Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 10: 273, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30949055

RESUMO

Recently, our group along with another demonstrated that GPR139 can be activated by L-phenylalanine (L-Phe) and L-tryptophan (L-Trp) at physiologically relevant concentrations. GPR139 is discretely expressed in brain, with highest expression in medial habenula. Not only are the endogenous ligands catecholamine/serotonin precursors, but GPR139 expressing areas can directly/indirectly regulate the activity of catecholamine/serotonin neurons. Thus, GPR139 appears expressed in an interconnected circuit involved in mood, motivation, and anxiety. The aim of this study was to characterize a selective and brain penetrant GPR139 agonist (JNJ-63533054) in relevant in vivo models. JNJ-63533054 was tested for its effect on c-fos activation in the habenula and dorsal striatum. In vivo microdialysis experiments were performed in freely moving rats to measure basal levels of serotonin or dopamine (DA) in prefrontal cortex (mPFC) and nucleus accumbens (NAc). Finally, the compound was profiled in behavioral models of anxiety, despair, and anhedonia. The agonist (10-30 mg/kg, p.o.) did not alter c-fos expression in medial habenula or dorsal striatum nor neurotransmitter levels in mPFC or NAc. JNJ-63533054 (10 mg/kg p.o.) produced an anhedonic-like effect on urine sniffing, but had no significant effect in tail suspension, with no interaction with imipramine, no effect on naloxone place aversion, and no effect on learned helplessness. In the marble burying test, the agonist (10 mg/kg p.o.) produced a small anxiolytic-like effect, with no interaction with fluoxetine, and no effect in elevated plus maze (EPM). Despite GPR139 high expression in medial habenula, an area with connections to limbic and catecholaminergic/serotoninergic areas, the GPR139 agonist had no effect on c-fos in medial habenula. It did not alter catecholamine/serotonin levels and had a mostly silent signal in in vivo models commonly associated with these pathways. The physiological function of GPR139 remains elusive.

2.
J Pharmacol Exp Ther ; 351(3): 628-41, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25271258

RESUMO

In the central nervous system, the ATP-gated Purinergic receptor P2X ligand-gated ion channel 7 (P2X7) is expressed in glial cells and modulates neurophysiology via release of gliotransmitters, including the proinflammatory cytokine interleukin (IL)-1ß. In this study, we characterized JNJ-42253432 [2-methyl-N-([1-(4-phenylpiperazin-1-yl)cyclohexyl]methyl)-1,2,3,4-tetrahydroisoquinoline-5-carboxamide] as a centrally permeable (brain-to-plasma ratio of 1), high-affinity P2X7 antagonist with desirable pharmacokinetic and pharmacodynamic properties for in vivo testing in rodents. JNJ-42253432 is a high-affinity antagonist for the rat (pKi 9.1 ± 0.07) and human (pKi 7.9 ± 0.08) P2X7 channel. The compound blocked the ATP-induced current and Bz-ATP [2'(3')-O-(4-benzoylbenzoyl)adenosine-5'-triphosphate tri(triethylammonium)]-induced release of IL-1ß in a concentration-dependent manner. When dosed in rats, JNJ-42253432 occupied the brain P2X7 channel with an ED50 of 0.3 mg/kg, corresponding to a mean plasma concentration of 42 ng/ml. The compound blocked the release of IL-1ß induced by Bz-ATP in freely moving rat brain. At higher doses/exposure, JNJ-42253432 also increased serotonin levels in the rat brain, which is due to antagonism of the serotonin transporter (SERT) resulting in an ED50 of 10 mg/kg for SERT occupancy. JNJ-42253432 reduced electroencephalography spectral power in the α-1 band in a dose-dependent manner; the compound also attenuated amphetamine-induced hyperactivity. JNJ-42253432 significantly increased both overall social interaction and social preference, an effect that was independent of stress induced by foot-shock. Surprisingly, there was no effect of the compound on either neuropathic pain or inflammatory pain behaviors. In summary, in this study, we characterize JNJ-42253432 as a novel brain-penetrant P2X7 antagonist with high affinity and selectivity for the P2X7 channel.


Assuntos
Fármacos do Sistema Nervoso Central/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Isoquinolinas/metabolismo , Isoquinolinas/farmacologia , Piperazinas/metabolismo , Piperazinas/farmacologia , Antagonistas do Receptor Purinérgico P2X/metabolismo , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Animais , Animais Recém-Nascidos , Fármacos do Sistema Nervoso Central/uso terapêutico , Relação Dose-Resposta a Droga , Humanos , Isoquinolinas/uso terapêutico , Masculino , Camundongos , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Piperazinas/uso terapêutico , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Ratos , Ratos Sprague-Dawley
3.
J Nucl Med ; 55(4): 635-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24614224

RESUMO

UNLABELLED: Neuropeptide Y2 (NPY2) receptors are implicated in diverse brain disorders, but no suitable PET radiotracers are currently available for studying NPY2 receptors in the living brain. We developed a novel positron-emitting radioligand based on the NPY2 receptor antagonist JNJ-31020028 (N-(4-(4-[2-(diethylamino)-2-oxo-1-phenylethyl]piperazin-1-yl)-3-fluorophenyl)-2-pyridin-3-ylbenzamide) and used the radiotracer for PET brain imaging in pigs. METHODS: In vitro receptor autoradiography studies were performed to establish the anatomic distribution of NPY2 receptors in the pig brain. In vivo, baseline 90-min PET recordings of N-(11)C-methyl-JNJ-31020028 were conducted in anesthetized Yorkshire x Landrace pigs, concurrent with arterial blood sampling. Postchallenge scans were conducted after injection of unlabeled JNJ-31020028 as a pharmacologic intervention. Cyclosporine A was used to enhance levels of the PET radiotracer in the brain. The PET images were manually coregistered to a MR imaging atlas of the pig brain. Maps of the N-(11)C-methyl-JNJ-31020028 volume of distribution in the brain were prepared, and regional binding potentials of NPY2 receptors toward the radioligand were calculated using the simplified reference tissue method. RESULTS: In autoradiography studies, N-(11)C-methyl-JNJ-31020028 receptor binding sites were observed primarily in the hippocampus and were inhibited by unlabeled JNJ-31020028. In PET studies, N-(11)C-methyl-JNJ-31020028 was metabolized slowly in the bloodstream, with 25% of the (11)C-labeled parent compound remaining 30 min after injection. PET imaging showed baseline binding potentials of 0.64 ± 0.07 in the thalamus, 0.55 ± 0.02 in the caudate, and 0.49 ± 0.03 in the hippocampus. Graphical reference region analyses demonstrated that N-(11)C-methyl-JNJ-31020028 binding was reversible; infusion of unlabeled JNJ-31020028 markedly displaced the PET radioligand from binding sites in the hippocampus, thalamus, caudate nucleus, and cerebellum but not in the corpus callosum, which served as reference region for nonspecific binding. CONCLUSION: N-(11)C-methyl-JNJ-31020028 has several suitable properties for PET neuroimaging of NPY2 receptors. First, it is metabolized slowly in the bloodstream of pigs. Second, using cyclosporine, the target-to-background ratio of N-(11)C-methyl-JNJ-31020028 is sufficient for estimating pharmacokinetic parameters. Third, N-(11)C-methyl-JNJ-31020028 binds reversibly and competitively to cerebral sites known to contain relatively high numbers of NPY2 receptors, such as the hippocampus, thalamus, caudate nucleus, and cerebellum. Fourth, white matter such as corpus callosum, known to contain negligible numbers of NPY2 receptors, can serve as a reference region for estimating binding potentials in brain regions. To our knowledge, there is no other radioligand with these favorable properties and with this specificity for NPY2 receptors, which makes N-(11)C-methyl-JNJ-31020028 the first candidate radioligand for PET investigations of NPY2 receptors in the living brain.


Assuntos
Benzamidas , Encéfalo/diagnóstico por imagem , Piperazinas , Compostos Radiofarmacêuticos , Receptores de Neuropeptídeo Y/metabolismo , Animais , Autorradiografia , Benzamidas/farmacocinética , Sítios de Ligação , Ligação Competitiva/efeitos dos fármacos , Química Encefálica , Ciclosporina/farmacologia , Feminino , Marcação por Isótopo/métodos , Imageamento por Ressonância Magnética , Piperazinas/farmacocinética , Tomografia por Emissão de Pósitrons , Ligação Proteica , Compostos Radiofarmacêuticos/farmacocinética , Sus scrofa , Suínos
4.
Br J Pharmacol ; 170(3): 624-40, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23889535

RESUMO

BACKGROUND AND PURPOSE: An increasing body of evidence suggests that the purinergic receptor P2X, ligand-gated ion channel, 7 (P2X7) in the CNS may play a key role in neuropsychiatry, neurodegeneration and chronic pain. In this study, we characterized JNJ-47965567, a centrally permeable, high-affinity, selective P2X7 antagonist. EXPERIMENTAL APPROACH: We have used a combination of in vitro assays (calcium flux, radioligand binding, electrophysiology, IL-1ß release) in both recombinant and native systems. Target engagement of JNJ-47965567 was demonstrated by ex vivo receptor binding autoradiography and in vivo blockade of Bz-ATP induced IL-1ß release in the rat brain. Finally, the efficacy of JNJ-47965567 was tested in standard models of depression, mania and neuropathic pain. KEY RESULTS: JNJ-47965567 is potent high affinity (pKi 7.9 ± 0.07), selective human P2X7 antagonist, with no significant observed speciation. In native systems, the potency of the compound to attenuate IL-1ß release was 6.7 ± 0.07 (human blood), 7.5 ± 0.07 (human monocytes) and 7.1 ± 0.1 (rat microglia). JNJ-47965567 exhibited target engagement in rat brain, with a brain EC50 of 78 ± 19 ng·mL(-1) (P2X7 receptor autoradiography) and functional block of Bz-ATP induced IL-1ß release. JNJ-47965567 (30 mg·kg(-1) ) attenuated amphetamine-induced hyperactivity and exhibited modest, yet significant efficacy in the rat model of neuropathic pain. No efficacy was observed in forced swim test. CONCLUSION AND IMPLICATIONS: JNJ-47965567 is centrally permeable, high affinity P2X7 antagonist that can be used to probe the role of central P2X7 in rodent models of CNS pathophysiology.


Assuntos
Encéfalo/efeitos dos fármacos , Niacinamida/análogos & derivados , Piperazinas/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/efeitos dos fármacos , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Analgésicos/metabolismo , Analgésicos/farmacologia , Animais , Antidepressivos/farmacologia , Antimaníacos/farmacologia , Comportamento Animal/efeitos dos fármacos , Ligação Competitiva , Transtorno Bipolar/metabolismo , Transtorno Bipolar/prevenção & controle , Transtorno Bipolar/psicologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Permeabilidade Capilar , Linhagem Celular , Depressão/metabolismo , Depressão/prevenção & controle , Depressão/psicologia , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Humanos , Interleucina-1beta/metabolismo , Macaca , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/metabolismo , Neuralgia/prevenção & controle , Neuralgia/psicologia , Niacinamida/metabolismo , Niacinamida/farmacologia , Piperazinas/metabolismo , Ligação Proteica , Antagonistas do Receptor Purinérgico P2X/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7/metabolismo , Fatores de Tempo , Transfecção
5.
Psychopharmacology (Berl) ; 214(4): 829-41, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21086115

RESUMO

RATIONALE: A few recent studies suggest that brain histamine levels and signaling via H(3) receptors play an important role in modulation of alcohol stimulation and reward in rodents. OBJECTIVE: The present study characterized the effects of a novel, selective, and brain penetrant H(3) receptor antagonist (JNJ-39220675) on the reinforcing effects of alcohol in rats. METHODS: The effect of JNJ-39220675 on alcohol intake and alcohol relapse-like behavior was evaluated in selectively bred alcohol-preferring (P) rats using the standard two-bottle choice method. The compound was also tested on operant alcohol self administration in non-dependent rats and on alcohol-induced ataxia using the rotarod apparatus. In addition, alcohol-induced dopamine release in the nucleus accumbens was tested in freely moving rats. RESULTS: Subcutaneous administration of the selective H(3) receptor antagonist dose-dependently reduced both alcohol intake and preference in alcohol-preferring rats. JNJ-39220675 also reduced alcohol preference in the same strain of rats following a 3-day alcohol deprivation. The compound significantly and dose-dependently reduced alcohol self-administration without changing saccharin self-administration in alcohol non-dependent rats. Furthermore, the compound did not change the ataxic effects of alcohol, alcohol elimination rate, nor alcohol-induced dopamine release in nucleus accumbens. CONCLUSIONS: These results indicate that blockade of H(3) receptor should be considered as a new attractive mechanism for the treatment of alcoholism.


Assuntos
Alcoolismo/tratamento farmacológico , Azepinas/uso terapêutico , Antagonistas dos Receptores Histamínicos H3/uso terapêutico , Piridinas/uso terapêutico , Alcoolismo/metabolismo , Alcoolismo/psicologia , Animais , Autorradiografia , Azepinas/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Antagonistas dos Receptores Histamínicos H3/farmacocinética , Antagonistas dos Receptores Histamínicos H3/farmacologia , Injeções Subcutâneas , Masculino , Microdiálise , Estrutura Molecular , Atividade Motora/efeitos dos fármacos , Ligação Proteica , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Histamínicos H3/metabolismo , Reforço Psicológico , Autoadministração
6.
Psychopharmacology (Berl) ; 215(1): 191-203, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21181123

RESUMO

RATIONALE: Orexin-1 receptor antagonists have been shown to block the reinforcing effects of drugs of abuse and food. However, whether blockade of orexin-2 receptor has similar effects has not been determined. We have recently described the in vitro and in vivo effects of JNJ-10397049, a selective and brain penetrant orexin-2 receptor antagonist. OBJECTIVE: The goal of these studies was to evaluate whether systemic administration of JNJ-10397049 blocks the rewarding effects of ethanol and reverses ethanol withdrawal in rodents. As a comparison, SB-408124, a selective orexin-1 receptor antagonist, was also evaluated. METHODS: Rats were trained to orally self-administer ethanol (8% v/v) or saccharin (0.1% v/v) under a fixed-ratio 3 schedule of reinforcement. A separate group of rats received a liquid diet of ethanol (8% v/v) and withdrawal signs were evaluated 4 h after ethanol discontinuation. In addition, ethanol-induced increases in extracellular dopamine levels in the nucleus accumbens were tested. In separate experiments, the acquisition, expression, and reinstatement of conditioned place preference (CPP) were evaluated in mice. RESULTS: Our results indicate that JNJ-10397049 (1, 3, and 10 mg/kg, sc) dose-dependently reduced ethanol self-administration without changing saccharin self-administration, dopamine levels, or withdrawal signs in rats. Treatment with JNJ-10397049 (10 mg/kg, sc) attenuated the acquisition, expression, and reinstatement of ethanol CPP and ethanol-induced hyperactivity in mice. Surprisingly, SB-408124 (3, 10 and 30 mg/kg, sc) did not have any effect in these procedures. CONCLUSIONS: Collectively, these results indicate, for the first time, that blockade of orexin-2 receptors is effective in reducing the reinforcing effects of ethanol.


Assuntos
Comportamento Aditivo/prevenção & controle , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Dioxanos/uso terapêutico , Etanol/administração & dosagem , Compostos de Fenilureia/uso terapêutico , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Neuropeptídeos/antagonistas & inibidores , Alcoolismo/metabolismo , Alcoolismo/prevenção & controle , Alcoolismo/psicologia , Animais , Comportamento Aditivo/psicologia , Dioxanos/administração & dosagem , Dioxanos/farmacologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos DBA , Atividade Motora/efeitos dos fármacos , Receptores de Orexina , Compostos de Fenilureia/administração & dosagem , Compostos de Fenilureia/farmacologia , Ratos , Ratos Wistar , Esquema de Reforço , Reforço Psicológico , Autoadministração
7.
Psychopharmacology (Berl) ; 208(2): 265-77, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19953226

RESUMO

RATIONALE: The lack of potent, selective, brain penetrant Y(2) receptor antagonists has hampered in vivo functional studies of this receptor. OBJECTIVE: Here, we report the in vitro and in vivo characterization of JNJ-31020028 (N-(4-{4-[2-(diethylamino)-2-oxo-1-phenylethyl]piperazin-1-yl}-3-fluorophenyl)-2-pyridin-3-ylbenzamide), a novel Y(2) receptor antagonist. METHODS: The affinity of JNJ-31020028 was determined by inhibition of the PYY binding to human Y(2) receptors in KAN-Ts cells and rat Y(2) receptors in rat hippocampus. The functional activity was determined by inhibition of PYY-stimulated calcium responses in KAN-Ts cells expressing a chimeric G protein Gqi5 and in the rat vas deferens (a prototypical Y(2) bioassay). Ex vivo receptor occupancy was revealed by receptor autoradiography. JNJ-31020028 was tested in vivo with microdialysis, in anxiety models, and on corticosterone release. RESULTS: JNJ-31020028 bound with high affinity (pIC(50) = 8.07 +/- 0.05, human, and pIC(50) = 8.22 +/- 0.06, rat) and was >100-fold selective versus human Y(1), Y(4), and Y(5) receptors. JNJ-31020028 was demonstrated to be an antagonist (pK(B) = 8.04 +/- 0.13) in functional assays. JNJ-31020028 occupied Y(2) receptor binding sites (approximately 90% at 10 mg/kg) after subcutaneous administration in rats. JNJ-31020028 increased norepinephrine release in the hypothalamus, consistent with the colocalization of norepinephrine and neuropeptide Y. In a variety of anxiety models, JNJ-31020028 was found to be ineffective, although it did block stress-induced elevations in plasma corticosterone, without altering basal levels, and normalized food intake in stressed animals without affecting basal food intake. CONCLUSION: These results suggest that Y(2) receptors may not be critical for acute behaviors in rodents but may serve modulatory roles that can only be elucidated under specific situational conditions.


Assuntos
Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Benzamidas/farmacologia , Hipocampo/efeitos dos fármacos , Piperazinas/farmacologia , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Administração Oral , Animais , Anorexia/metabolismo , Anorexia/prevenção & controle , Anorexia/psicologia , Ansiolíticos/administração & dosagem , Ansiolíticos/farmacocinética , Ansiedade/metabolismo , Ansiedade/psicologia , Autorradiografia , Benzamidas/administração & dosagem , Benzamidas/farmacocinética , Ligação Competitiva , Células CHO , Cálcio/metabolismo , Corticosterona/sangue , Cricetinae , Cricetulus , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Injeções Intravenosas , Injeções Subcutâneas , Masculino , Camundongos , Microdiálise , Norepinefrina/metabolismo , Peptídeo YY/metabolismo , Permeabilidade , Piperazinas/administração & dosagem , Piperazinas/farmacocinética , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores de Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/metabolismo , Transfecção , Ducto Deferente/efeitos dos fármacos , Ducto Deferente/metabolismo
8.
Psychopharmacology (Berl) ; 207(3): 433-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19795107

RESUMO

RATIONALE: Histamine H3 receptors (H3R) are presynaptic heteroreceptors that negatively modulate the release of histamine and other neurotransmitters such as acetylcholine. Blocking H3 receptors with antagonists/inverse agonists has been shown to be procognitive and this effect has often been associated with increases in acetylcholine transmission. H3 receptors are abundantly expressed in the prefrontal cortex, an area associated with cognitive performance. While the procognitive effects of H3 receptor antagonists/inverse agonists may depend on alterations to acetylcholine or histamine release, other transmitters involved in cognitive processing such as glutamate and gamma-aminobutyric acid (GABA) may also be involved. OBJECTIVE: The purpose of the present study was to examine the effects of thioperamide, an H3 receptor antagonist, on extracellular levels of glutamate and GABA in the prefrontal cortex. MATERIALS AND METHODS: By means of in vivo microdialysis on freely moving Sprague Dawley rats, samples were collected and assayed via high-performance liquid chromatography coupled to electrochemical detection. RESULTS: Replacement of calcium with magnesium revealed that the release of GABA, but not glutamate, was calcium-dependent. Thioperamide (10-20 mg/kg) did not affect basal glutamate or GABA release. Perfusion with a high concentration of potassium (100 mM) increased GABA, but not glutamate, release and thioperamide (20 mg/kg) attenuated the effects of high potassium on GABA release. CONCLUSION: These data indicate that H3 receptors in the prefrontal cortex can enhance stimulated GABA release, but do not regulate basal levels of glutamate or GABA.


Assuntos
Espaço Extracelular/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Antagonistas dos Receptores Histamínicos H3/farmacologia , Piperidinas/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Espaço Extracelular/metabolismo , Masculino , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley
9.
CNS Drug Rev ; 13(1): 107-36, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17461893

RESUMO

The NOP receptor (formerly referred to as opiate receptor-like 1, ORL-1, LC132, OP(4), or NOP(1)) is a G protein-coupled receptor that shares high homology to the classic opioid MOP, DOP, and KOP (mu, delta, and kappa, respectively) receptors and was first cloned in 1994 by several groups. The NOP receptor remained an orphan receptor until 1995, when the endogenous neuropeptide agonist, known as nociceptin or orphanin FQ (N/OFQ) was isolated. Five years later, a group at Hoffmann-La Roche reported on the selective, nonpeptide NOP agonist Ro 64-6198, which became the most extensively published nonpeptide NOP agonist and a valuable pharmacological tool in determining the potential of the NOP receptor as a therapeutic target. Ro 64-6198 is systemically active and achieves high brain penetration. It has subnanomolar affinity for the NOP receptor and is at least 100 times more selective for the NOP receptor over the classic opioid receptors. Ro 64-6198 ranges from partial to full agonist, depending on the assay. Preclinical data indicate that Ro 64-6198 may have broad clinical uses, such as in treating stress and anxiety, addiction, neuropathic pain, cough, and anorexia. This review summarizes the pharmacology and preclinical data of Ro 64-6198.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Imidazóis/farmacologia , Receptores Opioides/agonistas , Compostos de Espiro/farmacologia , Animais , Ansiedade/tratamento farmacológico , Humanos , Imidazóis/efeitos adversos , Imidazóis/química , Imidazóis/uso terapêutico , Dor/tratamento farmacológico , Compostos de Espiro/efeitos adversos , Compostos de Espiro/química , Compostos de Espiro/uso terapêutico , Estresse Fisiológico/tratamento farmacológico , Receptor de Nociceptina
10.
Neuropsychopharmacology ; 31(1): 171-7, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15956992

RESUMO

Naloxone administration produces a robust conditioned place aversion (CPA) in opiate-naive rodents by blocking the action of enkephalins at mu opioid receptors. This aversive response is potentiated by prior exposure to morphine. In vitro studies indicate that morphine treatment may promote constitutive activity of mu opioid receptors. We hypothesized that such enhanced constitutive activity in vivo may underlie the increased aversive property of naloxone by uncovering the inverse agonist property of this drug. The CPA produced by naloxone was compared with that produced by the neutral antagonists 6-alpha- and 6-beta-naloxol in mice with and without prior morphine exposure. While all three drugs produced CPA, only naloxone CPA was enhanced by morphine given 20 h prior to each naloxone injection. Furthermore, only naloxone produced withdrawal jumping when given 20 h after morphine, even though 6-alpha-naloxol was able to produce jumping when given 4 h after morphine. These data suggest that morphine may enhance naloxone CPA by increasing levels of constitutively active mu receptors and further support the role of such constitutive activity in mediating naloxone-precipitated physical withdrawal. Such long-term changes in constitutive activity of the mu receptor induced by exogenous opiate exposure may thus be an important factor in hedonic homeostatic dysregulation proposed to underlie the addictive process.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Dependência de Morfina/psicologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Animais , Condicionamento Operante/efeitos dos fármacos , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Habituação Psicofisiológica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Naloxona/análogos & derivados , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Síndrome de Abstinência a Substâncias/psicologia
11.
Neuropharmacology ; 49(4): 439-46, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15919100

RESUMO

ORL-1 agonists have been proposed as potential therapeutics for substance abuse based on their propensity to counter the effects of mu opioid agonists in several systems, and to inhibit mesolimbic dopamine release, while mostly being devoid of aversive properties. In support of this, ORL-1 agonists have been shown to block the acquisition of morphine conditioned place preference (CPP). We investigated the effect of Ro 64-6198, a systemically active ORL-1 agonist, on the acquisition, expression, extinction, and reinstatement of morphine (20 mg/kg, s.c.) CPP in C57BL6/J mice. Similar to effects obtained with nociceptin/orphanin FQ, Ro 64-6198 (1 mg/kg, i.p.) blocked the acquisition of morphine CPP when given 15 min prior to each drug and vehicle conditioning session. This effect was not due to state dependent learning, since when tested again in the presence of Ro 64-6198 or vehicle no CPP was observed. Administration of Ro 64-6198 (0.3 or 1 mg/kg, i.p.) on the test day alone, in a separate group of animals, failed to block the expression of morphine CPP. Another group of mice was conditioned to morphine to develop CPP, and then exposed to the CPP chambers in the absence of drug once a day for 30 min to extinguish the CPP. Ro 64-6198 (1 mg/kg, i.p.) given 15 min prior to each session during extinction did not affect the rate of extinction. Finally, another group was conditioned to morphine, their CPP extinguished and subsequently reinstated by a priming injection of morphine (20 mg/kg, s.c.). Ro 64-6198 (1 mg/kg, i.p.), given 15 min prior to the priming injection, blocked reinstatement of morphine CPP. These results suggest that Ro 64-6198's effects may be limited to attenuation of the acute rewarding effects of morphine.


Assuntos
Analgésicos Opioides/farmacologia , Condicionamento Operante/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Imidazóis/farmacologia , Morfina/farmacologia , Reforço Psicológico , Compostos de Espiro/farmacologia , Análise de Variância , Animais , Comportamento Animal , Relação Dose-Resposta a Droga , Interações Medicamentosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Relacionados ao Uso de Substâncias/psicologia , Fatores de Tempo
12.
Neurochem Res ; 29(7): 1437-42, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15202777

RESUMO

Amphetamine is more effective than methamphetamine at raising dopamine levels in the prefrontal cortex. The current study tested the hypothesis that norepinephrine transporters are involved in this difference. Using microdialysis, dopamine, norepinephrine, and serotonin were measured in the rat prefrontal cortex after administration of methamphetamine or amphetamine, with and without perfusion of desipramine. Amphetamine raised norepinephrine levels more than methamphetamine did. Desipramine raised dopamine and serotonin levels but did not alter metabolite levels. Desipramine attenuated the increase in dopamine by amphetamine while increasing the dopamine released by methamphetamine. These data suggest that methamphetamine and amphetamine differ in altering prefrontal cortical dopamine levels and in interacting with norepinephrine transporters. It is proposed that amphetamine releases dopamine in the prefrontal cortex primarily through norepinephrine transporters, whereas methamphetamine interacts minimally with norepinephrine transporters.


Assuntos
Anfetamina/farmacologia , Desipramina/farmacologia , Dopamina/metabolismo , Metanfetamina/farmacologia , Neurônios/fisiologia , Norepinefrina/metabolismo , Córtex Pré-Frontal/fisiologia , Animais , Feminino , Cinética , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo
13.
Psychopharmacology (Berl) ; 170(2): 150-6, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12774190

RESUMO

RATIONALE: Previously, we have shown that d-amphetamine (AMPH) was more potent than d-methamphetamine (METH) at increasing extracellular levels of dopamine (DA) in the prefrontal cortex (PFC) at doses that had similar effects in the nucleus accumbens. Since working memory depends on PFC DA, it was postulated that AMPH would also be more potent than METH at affecting working memory. OBJECTIVE: To determine if AMPH is more potent than METH at affecting working memory. METHODS: Working memory was measured in adult female Sprague-Dawley rats using a delayed-alternation T-maze task with multiple delays (1, 10, 60 s) and food rewards. The percentage of food rewards consumed was also recorded. Animals were tested with METH and AMPH before and after a chronic protocol, with measurements of locomotor activity used to test for pharmacological tolerance or sensitization. The effects of METH and AMPH on extinction were also examined by omitting the food rewards from the T-maze. RESULTS. Both METH and AMPH produced dose-related bimodal effects on working memory at the intermediate delay (10 s); however, AMPH was more potent than METH. Both METH and AMPH initially also decreased the percentage of food rewards consumed in the T-maze. After chronic testing, animals displayed tolerance to both the working memory impairments and the reduction in food reward intake produced by AMPH. Animals did not display significant tolerance to the effects of METH on food reward consumption and performed worse in the T-maze after chronic testing. METH, but not AMPH, interfered with extinction. CONCLUSIONS: These results indicate that METH and AMPH differ in altering working memory and the expression of tolerance, perhaps due to differences in behavioral inhibition.


Assuntos
Anfetaminas/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Tolerância a Medicamentos , Extinção Psicológica/efeitos dos fármacos , Memória de Curto Prazo/efeitos dos fármacos , Metanfetamina/farmacologia , Análise de Variância , Animais , Relação Dose-Resposta a Droga , Feminino , Habituação Psicofisiológica , Aprendizagem em Labirinto , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Fatores de Tempo
14.
Psychopharmacology (Berl) ; 165(4): 359-69, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12491026

RESUMO

RATIONALE: Methamphetamine (METH) and amphetamine (AMPH) are both abused psychostimulants. Although METH is generally accepted to be more addictive and potent than its analogue AMPH, there are no known neurobiological differences in action between the two drugs that may account for such differences. OBJECTIVE: METH and AMPH were compared to determine potential mechanisms for such differences between the two drugs in order to provide new targets for the treatment of METH addiction. METHODS: Using in vivo microdialysis on rats, dopamine (DA), DA metabolites, and glutamate (GLU) release in the nucleus accumbens (NAC) and prefrontal cortex (PFC) were measured after administration of 2 mg/kg, IP, of METH or AMPH. Based on the neurochemical differences between METH and AMPH, a locomotor activity study was designed to assess differences in locomotor activation for a range of doses (1-4 mg/kg, IP) of METH and AMPH and after pretreatment with intra-accumbens GLU antagonists. RESULTS: METH and AMPH raised NAC DA levels to a similar degree. In the PFC, both METH and AMPH raised DA levels, but METH was less effective than AMPH. In the NAC, AMPH raised GLU levels but METH did not. In the PFC, METH raised GLU levels but AMPH did not. The locomotor activity dose response curve for METH had a lower peak than that of AMPH. This difference was blocked by pretreatment with either the GLU NMDA antagonist AP5 or the GLU AMPA antagonist DNQX locally in the NAC. CONCLUSIONS: This study reveals several previously unknown neurochemical and behavioral differences between METH and AMPH. Based on these results, it is suggested that new pharmacotherapeutic agents that produce augmentations of NAC GLU or PFC DA activity, or perhaps inhibition of PFC GLU activity, may someday be useful for the treatment of METH addiction.


Assuntos
Anfetamina/farmacologia , Comportamento Animal/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Metanfetamina/farmacologia , Valina/análogos & derivados , Ácido 3,4-Di-Hidroxifenilacético/análise , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Ácido Glutâmico/análise , Ácido Homovanílico/análise , Microdiálise , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Quinoxalinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Valina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...