Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Neuron ; 99(6): 1260-1273.e4, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30146308

RESUMEN

Reward-seeking behavior is regulated by a diverse collection of inputs to the nucleus accumbens (NAc). The information encoded in each excitatory afferent to the NAc is unknown, in part because it is unclear when these pathways are active in relation to behavior. Here we compare the activity profiles of amygdala, hippocampal, and thalamic inputs to the NAc shell in mice performing a cued reward-seeking task using GCaMP-based fiber photometry. We find that the rostral and caudal ends of the NAc shell are innervated by distinct but intermingled populations of forebrain neurons that exhibit divergent feeding-related activity. In the rostral NAc shell, a coordinated network-wide reduction in excitatory drive correlates with feeding, and reduced input from individual pathways is sufficient to promote it. Overall, the data suggest that pathway-specific input activity at a population level may vary more across the NAc than between pathways.


Asunto(s)
Amígdala del Cerebelo/fisiología , Conducta Animal/fisiología , Hipocampo/fisiología , Núcleo Accumbens/fisiología , Animales , Señales (Psicología) , Masculino , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Neuronas/fisiología , Recompensa , Tálamo/fisiología
2.
Anesth Analg ; 122(6): 1818-25, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26836135

RESUMEN

BACKGROUND: Thalamocortical electroencephalographic rhythms in gamma (30-80 Hz) and high-gamma (80-200 Hz) ranges have been linked to arousal and conscious processes. We have recently shown that propofol causes a concentration-dependent attenuation of the power of thalamocortical rhythms in the 50 to 200 Hz range and that this effect is far more pronounced for the thalamus. To determine whether similar attenuation occurs with other anesthetics, we characterized the concentration-effect relationship of the inhaled anesthetic isoflurane on the spectral power of these rhythms. METHODS: Local field potentials were recorded from the barrel cortex and ventroposteromedial thalamic nucleus in 9 chronically instrumented rats to measure spectral power in the gamma/high-gamma range (30-200 Hz). Rats were placed in an airtight chamber and isoflurane was administered at 0.75%, 1.1%, and 1.5% concentrations. Spectral power was assessed during baseline, at the 3 isoflurane concentrations after 30 minutes for equilibration, and during recovery over 4 frequency bands (30-50, 51-75, 76-125, and 126-200 Hz). Unconsciousness was defined as sustained loss of righting reflex. Multiple linear regression was used to model the change in power (after logarithmic transformation) as a function of concentration and recording site. P values were corrected for multiple comparisons. RESULTS: Unconsciousness occurred at the 1.1% concentration in all animals. Isoflurane caused a robust (P ≤ 0.008) linear concentration-dependent attenuation of cortical and thalamic power in the 30 to 200 Hz range. The concentration-effect slope for the thalamus was steeper than for the cortex in the 51 to 75 Hz (P = 0.029) and 76 to 200 Hz (P < 0.001) ranges but not for the 30 to 50 Hz range (P = 0.320). Comparison with our previously published propofol data showed that slope for cortical power was steeper with isoflurane than with propofol for all frequency bands (P = 0.033). For thalamic power, the slope differences between isoflurane and propofol were not statistically significant (0.087 ≤ P ≤ 0.599). CONCLUSIONS: Isoflurane causes a concentration-dependent attenuation of the power of thalamocortical rhythms in the 30 to 200 Hz range, and this effect is more pronounced for the thalamus than for the cortex for frequencies >50 Hz. In comparison with propofol, isoflurane caused a greater attenuation in the cortex, but the effects on the thalamus were similar. Isoflurane and propofol cause common alterations of fast thalamocortical rhythms that may constitute an electrophysiologic signature of the anesthetized state.


Asunto(s)
Anestésicos por Inhalación/administración & dosificación , Corteza Cerebral/efectos de los fármacos , Electroencefalografía , Ritmo Gamma/efectos de los fármacos , Isoflurano/administración & dosificación , Tálamo/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Corteza Cerebral/fisiología , Estado de Conciencia/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Ratas Long-Evans , Reflejo de Enderezamiento/efectos de los fármacos , Procesamiento de Señales Asistido por Computador , Tálamo/fisiología , Factores de Tiempo
4.
PLoS One ; 10(4): e0123287, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875024

RESUMEN

BACKGROUND: Thalamocortical EEG rhythms in gamma (30-80 Hz) and high-gamma (80-200 Hz) ranges have been linked to arousal and conscious processes. To test the hypothesis that general anesthetics attenuate these rhythms, we characterized the concentration-effect relationship of propofol on the spectral power of these rhythms. In view of the ongoing debate about cortex versus thalamus as the primary site of anesthetic action for unconsciousness, we also compared the relative sensitivity of cortex and thalamus to this effect propofol. METHODS: Adult male Long-Evans rats were chronically implanted with electrodes in somatosensory (barrel) cortex and ventroposteromedial thalamus. Propofol was delivered by a computer-controlled infusion using real-time pharmacokinetic modeling to obtain the desired plasma concentration. Spectral power was assessed during baseline, at four stable propofol plasma-concentrations (0, 3,6,9,12 µg/ml) and during recovery over four frequency ranges (30-50, 51-75, 76-125, 126-200 Hz). Unconsciousness was defined as complete loss of righting reflex. Multiple regression was used to model the change of power (after logarithmic transformation) as a function of propofol concentration and recording site. RESULTS: Unconsciousness occurred at the 9 µg/ml concentration in all animals. Propofol caused a robust linear concentration-dependent attenuation of cortical power in the 76-200 Hz range and of thalamic power in the 30-200 Hz range. In all instances the concentration-effect slope for the thalamus was markedly steeper than for the cortex. Furthermore the lowest concentration causing unconsciousness significantly reduced cortical power in the 126-200 Hz range and thalamic power in the 30-200 Hz range. CONCLUSIONS: Propofol causes a concentration-dependent attenuation of the power of thalamocortical rhythms in the 30-200 Hz range and this effect is far more pronounced for the thalamus, where the attenuation provides a robust correlate of the hypnotic action of propofol [corrected].


Asunto(s)
Anestésicos Intravenosos/sangre , Corteza Cerebral/efectos de los fármacos , Ritmo Gamma/efectos de los fármacos , Propofol/sangre , Tálamo/efectos de los fármacos , Anestésicos Intravenosos/farmacocinética , Anestésicos Intravenosos/farmacología , Animales , Corteza Cerebral/fisiología , Relación Dosis-Respuesta a Droga , Electrodos Implantados , Ritmo Gamma/fisiología , Masculino , Especificidad de Órganos , Propofol/farmacocinética , Propofol/farmacología , Ratas , Ratas Long-Evans , Tálamo/fisiología , Inconsciencia/sangre , Inconsciencia/inducido químicamente
5.
Neuropsychiatr Dis Treat ; 10: 1369-79, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25092982

RESUMEN

There have been significant advances in the treatment of psychiatric disease in the last half century, but it is still unclear which neural circuits are ultimately responsible for specific disease states. Fortunately, technical limitations that have constrained this research have recently been mitigated by advances in research tools that facilitate circuit-based analyses. The most prominent of these tools is optogenetics, which refers to the use of genetically encoded, light-sensitive proteins that can be used to manipulate discrete neural circuits with temporal precision. Optogenetics has recently been used to examine the neural underpinnings of both psychiatric disease and symptom relief, and this research has rapidly identified novel therapeutic targets for what could be a new generation of rational drug development. As these and related methodologies for controlling neurons ultimately make their way into the clinic, circuit-based strategies for alleviating psychiatric symptoms could become a remarkably refined approach to disease treatment.

6.
Can J Anaesth ; 61(3): 254-62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24449402

RESUMEN

PURPOSE: Brain imaging studies suggest that loss of consciousness induced by general anesthetics is associated with impairment of thalamic function. There is, however, limited information on the time course of these changes. We recently obtained intracranial electroencephalogram (EEG) recordings from the ventroposterolateral (VPL) nucleus of the thalamus and from the motor cortex during induction of anesthesia in three patients to study the time course of the alterations of cortical and thalamic function. CLINICAL FEATURES: The patients were American Society of Anesthesiologists physical status I-II males aged 33-57 yr with intractable central pain caused by brachial plexus injury (patient 1 and 2) or insular infarct (patient 3). Anesthesia was induced with propofol (2.5-3.1 mg·kg(-1) over 30-45 sec) followed, after loss of consciousness, by rocuronium for tracheal intubation. The data retained for analysis are from one minute before the start of propofol to 110 sec later during ventilation of the patients' lungs before tracheal intubation. Spectral analysis was used to measure absolute EEG power. Propofol caused significant increases of cortical and thalamic power in the delta to beta frequency bands (1-30 Hz). These increases of cortical and thalamic power occurred either concomitantly or within seconds of each other. Propofol also caused a decrease in cortical and thalamic high-gamma (62-200 Hz) power that also followed a similar time course. CONCLUSION: We conclude that induction of anesthesia with propofol in these patients was associated with concurrent alterations of cortical and sensory thalamic activity.


Asunto(s)
Anestésicos Intravenosos/farmacología , Corteza Motora/efectos de los fármacos , Propofol/farmacología , Tálamo/efectos de los fármacos , Adulto , Anestésicos Intravenosos/administración & dosificación , Electroencefalografía , Humanos , Masculino , Persona de Mediana Edad , Corteza Motora/metabolismo , Dolor Intratable/terapia , Propofol/administración & dosificación , Tálamo/metabolismo
7.
Nat Neurosci ; 16(11): 1637-43, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24056699

RESUMEN

Rapid-eye movement (REM) sleep correlates with neuronal activity in the brainstem, basal forebrain and lateral hypothalamus. Lateral hypothalamus melanin-concentrating hormone (MCH)-expressing neurons are active during sleep, but their effects on REM sleep remain unclear. Using optogenetic tools in newly generated Tg(Pmch-cre) mice, we found that acute activation of MCH neurons (ChETA, SSFO) at the onset of REM sleep extended the duration of REM, but not non-REM, sleep episodes. In contrast, their acute silencing (eNpHR3.0, archaerhodopsin) reduced the frequency and amplitude of hippocampal theta rhythm without affecting REM sleep duration. In vitro activation of MCH neuron terminals induced GABAA-mediated inhibitory postsynaptic currents in wake-promoting histaminergic neurons of the tuberomammillary nucleus (TMN), and in vivo activation of MCH neuron terminals in TMN or medial septum also prolonged REM sleep episodes. Collectively, these results suggest that activation of MCH neurons maintains REM sleep, possibly through inhibition of arousal circuits in the mammalian brain.


Asunto(s)
Hipotálamo/fisiología , Red Nerviosa/fisiología , Vías Nerviosas/fisiología , Optogenética , Sueño REM/fisiología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Animales , Animales Recién Nacidos , Bicuculina/farmacología , Channelrhodopsins , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Regulación de la Expresión Génica , Hormonas Hipotalámicas/genética , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Melaninas/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Hormonas Hipofisarias/genética , Ritmo Teta/efectos de los fármacos , Ritmo Teta/genética , Transducción Genética , Valina/análogos & derivados , Valina/farmacología
8.
Neurosci Lett ; 426(1): 6-11, 2007 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-17869420

RESUMEN

Synaptic plasticity in olfactory inputs to the lateral entorhinal cortex may result in lasting changes in the processing of olfactory stimuli. Changes in dopaminergic tone can have strong effects on basal evoked synaptic responses in the superficial layers of the entorhinal cortex, and the current study investigated whether dopamine may modulate the induction of long-term potentiation (LTP) and depression (LTD) in piriform cortex inputs to layer II of the lateral entorhinal cortex in awake rats. Groups of animals were pretreated with either saline or the selective dopamine reuptake inhibitor GBR12909 prior to low or high frequency stimulation to induce LTD or LTP. In saline-treated groups, synaptic responses were potentiated to 122.4+/-6.4% of baseline levels following LTP induction, and were reduced to 84.5+/-4.9% following induction of LTD. Changes in synaptic responses were maintained for up to 60min and returned to baseline levels within 24h. In contrast, induction of both LTP and LTD was blocked in rats pretreated with GBR12909. Dopaminergic suppression of synaptic plasticity in the entorhinal cortex may serve to restrain activity-dependent plasticity during reward-relevant behavioral states or during processing of novel stimuli.


Asunto(s)
Vías Aferentes/metabolismo , Dopamina/metabolismo , Corteza Entorrinal/metabolismo , Potenciación a Largo Plazo/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Vías Aferentes/efectos de los fármacos , Animales , Inhibidores de Captación de Dopamina/farmacología , Estimulación Eléctrica , Corteza Entorrinal/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Vías Olfatorias/efectos de los fármacos , Vías Olfatorias/fisiología , Piperazinas/farmacología , Ratas , Ratas Long-Evans , Recompensa , Transmisión Sináptica/fisiología , Área Tegmental Ventral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA