Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neurovirol ; 16(4): 268-78, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20608774

RESUMO

Use of methamphetamine is increasingly a significant factor for the spread of human immunodeficiency virus type 1, for in certain populations, there is a convergence of methamphetamine abuse with human immunodeficiency virus type 1 infection. Methamphetamine and human immunodeficiency virus type 1 are both individually neuropathogenic, and the neuropathology caused by these two agents occurs in overlapping brain regions. However, the biological interaction of methamphetamine with lentiviruses remains unknown. Here, we investigate the effects of simultaneous exposure of these two agents on disease progression using the feline immunodeficiency virus model. The study models the bingeing methamphetamine user with sequential and repeated episodes of use, which were interrupted by periods of abstinence. Methamphetamine exposure significantly accelerated and enhanced the severity of the feline immunodeficiency virus model-induced central nervous system functional pathology, as measured in delays in brainstem auditory evoked potentials. Reciprocally, feline immunodeficiency virus enhanced the severity of the methamphetamine-induced effects on brain monoamine neurotransmitter and dopamine transporter levels. The results of this study indicate that a dual potentiation occurred. That is, methamphetamine enhanced feline immunodeficiency virus model-induced central nervous system disease and feline immunodeficiency virus model enhanced the toxic effects of methamphetamine, heralding a significant concern for those individuals that are exposed to both agents.


Assuntos
Encefalopatias/etiologia , Estimulantes do Sistema Nervoso Central/toxicidade , Síndrome de Imunodeficiência Adquirida Felina/complicações , Metanfetamina/toxicidade , Animais , Astrócitos/efeitos dos fármacos , Monoaminas Biogênicas/análise , Monoaminas Biogênicas/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encefalopatias/patologia , Encefalopatias/fisiopatologia , Gatos , Proteínas da Membrana Plasmática de Transporte de Dopamina/análise , Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Potenciais Evocados Auditivos/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida Felina/patologia , Síndrome de Imunodeficiência Adquirida Felina/fisiopatologia , Feminino , Proteína Glial Fibrilar Ácida/análise , Proteína Glial Fibrilar Ácida/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Vírus da Imunodeficiência Felina , Imuno-Histoquímica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Neurobiol Learn Mem ; 92(3): 429-38, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19531380

RESUMO

Changes in the state of CREB phosphorylation and in LTP in the hippocampus have been associated with learning and memory. Here we show that galanin, the neuropeptide released in the hippocampal formation from cholinergic and noradrenergic fibers, that has been shown to produce impairments in memory consolidation in the Morris water maze task inhibits both LTP and CREB phosphorylation in the rat hippocampus in vivo. While there are many transmitters regulating CREB phosphorylation none has been shown to suppress behaviorally-induced hippocampal CREB phosphorylation as potently as galanin. The in vivo inhibition of dentate gyrus-LTP and of CREB phosphorylation by the agonist occupancy of GalR1 and GalR2-type galanin receptors provides strong in vivo cellular and molecular correlates to galanin-induced learning deficits and designates galanin as a major regulator of the memory consolidation process.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Galanina/metabolismo , Hipocampo/fisiopatologia , Potenciação de Longa Duração/fisiologia , Transtornos da Memória/fisiopatologia , Neurônios/fisiologia , Precursores de Proteínas/metabolismo , Animais , Western Blotting , Giro Denteado/fisiopatologia , Eletrodos Implantados , Potenciais Evocados , Imuno-Histoquímica , Masculino , Memória/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
3.
Eur J Neurosci ; 26(3): 729-38, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17686045

RESUMO

Sleep need is characterized by the level of slow-wave activity (SWA) and increases with time spent awake. The molecular nature of this sleep homeostatic process is practically unknown. Here, we show that intracerebroventricular administration of the neuropeptide, cortistatin (CST-14), enhances EEG synchronization by selectively promoting deep slow-wave sleep (SWS) during both the light and dark period in rats. CST-14 also increases the level of slow-wave activity (SWA) within deep SWS during the first two hours following CST-14 administration. Steady-state levels of preprocortistatin mRNA oscillate during the light:dark cycle and are four-fold higher upon total 24-h sleep deprivation, returning progressively to normal levels after eight hours of sleep recovery. Preprocortistatin mRNA is expressed upon sleep deprivation in a particular subset of cortical interneurons that colocalize with c-fos. In contrast, the number of CST-positive cells coexpressing pERK1/2 decreases under sleep deprivation. The capacity of CST-14 to increase SWA, together with preprocortistatin's inverse correlation with time spent in SWS, suggests a potential role in sleep homeostatic processes.


Assuntos
Encéfalo/metabolismo , Ritmo Circadiano/fisiologia , Sincronização Cortical/efeitos dos fármacos , Neuropeptídeos/fisiologia , Sono/fisiologia , Animais , Relógios Biológicos/efeitos dos fármacos , Relógios Biológicos/fisiologia , Encéfalo/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Injeções Intraventriculares , Masculino , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neuropeptídeos/farmacologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/genética , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos
4.
Brain Res ; 1156: 46-58, 2007 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-17524371

RESUMO

The neural substrate of brain stimulation reward (BSR) has eluded identification since its discovery more than a half-century ago. Notwithstanding the difficulties in identifying the neuronal integrator of BSR, the mesocorticolimbic dopamine (DA) system originating in the ventral tegmental area (VTA) of the midbrain has been implicated. We have previously demonstrated that the firing rate of a subpopulation of gamma-aminobutyric acid (GABA) neurons in the VTA increases in anticipation of BSR. We show here that GABA neurons in the VTA, midbrain, hypothalamus, and thalamus of rats express connexin-36 (Cx36) gap junctions (GJs) and couple electrically upon DA application or by stimulation of the internal capsule (IC), which also supports self-stimulation. The threshold for responding for IC self-stimulation was the threshold for electrical coupling between GABA neurons, the degree of responding for IC self-stimulation was proportional to the magnitude of electrical coupling between GABA neurons, and GJ blockers increased the threshold for IC self-stimulation without affecting performance. Thus, a network of electrically coupled GABA neurons in the ventral brain may form the elusive neural integrator of BSR.


Assuntos
Encéfalo/fisiologia , Neurônios/fisiologia , Recompensa , Ácido gama-Aminobutírico/fisiologia , Animais , Conexinas/genética , Estimulação Elétrica , Regulação da Expressão Gênica , Hibridização in Situ Fluorescente , Ratos , Autoestimulação , Área Tegmentar Ventral/fisiologia , Proteína delta-2 de Junções Comunicantes
5.
Exp Neurol ; 202(1): 139-51, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16814775

RESUMO

Opiate activation of mu-opioid receptors (muORs) in the ventral tegmental area (VTA) modulates gamma-aminobutyric acid (GABA) neurotransmission within the mesocorticolimbic dopamine (DA) reward system. We combined in vivo extracellular electrophysiological recordings in anesthetized and freely behaving rats with intracellular Neurobiotin filling and immunocytochemistry to characterize the effects of opiates on VTA GABA neurons, evaluate their discharge activity during opiate self-administration, and identify the cellular sites for opiate activation. We identified a subpopulation of VTA GABA neurons that was characterized by location, spike discharge profile, activation by microelectrophoretic DA, and response to internal capsule (IC) stimulation. Systemic administration of heroin or microelectrophoretic application of the selective muOR agonist [d-Ala2, N-Me-Phe4, Gly-ol]-Enkephalin (DAMGO) reduced VTA GABA neuron firing rate (heroin IC(50) = 0.35 mg/kg) and was blocked by the muOR antagonist naloxone. Heroin also reduced IC-evoked post-stimulus spike discharges, a manifestation of gap-junction-mediated electrical coupling between VTA GABA neurons. The baseline firing rate of VTA GABA neurons significantly increased (239%) following the acquisition of heroin self-administration behavior and transiently increased during each response for heroin (105%), but decreased (49%) following heroin, similar to non-contingent heroin. Electrophysiologically characterized VTA GABA neurons were filled with Neurobiotin and labeled dendrites contained plasmalemmal muOR immunoreactivity. Dually labeled muOR dendrites contained dendrodendritic appositions characteristic of gap junctions. These findings indicate that inhibition of this population of GABAergic neurons by opiates acting on dendritic muORs has implications for modulation of electrical coupling between VTA GABA neurons and dopamine (DA) neurotransmission in the VTA and terminal field regions.


Assuntos
Heroína/administração & dosagem , Entorpecentes/administração & dosagem , Neurônios/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Área Tegmentar Ventral/citologia , Ácido gama-Aminobutírico/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Comportamento Animal , Biotina/análogos & derivados , Biotina/metabolismo , Dopamina/farmacologia , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Interações Medicamentosas , Estimulação Elétrica/métodos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Heroína/farmacologia , Imuno-Histoquímica/métodos , Masculino , Microscopia Imunoeletrônica/métodos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Entorpecentes/farmacologia , Neurônios/fisiologia , Neurônios/efeitos da radiação , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley , Autoadministração/métodos , Vigília
6.
Synapse ; 60(1): 20-31, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16575850

RESUMO

Communication between neurons in the mammalian brain is primarily through chemical synapses; however, evidence is accumulating in support of electrical synaptic transmission between some neuronal types in the mature nervous system. The authors have recently demonstrated that the gap junction (GJ) blocker quinidine suppresses stimulus-induced and dopamine-evoked coupling of gamma amino butyric acid (GABA) neurons in the ventral tegmental area (VTA) of mature rats (Stobbs et al., 2004). The aim of this study was to evaluate the role of connexin-36 (Cx36) GJs in mediating electrical coupling between VTA GABA neurons in P50-80 rats in vivo and P25-50 rats in vitro. Single stimulation of the internal capsule (IC) evoked VTA GABA neuron spike couplets in mature rats when activated antidromically, and multiple poststimulus spike discharges (PSDs) when activated with brief high-frequency stimulation of the IC (ICPSDs). The Cx36 GJ blocker mefloquine (30 mg/kg) suppressed VTA GABA neuron ICPSDs in mature freely behaving rats. VTA GABA neurons recorded via whole-cell patch clamp in the midbrain slice preparation of P25-50 rats showed robust expression of Cx36 transcripts when tested with single-cell quantitative reverse transcription polymerase chain reaction. In P50-80 rats, Cx36 protein immunoreactivity was evident in the VTA and surrounding structures. Dye-coupling between VTA neurons was observed following Neurobiotin labeling of VTA GABA neurons, as well as with the fluorochrome Alexa Fluor 488 using real-time video fluorescent microscopy. Thus, mature VTA GABA neurons appear to be connected by electrical synapses via Cx36 GJs, whose coupling is enhanced by corticotegmental input and by dopamine.


Assuntos
Comunicação Celular/fisiologia , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Neurônios/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Antimaláricos/farmacologia , Conexinas/efeitos dos fármacos , Estimulação Elétrica , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Junções Comunicantes/efeitos dos fármacos , Masculino , Mefloquina/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transmissão Sináptica/fisiologia , Área Tegmentar Ventral/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Proteína delta-2 de Junções Comunicantes
7.
J Neurosci ; 26(17): 4577-85, 2006 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-16641237

RESUMO

CNS abnormalities can be detected during chronic human immunodeficiency virus (HIV) infection, before the development of opportunistic infections or other sequelae of immunodeficiency. However, although end-stage dementia caused by HIV has been linked to the presence of infected and activated macrophages and microglia in the brain, the nature of the changes resulting in the motor and cognitive disorders in the chronic stage is unknown. Using simian immunodeficiency virus-infected rhesus monkeys, we sought the molecular basis for CNS dysfunction. In the chronic stable stage, nearly 2 years after infection, all animals had verified CNS functional abnormalities. Both virus and infiltrating lymphocytes (CD8+ T-cells) were found in the brain. Molecular analysis revealed that the expression of several immune response genes was increased, including CCL5, which has pleiotropic effects on neurons as well as immune cells. CCL5 was significantly upregulated throughout the course of infection, and in the chronic phase was present in the infiltrating lymphocytes. We have identified an altered state of the CNS at an important stage of the viral-host interaction, likely arising to protect against the virus but in the long term leading to damaging processes.


Assuntos
Quimiocinas/imunologia , Encefalite Viral/diagnóstico , Encefalite Viral/imunologia , Hospedeiro Imunocomprometido/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/diagnóstico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/isolamento & purificação , Animais , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/imunologia , Encefalite Viral/etiologia , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Síndrome de Imunodeficiência Adquirida dos Símios/virologia
8.
J Immunol ; 175(10): 6615-23, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16272316

RESUMO

Under noninflammatory conditions, both naive and central memory CD8 T cells can be eliminated in the periphery with either soluble peptide or cross-presented Ag. Here, we assess the tolerance susceptibility of tissue-resident memory CD8 T cells in mice to these two forms of tolerogen. Soluble peptide specifically eliminated the majority of memory CD8 cells present in both lymphoid and extralymphoid tissues including lung and liver, but was unable to reduce the number present in the CNS. In contrast, systemic cross-presentation of Ag by dendritic cells resulted in successful elimination of memory cells only from the spleen, with no significant reduction in the numbers of tissue-resident memory cells in the lung. The fact that tissue-resident memory cells were unable to access cross-presented Ag suggests that either the memory cells in the lung do not freely circulate out of the tissue, or that they circulate through a region in the spleen devoid of cross-presented Ag. Thus, although tissue-resident memory cells are highly susceptible to tolerance induction, both the form of tolerogen and location of the T cells can determine their accessibility to tolerogen and the degree to which they are successfully deleted from specific tissues.


Assuntos
Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Animais , Antígenos Virais , Reações Cruzadas , Feminino , Tolerância Imunológica , Vírus da Influenza A/imunologia , Tecido Linfoide/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Nucleocapsídeo , Nucleoproteínas/imunologia , Infecções por Orthomyxoviridae/imunologia , Proteínas de Ligação a RNA/imunologia , Solubilidade , Linfócitos T Citotóxicos/imunologia , Proteínas do Core Viral/imunologia
9.
Mol Cell Neurosci ; 30(3): 465-75, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16182561

RESUMO

Cortistatin-14 (CST) is a neuropeptide expressed in cortical and hippocampal interneurons that shares 11 of 14 residues with somatostatin. In contrast to somatostatin, infusion of CST decreases locomotor activity and selectively enhances slow wave sleep. Here, we show that transgenic mice that overexpress cortistatin under the control of neuron-specific enolase promoter do not express long-term potentiation in the dentate gyrus. This blockade of dentate LTP correlates with profound impairment of hippocampal-dependent spatial learning. Exogenously applied CST to slices of wild-type mice also blocked induction of LTP in the dentate gyrus. Our findings implicate cortistatin in the modulation of synaptic plasticity and cognitive function. Thus, increases in hippocampal cortistatin expression during aging could have an impact on age-related cognitive deficits.


Assuntos
Hipocampo/metabolismo , Deficiências da Aprendizagem/genética , Aprendizagem/fisiologia , Potenciação de Longa Duração/genética , Peptídeos/metabolismo , Transmissão Sináptica/genética , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Giro Denteado/metabolismo , Giro Denteado/fisiopatologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Peptídeos e Proteínas de Sinalização Intercelular , Deficiências da Aprendizagem/metabolismo , Deficiências da Aprendizagem/fisiopatologia , Masculino , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Transgênicos , Peptídeos/genética , Regiões Promotoras Genéticas/genética
10.
Biol Psychiatry ; 58(10): 831-7, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16018977

RESUMO

BACKGROUND: The 5-hydroxytryptamine7 receptor (5-HT7) is implicated in circadian rhythm phase resetting, and 5-HT7 receptor-selective antagonists alter rapid eye movement (REM) sleep parameters in a pattern opposite from those in patients with clinical depression. METHODS: As sleep, circadian rhythm, and mood regulation are related, we examined 5-HT7 receptor knockout mice in two behavioral models of depression. The forced swim and tail suspension tests are highly predictive for antidepressant drug activity. RESULTS: Unmedicated 5-HT7-/- mice showed decreased immobility in both tests, consistent with an antidepressantlike behavior. The selective 5-HT7 receptor antagonist SB-269970 also decreased immobility. The selective serotonin reuptake inhibitor citalopram, a widely used antidepressant, decreased immobility in both 5-HT7+/+ and 5-HT7-/- mice in the tail suspension test, suggesting that it utilizes an independent mechanism. The 5-HT7-/- mice spent less time in and had less frequent episodes of REM sleep, also consistent with an antidepressantlike state. CONCLUSIONS: The 5-HT7 receptor might have a role in mood disorders and antagonists might have therapeutic value as antidepressants.


Assuntos
Antidepressivos/farmacologia , Fenóis/farmacologia , Receptores de Serotonina/efeitos dos fármacos , Receptores de Serotonina/fisiologia , Sono/fisiologia , Sulfonamidas/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Transtorno Depressivo/fisiopatologia , Modelos Animais de Doenças , Resposta de Imobilidade Tônica/efeitos dos fármacos , Resposta de Imobilidade Tônica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sono REM/efeitos dos fármacos
11.
J Neurosci ; 25(23): 5465-74, 2005 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-15944374

RESUMO

Urotensin II (UII) is a cyclic neuropeptide with strong vasoconstrictive activity in the peripheral vasculature. UII receptor mRNA is also expressed in the CNS, in particular in cholinergic neurons located in the mesopontine tegmental area, including the pedunculopontine tegmental (PPT) and lateral dorsal tegmental nuclei. This distribution suggests that the UII system is involved in functions regulated by acetylcholine, such as the sleep-wake cycle. Here, we tested the hypothesis that UII influences cholinergic PPT neuron activity and alters rapid eye movement (REM) sleep patterns in rats. Local administration of UII into the PPT nucleus increases REM sleep without inducing changes in the cortical blood flow. Intracerebroventricular injection of UII enhances both REM sleep and wakefulness and reduces slow-wave sleep 2. Intracerebroventricular, but not local, administration of UII increases cortical blood flow. Moreover, whole-cell recordings from rat-brain slices show that UII selectively excites cholinergic PPT neurons via an inward current and membrane depolarization that were accompanied by membrane conductance decreases. This effect does not depend on action potential generation or fast synaptic transmission because it persisted in the presence of TTX and antagonists of ionotropic glutamate, GABA, and glycine receptors. Collectively, these results suggest that UII plays a role in the regulation of REM sleep independently of its cerebrovascular actions by directly activating cholinergic brainstem neurons.


Assuntos
Acetilcolinesterase/metabolismo , Neurônios/fisiologia , Sono REM/fisiologia , Tegmento Mesencefálico/fisiologia , Urotensinas/fisiologia , Animais , Circulação Cerebrovascular , Eletroencefalografia , Eletromiografia , Técnicas In Vitro , Injeções Intraventriculares , Masculino , Neurônios/metabolismo , Técnicas de Patch-Clamp , Núcleo Tegmental Pedunculopontino/citologia , Núcleo Tegmental Pedunculopontino/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Tegmento Mesencefálico/irrigação sanguínea , Tegmento Mesencefálico/citologia , Urotensinas/farmacologia , Vigília
12.
Neurobiol Dis ; 19(1-2): 255-65, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15837581

RESUMO

Prion protein (PrP(C)) is a constituent of most normal mammalian cells and plays an essential role in the pathogenesis of transmissible spongiform encephalopathies (TSE). However, the normal cellular function of PrP(C) remains unclear. Here, we document that mice with a selective deletion of PrP(C) exhibited deficits in hippocampal-dependent spatial learning, but non-spatial learning remained intact. mPrP-/- mice also showed reduction in paired-pulse facilitation and long-term potentiation in the dentate gyrus in vivo. These deficits were rescued in transgenic mPrP-/- mice expressing PrP(C) in neurons under control of the neuron-specific enolase (NSE) promoter indicating that they were due to lack of PrP(C) function in neurons. The deficits were seen in mPrP-/- mice with a homogeneous 129/Ola background and in mPrP-/- mice in the mixed (129/Ola x C57BL/10) background indicating that these abnormalities were unlikely due to variability of background genes or alteration of the nearby Prnd (doppel) gene.


Assuntos
Amiloide/deficiência , Transtornos Cognitivos/metabolismo , Neurônios/metabolismo , Precursores de Proteínas/deficiência , Amiloide/genética , Amiloide/fisiologia , Animais , Transtornos Cognitivos/genética , Transtornos Cognitivos/terapia , Feminino , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Priônicas , Príons , Precursores de Proteínas/genética , Precursores de Proteínas/fisiologia
13.
Neuropsychopharmacology ; 30(2): 350-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15483561

RESUMO

Acute high dose methamphetamine (METH) dosing regimens are frequently used in animal studies, however, these regimens can lead to considerable toxicity and even death in experimental animals. Acute high dosing regimens are quite distinct from the chronic usage patterns found in many human METH abusers. Furthermore, such doses, especially in nonhuman primates, can result in unexpected death, which is unacceptable, especially when such deaths fail to accurately model effects of human usage. As a model of chronic human METH abuse we have developed a nonlethal chronic METH administration procedure for the rhesus macaque that utilizes an escalating dose protocol. This protocol slowly increases the METH dosage from 0.1 to 0.7 mg/kg b.i.d. over a period of 4 weeks, followed by a period of chronic METH administration at 0.75 mg/kg b.i.d. (= total daily METH administration of 1.5 mg/kg). In parallel to human usage patterns, METH injections were given 20-23 times a month. This regimen produced a number of behavioral and physiological effects including decreased food intake and a significant increase in urinary cortisol excretion.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/psicologia , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Metanfetamina/farmacologia , Transtornos Relacionados ao Uso de Anfetaminas/urina , Animais , Temperatura Corporal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/administração & dosagem , Estimulantes do Sistema Nervoso Central/urina , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Hidrocortisona/urina , Macaca mulatta , Masculino , Metanfetamina/administração & dosagem , Metanfetamina/urina , Síndrome de Abstinência a Substâncias/psicologia
14.
Sleep ; 27(5): 857-65, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15453543

RESUMO

STUDY OBJECTIVES: Oleamide and anandamide are fatty acid amides implicated in the regulatory mechanisms of sleep processes. However, due to their prompt catabolism by fatty acid amide hydrolase (FAAH), their pharmacologic and behavioral effects, in vivo, disappear rapidly. To determine if, in the absence of FAAH, the hypnogenic fatty acid amides induce an increase of sleep, we characterized the sleep-wake patters in FAAH-knockout mice [FAAH (-/-)] before and after sleep deprivation. DESIGN: FAAH (-/-), FAAH (+/-), and FAAH (+/+) mice were implanted chronically for sleep, body temperature (Tb), and locomotor activity (LMA) recordings. Sleep-wake states were recorded during a 24-hour baseline session followed by 8 hours of sleep deprivation. Recovery recordings were done during the 16 hours following sleep deprivation. Total amount of wake, slow-wave sleep, and rapid eye movement sleep were calculated and compared between genotypes. The electroencephalographic spectral analysis was performed by fast Fourier transform analysis. Telemetry recordings of Tb and LMA were carried out continuously during 4 days under baseline conditions. SETTING: N/A. PATIENTS OR PARTICIPANTS: FAAH (-/-) mice and their heterozygote (+/-) and control (+/+) littermates were used. INTERVENTIONS: Sleep deprivation. MEASUREMENTS AND RESULTS: FAAH (-/-) mice possess higher values of slow-wave sleep and more intense episodes of slow-wave sleep than do control littermates under baseline conditions that are not related to differences in Tb and LMA. A rebound of slow-wave sleep and rapid eye movement sleep as well an increase in the levels of slow-wave activity were observed after sleep deprivation in all genotypes. CONCLUSION: These findings support the role of fatty acid amides as possible modulators of sleep and indicate that the homeostatic mechanisms of sleep in FAAH (-/-) mice are not disrupted.


Assuntos
Amidoidrolases/fisiologia , Privação do Sono/metabolismo , Sono REM/fisiologia , Vigília/fisiologia , Amidoidrolases/deficiência , Amidoidrolases/genética , Animais , Ácidos Araquidônicos/administração & dosagem , Ácidos Araquidônicos/análise , Ácidos Araquidônicos/farmacologia , Nível de Alerta/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Química Encefálica , Eletroencefalografia , Endocanabinoides , Genótipo , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/análise , Hipnóticos e Sedativos/farmacologia , Camundongos , Camundongos Knockout , Ácidos Oleicos/administração & dosagem , Ácidos Oleicos/análise , Ácidos Oleicos/farmacologia , Alcamidas Poli-Insaturadas , Sono REM/efeitos dos fármacos , Fatores de Tempo , Vigília/efeitos dos fármacos
15.
Neuron ; 43(4): 487-97, 2004 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-15312648

RESUMO

Arousal and anxiety are behavioral responses that involve complex neurocircuitries and multiple neurochemical components. Here, we report that a neuropeptide, neuropeptide S (NPS), potently modulates wakefulness and could also regulate anxiety. NPS acts by activating its cognate receptor (NPSR) and inducing mobilization of intracellular Ca2+. The NPSR mRNA is widely distributed in the brain, including the amygdala and the midline thalamic nuclei. Central administration of NPS increases locomotor activity in mice and decreases paradoxical (REM) sleep and slow wave sleep in rats. NPS was further shown to produce anxiolytic-like effects in mice exposed to four different stressful paradigms. Interestingly, NPS is expressed in a previously undefined cluster of cells located between the locus coeruleus (LC) and Barrington's nucleus. These results indicate that NPS could be a new modulator of arousal and anxiety. They also show that the LC region encompasses distinct nuclei expressing different arousal-promoting neurotransmitters.


Assuntos
Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Nível de Alerta/fisiologia , Neuropeptídeos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Sequência de Aminoácidos , Animais , Ansiolíticos/metabolismo , Ansiolíticos/uso terapêutico , Ansiedade/metabolismo , Nível de Alerta/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiologia , Células CHO , Sequência Conservada , Cricetinae , Evolução Molecular , Feminino , Humanos , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Atividade Motora/fisiologia , Neuropeptídeos/biossíntese , Neuropeptídeos/farmacologia , Neuropeptídeos/uso terapêutico , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/uso terapêutico , Receptores de Neuropeptídeos/metabolismo , Homologia de Sequência de Aminoácidos
16.
Neurosci Lett ; 364(1): 1-6, 2004 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15193744

RESUMO

Oleamide is a lipid with diverse properties, including cannabinoid-like activity. For example, it induces the classic triad of effects attributable to these molecules: decrease in core temperature, hypolocomotion, and reduction in pain perception. However, as it binds to the cannabinoid receptors (CB1) only at high concentrations, it is not considered an actual endocannabinoid. In this study, we tested the effect of oleamide on food intake and sexual behavior and compared it to the effect induced by anandamide. Results indicate that oleamide and anandamide increased food intake during the 3h post-injection. In addition, anandamide but not oleamide induced changes in sexual performance. This study further supports the role of endocannabinoids in food ingestion and male sexual behavior and gives additional support to the notion that, although oleamide might not be an endocannabinoid, it shares some effects with them.


Assuntos
Ácidos Araquidônicos/farmacologia , Moduladores de Receptores de Canabinoides/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Ácidos Oleicos/farmacologia , Comportamento Sexual/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Endocanabinoides , Hipnóticos e Sedativos/farmacologia , Masculino , Alcamidas Poli-Insaturadas , Ratos , Ratos Wistar
17.
J Neurovirol ; 10 Suppl 1: 58-66, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14982741

RESUMO

The duration and severity of the symptomatology present during the early phase of human immunodeficiency virus (HIV) infection (known as the acute retroviral syndrome) is associated with alterations in the clinical profile of infection, such as a shortening of duration between infection with HIV and the onset of neurocognitive impairment and acquired immunodeficiency syndrome (AIDS). Viral-specific CD8+ cytotoxic T lymphocytes (CTLs) and CD8+ natural killer (NK) cells play a key role in antiviral immunity. Loss of CD8+ cells or their functional impairment during the early period of infection is associated with a rapid progression to AIDS in nonhuman primate studies. However, no studies have determined whether CD8+ cell loss or impairment is associated with symptoms of acute retroviral illness such as fever. In this study, the authors compared the early phase of simian immunodeficiency virus (SIV) infection in animals that were treated with the anti-CD8 monoclonal antibody cM-T807 to deplete CD8+ cells during the early period of infection (SIV+ CD8- group) to those with intact CD8+ cells (SIV+ CD8+ group). The SIV+ CD8- group had an enhanced acute retroviral syndrome when compared to the SIV+ CD8+ group. The SIV+ CD8- group also had prolonged high viral loads and distinct alterations in the proinflammatory cytokines interleukin (IL)-6 and interferon (IFN)-alpha, as well as in monocyte chemoattractant protein (MCP)-1. CD8+ cell depletion, therefore, appears to enhance symptoms of the acute retroviral syndrome and alters several of the immunological factors associated with the early phase of infection.


Assuntos
Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/fisiopatologia , Doença Aguda , Animais , Quimiocina CCL2/sangue , Febre/imunologia , Febre/mortalidade , Febre/virologia , Interferon-alfa/sangue , Interleucina-6/sangue , Macaca mulatta , Masculino , Atividade Motora , Síndrome de Imunodeficiência Adquirida dos Símios/mortalidade , Análise de Sobrevida , Carga Viral
18.
Nat Neurosci ; 7(2): 160-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14730310

RESUMO

The neural mechanisms that mediate the transition from a drug-naive state to a state of drug dependence and addiction are not yet known. Here we show that a discrete population of GABA(A) receptors in the mammalian ventral tegmental area (VTA) serves as a potential addiction switching mechanism by gating reward transmission through one of two neural motivational systems: either a dopamine-independent (opiate-naive) or a dopaminergic (opiate-dependent or opiate-withdrawn) system. Bi-directional transmission of reward signals through this GABA(A) receptor substrate is dynamically controlled by the opiate state of the organism and involves a molecular alteration of the GABA(A) receptor. After opiate exposure and subsequent withdrawal, the functional conductance properties of the rat VTA GABA(A) receptor switch from an inhibitory to an excitatory signaling mode.


Assuntos
Dopamina/metabolismo , Transtornos Relacionados ao Uso de Opioides , Receptores de GABA-A/fisiologia , Recompensa , Área Tegmentar Ventral/fisiologia , Animais , Bicuculina/administração & dosagem , Anidrases Carbônicas/metabolismo , Condicionamento Clássico/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Agonistas GABAérgicos/administração & dosagem , Antagonistas GABAérgicos/administração & dosagem , Heroína/farmacologia , Injeções Intraventriculares , Masculino , Muscimol/administração & dosagem , Entorpecentes/farmacologia , Ratos , Ratos Wistar , Receptores de GABA-A/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/fisiopatologia , Área Tegmentar Ventral/efeitos dos fármacos
19.
Pharmacol Biochem Behav ; 77(2): 365-70, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14751466

RESUMO

We investigated the effects of methamphetamine (METH) on core body temperature (Tb) and motor activity (MA) with or without exposure to a peripheral immune challenge. Mice were exposed to an escalating METH treatment and then to a METH treatment known to cause neurotoxicity (binge METH treatment). This was followed by a challenge with lipopolysaccharide (LPS). Three days later, METH and saline-treated control groups were challenged with an acute test dose of METH (METH test). Animals exposed to the escalating METH treatment exhibited a significant increase in Tb only after the initial exposure to METH (Day 1) and following the METH test (Day 7). The hyperthermic effect produced by the METH test (Day 7) was reduced in mice previously exposed to combined exposure to binge METH and LPS treatments. The escalating METH treatment produced MA sensitization to the METH test. Animals treated with the binge METH, LPS injection or both treatments combined prevented MA sensitization to the METH test. These findings suggest that induction of peripheral endotoxemia in animals with a history of METH reduced the hyperthermic response to a subsequent challenge with METH.


Assuntos
Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Endotoxemia/fisiopatologia , Endotoxemia/psicologia , Metanfetamina/farmacologia , Animais , Temperatura Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos
20.
Drug Alcohol Depend ; 72(2): 141-9, 2003 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-14636969

RESUMO

Opiate abuse is a risk factor for human immunodeficiency virus (HIV) infection. Because the direct effects of opiates on HIV infection are difficult to determine epidemiologically, animal models of lentivirus infection are relied upon to study the effects of opiates in the absence of confounding factors. Morphine, the predominant metabolite of heroin, is used in most experimental systems examining heroin abuse. In this study, morphine treatment of feline immunodeficiency virus (FIV)-infected cats modeled a typical pattern of escalating drug use interspersed with withdrawals. Plasma cortisol levels were measured for evidence of stress associated with morphine withdrawal. In the morphine-treated cats, cortisol levels peaked at time points corresponding to morphine withdrawal and returned to baseline levels during treatment and several weeks after the final withdrawal. Morphine-treated cats displayed clear behavioral and physical signs of opiate exposure and evidence of withdrawal when the drug was stopped. Morphine-exposed cats did not experience enhanced severity of FIV-related disease; in fact, morphine demonstrated a protective effect on FIV-associated changes in brainstem auditory evoked potentials. Our research suggests that opiate exposure is unlikely to adversely affect the progression of acute lentivirus infection and might be beneficial in controlling associated neurological disease.


Assuntos
Modelos Animais de Doenças , Infecções por HIV/complicações , Vírus da Imunodeficiência Felina , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/virologia , Morfina/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/complicações , Síndrome de Abstinência a Substâncias/etiologia , Animais , Gatos , Doença Crônica , Esquema de Medicação , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Feminino , Humanos , Hidrocortisona/sangue , Morfina/administração & dosagem , Morfina/efeitos adversos , Índice de Gravidade de Doença , Síndrome de Abstinência a Substâncias/sangue , Fatores de Tempo , Viremia/sangue , Viremia/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA