Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Headache ; 60(9): 1961-1981, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32750230

RESUMO

OBJECTIVE: A hallmark of migraine is photophobia. In mice, photophobia-like behavior is induced by calcitonin gene-related peptide (CGRP), a neuropeptide known to be a key player in migraine. In this study, we sought to identify sites within the brain from which CGRP could induce photophobia. DESIGN: We focused on the posterior thalamic region, which contains neurons responsive to both light and dural stimulation and has CGRP binding sites. We probed this area with both optogenetic stimulation and acute CGRP injections in wild-type mice. Since the light/dark assay has historically been used to investigate anxiety-like responses in animals, we measured anxiety in a light-independent open field assay and asked if stimulation of a brain region, the periaqueductal gray, that induces anxiety would yield similar results to posterior thalamic stimulation. The hippocampus was used as an anatomical control to ensure that light-aversive behaviors could not be induced by the stimulation of any brain region. RESULTS: Optogenetic activation of neuronal cell bodies in the posterior thalamic nuclei elicited light aversion in both bright and dim light without an anxiety-like response in an open field assay. Injection of CGRP into the posterior thalamic region triggered similar light-aversive behavior without anxiety. In contrast to the posterior thalamic nuclei, optogenetic stimulation of dorsal periaqueductal gray cell bodies caused both light aversion and an anxiety-like response, while CGRP injection had no effect. In the dorsal hippocampus, neither optical stimulation nor CGRP injection affected light aversion or open field behaviors. CONCLUSION: Stimulation of posterior thalamic nuclei is able to initiate light-aversive signals in mice that may be modulated by CGRP to cause photophobia in migraine.


Assuntos
Comportamento Animal , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Optogenética , Fotofobia/etiologia , Núcleos Posteriores do Tálamo , Animais , Comportamento Animal/efeitos dos fármacos , Peptídeo Relacionado com Gene de Calcitonina/administração & dosagem , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fotofobia/induzido quimicamente , Núcleos Posteriores do Tálamo/efeitos dos fármacos
2.
Front Neural Circuits ; 11: 69, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29021744

RESUMO

Rodents move rhythmically their facial whiskers and compute differences between signals predicted and those resulting from the movement to infer information about objects near their head. These computations are carried out by a large network of forebrain structures that includes the thalamus and the primary somatosensory (S1BF) and motor (M1wk) cortices. Spatially and temporally precise mechanorreceptive whisker information reaches the S1BF cortex via the ventroposterior medial thalamic nucleus (VPM). Other whisker-related information may reach both M1wk and S1BF via the axons from the posterior thalamic nucleus (Po). However, Po axons may convey, in addition to direct sensory signals, the dynamic output of computations between whisker signals and descending motor commands. It has been proposed that this input may be relevant for adjusting cortical responses to predicted vs. unpredicted whisker signals, but the effects of Po input on M1wk and S1BF function have not been directly tested or compared in vivo. Here, using electrophysiology, optogenetics and pharmacological tools, we compared in adult rats M1wk and S1BF in vivo responses in the whisker areas of the motor and primary somatosensory cortices to passive multi-whisker deflection, their dependence on Po activity, and their changes after a brief intense activation of Po axons. We report that the latencies of the first component of tactile-evoked local field potentials in M1wk and S1BF are similar. The evoked potentials decrease markedly in M1wk, but not in S1BF, by injection in Po of the GABAA agonist muscimol. A brief high-frequency electrical stimulation of Po decreases the responsivity of M1wk and S1BF cells to subsequent whisker stimulation. This effect is prevented by the local application of omega-agatoxin, suggesting that it may in part depend on GABA release by fast-spiking parvalbumin (PV)-expressing cortical interneurons. Local optogenetic activation of Po synapses in different cortical layers also diminishes M1wk and S1BF responses. This effect is most pronounced in the superficial layers of both areas, known to be the main source and target of their reciprocal cortico-cortical connections.


Assuntos
Córtex Motor/fisiologia , Núcleos Posteriores do Tálamo/fisiologia , Córtex Somatossensorial/fisiologia , Percepção do Tato/fisiologia , Animais , Feminino , Masculino , Microeletrodos , Córtex Motor/citologia , Córtex Motor/efeitos dos fármacos , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Optogenética , Estimulação Física , Núcleos Posteriores do Tálamo/citologia , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Ratos Sprague-Dawley , Receptores de GABA-A/metabolismo , Córtex Somatossensorial/citologia , Córtex Somatossensorial/efeitos dos fármacos , Percepção do Tato/efeitos dos fármacos , Vibrissas/fisiologia
3.
J Neurophysiol ; 118(1): 507-519, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28331010

RESUMO

The posterior nucleus of thalamus (PO) is a higher-order nucleus involved in sensorimotor processing, including nociception. An important characteristic of PO is its wide range of activity profiles that vary across states of arousal, thought to underlie differences in somatosensory perception subject to attention and degree of consciousness. Furthermore, PO loses the ability to downregulate its activity level in some forms of chronic pain, suggesting that regulatory mechanisms underlying the normal modulation of PO activity may be pathologically altered. However, the mechanisms responsible for regulating such a wide dynamic range of activity are unknown. Here, we test a series of hypotheses regarding the function of several presynaptic receptors on both GABAergic and glutamatergic afferents targeting PO in mouse, using acute slice electrophysiology. We found that presynaptic GABAB receptors are present on both GABAergic and glutamatergic terminals in PO, but only those on GABAergic terminals are tonically active. We also found that release from GABAergic terminals, but not glutamatergic terminals, is suppressed by cholinergic activation and that a subpopulation of GABAergic terminals is regulated by cannabinoids. Finally, we discovered the presence of tonic currents mediated by extrasynaptic GABAA receptors in PO that are heterogeneously distributed across the nucleus. Thus we demonstrate that multiple regulatory mechanisms concurrently exist in PO, and we propose that regulation of inhibition, rather than excitation, is the more consequential mechanism by which PO activity can be regulated.NEW & NOTEWORTHY The posterior nucleus of thalamus (PO) is a key sensorimotor structure, whose activity is tightly regulated by inhibition from several nuclei. Maladaptive plasticity in this inhibition leads to severe pathologies, including chronic pain. We reveal here, for the first time in PO, multiple regulatory mechanisms that modulate synaptic transmission within PO. These findings may lead to targeted therapies for chronic pain and other disorders.


Assuntos
Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Núcleos Posteriores do Tálamo/metabolismo , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Feminino , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurotransmissores/farmacologia , Técnicas de Patch-Clamp , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Receptores de GABA-B/metabolismo , Sinapses/efeitos dos fármacos , Técnicas de Cultura de Tecidos
4.
Neurotherapeutics ; 10(3): 520-38, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23775067

RESUMO

Spinal cord injury (SCI) causes not only sensorimotor and cognitive deficits, but frequently also severe chronic pain that is difficult to treat (SCI pain). We previously showed that hyperesthesia, as well as spontaneous pain induced by electrolytic lesions in the rat spinothalamic tract, is associated with increased spontaneous and sensory-evoked activity in the posterior thalamic nucleus (PO). We have also demonstrated that rodent impact SCI increases cell cycle activation (CCA) in the injury region and that post-traumatic treatment with cyclin dependent kinase inhibitors reduces lesion volume and motor dysfunction. Here we examined whether CCA contributes to neuronal hyperexcitability of PO and hyperpathia after rat contusion SCI, as well as to microglial and astroglial activation (gliopathy) that has been implicated in delayed SCI pain. Trauma caused enhanced pain sensitivity, which developed weeks after injury and was correlated with increased PO neuronal activity. Increased CCA was found at the thoracic spinal lesion site, the lumbar dorsal horn, and the PO. Increased microglial activation and cysteine-cysteine chemokine ligand 21 expression was also observed in the PO after SCI. In vitro, neurons co-cultured with activated microglia showed up-regulation of cyclin D1 and cysteine-cysteine chemokine ligand 21 expression. In vivo, post-injury treatment with a selective cyclin dependent kinase inhibitor (CR8) significantly reduced cell cycle protein induction, microglial activation, and neuronal activity in the PO nucleus, as well as limiting chronic SCI-induced hyperpathia. These results suggest a mechanistic role for CCA in the development of SCI pain, through effects mediated in part by the PO nucleus. Moreover, cell cycle modulation may provide an effective therapeutic strategy to improve reduce both hyperpathia and motor dysfunction after SCI.


Assuntos
Ciclo Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Hiperestesia/etiologia , Hiperestesia/patologia , Núcleos Posteriores do Tálamo/fisiopatologia , Traumatismos da Medula Espinal/complicações , Potenciais de Ação/efeitos dos fármacos , Animais , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ciclinas/farmacologia , Ciclinas/uso terapêutico , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Seguimentos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/tratamento farmacológico , Gliose/etiologia , Masculino , Microglia/química , Microglia/metabolismo , Microglia/patologia , Fibras Nervosas Amielínicas/patologia , Neurônios/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Núcleos Posteriores do Tálamo/patologia , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/patologia , Fatores de Tempo
5.
Epilepsy Behav ; 28(1): 104-7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23692689

RESUMO

The lateral-posterior thalamic nuclei (LP) have been shown to play an important role in controlling epileptic activity. In addition, thalamic atrophy and neuronal loss have been observed in epilepsy. The objective of this study was to investigate whether lateral-posterior neuronal activation may be observed shortly after a single generalized seizure in rats submitted to the pilocarpine model of epilepsy. The results showed an increased lateral-posterior activation as soon as the seizure occurred, suggesting that neuronal loss in the thalamus is not only the consequence of chronic epilepsy.


Assuntos
Epilepsia Generalizada/patologia , Núcleos Posteriores do Tálamo/patologia , Animais , Modelos Animais de Doenças , Epilepsia Generalizada/induzido quimicamente , Masculino , Agonistas Muscarínicos/toxicidade , Neurônios/metabolismo , Pilocarpina/toxicidade , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar
6.
J Pharmacol Exp Ther ; 340(1): 73-82, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21984836

RESUMO

3,4-Methylenedioxymethamphetamine (MDMA) is known to enhance tactile sensory perception, an effect that contributes to its popularity as a recreational drug. The neurophysiological basis for the effects of MDMA on somatosensation are unknown. However, MDMA interactions with the serotonin transporter (SERT) and subsequent enhancement of serotonin neurotransmission are well known. The rat trigeminal somatosensory system receives serotonergic afferents from the dorsal raphe nucleus. Because these fibers express SERT, they should be vulnerable to MDMA-induced effects. We found that administration of a challenge injection of MDMA (3 mg/kg i.p.) after repeated MDMA treatment (3 mg/kg per day for 4 days) elicits both serotonin and norepinephrine efflux in the ventral posterior medial (VPM) thalamus of Long-Evans hooded rats, the main relay along the lemniscal portion of the rodent trigeminal somatosensory pathway. We evaluated the potential for repeated MDMA administration to modulate whisker-evoked discharge of individual neurons in this region. After surgically implanting stainless steel eight-wire multichannel electrode bundles, we recorded spike train activity of single cells while activating the whisker pathway using a piezoelectric mechanical stimulator. We found that repeated MDMA administration increased the spontaneous firing rate but reduced both the magnitude and duration of whisker-evoked discharge in individual VPM thalamic neurons. The time course of drug action on neuronal firing patterns was generally consistent with fluctuations in neurotransmitter efflux as shown from our microdialysis studies. On the basis of these results, we propose that single use and repeated administration of MDMA may "distort," rather than enhance, tactile experiences in humans, in part, by disrupting normal spike firing patterns through somatosensory thalamic relay circuits.


Assuntos
N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Neurotransmissores/metabolismo , Estimulação Física , Núcleos Posteriores do Tálamo/metabolismo , Serotoninérgicos/farmacologia , Animais , Cromatografia Líquida de Alta Pressão , Fenômenos Eletrofisiológicos , Potenciais Somatossensoriais Evocados/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Masculino , Microdiálise , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , N-Metil-3,4-Metilenodioxianfetamina/farmacocinética , Norepinefrina/análise , Norepinefrina/metabolismo , Técnicas de Patch-Clamp , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Ratos , Ratos Long-Evans , Serotonina/análise , Serotonina/metabolismo , Serotoninérgicos/administração & dosagem , Serotoninérgicos/farmacocinética , Vibrissas/fisiologia
7.
Neurosci Res ; 64(3): 323-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19376165

RESUMO

Tactile information received by the whiskers of rodents is processed along several parallel pathways. A pathway that particularly includes the principal trigeminal nucleus (Pr5) and the thalamic ventral posterior medial nucleus (VPm) is called "lemniscal", and a pathway that includes the spinal trigeminal subnucleus interpolaris (Sp5i) and the thalamic posterior medial nucleus (POm) is called "paralemniscal". We trained rats to discriminate between two directions of stimulation applied to their single whiskers (forward or backward) to investigate how these pathways contributed to their perception of the direction of the single-whisker stimulation, and injected muscimol into either the lemniscal or paralemniscal nucleus while rats performed this task. The correct rate dropped significantly after muscimol injections into Pr5 or VPm, whereas we found no significant effects on discrimination after muscimol injections into Sp5i or POm. These results suggest that the lemniscal system is involved in enabling the direction of the single-whisker stimulus to be discriminated than the paralemniscal.


Assuntos
Vias Aferentes/fisiologia , Muscimol/administração & dosagem , Percepção do Tato/efeitos dos fármacos , Vibrissas/fisiologia , Vias Aferentes/efeitos dos fármacos , Animais , Discriminação Psicológica , Agonistas GABAérgicos/administração & dosagem , Masculino , Microinjeções , Estimulação Física , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Núcleos Posteriores do Tálamo/fisiologia , Ratos , Ratos Wistar , Núcleos do Trigêmeo/efeitos dos fármacos , Núcleos do Trigêmeo/fisiologia , Núcleos Ventrais do Tálamo/efeitos dos fármacos , Núcleos Ventrais do Tálamo/fisiologia
9.
Brain Res ; 1046(1-2): 38-44, 2005 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-15869749

RESUMO

Our previous studies have indicated that the thalamic nucleus submedius (Sm) is involved in modulation of nociception as part of an ascending component of an endogenous analgesic system consisting of spinal cord-Sm-ventrolateral orbital cortex (VLO)-periaqueductal gray (PAG)-spinal cord loop. Microinjection of 5-hydroxytryptamine (5-HT) into Sm produces antinociception and this effect is blocked by 5-HT(2) receptor antagonist. The aim of the present study was to examine whether the 5-HT(1) and 5-HT(3) receptors were also involved in the Sm 5-HT-evoked antinociception. Nociception was assessed in lightly anesthetized rats with radiant-heat-evoked tail flick (TF). 5-HT(1A) and 5-HT(3) receptor antagonists were microinjected into the Sm alone or in combination with a microinjection of 5-HT into the same Sm site. 5-HT(1A) receptor antagonist p-MPPI (0.87 nmol) facilitated the TF reflex; a lower dose (0.43 nmol) of p-MPPI significantly attenuated the Sm 5-HT-evoked inhibition of TF reflex. Microinjection of the 5-HT(3) receptor antagonist LY-278,584 (12 nmol) had no effect either on the TF reflex or on the Sm 5-HT-evoked inhibition. These results suggest that 5-HT(1A) receptor but not 5-HT(3) receptor is involved in mediating the 5-HT-evoked antinociception. Possible mechanisms of Sm 5-HT-induced descending antinociception are discussed.


Assuntos
Limiar da Dor/fisiologia , Núcleos Posteriores do Tálamo/fisiologia , Receptor 5-HT1A de Serotonina/fisiologia , Receptores 5-HT3 de Serotonina/fisiologia , Analgesia , Animais , Microinjeções , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia , Limiar da Dor/efeitos dos fármacos , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Receptores 5-HT3 de Serotonina/efeitos dos fármacos , Valores de Referência , Reflexo/efeitos dos fármacos , Reflexo/fisiologia , Antagonistas da Serotonina/administração & dosagem
10.
Neuropharmacology ; 48(4): 607-16, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15755488

RESUMO

The aim of the present study was to compare the effects of morphine on thalamic neuronal responses in naive rats and rats with carrageenan-induced hindpaw inflammation. Multiple single unit ventral posterolateral (VPL) and posterior complex (Po) activity was recorded and mechanically- (7 g, 14 g, 21 g, 60 g and 80 g) evoked responses of VPL and Po neurones were measured in naive rats and rats with carrageenan (100 microl, 2%)-induced hindpaw inflammation. Effects of systemic (0.5 mg kg(-1)) and intra-thalamic (66 microM, 250 nL) morphine on neuronal responses were determined. Mechanically-evoked (60 g) nociceptive responses of VPL neurones were significantly larger in inflamed rats (29 +/- 4 spikes s(-1)) compared to naive rats (19 +/- 2 spikes s(-1), P < 0.05). Systemic morphine inhibited 7 g-evoked responses of VPL neurones in inflamed (24 +/- 8% control, P < 0.01), but not in naive rats (123 +/- 3% control). Frank noxious-evoked responses of VPL neurones in inflamed rats were less sensitive to the effects of systemic and intra-thalamic morphine, compared to naive rats (P < 0.05 for both). These data provide evidence for altered evoked responses of neurones at the level of VPL, but not at Po, during hindpaw inflammation and suggest that thalamic sites of action contribute to the effects of systemic morphine.


Assuntos
Inflamação/fisiopatologia , Inibição Neural/fisiologia , Núcleos Posteriores do Tálamo/fisiologia , Receptores Opioides mu/fisiologia , Núcleos Ventrais do Tálamo/fisiologia , Animais , Membro Posterior/efeitos dos fármacos , Membro Posterior/fisiologia , Inflamação/tratamento farmacológico , Masculino , Morfina/farmacologia , Morfina/uso terapêutico , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Núcleos Ventrais do Tálamo/efeitos dos fármacos
11.
Behav Brain Res ; 153(2): 367-76, 2004 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-15265631

RESUMO

The early postnatal brain development, when many potentially sensitive processes occur, has been shown to be vulnerable to different pharmacological and environmental compounds. In the present investigation, four groups of neonatal NMRI male mice were administered the glutamate NMDA receptor antagonist ketamine (50 mg/kg, s.c.), or the GABA(A) receptor agonist diazepam (5 mg/kg, s.c.), or co-administered ketamine (50 mg/kg, s.c.) and diazepam (5 mg/kg, s.c.), or vehicle (0.9% saline, s.c.) on day 10 after birth. On day 11, mice from each treatment group were sacrificed and brains were taken for analysis of neuronal cell degeneration, using Fluoro-Jade staining technique. Ketamine, but not diazepam, induced a severe degeneration of cells in the parietal cortex. The opposite was observed for diazepam in the laterodorsal thalamus. The most pronounced cell degeneration was seen in parietal cortex of mice exposed to both ketamine and diazepam. At 2 months of age each treatment group was tested for motor activity and learning performance. Ketamine and ketamine + diazepam treated mice displayed severe deficits of habituation to the test chamber in the spontaneous motor activity test, marked deficits of acquisition learning and retention memory in the radial arm maze-learning task and less shift learning in the circular swim maze-learning task. This study indicates that the observed functional deficits can be related to cell degeneration induced during a critical stage of neonatal brain development. The potentiated apoptosis induced by ketamine and diazepam may have implications for the selection of drugs used in neonatal paediatric anaesthesia.


Assuntos
Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Ketamina/toxicidade , Aprendizagem em Labirinto/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Encéfalo/crescimento & desenvolvimento , Diazepam/farmacologia , Interações Medicamentosas , Feminino , Agonistas de Receptores de GABA-A , Injeções Subcutâneas , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Lobo Parietal/efeitos dos fármacos , Lobo Parietal/crescimento & desenvolvimento , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Núcleos Posteriores do Tálamo/crescimento & desenvolvimento , Gravidez
12.
Neurosci Lett ; 328(2): 141-4, 2002 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-12133575

RESUMO

Our previous studies have indicated that the thalamic nucleus submedius (Sm) is involved in modulation of nociception and plays an important role in an endogenous analgesic system (a feedback loop) consisting of spinal cord-Sm-ventrolateral orbital cortex-periaqueductal gray-spinal cord. To investigate whether opioids are involved in this antinociception pathway, the effects of microinjection of morphine and naloxone into the Sm on the nociceptive behavior (agitation) evoked in the formalin test were investigated in the awake rat using an automated movement detection system. The results indicate that a unilateral microinjection of morphine (5 micro g, 0.5 microl) into the Sm suppresses the formalin-induced agitation response, but does not influence spontaneous motor activity, and that the morphine-induced depression can be reversed by microinjection of the opioid receptor antagonist naloxone (1.0 micro g, 0.5 microl) into the same Sm site. The results suggest that opioid receptors in the Sm may be involved in the Sm-mediated depression of persistent inflammatory pain.


Assuntos
Vias Aferentes/metabolismo , Vias Eferentes/metabolismo , Morfina/farmacologia , Inibição Neural/efeitos dos fármacos , Nociceptores/metabolismo , Dor/metabolismo , Núcleos Posteriores do Tálamo/metabolismo , Vias Aferentes/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Vias Eferentes/efeitos dos fármacos , Feminino , Masculino , Microinjeções , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Inibição Neural/fisiologia , Nociceptores/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Dor/fisiopatologia , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Agitação Psicomotora/tratamento farmacológico , Agitação Psicomotora/metabolismo , Agitação Psicomotora/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides/agonistas , Receptores Opioides/metabolismo
13.
Eur J Neurosci ; 11(10): 3701-14, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10564377

RESUMO

In the present positron emission tomography (PET) study, we examine the effect of a scopolamine-induced challenge to encoding upon the pattern of regional cerebral blood flow during recognition of a list of abstract visual shapes 3 days after encoding of these shapes. This study was conducted to test hypotheses concerning the fusiform and thalamic contributions to object recognition arising from a previous imaging study of impaired recognition. In that study, we demonstrated that activity in the fusiform cortex and the thalamus during shape recognition was modulated by memory challenges. These memory challenges included, on one hand, impaired storage as a consequence of diazepam administration during encoding, and, on the other hand, impaired retrieval caused by a perceptual challenge. Activation in the fusiform cortex decreased during impaired recognition, irrespective of the type of challenge. In contrast, thalamic activation increased only when the recognition deficit resulted from impaired memory storage. Based on these results, we hypothesized that fusiform activation during recognition reflects the matching of an incoming stimulus with a stored one, whereas thalamic activation reflects retrieval attempts. These hypotheses would receive considerable support if scopolamine, which also impairs memory storage, induced similar modulations of fusiform and thalamic activation. In the present study, we observed that a scopolamine challenge to encoding does indeed modulate the activity in the very same regions that were previously modulated by a diazepam challenge. Hence, a similar memory deficit, although primarily effected through different neurochemical pathways, was paralleled by a similar modulation of activity in the same set of nodes in the shape recognition network. In the fusiform cortex, scopolamine decreased recognition-related activity, as did the sensory challenge of retrieval. Furthermore, covariate analysis demonstrated that the level of fusiform activity linearly correlates with behavioural performance. In the thalamus, activation increased following impaired encoding. This is in accordance with the idea that enhanced thalamic activity reflects increased effort expended in retrieval. In addition, in the intraparietal sulcus, differential activation also increased following impaired memory storage, possibly reflecting enhanced visuospatial attention in an effort to compensate for impaired performance.


Assuntos
Antagonistas Muscarínicos/administração & dosagem , Reconhecimento Visual de Modelos/efeitos dos fármacos , Escopolamina/administração & dosagem , Administração Oral , Adulto , Circulação Cerebrovascular , Condicionamento Psicológico/fisiologia , Diazepam/administração & dosagem , Moduladores GABAérgicos/administração & dosagem , Humanos , Imageamento por Ressonância Magnética , Masculino , Memória/fisiologia , Lobo Parietal/irrigação sanguínea , Lobo Parietal/diagnóstico por imagem , Lobo Parietal/efeitos dos fármacos , Núcleos Posteriores do Tálamo/irrigação sanguínea , Núcleos Posteriores do Tálamo/diagnóstico por imagem , Núcleos Posteriores do Tálamo/efeitos dos fármacos , Tomografia Computadorizada de Emissão , Córtex Visual/irrigação sanguínea , Córtex Visual/diagnóstico por imagem , Córtex Visual/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA