parvalbumin (PV), somatostatin (SOM), and vasoactive intestinal peptide (VIP) in mouse auditory cortex, in vitro. Bath application of nicotine strongly depolarized and excited VIP neurons, weakly depolarized Pyr neurons, and had no effect on the membrane potential of SOM or PV neurons. The use of receptor antagonists showed that nicotine's effects on VIP and Pyr neurons were direct and indirect, respectively. Nicotine also enhanced the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) in Pyr, VIP, and SOM, but not PV, cells. Using Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), we show that chemogenetic inhibition of VIP neurons prevents nicotine's effects on Pyr neurons. Since VIP cells preferentially contact other inhibitory interneurons, we suggest that nicotine drives VIP cell firing to disinhibit Pyr cell somata, potentially making Pyr cells more responsive to auditory stimuli. In parallel, activation of VIP cells also directly inhibits Pyr neurons, likely altering integration of other synaptic inputs. These cellular and synaptic mechanisms likely contribute to nicotine's beneficial effects on cognitive and sensory function.


Asunto(s)
Corteza Auditiva/efectos de los fármacos , Interneuronas/efectos de los fármacos , Nicotina/farmacología , Células Piramidales/efectos de los fármacos , Animales , Corteza Auditiva/fisiología , Femenino , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Interneuronas/metabolismo , Masculino , Ratones , Agonistas Nicotínicos/farmacología , Células Piramidales/fisiología , Péptido Intestinal Vasoactivo/metabolismo
5.
Nicotine Tob Res ; 21(3): 377-382, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30137439

RESUMEN

Activation of nicotinic acetylcholine receptors (nAChRs) enhances sensory-cognitive function in human subjects and animal models, yet the neural mechanisms are not fully understood. This review summarizes recent studies on nicotinic regulation of neural processing in the cerebral cortex that point to potential mechanisms underlying enhanced cognitive function. Studies from our laboratory focus on nicotinic regulation of auditory cortex and implications for auditory-cognitive processing, but relevant emerging insights from multiple brain regions are discussed. Although the major contributions of the predominant nAChRs containing α7 (homomeric receptors) or α4 and ß2 (heteromeric) subunits are well recognized, recent results point to additional, potentially critical contributions from α2 subunits that are relatively sparse in cortex. Ongoing studies aim to elucidate the specific contributions to cognitive and cortical function of diverse nAChRs. IMPLICATIONS: This review highlights the therapeutic potential of activating nAChRs in the cerebral cortex to enhance cognitive function. Future work also must determine the contributions of relatively rare but important nAChR subtypes, potentially to develop more selective treatments for cognitive deficits.


Asunto(s)
Trastornos del Conocimiento/prevención & control , Cognición/efectos de los fármacos , Agonistas Nicotínicos/uso terapéutico , Receptores Nicotínicos/metabolismo , Trastornos de la Sensación/prevención & control , Animales , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Humanos , Trastornos de la Sensación/metabolismo , Trastornos de la Sensación/patología
6.
Bioorg Med Chem Lett ; 28(3): 371-377, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29277457

RESUMEN

Nicotinic acetylcholine α4ß2∗ receptors (nAChRs) are implicated in various neurodegenerative diseases and smoking addiction. Imaging of brain high-affinity α4ß2∗ nAChRs at the cellular and subcellular levels would greatly enhance our understanding of their functional role. Since better resolution could be achieved with fluorescent probes, using our previously developed positron emission tomography (PET) imaging agent [18F]nifrolidine, we report here design, synthesis and evaluation of two fluorescent probes, nifrodansyl and nifrofam for imaging α4ß2∗ nAChRs. The nifrodansyl and nifrofam exhibited nanomolar affinities for the α4ß2∗ nAChRs in [3H]cytisine-radiolabeled rat brain slices. Nifrofam labeling was observed in α4ß2∗ nAChR-expressing HEK cells and was upregulated by nicotine exposure. Nifrofam co-labeled cell-surface α4ß2∗ nAChRs, labeled with antibodies specific for a ß2 subunit extracellular epitope indicating that nifrofam labels α4ß2∗ nAChR high-affinity binding sites. Mouse brain slices exhibited discrete binding of nifrofam in the auditory cortex showing promise for examining cellular distribution of α4ß2∗ nAChRs in brain regions.


Asunto(s)
Colorantes Fluorescentes/química , Imagen Óptica , Receptores Nicotínicos/análisis , Animales , Sitios de Unión , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/farmacocinética , Células HEK293 , Humanos , Ratones , Estructura Molecular , Tomografía de Emisión de Positrones , Relación Estructura-Actividad , Distribución Tisular
7.
J Comp Neurol ; 526(1): 80-95, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28875553

RESUMEN

Nicotinic acetylcholinergic receptors (nAChR's) have been implicated in several brain disorders, including addiction, Parkinson's disease, Alzheimer's disease and schizophrenia. Here we report in vitro selectivity and functional properties, toxicity in rats, in vivo evaluation in humans, and comparison across species of [18 F]Nifene, a fast acting PET imaging agent for α4ß2* nAChRs. Nifene had subnanomolar affinities for hα2ß2 (0.34 nM), hα3ß2 (0.80 nM) and hα4ß2 (0.83 nM) nAChR but weaker (27-219 nM) for hß4 nAChR subtypes and 169 nM for hα7 nAChR. In functional assays, Nifene (100 µM) exhibited 14% agonist and >50% antagonist characteristics. In 14-day acute toxicity in rats, the maximum tolerated dose (MTD) and the no observed adverse effect level (NOAEL) were estimated to exceed 40 µg/kg/day (278 µg/m2 /day). In human PET studies, [18 F]Nifene (185 MBq; <0.10 µg) was well tolerated with no adverse effects. Distribution volume ratios (DVR) of [18 F]Nifene in white matter thalamic radiations were ∼1.6 (anterior) and ∼1.5 (superior longitudinal fasciculus). Habenula known to contain α3ß2 nAChR exhibited low levels of [18 F]Nifene binding while the red nucleus with α2ß2 nAChR had DVR ∼1.6-1.7. Females had higher [18 F]Nifene binding in all brain regions, with thalamus showing >15% than males. No significant aging effect was observed in [18 F]Nifene binding over 5 decades. In all species (mice, rats, monkeys, and humans) thalamus showed highest [18 F]Nifene binding with reference region ratios >2 compared to extrathalamic regions. Our findings suggest that [18 F]Nifene PET may be used to study α4ß2* nAChRs in various CNS disorders and for translational research.


Asunto(s)
Envejecimiento/fisiología , Encéfalo , Piridinas/farmacocinética , Pirroles/farmacocinética , Receptores Nicotínicos/metabolismo , Caracteres Sexuales , Adulto , Anciano , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Persona de Mediana Edad , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Tomografía de Emisión de Positrones , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Adulto Joven
8.
eNeuro ; 4(3)2017.
Artículo en Inglés | MEDLINE | ID: mdl-28660244

RESUMEN

Nicotine enhances sensory and cognitive processing via actions at nicotinic acetylcholine receptors (nAChRs), yet the precise circuit- and systems-level mechanisms remain unclear. In sensory cortex, nicotinic modulation of receptive fields (RFs) provides a model to probe mechanisms by which nAChRs regulate cortical circuits. Here, we examine RF modulation in mouse primary auditory cortex (A1) using a novel electrophysiological approach: current-source density (CSD) analysis of responses to tone-in-notched-noise (TINN) acoustic stimuli. TINN stimuli consist of a tone at the characteristic frequency (CF) of the recording site embedded within a white noise stimulus filtered to create a spectral "notch" of variable width centered on CF. Systemic nicotine (2.1 mg/kg) enhanced responses to the CF tone and to narrow-notch stimuli, yet reduced the response to wider-notch stimuli, indicating increased response gain within a narrowed RF. Subsequent manipulations showed that modulation of cortical RFs by systemic nicotine reflected effects at several levels in the auditory pathway: nicotine suppressed responses in the auditory midbrain and thalamus, with suppression increasing with spectral distance from CF so that RFs became narrower, and facilitated responses in the thalamocortical pathway, while nicotinic actions within A1 further contributed to both suppression and facilitation. Thus, multiple effects of systemic nicotine integrate along the ascending auditory pathway. These actions at nAChRs in cortical and subcortical circuits, which mimic effects of auditory attention, likely contribute to nicotinic enhancement of sensory and cognitive processing.


Asunto(s)
Corteza Auditiva/efectos de los fármacos , Percepción Auditiva/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Estimulación Acústica , Análisis de Varianza , Animales , Corteza Auditiva/fisiología , Vías Auditivas/efectos de los fármacos , Vías Auditivas/fisiología , Percepción Auditiva/fisiología , Potenciales Evocados Auditivos/efectos de los fármacos , Agonistas de Receptores de GABA-A/farmacología , Masculino , Ratones , Microinyecciones , Muscimol/farmacología , Oxadiazoles/farmacología , Piridinas/farmacología , Receptores Nicotínicos/metabolismo
9.
J Neurophysiol ; 115(4): 2083-94, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26888102

RESUMEN

The GABAergic agonist muscimol is used to inactivate brain regions in order to reveal afferent inputs in isolation. However, muscimol's use in primary auditory cortex (A1) has been questioned on the grounds that it may unintentionally suppress thalamocortical inputs. We tested whether muscimol can preferentially suppress cortical, but not thalamocortical, circuits in urethane-anesthetized mice. We recorded tone-evoked current source density profiles to determine frequency receptive fields (RFs) for three current sinks: the "layer 4" sink (fastest onset, middle-layer sink) and current sinks 100 µm above ("layer 2/3") and 300 µm below ("layer 5/6") the main input. We first determined effects of muscimol dose (0.01-1 mM) on the characteristic frequency (CF) tone-evoked layer 4 sink. An "ideal" dose (100 µM) had no effect on CF-evoked sink onset latency or initial response but reduced peak amplitude by >80%, implying inhibition of intracortical, but not thalamocortical, activity. We extended the analysis to current sinks in layers 2/3 and 5/6 and for all three sinks determined RF breadth (quarter-octave steps, 20 dB above CF threshold). Muscimol reduced RF breadth 42% in layer 2/3 (from 2.4 ± 0.14 to 1.4 ± 0.11 octaves), 14% in layer 4 (2.2 ± 0.12 to 1.9 ± 0.10 octaves), and not at all in layer 5/6 (1.8 ± 0.10 to 1.7 ± 0.12 octaves). The results provide an estimate of the laminar and spectral extent of thalamocortical projections and support the hypothesis that intracortical pathways contribute to spectral integration in A1.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Neuronas/efectos de los fármacos , Tálamo/efectos de los fármacos , Potenciales de Acción , Animales , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Agonistas de Receptores de GABA-A/farmacología , Masculino , Ratones , Muscimol/farmacología , Vías Nerviosas/citología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Neuronas/fisiología , Tálamo/citología , Tálamo/fisiología
10.
Biol Psychol ; 116: 4-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26555718

RESUMEN

This Special Issue focuses on the auditory-evoked mismatch negativity (MMN), an electrophysiological index of change, and its reduction in schizophrenia. The following brief review is an attempt to complement the behavioral and clinical contributions to the Special Issue by providing basic information on synaptic interactions and processing in auditory cortex. A key observation in previous studies is that the MMN involves activation of cortical N-methyl-D-aspartate (NMDA) receptors. Yet, NMDA receptor activation is regulated by a number of synaptic events, which also may contribute to the MMN reduction in schizophrenia. Accordingly, this review will focus on synaptic interactions, notably inhibitory regulation of NMDA receptor-mediated activity, in auditory cortex.


Asunto(s)
Corteza Auditiva/fisiología , Sinapsis Eléctricas/fisiología , Potenciales Evocados Auditivos/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , GABAérgicos/metabolismo , Humanos , Interneuronas/fisiología , Ratones , Ratas , Receptores de GABA-B/metabolismo , Receptores de Glutamato/metabolismo , Esquizofrenia/fisiopatología
11.
Synapse ; 67(8): 455-68, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23401204

RESUMEN

Enhancement of sound-evoked responses in auditory cortex (ACx) following administration of systemic nicotine is known to depend on activation of extracellular-signaling regulated kinase (ERK), but the nature of this enhancement is not clear. Here, we show that systemic nicotine increases the density of cells immunolabeled for phosphorylated (activated) ERK (P-ERK) in mouse primary ACx (A1). Cortical injection of dihydro-ß-erythroidine reduced nicotine-induced P-ERK immunolabel, suggesting a role for nicotinic acetylcholine receptors located in A1 and containing α4 and ß2 subunits. P-ERK expressing cells were distributed mainly in layers 2/3 and more sparsely in lower layers, with many cells exhibiting immunolabel within pyramidal-shaped somata and proximal apical dendrites. About one-third of P-ERK positive cells also expressed calbindin. In the thalamus, P-ERK immunopositive cells were found in the nonlemniscal medial geniculate (MG) and adjacent nuclei, but were absent in the lemniscal MG. Pairing broad spectrum acoustic stimulation (white noise) with systemic nicotine increased P-ERK immunopositive cell density in ACx as well as the total amount of P-ERK protein, particularly the phosphorylated form of ERK2. However, narrow spectrum (tone) stimulation paired with nicotine increased P-ERK immunolabel preferentially at a site within A1 where the paired frequency was characteristic frequency (CF), relative to a second site with a spectrally distant CF (two octaves above or below the paired frequency). Together, these results suggest that ERK is activated optimally where nicotinic signaling and sound-evoked neural activity converge.


Asunto(s)
Corteza Auditiva/fisiología , Potenciales Evocados Auditivos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Nicotina/farmacología , Estimulación Acústica , Animales , Corteza Auditiva/citología , Corteza Auditiva/metabolismo , Calbindinas , Dihidro-beta-Eritroidina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Fosforilación , Subunidades de Proteína/metabolismo , Receptores Nicotínicos/metabolismo , Proteína G de Unión al Calcio S100/genética , Proteína G de Unión al Calcio S100/metabolismo , Tálamo/citología , Tálamo/metabolismo
12.
Neurobiol Learn Mem ; 101: 75-84, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23357284

RESUMEN

Auditory-cued behavioral training can alter neural circuits in primary auditory cortex (A1), but the mechanisms and consequences of experience-dependent cortical plasticity are not fully understood. To address this issue, we trained adult rats to detect a 5 kHz target in order to receive a food reward. After 14 days training we identified three locations within A1: (i) the region representing the characteristic frequency (CF) 5 kHz, (ii) a nearby region with CF ∼10 kHz, and (iii) a more distant region with CF ∼20 kHz. In order to compare functional connectivity in A1 near to, vs. far from, the representation of the target frequency, we placed a 16-channel multiprobe in middle- (∼10 kHz) and high- (∼20 kHz) CF regions and obtained current-source density (CSD) profiles evoked by a range of tone stimuli (CF±1-3 octaves in quarter-octave steps). Our aim was to construct "CSD receptive fields" (CSD RFs) in order to determine the laminar and spectral profile of tone-evoked current sinks, and infer changes to thalamocortical and intracortical inputs. Behavioral training altered CSD RFs at the 10 kHz, but not 20 kHz, site relative to CSD RFs in untrained control animals. At the 10 kHz site, current sinks evoked by the target frequency were enhanced in layer 2/3, but the initial current sink in layer 4 was not altered. The results imply training-induced plasticity along intracortical pathways connecting the target representation with nearby cortical regions. Finally, we related behavioral performance (sensitivity index, d') to CSD responses in individual animals, and found a significant correlation between the development of d' over training and the amplitude of the target-evoked current sink in layer 2/3. The results suggest that plasticity along intracortical pathways is important for auditory learning.


Asunto(s)
Corteza Auditiva/fisiología , Condicionamiento Operante/fisiología , Potenciales Evocados Auditivos/fisiología , Estimulación Acústica , Animales , Masculino , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley
13.
Front Behav Neurosci ; 6: 44, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22833720

RESUMEN

Although it has been known for decades that the drug nicotine can improve cognitive function, the nature of its effects and the underlying mechanisms are not well understood. Nicotine activates nicotinic acetylcholine (ACh) receptors (nAChRs) that normally are activated by endogenous ACh, presumably "hijacking" the cholinergic contribution to multiple cognitive functions, notably attention. Thus, studying nicotine's effects helps to better understand a commonly used drug as well as functions of nAChRs. Moreover, nicotinic agonists are being developed to treat a variety of disorders that involve attention-related or age-related cognitive dysfunction. Studies have shown that nicotine can enhance processing of attended stimuli and/or reduce processing of distracters; that is, nicotine enhances attentional filtering. To examine potential mechanisms within sensory cortex that may contribute to cognitive functions, here we describe nicotinic actions in primary auditory cortex, where well-characterized neural "filters"-frequency receptive fields-can be exploited to examine nicotinic regulation of cortical processing. Using tone-evoked current-source density (CSD) profiles, we show that nicotine produces complex, layer-dependent effects on spectral and temporal processing that, broadly speaking, enhance responses to characteristic frequency (optimal) stimuli while simultaneously suppressing responses to spectrally distant stimuli. That is, nicotine appears to narrow receptive fields and enhances processing within the narrowed receptive field. Since basic cortical circuitry and nAChR distributions are similar across neocortex, these findings may generalize to neural processing in other sensory regions, and to non-sensory regions where afferent inputs are more difficult to manipulate experimentally. Similar effects across sensory and non-sensory cortical circuits could contribute to nicotinic enhancement of cognitive functions.

14.
J Neurophysiol ; 107(10): 2782-93, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22357798

RESUMEN

Activation of nicotinic acetylcholine receptors (nAChRs) by systemic nicotine enhances sensory-cognitive function and sensory-evoked cortical responses. Although nAChRs mediate fast neurotransmission at many synapses in the nervous system, nicotinic regulation of cortical processing is neuromodulatory. To explore potential mechanisms of nicotinic neuromodulation, we examined whether intracellular signal transduction involving mitogen-activated protein kinase (MAPK) contributes to regulation of tone-evoked responses in primary auditory cortex (A1) in the mouse. Systemic nicotine enhanced characteristic frequency (CF) tone-evoked current-source density (CSD) profiles in A1, including the shortest-latency (presumed thalamocortical) current sink in layer 4 and longer-latency (presumed intracortical) sinks in layers 2-4, by increasing response amplitudes and decreasing response latencies. Microinjection of the MAPK kinase (MEK) inhibitor U0126 into the thalamus, targeting the auditory thalamocortical pathway, blocked the effect of nicotine on the initial (thalamocortical) CSD component but did not block enhancement of longer-latency (intracortical) responses. Conversely, microinjection of U0126 into supragranular layers of A1 blocked nicotine's effect on intracortical, but not thalamocortical, CSD components. Simultaneously with enhancement of CF-evoked responses, responses to spectrally distant (nonCF) stimuli were reduced, implying nicotinic "sharpening" of frequency receptive fields, an effect also blocked by MEK inhibition. Consistent with these physiological results, acoustic stimulation with nicotine produced immunolabel for activated MAPK in A1, primarily in layer 2/3 cell bodies. Immunolabel was blocked by intracortical microinjection of the nAChR antagonist dihydro-ß-erythroidine, but not methyllycaconitine, implicating α4ß2*, but not α7, nAChRs. Thus activation of MAPK in functionally distinct forebrain circuits--thalamocortical, local intracortical, and long-range intracortical--underlies nicotinic neuromodulation of A1.


Asunto(s)
Corteza Auditiva/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Nicotina/farmacología , Receptores Nicotínicos/metabolismo , Tálamo/efectos de los fármacos , Estimulación Acústica , Animales , Corteza Auditiva/metabolismo , Butadienos/farmacología , Inhibidores Enzimáticos/farmacología , Potenciales Evocados Auditivos/efectos de los fármacos , Potenciales Evocados Auditivos/fisiología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Masculino , Ratones , Vías Nerviosas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nitrilos/farmacología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Tálamo/metabolismo
15.
Synapse ; 66(5): 418-34, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22213342

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) in the brain are important for cognitive function; however, their specific role in relevant brain regions remains unclear. In this study, we used the novel compound ¹8F-nifene to examine the distribution of nAChRs in the rat forebrain, and for individual animals related the results to behavioral performance on an auditory-cognitive task. We first show negligible binding of ¹8F-nifene in mice lacking the ß2 nAChR subunit, consistent with previous findings that ¹8F-nifene binds to α4ß2* nAChRs. We then examined the distribution of ¹8F-nifene in rat using three methods: in vivo PET, ex vivo PET and autoradiography. Generally, ¹8F-nifene labeled forebrain regions known to contain nAChRs, and the three methods produced similar relative binding among regions. Importantly, ¹8F-nifene also labeled some white matter (myelinated axon) tracts, most prominently in the temporal subcortical region that contains the auditory thalamocortical pathway. Finally, we related ¹8F-nifene binding in several forebrain regions to each animal's performance on an auditory-cued, active avoidance task. The strongest correlations with performance after 14 days training were found for ¹8F-nifene binding in the temporal subcortical white matter, subiculum, and medial frontal cortex (correlation coefficients, r > 0.8); there was no correlation with binding in the auditory thalamus or auditory cortex. These findings suggest that individual performance is linked to nicotinic functions in specific brain regions, and further support a role for nAChRs in sensory-cognitive function.


Asunto(s)
Reacción de Prevención/fisiología , Radioisótopos de Flúor/farmacocinética , Prosencéfalo/metabolismo , Piridinas/farmacocinética , Pirroles/farmacocinética , Receptores Nicotínicos/metabolismo , Animales , Autorradiografía , Cerebelo/diagnóstico por imagen , Cerebelo/metabolismo , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/metabolismo , Lóbulo Frontal/diagnóstico por imagen , Lóbulo Frontal/metabolismo , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Fibras Nerviosas Mielínicas/diagnóstico por imagen , Fibras Nerviosas Mielínicas/metabolismo , Tomografía de Emisión de Positrones , Prosencéfalo/diagnóstico por imagen , Radiofármacos , Ratas , Ratas Sprague-Dawley , Tálamo/diagnóstico por imagen , Tálamo/metabolismo
16.
J Neurosci ; 31(40): 14367-77, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21976522

RESUMEN

Adolescent smoking is associated with auditory-cognitive deficits and structural alterations to auditory thalamocortical systems, suggesting that higher auditory function is vulnerable to nicotine exposure during adolescence. Although nicotinic acetylcholine receptors (nAChRs) regulate thalamocortical processing in adults, it is not known whether they regulate processing at earlier ages since their expression pattern changes throughout postnatal development. Here we investigate nicotinic regulation of tone-evoked current source density (CSD) profiles in mouse primary auditory cortex from just after hearing onset until adulthood. At the youngest ages, systemic nicotine did not affect CSD profiles. However, beginning in early adolescence nicotine enhanced characteristic frequency (CF)-evoked responses in layers 2-4 by enhancing thalamocortical, early intracortical, and late intracortical response components. Nicotinic responsiveness developed rapidly and peaked over the course of adolescence, then declined thereafter. Generally, responsiveness in females developed more quickly, peaked earlier, and declined more abruptly and fully than in males. In contrast to the enhancement of CF-evoked responses, nicotine suppressed shorter-latency intracortical responses to spectrally distant (non-CF) stimuli while enhancing longer-latency responses. Intracortical infusion of nAChR antagonists showed that enhancement of CF-evoked intracortical processing involves α4ß2*, but not α7, nAChRs, whereas both receptor subtypes regulate non-CF-evoked late intracortical responses. Notably, antagonist effects in females implied regulation by endogenous acetylcholine. Thus, nicotinic regulation of cortical processing varies with age and sex, with peak effects during adolescence that may contribute to the vulnerability of adolescents to smoking.


Asunto(s)
Estimulación Acústica/métodos , Corteza Auditiva/fisiología , Potenciales Evocados Auditivos/fisiología , Nicotina/administración & dosificación , Factores de Edad , Animales , Corteza Auditiva/efectos de los fármacos , Potenciales Evocados Auditivos/efectos de los fármacos , Femenino , Inyecciones Intraventriculares , Masculino , Ratones , Antagonistas Nicotínicos/administración & dosificación , Receptores Nicotínicos/fisiología , Receptor Nicotínico de Acetilcolina alfa 7
17.
PLoS One ; 6(10): e26192, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022561

RESUMEN

Primary auditory cortex (A1) exhibits a tonotopic representation of characteristic frequency (CF). The receptive field properties of A1 neurons emerge from a combination of thalamic inputs and intracortical connections. However, the mechanisms that guide growth of these inputs during development and shape receptive field properties remain largely unknown. We previously showed that Eph family proteins help establish tonotopy in the auditory brainstem. Moreover, other studies have shown that these proteins shape topography in visual and somatosensory cortices. Here, we examined the contribution of Eph proteins to cortical organization of CF, response thresholds and sharpness of frequency tuning. We examined mice with null mutations in EphB2 and EphB3, as these mice show significant changes in auditory brainstem connectivity. We mapped A1 using local field potential recordings in adult EphB2(-/-);EphB3(-/-) and EphB3(-/-) mice, and in a central A1 location inserted a 16-channel probe to measure tone-evoked current-source density (CSD) profiles. Based on the shortest-latency current sink in the middle layers, which reflects putative thalamocortical input, we determined frequency receptive fields and sharpness of tuning (Q(20)) for each recording site. While both mutant mouse lines demonstrated increasing CF values from posterior to anterior A1 similar to wild type mice, we found that the double mutant mice had significantly lower Q(20) values than either EphB3(-/-) mice or wild type mice, indicating broader tuning. In addition, we found that the double mutants had significantly higher CF thresholds and longer onset latency at threshold than mice with wild type EphB2. These results demonstrate that EphB receptors influence auditory cortical responses, and suggest that EphB signaling has multiple functions in auditory system development.


Asunto(s)
Corteza Auditiva/fisiología , Umbral Auditivo/fisiología , Mutación/genética , Receptores de la Familia Eph/genética , Animales , Vías Auditivas/fisiología , Potenciales Evocados Auditivos , Masculino , Ratones , Receptores de la Familia Eph/metabolismo
18.
Neurosci Biobehav Rev ; 35(10): 2058-63, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21144860

RESUMEN

Although it is known that primary auditory cortex (A1) contributes to the processing and perception of sound, its precise functions and the underlying mechanisms are not well understood. Recent studies point to a remarkably broad spectral range of largely subthreshold inputs to individual neurons in A1--seemingly encompassing, in some cases, the entire audible spectrum--as evidence for potential, and potentially unique, cortical functions. We have proposed a general mechanism for spectral integration by which information converges on neurons in A1 via a combination of thalamocortical pathways and intracortical long-distance, "horizontal", pathways. Here, this proposal is briefly reviewed and updated with results from multiple laboratories. Since spectral integration in A1 is dynamically regulated, we also show how one regulatory mechanism--modulation by the neurotransmitter acetylcholine (ACh)--could act within the hypothesized framework to alter integration in single neurons. The results of these studies promote a cellular understanding of information processing in A1.


Asunto(s)
Corteza Auditiva/fisiología , Vías Auditivas/fisiología , Neuronas Colinérgicas/fisiología , Tálamo/fisiología , Animales , Corteza Auditiva/efectos de los fármacos , Agonistas Colinérgicos/farmacología , Antagonistas Colinérgicos/farmacología , Humanos , Modelos Neurológicos , Tálamo/efectos de los fármacos
19.
J Neurosci ; 29(13): 4004-15, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19339596

RESUMEN

Soluble amyloid beta oligomers (AbetaOs) interfere with synaptic function and bind with high affinity to synapses, but the mechanism underlying AbetaO synaptic targeting is not known. Here, we show that the accumulation of synthetic or native Alzheimer's disease (AD)-brain oligomers at synapses is regulated by synaptic activity. Electrical or chemical stimulation increased AbetaO synaptic localization and enhanced oligomer formation at synaptic terminals, whereas inhibition with TTX blocked AbetaO synaptic localization and reduced AbetaO synaptic load. The zinc-binding 8-OH-quinoline clioquinol markedly reduced AbetaO synaptic targeting, which was also reduced in brain sections of animals deficient in the synaptic vesicle zinc transporter ZnT3, indicating that vesicular zinc released during neurotransmission is critical for AbetaO synaptic targeting. Oligomers were not internalized in recycled vesicles but remained at the cell surface, where they colocalized with NR2B NMDA receptor subunits. Furthermore, NMDA antagonists blocked AbetaO synaptic targeting, implicating excitatory receptor activity in oligomer formation and accumulation at synapses. In AD brains, oligomers of different size colocalized with synaptic markers in hippocampus and cortex, where oligomer synaptic accumulation correlated with synaptic loss.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Sinapsis/metabolismo , Zinc/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Animales , Encéfalo/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Proteínas Portadoras , Proteínas de Transporte de Catión , Células Cultivadas , Interacciones Farmacológicas , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Ácido Glutámico/farmacología , Hipocampo/citología , Humanos , Técnicas In Vitro , Proteínas de la Membrana/deficiencia , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados , Neuronas , Cloruro de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos , Sinaptofisina/metabolismo , Tetrodotoxina/farmacología , Factores de Tiempo , Zinc/farmacología
20.
Neurobiol Learn Mem ; 90(1): 138-46, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18378471

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) contribute to sensory-cognitive function, as demonstrated by evidence that nAChR activation enhances, and nAChR blockade impairs, neural processing of sensory stimuli and sensory-cognitive behavior. To better understand the relationship between nAChR function and behavior, here we compare the strength of nAChR-mediated physiology in individual animals to their prior auditory behavioral performance. Adult rats were trained on an auditory-cued, active avoidance task over 4 days and classified as "good," "intermediate" or "poor" performers based on their initial rate of learning and eventual level of performance. Animals were then anesthetized, and tone-evoked local field potentials (LFPs) recorded in layer 4 of auditory cortex (ACx) before and after a test dose of nicotine (0.7mg/kg, s.c.) or saline. In "good" performers, nicotine enhanced LFP amplitude and decreased response threshold to characteristic frequency (CF) stimuli, yet had opposite effects (decreased amplitude, increased threshold) on responses to spectrally distant stimuli; i.e., cortical receptive fields became more selective for CF stimuli. In contrast, nicotine had little effect on LFP amplitude in "intermediate" or "poor" performing animals. Nicotine did, however, reduce LFP onset latency in all three groups, indicating that all received an effective dose of the drug. Our findings suggest that nicotinic regulation of cortical receptive fields may be a distinguishing feature of the best-performing animals, and may facilitate sensory-related learning by enhancing receptive field selectivity.


Asunto(s)
Corteza Auditiva/fisiología , Reacción de Prevención/fisiología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Receptores Nicotínicos/fisiología , Acetilcolina/fisiología , Estimulación Acústica , Animales , Corteza Auditiva/efectos de los fármacos , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Potenciales Evocados Auditivos/efectos de los fármacos , Potenciales Evocados Auditivos/fisiología , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA