Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.725
Filtrar
1.
Schizophr Res ; 267: 451-461, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38643726

RESUMEN

The methylazoxymethanol acetate (MAM) rodent model is used to study aspects of schizophrenia. However, numerous studies that have employed this model have used only males, resulting in a dearth of knowledge on sex differences in brain function and behaviour. The purpose of this study was to determine whether differences exist between male and female MAM rats in neuronal oscillatory function within and between the prefrontal cortex (PFC), ventral hippocampus (vHIP) and thalamus, behaviour, and in proteins linked to schizophrenia neuropathology. We showed that female MAM animals exhibited region-specific alterations in theta power, elevated low and high gamma power in all regions, and elevated PFC-thalamus high gamma coherence. Male MAM rats had elevated beta and low gamma power in PFC, and elevated vHIP-thalamus coherence. MAM females displayed impaired reversal learning whereas MAM males showed impairments in spatial memory. Glycogen synthase kinase-3 (GSK-3) was altered in the thalamus, with female MAM rats displaying elevated GSK-3α phosphorylation. Male MAM rats showed higher expression and phosphorylation GSK-3α, and higher expression of GSK-ß. Sex-specific changes in phosphorylated Tau levels were observed in a region-specific manner. These findings demonstrate there are notable sex differences in behaviour, oscillatory network function, and GSK-3 signaling in MAM rats, thus highlighting the importance of inclusion of both sexes when using this model to study schizophrenia.


Asunto(s)
Modelos Animales de Enfermedad , Acetato de Metilazoximetanol , Esquizofrenia , Caracteres Sexuales , Animales , Acetato de Metilazoximetanol/farmacología , Esquizofrenia/fisiopatología , Esquizofrenia/inducido químicamente , Esquizofrenia/metabolismo , Femenino , Masculino , Ratas , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Corteza Prefrontal/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatología , Tálamo/efectos de los fármacos , Tálamo/fisiopatología , Tálamo/metabolismo , Fosforilación/efectos de los fármacos , Proteínas tau/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Neuronas/patología , Ratas Sprague-Dawley
2.
Epilepsy Res ; 202: 107359, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582072

RESUMEN

PURPOSE: In developmental and epileptic encephalopathy with spike-and-wave activation in sleep (DEE-SWAS), the thalamocortical network is suggested to play an important role in the pathophysiology of the progression from focal epilepsy to DEE-SWAS. Ethosuximide (ESM) exerts effects by blocking T-type calcium channels in thalamic neurons. With the thalamocortical network in mind, we studied the prediction of ESM effectiveness in DEE-SWAS treatment using phase-amplitude coupling (PAC) analysis. METHODS: We retrospectively enrolled children with DEE-SWAS who had an electroencephalogram (EEG) recorded between January 2009 and September 2022 and were prescribed ESM at Okayama University Hospital. Only patients whose EEG showed continuous spike-and-wave during sleep were included. We extracted 5-min non-rapid eye movement sleep stage N2 segments from EEG recorded before starting ESM. We calculated the modulation index (MI) as the measure of PAC in pair combination comprising one of two fast oscillation types (gamma, 40-80 Hz; ripples, 80-150 Hz) and one of five slow-wave bands (delta, 0.5-1, 1-2, 2-3, and 3-4 Hz; theta, 4-8 Hz), and compared it between ESM responders and non-responders. RESULTS: We identified 20 children with a diagnosis of DEE-SWAS who took ESM. Fifteen were ESM responders. Regarding gamma oscillations, significant differences were seen only in MI with 0.5-1 Hz slow waves in the frontal pole and occipital regions. Regarding ripples, ESM responders had significantly higher MI in coupling with all slow waves in the frontal pole region, 0.5-1, 3-4, and 4-8 Hz slow waves in the frontal region, 3-4 Hz slow waves in the parietal region, 0.5-1, 2-3, 3-4, and 4-8 Hz slow waves in the occipital region, and 3-4 Hz slow waves in the anterior-temporal region. SIGNIFICANCE: High MI in a wider area of the brain may represent the epileptic network mediated by the thalamus in DEE-SWAS and may be a predictor of ESM effectiveness.


Asunto(s)
Anticonvulsivantes , Electroencefalografía , Etosuximida , Sueño , Humanos , Etosuximida/uso terapéutico , Etosuximida/farmacología , Masculino , Femenino , Electroencefalografía/métodos , Estudios Retrospectivos , Anticonvulsivantes/uso terapéutico , Anticonvulsivantes/farmacología , Preescolar , Niño , Sueño/efectos de los fármacos , Sueño/fisiología , Lactante , Ondas Encefálicas/efectos de los fármacos , Ondas Encefálicas/fisiología , Tálamo/efectos de los fármacos , Tálamo/fisiopatología , Espasmos Infantiles/tratamiento farmacológico , Espasmos Infantiles/fisiopatología
3.
Eur J Pharmacol ; 972: 176561, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38580182

RESUMEN

Neuronal depression in the thalamus underlies anesthetic-induced loss of consciousness, while the precise sub-thalamus nuclei and molecular targets involved remain to be elucidated. The present study investigated the role of extrasynaptic GABAA receptors in the central medial thalamic nucleus (CM) in anesthesia induced by gaboxadol (THIP) and diazepam (DZP) in rats. Local lesion of the CM led to a decrease in the duration of loss of righting reflex induced by THIP and DZP. CM microinjection of THIP but not DZP induced anesthesia. The absence of righting reflex in THIP-treated rats was consistent with the increase of low frequency oscillations in the delta band in the medial prefrontal cortex. CM microinjection of GABAA receptor antagonist SR95531 significantly attenuated the anesthesia induced by systemically-administered THIP, but not DZP. Moreover, the rats with declined expression of GABAA receptor δ-subunit in the CM were less responsive to THIP or DZP. These findings explained a novel mechanism of THIP-induced loss of consciousness and highlighted the role of CM extrasynaptic GABAA receptors in mediating anesthesia.


Asunto(s)
Anestesia , Isoxazoles , Receptores de GABA-A , Animales , Receptores de GABA-A/metabolismo , Masculino , Ratas , Isoxazoles/farmacología , Diazepam/farmacología , Ratas Sprague-Dawley , Núcleo Talámico Mediodorsal/efectos de los fármacos , Núcleo Talámico Mediodorsal/metabolismo , Núcleo Talámico Mediodorsal/fisiología , Reflejo de Enderezamiento/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Tálamo/efectos de los fármacos , Tálamo/metabolismo
4.
Mult Scler ; 30(6): 687-695, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38469809

RESUMEN

BACKGROUND: Loss of brain gray matter fractional volume predicts multiple sclerosis (MS) progression and is associated with worsening physical and cognitive symptoms. Within deep gray matter, thalamic damage is evident in early stages of MS and correlates with physical and cognitive impairment. Natalizumab is a highly effective treatment that reduces disease progression and the number of inflammatory lesions in patients with relapsing-remitting MS (RRMS). OBJECTIVE: To evaluate the effect of natalizumab on gray matter and thalamic atrophy. METHODS: A combination of deep learning-based image segmentation and data augmentation was applied to MRI data from the AFFIRM trial. RESULTS: This post hoc analysis identified a reduction of 64.3% (p = 0.0044) and 64.3% (p = 0.0030) in mean percentage gray matter volume loss from baseline at treatment years 1 and 2, respectively, in patients treated with natalizumab versus placebo. The reduction in thalamic fraction volume loss from baseline with natalizumab versus placebo was 57.0% at year 2 (p < 0.0001) and 41.2% at year 1 (p = 0.0147). Similar findings resulted from analyses of absolute gray matter and thalamic fraction volume loss. CONCLUSION: These analyses represent the first placebo-controlled evidence supporting a role for natalizumab treatment in mitigating gray matter and thalamic fraction atrophy among patients with RRMS. CLINICALTRIALS.GOV IDENTIFIER: NCT00027300URL: https://clinicaltrials.gov/ct2/show/NCT00027300.


Asunto(s)
Atrofia , Sustancia Gris , Factores Inmunológicos , Imagen por Resonancia Magnética , Esclerosis Múltiple Recurrente-Remitente , Natalizumab , Tálamo , Humanos , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/patología , Esclerosis Múltiple Recurrente-Remitente/diagnóstico por imagen , Natalizumab/farmacología , Natalizumab/uso terapéutico , Sustancia Gris/patología , Sustancia Gris/diagnóstico por imagen , Sustancia Gris/efectos de los fármacos , Adulto , Tálamo/patología , Tálamo/diagnóstico por imagen , Tálamo/efectos de los fármacos , Masculino , Femenino , Factores Inmunológicos/farmacología , Atrofia/patología , Persona de Mediana Edad , Aprendizaje Profundo
5.
Br J Anaesth ; 132(2): 334-342, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38044237

RESUMEN

BACKGROUND: Delayed emergence from general anaesthesia poses a significant perioperative safety hazard. Subanaesthetic doses of ketamine not only deepen anaesthesia but also accelerate recovery from isoflurane anaesthesia; however, the mechanisms underlying this phenomenon remain elusive. Esketamine exhibits a more potent receptor affinity and fewer adverse effects than ketamine and exhibits shorter recovery times after brief periods of anaesthesia. As the paraventricular thalamus (PVT) plays a pivotal role in regulating wakefulness, we studied its role in the emergence process during combined esketamine and isoflurane anaesthesia. METHODS: The righting reflex and cortical electroencephalography were used as measures of consciousness in mice during isoflurane anaesthesia with coadministration of esketamine. The expression of c-Fos was used to determine neuronal activity changes in PVT neurones after esketamine administration. The effect of esketamine combined with isoflurane anaesthesia on PVT glutamatergic (PVTGlu) neuronal activity was monitored by fibre photometry, and chemogenetic technology was used to manipulate PVTGlu neuronal activity. RESULTS: A low dose of esketamine (5 mg kg-1) accelerated emergence from isoflurane general anaesthesia (474 [30] s vs 544 [39] s, P=0.001). Esketamine (5 mg kg-1) increased PVT c-Fos expression (508 [198] vs 258 [87], P=0.009) and enhanced the population activity of PVTGlu neurones (0.03 [1.7]% vs 6.9 [3.4]%, P=0.002) during isoflurane anaesthesia (1.9 [5.7]% vs -5.1 [5.3]%, P=0.016) and emergence (6.1 [6.2]% vs -1.1 [5.0]%, P=0.022). Chemogenetic suppression of PVTGlu neurones abolished the arousal-promoting effects of esketamine (459 [33] s vs 596 [33] s, P<0.001). CONCLUSIONS: Our results suggest that esketamine promotes recovery from isoflurane anaesthesia by activating PVTGlu neurones. This mechanism could explain the rapid arousability exhibited upon treatment with a low dose of esketamine.


Asunto(s)
Anestésicos por Inhalación , Isoflurano , Ketamina , Tálamo , Animales , Ratones , Anestesia General , Anestésicos por Inhalación/farmacología , Isoflurano/farmacología , Ketamina/farmacología , Tálamo/efectos de los fármacos
6.
Front Neural Circuits ; 15: 659280, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34322001

RESUMEN

Corticofugal projections outnumber subcortical input projections by far. However, the specific role for signal processing of corticofugal feedback is still less well understood in comparisonto the feedforward projection. Here, we lesioned corticothalamic (CT) neurons in layers V and/or VI of the auditory cortex of Mongolian gerbils by laser-induced photolysis to investigate their contribution to cortical activation patterns. We have used laminar current-source density (CSD) recordings of tone-evoked responses and could show that, particularly, lesion of CT neurons in layer VI affected cortical frequency processing. Specifically, we found a decreased gain of best-frequency input in thalamocortical (TC)-recipient input layers that correlated with the relative lesion of layer VI neurons, but not layer V neurons. Using cortical silencing with the GABA a -agonist muscimol and layer-specific intracortical microstimulation (ICMS), we found that direct activation of infragranular layers recruited a local recurrent cortico-thalamo-cortical loop of synaptic input. This recurrent feedback was also only interrupted when lesioning layer VI neurons, but not cells in layer V. Our study thereby shows distinct roles of these two types of CT neurons suggesting a particular impact of CT feedback from layer VI to affect the local feedforward frequency processing in auditory cortex.


Asunto(s)
Apoptosis/fisiología , Corteza Auditiva/fisiología , Retroalimentación Fisiológica/fisiología , Rayos Láser/efectos adversos , Neuronas/fisiología , Tálamo/fisiología , Estimulación Acústica/métodos , Animales , Apoptosis/efectos de los fármacos , Corteza Auditiva/efectos de los fármacos , Corteza Auditiva/patología , Retroalimentación Fisiológica/efectos de los fármacos , Agonistas de Receptores de GABA-A/farmacología , Gerbillinae , Masculino , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/patología , Vías Nerviosas/fisiología , Neuronas/efectos de los fármacos , Neuronas/patología , Tálamo/efectos de los fármacos , Tálamo/patología
7.
Neuropharmacology ; 196: 108676, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34216585

RESUMEN

The mouse model of beta-amyloid (Aß) deposition, APP/PS1-21, exhibits high brain uptake of the tau-tracer (S)-[18F]THK5117, although no neurofibrillary tangles are present in this mouse model. For this reason we investigated (S)-[18F]THK5117 off-target binding to Aß plaques and MAO-B enzyme in APP/PS1-21 transgenic (TG) mouse model of Aß deposition. APP/PS1-21 TG and wild-type (WT) control mice in four different age groups (2-26 months) were imaged antemortem by positron emission tomography with (S)-[18F]THK5117, and then brain autoradiography. Additional animals were used for immunohistochemical staining and MAO-B enzyme blocking study with deprenyl pre-treatment. Regional standardized uptake value ratios for the cerebellum revealed a significant temporal increase in (S)-[18F]THK5117 uptake in aged TG, but not WT, brain. Immunohistochemical staining revealed a similar increase in Aß plaques but not endogenous hyper-phosphorylated tau or MAO-B enzyme, and ex vivo autography showed that uptake of (S)-[18F]THK5117 co-localized with the amyloid pathology. Deprenyl hydrochloride pre-treatment reduced the binding of (S)-[18F]THK5117 in the neocortex, hippocampus, and thalamus. This study's findings suggest that increased (S)-[18F]THK5117 binding in aging APP/PS1-21 TG mice is mainly due to increasing Aß deposition, and to a lesser extent binding to MAO-B enzyme, but not hyper-phosphorylated tau.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides/metabolismo , Encéfalo/diagnóstico por imagen , Monoaminooxidasa/metabolismo , Placa Amiloide/diagnóstico por imagen , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Compuestos de Anilina , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hipocampo/diagnóstico por imagen , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Inhibidores de la Monoaminooxidasa/farmacología , Neocórtex/diagnóstico por imagen , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Placa Amiloide/metabolismo , Tomografía de Emisión de Positrones , Presenilina-1/genética , Quinolinas , Radiofármacos , Selegilina/farmacología , Tálamo/diagnóstico por imagen , Tálamo/efectos de los fármacos , Tálamo/metabolismo
8.
J Psychiatry Neurosci ; 46(4): E459-E471, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34318655

RESUMEN

Background: Orexin-A (OrxA) administration in the posterior paraventricular nucleus of the thalamus (pPVT) reinstates extinguished cocaine-seeking behaviour following extended access to the drug (a model of dependence). The pPVT receives and integrates information associated with emotionally salient events and sends excitatory inputs to brain regions involved in the expression of emotional states, such as those driving cocaine-seeking behaviour (i.e., the nucleus accumbens, the central nucleus of the amygdala [CeA], the basolateral amygdala, the bed nucleus of the stria terminalis [BNST] and the prefrontal cortex). Methods: We monitored the activation pattern of these regions (measured by Fos) during cocaine-seeking induced by OrxA administered to the pPVT. The BNST and CeA emerged as being selectively activated. To test whether the functionality of these regions was pivotal during OrxA-induced cocaine-seeking behaviour, we transiently inactivated these regions concomitantly with OrxA administration to the pPVT. We then tested the participation of corticotropin-releasing factor receptors (CRF1) in the CeA during OrxA-induced cocaine-seeking using the CRF1 antagonist CP154526. Results: We observed selective activation of the CeA and BNST during cocaine-seeking induced by OrxA administered to the pPVT, but only transient inactivation of the CeA prevented cocaine-seeking behaviour. Administration of CP154526 to the CeA prevented OrxAinduced cocaine-seeking behaviour. Limitations: The use of only male rats could have been a limitation. Other limitations could have been the use of an indirect approach to test the hypothesis that administration of OrxA to the pPVT drives cocaine-seeking via CRF1 signalling in the CeA, and a lack of analysis of the participation of CeA subregions. Conclusion: Cocaine-seeking behaviour induced by OrxA administered to the pPVT is driven by activation of the CeA via CRF1 signalling.


Asunto(s)
Núcleo Amigdalino Central/efectos de los fármacos , Trastornos Relacionados con Cocaína/prevención & control , Cocaína , Orexinas/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Tálamo/efectos de los fármacos , Animales , Cocaína/farmacología , Masculino , Orexinas/administración & dosificación , Ratas
9.
Sci Rep ; 11(1): 15060, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34301974

RESUMEN

Evidence suggests that selective serotonin reuptake inhibitors (SSRIs) reorganize neural networks via a transient window of neuroplasticity. While previous findings support an effect of SSRIs on intrinsic functional connectivity, little is known regarding the influence of SSRI-administration on connectivity during sequence motor learning. To investigate this, we administered 20 mg escitalopram or placebo for 1-week to 60 healthy female participants undergoing concurrent functional magnetic resonance imaging and sequence motor training in a double-blind randomized controlled design. We assessed task-modulated functional connectivity with a psycho-physiological interaction (PPI) analysis in the thalamus, putamen, cerebellum, dorsal premotor, primary motor, supplementary motor, and dorsolateral prefrontal cortices. Comparing an implicit sequence learning condition to a control learning condition, we observed decreased connectivity between the thalamus and bilateral motor regions after 7 days of escitalopram intake. Additionally, we observed a negative correlation between plasma escitalopram levels and PPI connectivity changes, with higher escitalopram levels being associated with greater thalamo-cortico decreases. Our results suggest that escitalopram enhances network-level processing efficiency during sequence motor learning, despite no changes in behaviour. Future studies in more diverse samples, however, with quantitative imaging of neurochemical markers of excitation and inhibition, are necessary to further assess neural responses to escitalopram.


Asunto(s)
Citalopram/administración & dosificación , Aprendizaje/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Adulto , Cerebelo/diagnóstico por imagen , Cerebelo/efectos de los fármacos , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Neuronas Motoras/efectos de los fármacos , Red Nerviosa/diagnóstico por imagen , Red Nerviosa/efectos de los fármacos , Putamen/diagnóstico por imagen , Putamen/efectos de los fármacos , Tálamo/diagnóstico por imagen , Tálamo/efectos de los fármacos , Adulto Joven
10.
Sci Rep ; 11(1): 12613, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131204

RESUMEN

Secondary injury following cortical stroke includes delayed gliosis and eventual neuronal loss in the thalamus. However, the effects of aging and the potential to ameliorate this gliosis with NMDA receptor (NMDAR) antagonism are not established. We used the permanent distal middle cerebral artery stroke model (pdMCAO) to examine secondary thalamic injury in young and aged mice. At 3 days post-stroke (PSD3), slight microgliosis (IBA-1) and astrogliosis (GFAP) was evident in thalamus, but no infarct. Gliosis increased dramatically through PSD14, at which point degenerating neurons were detected. Flow cytometry demonstrated a significant increase in CD11b+/CD45int microglia (MG) in the ipsilateral thalamus at PSD14. CCR2-RFP reporter mouse further demonstrated that influx of peripheral monocytes contributed to the MG/Mϕ population. Aged mice demonstrated reduced microgliosis and astrogliosis compared with young mice. Interestingly, astrogliosis demonstrated glial scar-like characteristics at two years post-stroke, but not by 6 weeks. Lastly, treatment with memantine (NMDAR antagonist) at 4 and 24 h after stroke significantly reduced gliosis at PSD14. These findings expand our understanding of gliosis in the thalamus following cortical stroke and demonstrate age-dependency of this secondary injury. Additionally, these findings indicate that delayed treatment with memantine (an FDA approved drug) provides significant reduction in thalamic gliosis.


Asunto(s)
Gliosis/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Memantina/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Gliosis/etiología , Gliosis/patología , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Ratones , Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/complicaciones , Tálamo/efectos de los fármacos , Tálamo/patología
11.
Int J Mol Sci ; 22(8)2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923533

RESUMEN

It has been established that the selective α2A adrenoceptor agonist guanfacine reduces hyperactivity and improves cognitive impairment in patients with attention-deficit/hyperactivity disorder (ADHD). The major mechanisms of guanfacine are considered to involve the activation of the postsynaptic α2A adrenoceptor of glutamatergic pyramidal neurons in the frontal cortex, but the effects of chronic guanfacine administration on catecholaminergic and glutamatergic transmissions associated with the orbitofrontal cortex (OFC) are yet to be clarified. The actions of guanfacine on catecholaminergic transmission, the effects of acutely local and systemically chronic (for 7 days) administrations of guanfacine on catecholamine release in pathways from the locus coeruleus (LC) to OFC, the ventral tegmental area (VTA) and reticular thalamic-nucleus (RTN), from VTA to OFC, from RTN to the mediodorsal thalamic-nucleus (MDTN), and from MDTN to OFC were determined using multi-probe microdialysis with ultra-high performance liquid chromatography. Additionally, the effects of chronic guanfacine administration on the expression of the α2A adrenoceptor in the plasma membrane fraction of OFC, VTA and LC were examined using a capillary immunoblotting system. The acute local administration of therapeutically relevant concentrations of guanfacine into the LC decreased norepinephrine release in the OFC, VTA and RTN without affecting dopamine release in the OFC. Systemically, chronic administration of therapeutically relevant doses of guanfacine for 14 days increased the basal release of norepinephrine in the OFC, VTA, RTN, and dopamine release in the OFC via the downregulation of the α2A adrenoceptor in the LC, OFC and VTA. Furthermore, systemically, chronic guanfacine administration did not affect intrathalamic GABAergic transmission, but it phasically enhanced thalamocortical glutamatergic transmission. The present study demonstrated the dual actions of guanfacine on catecholaminergic transmission-acute attenuation of noradrenergic transmission and chronic enhancement of noradrenergic transmission and thalamocortical glutamatergic transmission. These dual actions of guanfacine probably contribute to the clinical effects of guanfacine against ADHD.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Guanfacina/farmacología , Corteza Prefrontal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Tálamo/efectos de los fármacos , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Animales , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Guanfacina/administración & dosificación , Guanfacina/uso terapéutico , Masculino , Norepinefrina/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Tálamo/metabolismo , Tálamo/fisiopatología , Ácido gamma-Aminobutírico/metabolismo
12.
Elife ; 102021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33904411

RESUMEN

The specific circuit mechanisms through which anesthetics induce unconsciousness have not been completely characterized. We recorded neural activity from the frontal, parietal, and temporal cortices and thalamus while maintaining unconsciousness in non-human primates (NHPs) with the anesthetic propofol. Unconsciousness was marked by slow frequency (~1 Hz) oscillations in local field potentials, entrainment of local spiking to Up states alternating with Down states of little or no spiking activity, and decreased coherence in frequencies above 4 Hz. Thalamic stimulation 'awakened' anesthetized NHPs and reversed the electrophysiologic features of unconsciousness. Unconsciousness is linked to cortical and thalamic slow frequency synchrony coupled with decreased spiking, and loss of higher-frequency dynamics. This may disrupt cortical communication/integration.


Asunto(s)
Anestésicos Intravenosos/farmacología , Corteza Cerebral/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Macaca mulatta/fisiología , Propofol/farmacología , Tálamo/efectos de los fármacos , Inconsciencia/inducido químicamente , Animales , Corteza Cerebral/fisiología , Femenino , Masculino , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Tálamo/fisiología
13.
J Psychiatry Neurosci ; 46(3): E319-E327, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33904667

RESUMEN

Background: Selective serotonin reuptake inhibitors (SSRIs) show acute effects on the neural processes associated with negative affective bias in healthy people and people with depression. However, whether and how SSRIs also affect reward and punishment processing on a similarly rapid time scale remains unclear. Methods: We investigated the effects of an acute and clinically relevant dose (20 mg) of the SSRI escitalopram on brain response during reward and punishment processing in 19 healthy participants. In a doubleblind, placebo-controlled study using functional MRI, participants performed a well-established monetary reward task at 3 time points: at baseline; after receiving placebo or escitalopram; and after receiving placebo or escitalopram following an 8-week washout period. Results: Acute escitalopram administration reduced blood-oxygen-level-dependent (BOLD) response during punishment feedback in the right thalamus (family-wise error corrected [FWE] p = 0.013 at peak level) and the right caudate head (pFWE = 0.011 at peak level) compared to placebo. We did not detect any significant BOLD changes during reward feedback. Limitations: We included only healthy participants, so interpretation of findings are limited to the healthy human brain and require future testing in patient populations. The paradigm we used was based on monetary stimuli, and results may not be generalizable to other forms of reward. Conclusion: Our findings extend theories of rapid SSRI action on the neural processing of rewarding and aversive stimuli and suggest a specific and acute effect of escitalopram in the punishment neurocircuitry.


Asunto(s)
Núcleo Caudado/efectos de los fármacos , Escitalopram/administración & dosificación , Escitalopram/farmacología , Neuronas/efectos de los fármacos , Castigo , Recompensa , Tálamo/efectos de los fármacos , Adulto , Núcleo Caudado/citología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Tálamo/citología , Adulto Joven
14.
Sci Rep ; 11(1): 6345, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737568

RESUMEN

The medial (DMS) and lateral (DLS) dorsal striatum differentially drive goal-directed and habitual/compulsive behaviors, respectively, and are implicated in a variety of neuropsychiatric disorders. These subregions receive distinct inputs from cortical and thalamic regions which uniquely determine dorsal striatal activity and function. Adenosine A1 receptors (A1Rs) are prolific within striatum and regulate excitatory glutamate transmission. Thus, A1Rs may have regionally-specific effects on neuroadaptive processes which may ultimately influence striatally-mediated behaviors. The occurrence of A1R-driven plasticity at specific excitatory inputs to dorsal striatum is currently unknown. To better understand how A1Rs may influence these behaviors, we first sought to understand how A1Rs modulate these distinct inputs. We evaluated A1R-mediated inhibition of cortico- and thalamostriatal transmission using in vitro whole-cell, patch clamp slice electrophysiology recordings in medium spiny neurons from both the DLS and DMS of C57BL/6J mice in conjunction with optogenetic approaches. In addition, conditional A1R KO mice lacking A1Rs at specific striatal inputs to DMS and DLS were generated to directly determine the role of these presynaptic A1Rs on the measured electrophysiological responses. Activation of presynaptic A1Rs produced significant and prolonged synaptic depression (A1R-SD) of excitatory transmission in the both the DLS and DMS of male and female animals. Our findings indicate that A1R-SD at corticostriatal and thalamostriatal inputs to DLS can be additive and that A1R-SD in DMS occurs primarily at thalamostriatal inputs. These findings advance the field's understanding of the functional roles of A1Rs in striatum and implicate their potential contribution to neuropsychiatric diseases.


Asunto(s)
Conducta Compulsiva/genética , Cuerpo Estriado/fisiología , Depresión Sináptica a Largo Plazo/genética , Receptor de Adenosina A1/genética , Adenosina/farmacología , Animales , Conducta Animal/fisiología , Potenciales Postsinápticos Excitadores , Femenino , Depresión Sináptica a Largo Plazo/fisiología , Masculino , Ratones , Ratones Noqueados , Neuronas/patología , Neuronas/fisiología , Técnicas de Placa-Clamp , Sinapsis/fisiología , Transmisión Sináptica , Tálamo/efectos de los fármacos , Tálamo/fisiología
15.
Clin Neurophysiol ; 132(5): 1049-1056, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33743300

RESUMEN

OBJECTIVE: Wedesignedalongitudinalcohortstudyon People with Epilepsy (PwE) with the aimofassessingthe effect of Perampanel (PER) oncortico-subcortical networks, as measured by high-frequency oscillations of somatosensory evoked potentials (SEP-HFOs). SEP-HFOs measure the excitability of both thalamo-corticalprojections(early HFOs) and intracortical GABAergic synapses (late HFOs), thus they could be used to study the anti-glutamatergic action of PER, a selective antagonist of the AMPA receptor. METHODS: 15 PwE eligible for PER add-on therapy, were enrolled prospectively. Subjects underwent SEPs recording from the dominant hand at two times: PwET0 (baseline, before PER titration) and PwET1 (therapeutic dose of 4 mg). HFOs were obtained by filtering N20 scalp response in the 400-800 Hz range. Patients were compared with a normative population of 15 healthy controls (HC) matched for age and sex. RESULTS: We found a significant reduction ofTotal HFOs and mostly early HFOs area between PwET0 and PwET1 (p = 0.05 and p = 0.045 respectively) and between HC and PwET1 (p = 0.01). Furthermore, we found a significant reduction of P24/N24 Amplitude between PwET0 and HC and between PwET0 and PwET1 (p = 0.006 and p = 0.032, respectively). CONCLUSIONS: Introduction of PER as add-on therapy reduced the area of total HFOs, acting mainly on the early burst, related to thalamo-cortical pathways. Furthermore P24/N24 amplitude, which seems to reflect a form of cortico-subcortical integration, resulted increased in PwE at T0 and normalized at T1. SIGNIFICANCE: Our findings suggest that PER acts on cortico-subcortical excitability. This could explain the broad spectrum of PER and its success in forms of epilepsy characterized by thalamo-cortical hyperexcitability.


Asunto(s)
Anticonvulsivantes/farmacología , Corteza Cerebral/efectos de los fármacos , Potenciales Evocados Somatosensoriales , Nitrilos/farmacología , Piridonas/farmacología , Receptores AMPA/antagonistas & inhibidores , Tálamo/efectos de los fármacos , Adulto , Corteza Cerebral/fisiología , Corteza Cerebral/fisiopatología , Epilepsia/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tálamo/fisiología , Tálamo/fisiopatología
16.
J Psychopharmacol ; 35(4): 469-482, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33645311

RESUMEN

BACKGROUND: The reticular thalamus gates thalamocortical information flow via finely tuned inhibition of thalamocortical cells in the mediodorsal thalamus. Brain imaging studies in humans show that the psychedelic lysergic acid diethylamide (LSD) modulates activity and connectivity within the cortico-striato-thalamo-cortical (CSTC) circuit, altering consciousness. However, the electrophysiological effects of LSD on the neurons in these brain areas remain elusive. METHODS: We employed in vivo extracellular single-unit recordings in anesthetized adult male mice to investigate the dose-response effects of cumulative LSD doses (5-160 µg/kg, intraperitoneal) upon reticular thalamus GABAergic neurons, thalamocortical relay neurons of the mediodorsal thalamus, and pyramidal neurons of the infralimbic prefrontal cortex. RESULTS: LSD decreased spontaneous firing and burst-firing activity in 50% of the recorded reticular thalamus neurons in a dose-response fashion starting at 10 µg/kg. Another population of neurons (50%) increased firing and burst-firing activity starting at 40 µg/kg. This modulation was accompanied by an increase in firing and burst-firing activity of thalamocortical neurons in the mediodorsal thalamus. On the contrary, LSD excited infralimbic prefrontal cortex pyramidal neurons only at the highest dose tested (160 µg/kg). The dopamine D2 receptor (D2) antagonist haloperidol administered after LSD increased burst-firing activity in the reticular thalamus neurons inhibited by LSD, decreased firing and burst-firing activity in the mediodorsal thalamus, and showed a trend towards further increasing the firing activity of neurons of the infralimbic prefrontal cortex. CONCLUSION: LSD modulates firing and burst-firing activity of reticular thalamus neurons and disinhibits mediodorsal thalamus relay neurons at least partially in a D2-mediated fashion. These effects of LSD on thalamocortical gating could explain its consciousness-altering effects in humans.


Asunto(s)
Relación Dosis-Respuesta a Droga , Fenómenos Electrofisiológicos , Dietilamida del Ácido Lisérgico/farmacología , Corteza Prefrontal , Tálamo , Animales , Trastornos de la Conciencia/inducido químicamente , Trastornos de la Conciencia/metabolismo , Antagonistas de los Receptores de Dopamina D2/farmacología , Alucinógenos/farmacología , Masculino , Ratones , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptores de Dopamina D2/metabolismo , Tálamo/efectos de los fármacos , Tálamo/metabolismo
17.
J Nippon Med Sch ; 88(5): 485-495, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-33692297

RESUMEN

BACKGROUND: Modafinil improves wakefulness and attention, is approved in Japan for treatment of narcolepsy, and was reported to be effective for attention-deficit/hyperactivity disorder. However, it was reported to induce emotional instability, including mania, depression, and suicidal ideation. Such side effects may be related to changes in cognitive behavior caused by the effects of modafinil on emotional recognition. However, the effects of modafinil on the neural basis of emotional processing have not been fully verified. We used functional magnetic resonance imaging to investigate the effects of modafinil on the neural basis of auditory emotional processing. METHODS: This study adopted a placebo-controlled within-subject crossover design. Data from 14 participants were analyzed. The effects of modafinil on cerebral activation and task performance during an emotional judgement task were analyzed. RESULTS: Task accuracy decreased significantly and response time of emotional judgement was significantly delayed by modafinil, as compared with placebo. Right thalamic activation in auditory emotional processing was significantly less in the modafinil condition than in the placebo condition. In addition, reduction of right thalamic activation by modafinil was positively correlated with accuracy of emotional judgement. CONCLUSIONS: Our findings suggest that modafinil acts on the right thalamus and changes behavior and brain function associated with auditory emotional processing. These results indicate that modafinil might change emotional recognition by reducing emotional activation related to social communication.


Asunto(s)
Afecto/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/uso terapéutico , Emociones/fisiología , Modafinilo/uso terapéutico , Tálamo/efectos de los fármacos , Estudios Cruzados , Potenciales Evocados Auditivos , Humanos , Imagen por Resonancia Magnética , Tálamo/diagnóstico por imagen
18.
Neuroimage ; 232: 117919, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33652141

RESUMEN

Unilateral damage to the frontoparietal network typically impairs saccade target selection within the contralesional visual hemifield. Severity of deficits and the degree of recovery have been associated with widespread network dysfunction, yet it is not clear how these behavioural and functional brain changes relate with the underlying structural white matter tracts. Here, we investigated whether recovery after unilateral prefrontal cortex (PFC) lesions was associated with changes in white matter microstructure across large-scale frontoparietal cortical and thalamocortical networks. Diffusion-weighted imaging was acquired in four male rhesus macaques at pre-lesion, week 1, and week 8-16 post-lesion when target selection deficits largely recovered. Probabilistic tractography was used to reconstruct cortical frontoparietal fiber tracts, including the superior longitudinal fasciculus (SLF) and transcallosal fibers connecting the PFC or posterior parietal cortex (PPC), as well as thalamocortical fiber tracts connecting the PFC and PPC to thalamic nuclei. We found that the two animals with small PFC lesions showed increased fractional anisotropy in both cortical and thalamocortical fiber tracts when behaviour had recovered. However, we found that fractional anisotropy decreased in cortical frontoparietal tracts after larger PFC lesions yet increased in some thalamocortical tracts at the time of behavioural recovery. These findings indicate that behavioural recovery after small PFC lesions may be supported by both cortical and subcortical areas, whereas larger PFC lesions may have induced widespread structural damage and hindered compensatory remodeling in the cortical frontoparietal network.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/métodos , Red Nerviosa/diagnóstico por imagen , Corteza Prefrontal/diagnóstico por imagen , Recuperación de la Función/fisiología , Tálamo/diagnóstico por imagen , Sustancia Blanca/diagnóstico por imagen , Animales , Macaca mulatta , Masculino , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Estimulación Luminosa/métodos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Recuperación de la Función/efectos de los fármacos , Tálamo/efectos de los fármacos , Tálamo/fisiología , Vasoconstrictores/toxicidad , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/fisiología
19.
Behav Brain Res ; 405: 113208, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33640395

RESUMEN

Individuals diagnosed with Fetal Alcohol Spectrum Disorders (FASD) often display behavioral impairments in executive functioning (EF). Specifically, the domains of working memory, inhibition, and set shifting are frequently impacted by prenatal alcohol exposure. Coordination between prefrontal cortex and hippocampus appear to be essential for these domains of executive functioning. The current study uses a rodent model of human third-trimester binge drinking to identify the extent of persistent executive functioning deficits following developmental alcohol by using a behavioral battery of hippocampus- and prefrontal cortex-dependent behavioral assays in adulthood. Alcohol added to milk formula was administered to Long Evans rat pups on postnatal days 4-9 (5.25 g/kg/day of ethanol; intragastric intubation), a period when rodent brain development undergoes comparable processes to human third-trimester neurodevelopment. Procedural control animals underwent sham intubation, without administration of any liquids (i.e., alcohol, milk solution). In adulthood, male rats were run on a battery of behavioral assays: novel object recognition, object-in-place associative memory, spontaneous alternation, and behavioral flexibility tasks. Alcohol-exposed rats demonstrated behavioral impairment in object-in-place preference and performed worse when the rule was switched on a plus maze task. All rats showed similar levels of novel object recognition, spontaneous alternation, discrimination learning, and reversal learning, suggesting alcohol-induced behavioral alterations are selective to executive functioning domains of spatial working memory and set-shifting in this widely-utilized rodent model. These specific behavioral alterations support the hypothesis that behavioral impairments in EF following prenatal alcohol exposure are caused by distributed damage to the prefrontal-thalamo-hippocampal circuit consisting of the medial prefrontal cortex, thalamic nucleus reuniens, and CA1 of hippocampus.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Disfunción Cognitiva , Etanol/farmacología , Función Ejecutiva , Trastornos del Espectro Alcohólico Fetal/fisiopatología , Hipotálamo , Red Nerviosa , Corteza Prefrontal , Tálamo , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Consumo Excesivo de Bebidas Alcohólicas/complicaciones , Depresores del Sistema Nervioso Central/administración & dosificación , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/fisiopatología , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Función Ejecutiva/efectos de los fármacos , Función Ejecutiva/fisiología , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiopatología , Masculino , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiopatología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Embarazo , Tercer Trimestre del Embarazo/efectos de los fármacos , Ratas , Ratas Long-Evans , Tálamo/efectos de los fármacos , Tálamo/fisiopatología
20.
Mol Pain ; 17: 1744806921992628, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33599155

RESUMEN

Electroconvulsive therapy (ECT) has been applied for chronic pain for decades. The amounts of opioids to treat pain are sometimes reduced after a series of ECT. The effect of ECT on morphine-induced analgesia and its mechanism underlying the reduction of morphine requirement has yet to be clarified. Therefore, we administered electroconvulsive shocks (ECS) to mice and investigated the antinociceptive effect of morphine in a hot plate test. We examined the expression level of µ-opioid receptor in the thalami of mice 25 h after administration of ECS compared to the thalami of mice without ECS administration using western blotting. ECS disturbed the development of a decrease in the percentage of maximal possible effect (%MPE), which was observed 24 h after a morphine injection, when ECS was applied 25, 23, 21, and 12 h before the second administration of morphine. We also examined the effect of ECS on the dose-response curve of %MPE to morphine-antinociception. Twenty-five hours after ECS, the dose-response curve was shifted to the left, and the EC50 of morphine given to ECS-pretreated mice decreased by 30.1% compared to the mice that were not pretreated with ECS. We also found that the expression level of µ-opioid receptors was significantly increased after ECS administration. These results confirm previous clinical reports showing that ECT decreased the required dose of opioids in neuropathic pain patients and suggest the hypothesis that this effect of ECT works through the thalamus.


Asunto(s)
Electrochoque , Morfina/farmacología , Nocicepción/fisiología , Animales , Masculino , Ratones Endogámicos C57BL , Nocicepción/efectos de los fármacos , Receptores Opioides mu/metabolismo , Tálamo/efectos de los fármacos , Tálamo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA