Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Brain Struct Funct ; 222(2): 1007-1026, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27376372

RESUMO

It is well established that central nervous system norepinephrine (NE) and corticotropin-releasing factor (CRF) systems are important mediators of behavioral responses to stressors. More recent studies have defined a role for delta opioid receptors (DOPR) in maintaining emotional valence including anxiety. The amygdala plays an important role in processing emotional stimuli, and has been implicated in the development of anxiety disorders. Activation of DOPR or inhibition of CRF in the amygdala reduces baseline and stress-induced anxiety-like responses. It is not known whether CRF- and DOPR-containing amygdalar neurons interact or whether they are regulated by NE afferents. Therefore, this study sought to better define interactions between the CRF, DOPR and NE systems in the basolateral (BLA) and central nucleus of the amygdala (CeA) of the male rat using anatomical and functional approaches. Irrespective of the amygdalar subregion, dual immunofluorescence microscopy showed that DOPR was present in CRF-containing neurons. Immunoelectron microscopy confirmed that DOPR was localized to both dendritic processes and axon terminals in the BLA and CeA. Semi-quantitative dual immunoelectron microscopy analysis of gold-silver labeling for DOPR and immunoperoxidase labeling for CRF revealed that 55 % of the CRF neurons analyzed contained DOPR in the BLA while 67 % of the CRF neurons analyzed contained DOPR in the CeA. Furthermore, approximately 41 % of DOPR-labeled axon terminals targeted BLA neurons that expressed CRF while 29 % of DOPR-labeled axon terminals targeted CeA neurons that expressed CRF. Triple label immunofluorescence microscopy revealed that DOPR and CRF were co-localized in common cellular profiles that were in close proximity to NE-containing fibers in both subregions. These anatomical results indicate significant interactions between DOPR and CRF in this critical limbic region and reveal that NE is poised to regulate these peptidergic systems in the amygdala. Functional studies were performed to determine if activation of DOPR could inhibit the anxiety produced by elevation of NE in the amygdala using the pharmacological stressor yohimbine. Administration of the DOPR agonist, SNC80, significantly attenuated elevated anxiogenic behaviors produced by yohimbine as measured in the rat on the elevated zero maze. Taken together, results from this study demonstrate the convergence of three important systems, NE, CRF, and DOPR, in the amygdala and provide insight into their functional role in modulating stress and anxiety responses.


Assuntos
Ansiedade/fisiopatologia , Complexo Nuclear Basolateral da Amígdala/metabolismo , Complexo Nuclear Basolateral da Amígdala/ultraestrutura , Núcleo Central da Amígdala/metabolismo , Núcleo Central da Amígdala/ultraestrutura , Hormônio Liberador da Corticotropina/metabolismo , Receptores Opioides delta/metabolismo , Neurônios Adrenérgicos/citologia , Neurônios Adrenérgicos/metabolismo , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/ultraestrutura , Animais , Benzamidas/administração & dosagem , Masculino , Neurônios/metabolismo , Neurônios/ultraestrutura , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Piperazinas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas
2.
Br J Pharmacol ; 172(22): 5414-24, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26375494

RESUMO

BACKGROUND AND PURPOSE: Ceftriaxone is a ß-lactam antibiotic and glutamate transporter activator that reduces the reinforcing effects of psychostimulants. Ceftriaxone also reduces locomotor activation following acute psychostimulant exposure, suggesting that alterations in dopamine transmission in the nucleus accumbens contribute to its mechanism of action. In the present studies we tested the hypothesis that pretreatment with ceftriaxone disrupts acute cocaine-evoked dopaminergic neurotransmission in the nucleus accumbens. EXPERIMENTAL APPROACH: Adult male Sprague-Dawley rats were pretreated with saline or ceftriaxone (200 mg kg(-1) , i.p. × 10 days) and then challenged with cocaine (15 mg kg(-1) , i.p.). Motor activity, dopamine efflux (via in vivo microdialysis) and protein levels of tyrosine hydroxylase (TH), the dopamine transporter and organic cation transporter as well as α-synuclein, Akt and GSK3ß were analysed in the nucleus accumbens. KEY RESULTS: Ceftriaxone-pretreated rats challenged with cocaine displayed reduced locomotor activity and accumbal dopamine efflux compared with saline-pretreated controls challenged with cocaine. The reduction in cocaine-evoked dopamine levels was not counteracted by excitatory amino acid transporter 2 blockade in the nucleus accumbens. Pretreatment with ceftriaxone increased Akt/GSK3ß signalling in the nucleus accumbens and reduced levels of dopamine transporter, TH and phosphorylated α-synuclein, indicating that ceftriaxone affects numerous proteins involved in dopaminergic transmission. CONCLUSIONS AND IMPLICATIONS: These results are the first evidence that ceftriaxone affects cocaine-evoked dopaminergic transmission, in addition to its well-described effects on glutamate, and suggest that its ability to attenuate cocaine-induced behaviours, such as psychomotor activity, is due in part to reduced dopaminergic neurotransmission in the nucleus accumbens.


Assuntos
Ceftriaxona/farmacologia , Dopamina/fisiologia , Núcleo Accumbens/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Animais , Cocaína , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Masculino , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley
3.
Brain Struct Funct ; 220(1): 541-58, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24271021

RESUMO

Amygdalar norepinephrine (NE) plays a key role in regulating neural responses to emotionally arousing stimuli and is involved in memory consolidation of emotionally charged events. Corticotropin-releasing factor (CRF) and dynorphin (DYN), two neuropeptides that mediate the physiological and behavioral responses to stress, are abundant in the central nucleus of the amygdala (CeA), and directly innervate brainstem noradrenergic locus coeruleus (LC) neurons. Whether the CRF- and DYN-containing amygdalar neurons receive direct noradrenergic innervation has not yet been elucidated. The present study sought to define cellular substrates underlying noradrenergic modulation of CRF- and DYN-containing neurons in the CeA using immunohistochemistry and electron microscopy. Ultrastructural analysis revealed that NE-labeled axon terminals form synapses with CRF- and DYN-containing neurons in the CeA. Semi-quantitative analysis showed that approximately 31 % of NET-labeled axon terminals targeted CeA neurons that co-expressed DYN and CRF. As a major source of CRF innervation to the LC, it is also not known whether CRF-containing CeA neurons are directly targeted by noradrenergic afferents. To test this, retrograde tract tracing using FluoroGold from the LC was combined with immunocytochemical detection of CRF and NET in the CeA. Our results revealed a population of LC-projecting CRF-containing CeA neurons that are directly innervated by NE afferents. Analysis showed that approximately 34 % of NET-labeled axon terminals targeted LC-projecting CeA neurons that contain CRF. Taken together, these results indicate significant interactions between NE, CRF and DYN in this critical limbic region and reveal direct synaptic interactions of NE with amygdalar CRF that influence the LC-NE arousal system.


Assuntos
Neurônios Adrenérgicos/fisiologia , Vias Aferentes/fisiologia , Tonsila do Cerebelo/citologia , Locus Cerúleo/citologia , Neurônios Adrenérgicos/metabolismo , Neurônios Adrenérgicos/ultraestrutura , Tonsila do Cerebelo/ultraestrutura , Animais , Hormônio Liberador da Corticotropina/metabolismo , Dopamina beta-Hidroxilase/metabolismo , Dinorfinas/metabolismo , Masculino , Microscopia Eletrônica , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/ultraestrutura , Precursores de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Coloração pela Prata , Estilbamidinas/metabolismo , Tirosina 3-Mono-Oxigenase
4.
Neuroscience ; 161(1): 13-22, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19303923

RESUMO

The role of chemokines in immune function is clearly established. Recent evidence suggests that these molecules also play an important role in the central nervous system as modulators of neuronal activity. The chemokine CXCL12 has been identified in several regions of the adult rat brain including the substantia nigra, ventral tegmental area and caudate putamen. CXCR4, a receptor activated by CXCL12, is expressed by dopaminergic neurons in the substantia nigra. The present study tested the effects of intracranial injections of CXCL12 on cocaine-induced locomotion and stereotypic activity in adult male Sprague-Dawley rats. Results demonstrate that intracerebral ventricular administration of CXCL12 (25 ng/4 microl) 15 min prior to cocaine (20 mg/kg intraperitoneal (i.p.)) produced a significant potentiation of both ambulatory and stereotypic activity as compared to cocaine alone. The effects of CXCL12 were blocked by administration of the selective CXCR4 antagonist, AMD 3100. Administration of CXCL12 into specific brain regions was performed to further understand the site of action of CXCL12. Bilateral administration of CXCL12 (25 ng/0.5 microl) into the ventral tegmental area 15 min prior to cocaine (20 mg/kg i.p.) significantly potentiated cocaine-induced ambulatory activity, whereas microinjections of CXCL12 into the caudate putamen selectively increased stereotypy. Conversely, administration of CXCL12 into the lateral shell of the nucleus accumbens resulted in an inhibition of cocaine-stimulated ambulatory activity. No alterations in ambulatory or stereotypic activity were observed following CXCL12 administration into the core of the nucleus accumbens. These results demonstrate that CXCL12 can modulate the behavioral effects produced by cocaine in a brain region-specific manner.


Assuntos
Quimiocina CXCL12/fisiologia , Cocaína/farmacologia , Atividade Motora/efeitos dos fármacos , Comportamento Estereotipado/efeitos dos fármacos , Animais , Benzilaminas , Quimiocina CXCL12/administração & dosagem , Quimiocina CXCL12/farmacologia , Ciclamos , Dopamina/fisiologia , Compostos Heterocíclicos/farmacologia , Injeções Intraventriculares , Masculino , Camundongos , Vias Neurais , Núcleo Accumbens/fisiologia , Putamen/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores CXCR4/antagonistas & inibidores , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Área Tegmentar Ventral/fisiologia
5.
Neuroscience ; 154(4): 1506-16, 2008 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-18550291

RESUMO

Opioid receptor agonists and antagonists have profound effects on cocaine-induced hyperactivity and conditioned reward. Recently, the role specifically of the mu opioid receptor has been demonstrated based on the finding that i.c.v. administration of the selective mu opioid receptor antagonist, D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP), can attenuate cocaine-induced behaviors. The purpose of the present study was to determine the location of mu opioid receptors that are critical for cocaine-induced reward and hyperactivity. Adult male Sprague-Dawley rats received injections of CTAP into the caudate putamen, the rostral or caudal ventral tegmental area (VTA) or the medial shell or core of the nucleus accumbens prior to cocaine to determine the role of mu opioid receptors in cocaine-induced reward and hyperactivity. Cocaine-induced reward was assessed using an unbiased conditioned place preference procedure. Results demonstrate that animals pre-treated with CTAP into the nucleus accumbens core or rostral VTA, but not the caudal VTA, caudate putamen or medial nucleus accumbens shell, during conditioning with cocaine showed an attenuation of the development of cocaine-induced place preference. In contrast, CTAP injected into the nucleus accumbens shell but not the core attenuated the expression of cocaine place preference. Intra-nucleus accumbens core, caudate putamen or caudal VTA CTAP significantly attenuated cocaine-induced hyperactivity. In addition, the number of cFos positive cells was increased in the motor cortex, medial and ventromedial aspects of the nucleus accumbens shell, basolateral amygdala and caudal VTA during the expression of cocaine place preference, and this increase was attenuated in the animals that received intra-accumbens core CTAP during daily cocaine conditioning. These results demonstrate the importance of mu opioid receptors in the nucleus accumbens and VTA in cocaine-induced reward and hyperactivity and suggest that some aspects of the behavioral effects of cocaine are mediated by endogenous activation of mu opioid receptors in these brain regions.


Assuntos
Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Núcleo Accumbens/metabolismo , Receptores Opioides mu/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Condicionamento Operante , Imuno-Histoquímica , Injeções Intraventriculares , Masculino , Núcleo Accumbens/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Proteínas Proto-Oncogênicas c-fos/metabolismo , Agitação Psicomotora/metabolismo , Ratos , Ratos Sprague-Dawley , Recompensa , Somatostatina/administração & dosagem , Área Tegmentar Ventral/efeitos dos fármacos
6.
Neuroscience ; 142(2): 481-91, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16893609

RESUMO

Numerous studies support a role for the endogenous opioid system in cocaine-influenced behavior. Few of these studies, however, selectively delineate a role for the mu opioid receptor (MOR) in this regard. This investigation examined if the MOR modulates cocaine-induced behavior in mice using a 17-base antisense oligodeoxynucleotide (AS ODN) directed against the MOR coding sequence 16-32. Specifically, cocaine-induced behavioral sensitization and conditioned reward were investigated. For the sensitization study, C57BL/6J mice received eight intermittent i.c.v. infusions of saline, mismatch oligodeoxynucleotide (ODN) (20 microg/4 microl) or AS ODN (20 microg/4 microl) over 20 days. Mice also received concomitant once daily i.p. injections of saline (4 ml/kg) or cocaine (15 mg/kg) for 10 days. There was a 7-day withdrawal period, after which all mice were challenged with cocaine (15 mg/kg) to test for behavioral sensitization. For the conditioned place preference (CPP) study, mice received five i.c.v. infusions of mismatch ODN or MOR AS ODN (days 1-5). An unbiased counterbalanced conditioning procedure was used where mice were conditioned with saline (4 ml/kg, i.p.) and cocaine (15 mg/kg, i.p.) on alternate days for four sessions (days 3-6). Mice were tested on day 7 for CPP. Immediately following testing, [3H]DAMGO (D-Ala2, N-Me-Phe4, Gly-ol5-enkephalin) receptor binding to brain homogenates was conducted. MOR AS attenuated cocaine-induced behavioral sensitization and conditioned reward. MOR AS ODN also reduced [3H]DAMGO binding. Collectively, these findings implicate the MOR as playing an important neuromodulatory role in the behavioral effects of cocaine in mice.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Oligodesoxirribonucleotídeos Antissenso/uso terapêutico , Receptores Opioides mu/fisiologia , Recompensa , Análise de Variância , Animais , Comportamento Animal , Transtornos Relacionados ao Uso de Cocaína/etiologia , Condicionamento Operante/efeitos dos fármacos , Vias de Administração de Medicamentos , Interações Medicamentosas , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Entorpecentes/farmacologia , Ligação Proteica/efeitos dos fármacos , Radiografia/métodos , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/genética , Fatores de Tempo , Trítio/farmacocinética
7.
Neuroscience ; 140(4): 1265-76, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16650615

RESUMO

We have previously demonstrated that GEC1 interacts with the kappa opioid receptor and GEC1 expression enhances cell surface expression of the receptor in Chinese hamster ovary cells. In this study, we generated an antiserum (PA629) directed against GEC1 in rabbits, characterized its specificity, and investigated distribution of GEC1 in tissues and in brain regions and spinal cord and its subcellular localization in hypothalamic neurons in the rat. Immunofluorescence staining demonstrated that PA629 recognized HA-GEC1 transfected into Chinese hamster ovary cells, but not HA-GABARAP or HA-GATE-16, although the three share high homology. Pre-incubation of PA629 with GST-GEC1, but not GST, abolished the staining. In immunoblotting, affinity-purified PA629 (PA629p) recognized GEC1, GABARAP and GATE-16. GEC1 migrated slower than GABARAP and GATE-16, with a M(r) of 16 kDa for GEC1 and M(r) of 14 kDa for GABARAP and GATE-16. Immunoblotting results showed that GEC1 level was higher in liver and brain than in lung and heart, and very low in kidney and skeletal muscle. GEC1 was present in all rat brain regions examined and spinal cord. Immunohistochemistry demonstrated that GEC1 immunoreactivity was distributed ubiquitously in the rat CNS with highly intense immunoreactivity in various brain nuclei and motor neurons of the spinal cord. Ultrastructural examination of neurons in the paraventricular nucleus of the hypothalamus showed that GEC1 was associated with endoplasmic reticulum and Golgi apparatus and distributed along plasma membranes and in cytosol. Coupled with our previous observation that GEC1 interacts with N-ethylmaleimide-sensitive factor, these findings strongly suggest that GEC1 functions in intracellular trafficking in the biosynthesis pathway and perhaps also the endocytic pathway. The widespread distribution of GEC1 suggests that GEC1 may be associated with many proteins, in addition to the kappa opioid receptor.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Química Encefálica/fisiologia , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Proteínas Associadas aos Microtúbulos/metabolismo , Medula Espinal/metabolismo , Medula Espinal/ultraestrutura , Animais , Sistema Nervoso Central/química , Sistema Nervoso Central/ultraestrutura , Masculino , Proteínas Associadas aos Microtúbulos/ultraestrutura , Ratos , Ratos Sprague-Dawley
8.
Neurosci Lett ; 399(3): 191-6, 2006 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-16517070

RESUMO

Cocaine's enhancement of dopaminergic neurotransmission in the mesolimbic pathway plays a critical role in the initial reinforcing properties of this drug. However, other neurotransmitter systems are also integral to the addiction process. A large body of data indicates that opioids and dopamine together mediate emotional and reinforced behaviors. In support of this, cocaine-mediated increases in activation of dopamine D1 receptors (D1R) results in a desensitization of delta-opioid receptor (DOR) signaling through adenylyl cyclase (AC) in striatal neurons. To further define cellular mechanisms underlying this effect, the subcellular distribution of DOR and D1R was examined in the rat dorsolateral striatum. Dual immunoperoxidase/gold-silver detection combined with electron microscopy was used to identify DOR and D1R immunoreactivities in the same section of tissue. Semi-quantitative analysis revealed that a subset of dendritic cellular profiles exhibited both DOR and D1R immunoreactivities. Of 198 randomly sampled D1R immunoreactive profiles, 43% contained DOR. Similarly of 165 DOR-labeled cellular profiles, 52% contained D1R. The present data provide ultrastructural evidence for co-existence between DOR and D1R in striatal neurons, suggesting a possible mechanism whereby D1R modulation may alter DOR function.


Assuntos
Corpo Estriado/citologia , Neurônios/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores Opioides delta/metabolismo , Animais , Imuno-Histoquímica/métodos , Masculino , Microscopia Imunoeletrônica/métodos , Modelos Biológicos , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley
9.
Anat Rec A Discov Mol Cell Evol Biol ; 279(1): 583-91, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15224400

RESUMO

Previous studies have shown significant changes in dopamine and opioid receptors in the basal ganglia following administration of cocaine. Cocaine administration results in a significant increase in the number of opioid receptors in dopamine-enriched brain regions. The aim of this study was to determine if dopamine D2 receptors (D2r) and micro-opioid receptors (microOr) are localized to the same neurons in the dorsolateral striatum. Immunoperoxidase and immunogold-silver labeling combined with electron microscopy was used to examine the ultrastructural localization of both receptors in the dorsolateral striatum. Approximately half of the microOr-labeled somatodendritic processes showed immunolabeling for the D2r. Similarly, about half of the D2r-labeled dendrites and cell bodies showed immunolabeling for the microOr. In conclusion, our results indicate that individual neurons in the rat dorsolateral striatum may be directly modulated by both dopaminergic and opioid ligands. These data also suggest that the molecular mechanism responsible for the up-regulation of microOrs in the caudate and putamen following cocaine exposure may depend, in part, on the co-existence of D2rs and micro-Ors in these cells.


Assuntos
Corpo Estriado/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores Opioides mu/metabolismo , Animais , Corpo Estriado/ultraestrutura , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
10.
Neuroscience ; 125(1): 211-20, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15051160

RESUMO

Cocaine-induced behavioral sensitization is a complex phenomenon involving a number of neuromodulator and neurotransmitter systems. To specifically investigate the role of the micro opioid receptor (MOR) in cocaine-induced behavioral sensitization in mice, both genetic and pharmacological approaches were undertaken. MOR-1 deficient mice of varying backgrounds (C57BL/6J, 129S6, F1 hybrid 129S6xC57BL/6J and 129S6xC57BL/6J) and wild-type C57BL/6J mice exposed continuously to naltrexone, an opioid receptor antagonist, received single daily injections of saline or cocaine for 10 days. All mice received a single cocaine challenge 7 days following the last saline or cocaine injection to test for the expression of sensitization. The locomotor-stimulating and sensitizing effects of cocaine observed in MOR-1 wild-type mice were absent in MOR-1 knockout mice maintained on the mixed 129S6xC57BL/6J background. In contrast, MOR-1 deficient mice developed on a C57BL/6J background showed an accentuated sensitivity to cocaine-induced locomotion. Cocaine's psychomotor activating effects were more pronounced in the MOR-1 C57BL/6J knockouts injected daily with cocaine than in the MOR-1 wild-type mice. Similar locomotor-stimulating and sensitizing effects were found in both F1 hybrid 129S6xC57BL/6J MOR-1 wild-type and MOR-1 knockout mice, while the 129S6 strain showed an overall indifference to cocaine. That is, both the locomotor-stimulating and sensitizing effects of cocaine were absent in both MOR-1 wild-type and MOR-1 knockout mice maintained on the 129S6 background. Lastly, the locomotor-stimulating and sensitizing effects of cocaine were attenuated in C57BL/6J wild-type mice exposed continuously to naltrexone. Collectively, these data support a role for opioidergic involvement in cocaine-influenced behavior in mice. Moreover, MORs appear to differentially modulate a sensitized response to cocaine in different strains of mice as delineated by MOR-1 gene deletion and pharmacological antagonism.


Assuntos
Adaptação Fisiológica/fisiologia , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Receptores Opioides mu/genética , Animais , Comportamento Animal , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/deficiência
11.
Synapse ; 47(4): 243-9, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12539196

RESUMO

Repeated intake of opioids is associated with dose escalation and alterations in signal transduction at the G-protein-coupled receptor level. The current study utilized two experiments to identify factors in rats that influence consumption rates such as daily intake of self-administered morphine and receptor desensitization. In Experiment 1, rats self-administered either 0.30, 1.00, or 3.00 mg/kg/infusion morphine sulfate (morphine) during 7 daily 4-h sessions. For Experiment 2, rats were assigned to groups that self-administered either 1) self-regulated escalating doses of morphine, 2) a fixed dose of morphine, or 3) saline during 18-h sessions for 7 days to determine if dose control would increase consumption without significantly decreasing response rate. We then assessed morphine-stimulated [(35)S]GTPgammaS binding in the amygdala and thalamus from these three groups in Experiment 2. Results from Experiment 1 demonstrated that 0.30 mg/kg/morphine did not support stable self-administration. For Experiment 2, the self-escalation group self-administered more morphine than the fixed-dose group, yet maintained similar response rates. Additionally, self-escalation rats demonstrated decreased morphine-stimulated [(35)S]GTPgammaS binding in membranes prepared from amygdalar and thalamic nuclei compared to the fixed-dose and control groups. Our results suggest that session length inversely affects consumption rates for fixed doses of morphine. Self-regulated dosing of morphine is also associated with rapid escalation of daily consumption and no significant alterations in consumption rates. These results suggest subject-regulated dosing is a useful approach for modeling dose escalation associated with opioid dependence.


Assuntos
Tolerância a Medicamentos/fisiologia , Morfina/administração & dosagem , Morfina/farmacologia , Entorpecentes/administração & dosagem , Entorpecentes/farmacologia , Tonsila do Cerebelo/metabolismo , Animais , Relação Dose-Resposta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Dependência de Morfina , Ratos , Autoadministração , Tálamo/metabolismo , Fatores de Tempo
12.
J Neuroendocrinol ; 13(9): 808-17, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11578531

RESUMO

The hypothalamic-pituitary-adrenal (HPA) axis is regulated by stress-related excitatory inputs, and various inhibitory and negative-feedback controls by glucocorticoids and opioids, including pro-opiomelanocortin (POMC)-derived peptides. The role of POMC-derived peptides of pituitary origin in the modulation of brain POMC mRNA expression and opioid receptor binding was investigated using a line of transgenic mice that express a fusion gene composed of the pituitary expression-specific promoter region of the POMC gene driving the herpes simplex viral-1 thymidine kinase (TK). Male adult mice were treated with the antiherpes agent ganciclovir that selectively ablates cells expressing TK. Following treatment, POMC mRNA levels, measured by quantitative solution hybridization/RNase protection assays, were decreased by 48% in the pituitary of the TK+/+ mice, reflecting an expected loss of the pituitary corticotrope POMC cells. This treatment also significantly lowered pituitary beta-endorphin immunoreactivity content and plasma concentrations of corticosterone. In contrast, POMC mRNA levels were increased by 79% in the hypothalamus of the TK+/+ mice with pituitary POMC cell ablation. Binding of [(3)H]DAMGO to mu opioid receptors, as measured by quantitative autoradiography, was significantly reduced in several brain regions including the central grey, median raphe and superficial grey layer of the superior colliculus. These regions are innervated by hypothalamic POMC neurones. No significant differences in binding to either kappa or delta opioid receptors were found in the brain regions studied. These results suggest that POMC-derived peptides of pituitary origin may exert a tonic negative-feedback effect on hypothalamic POMC neurones. In turn, the downregulation of central mu opioid receptors in this model may be mediated through a mechanism related to hypothalamic POMC overexpression.


Assuntos
Hipotálamo/metabolismo , Mesencéfalo/metabolismo , Hipófise/metabolismo , Pró-Opiomelanocortina/fisiologia , RNA Mensageiro/metabolismo , Receptores Opioides mu/metabolismo , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/metabolismo , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Lobo Frontal/metabolismo , Masculino , Camundongos , Camundongos Transgênicos/genética , Hipófise/citologia , Pró-Opiomelanocortina/genética , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Distribuição Tecidual
13.
Life Sci ; 69(9): 1093-102, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11508652

RESUMO

Results of numerous studies indicate that the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) modulates central dopamine systems, and that GABA(B) receptors may play a primary role in decreasing dopamine release. To determine if chronic cocaine administration alters the functional coupling of GABA(B) receptors to G-proteins in central dopamine systems, male F-344 rats received cocaine (15 mg/kg/injection) or saline three times a day at hourly intervals for fourteen consecutive days. Rats were decapitated one hour after the last injection and crude membrane preparations were made from the substantia nigra, caudate-putamen, ventral tegmental area, nucleus accumbens, and frontal cortex of individual rats. The ability of the specific GABA(B) receptor agonist baclofen to stimulate 35S-GTPgammaS binding in each of these regions was determined for individual animals. Additionally, baclofen-stimulated 35S-GTPgammaS binding in each of these regions in rats that received cocaine was compared to baclofen-stimulated 35S-GTPgammaS binding in rats that received control injections of saline. The EC50 of baclofen and maximal baclofen-stimulated 35S-GTPgammaS binding over basal levels were determined in each brain region in the saline group and in the cocaine group. Two-way ANOVA revealed a significant decrease in GABA(B) receptor-stimulated 35S-GTPgammaS binding in the ventral tegmental area of the cocaine group compared to the saline group. These data suggest that chronic exposure to cocaine decreases the functional coupling of GABA(B) receptors to G-proteins selectively in the ventral tegmental area. This finding may have implications in the augmented extracellular dopamine levels seen in the nucleus accumbens of rats that have been sensitized to cocaine.


Assuntos
Baclofeno/farmacologia , Cocaína/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Receptores de GABA-B/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Antagonistas de Receptores de GABA-B , Sistema Límbico/metabolismo , Masculino , Ratos , Ratos Endogâmicos F344 , Frações Subcelulares , Substância Negra/metabolismo , Radioisótopos de Enxofre , Membranas Sinápticas/efeitos dos fármacos , Membranas Sinápticas/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos
14.
Ann N Y Acad Sci ; 937: 74-92, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11458541

RESUMO

Cocaine is a widely abused psychostimulant. Its direct actions include inhibition of dopamine, serotonin, and norepinephrine reuptake into presynaptic nerve terminals, thereby potentiating the actions of these transmitters in the synapse. A variety of studies have demonstrated that cocaine can also have profound effects on the endogenous opioid system. Compelling evidence points to the importance of mu opioid receptors in human cocaine addiction and craving. Animal studies support these findings and demonstrate that chronic cocaine administration can result in alterations in opioid receptor expression and function as measured by changes in critical signal transduction pathways. This chapter reviews studies on the regulation of opioid receptors as the result of exposure to cocaine.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Receptores Opioides/efeitos dos fármacos , Receptores Opioides/fisiologia , Animais , AMP Cíclico/farmacologia , Humanos , Receptores Opioides/biossíntese , Transdução de Sinais
15.
Brain Res ; 900(1): 103-9, 2001 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-11325352

RESUMO

The present study investigated the impact of dosing schedule on cocaine-induced receptor alterations. Rats were injected with 30 mg/kg per day of cocaine given either as a single injection or in two equally divided doses for 14 days. The effects of these two dosing regimens were compared with our previous findings following administration of cocaine three times daily at 1-h intervals. Using receptor autoradiography, twice daily injections of cocaine produced an upregulation of mu opioid receptors in the rostral nucleus accumbens, rostral caudate putamen, and layer I of the rostral cingulate cortex, whereas single daily injections resulted in a significant increase in the nucleus accumbens only. Only small insignificant increases in kappa opioid receptor densities were found following either once or twice daily cocaine injections, whereas three daily injections produced an increase in kappa receptor density in the cingulate cortex, nucleus accumbens, and caudate putamen. Increased dopamine D1 receptor binding was found in the nucleus accumbens and olfactory tubercle following twice daily cocaine injections, but not after single daily injections of the same total daily dose. These results demonstrate that the same total daily dose of cocaine administered in multiple small injections produces a greater effect on receptor regulation than a single larger injection. This suggests that the interval between cocaine injections is an important variable when studying the effects of cocaine on neurochemistry.


Assuntos
Cocaína/administração & dosagem , Receptores de Dopamina D1/efeitos dos fármacos , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Núcleo Caudado/efeitos dos fármacos , Núcleo Caudado/metabolismo , Cocaína/farmacologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Giro do Cíngulo/efeitos dos fármacos , Giro do Cíngulo/metabolismo , Injeções Intraperitoneais , Masculino , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Condutos Olfatórios/efeitos dos fármacos , Condutos Olfatórios/metabolismo , Especificidade de Órgãos , Putamen/efeitos dos fármacos , Putamen/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores de Dopamina D1/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Regulação para Cima/efeitos dos fármacos
16.
Neuropharmacology ; 39(3): 372-81, 2000 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-10698003

RESUMO

Dopaminergic and opioidergic systems interact in the striatum to modulate locomotor and motivated behaviors. The present study investigated the molecular interactions of these two systems by determining the role of dopamine D1 and D2 receptors in the modulation of opioid receptor-mediated signal transduction. Male Fischer rats were injected daily for 10 days with either saline, the D1 receptor agonist SKF 82958, the D2 receptor agonist quinpirole, or both SKF 82958 and quinpirole. Administration of SKF 82958 alone or together with quinpirole attenuated the ability of the delta receptor agonist D-Pen2,D-Pen5-enkephalin (DPDPE) to inhibit adenylyl cyclase activity in the caudate putamen and nucleus accumbens. Quinpirole administration alone had no effect. The efficacy and potency of the mu opioid receptor agonist D-Ala2,N-Me-Phe4,Gly-ol5-enkephalin (DAMGO) to inhibit adenylyl cyclase activity was unaltered following administration of either dopamine receptor agonist. Administration of SKF 82958 had no affect on delta receptor binding, forskolin-stimulated adenylyl cyclase activity, or G protein/adenylyl cyclase coupling. However, the ability of DPDPE to stimulate binding of [35S]GTPgammaS to inhibitory G proteins was attenuated in animals that received SKF 82958. These results suggest that repeated activation of D1 receptors attenuates the functional coupling of delta opioid receptors with adenylyl cyclase due to decreased coupling between delta receptors and G proteins.


Assuntos
Adenilil Ciclases/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Receptores de Dopamina D1/efeitos dos fármacos , Receptores Opioides delta/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Analgésicos Opioides/farmacologia , Animais , Benzazepinas/farmacologia , Corpo Estriado/metabolismo , Agonistas de Dopamina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Ratos , Ratos Endogâmicos F344 , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/metabolismo , Receptores Opioides delta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
Nat Neurosci ; 2(2): 151-6, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10195199

RESUMO

Morphine produces analgesia by activating mu opioid receptors encoded by the MOR-1 gene. Although morphine-6 beta-glucuronide (M6G), heroin and 6-acetylmorphine also are considered mu opioids, recent evidence suggests that they act through a distinct receptor mechanism. We examined this question in knockout mice containing disruptions of either the first or second coding exon of MOR-1. Mice homozygous for either MOR-1 mutation were insensitive to morphine. Heroin, 6-acetylmorphine and M6G still elicited analgesia in the exon-1 MOR-1 mutant, which also showed specific M6G binding, whereas M6G and 6-acetylmorphine were inactive in the exon-2 MOR-1 mutant. These results provide genetic evidence for a unique receptor site for M6G and heroin analgesia.


Assuntos
Analgésicos Opioides/farmacologia , Éxons/genética , Heroína/farmacologia , Derivados da Morfina/farmacologia , Receptores Opioides mu/genética , Animais , Resistência a Medicamentos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Camundongos Knockout/fisiologia , Transcrição Gênica/fisiologia
18.
Synapse ; 30(2): 166-71, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9723786

RESUMO

The effect of chronic cocaine exposure on the central serotonergic system in the rat was investigated using a selective 5-HT1A receptor agonist, [3H]8-hydroxy-2-(di-N-propylamino) tetralin (8-OH-DPAT), and a 5-HT2A receptor antagonist, [3H]ketanserin, as tritiated ligands in a quantitative autoradiography study. Rats were administered cocaine in a "binge" pattern, 15 mg/kg/injection, three times a day, at 1-h intervals for 14 days to mimic the pattern often seen in human cocaine addicts. A significant decrease in the binding of [3H]8-OH-DPAT was found in the ventromedial hypothalamus (P < 0.001) and the dorsal dentate gyrus (P < 0.01) in rats administered cocaine as compared with rats injected with saline. No significant difference in the binding of [3H]ketanserin was found in frontal, parietal, agranular insular, and piriform cortices, caudate-putamen, olfactory tubercle, nucleus accumbens, thalamus, septohippocampal nucleus, and claustrum. Several studies have shown that 5-HT1A receptor agonists have antidepressant properties. Other studies, in animal models, have shown that 5-HT1A receptor agonists stimulate the hypothalamic-pituitary-adrenal axis, which is of interest, since chronic activation of this axis has been related to anxiety and depression. Our data show that the 5-HT1A component of the serotonergic system is altered following chronic "binge" pattern cocaine administration in an animal model and may be related to changes in the HPA axis and behavior.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/psicologia , Giro Denteado/metabolismo , Regulação para Baixo/fisiologia , Hipotálamo/metabolismo , Receptores de Serotonina/biossíntese , Animais , Autorradiografia , Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Masculino , Ratos , Receptor 5-HT2A de Serotonina , Receptores 5-HT1 de Serotonina
19.
Neuroscience ; 85(3): 897-905, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9639282

RESUMO

The present study utilized a newly developed quantitative immunohistochemical assay to measure changes in mu opioid receptor abundance following chronic administration of the opioid receptor antagonist naltrexone. These data were compared with those obtained from mu receptor radioligand binding on adjacent tissue sections, in order to determine whether the characteristic antagonist-induced increase in radioligand binding is due to an increase in the total number of mu receptors and/or to an increase in the proportion of receptors that are in an active binding conformation in the absence of a change in the total number of receptors. Adult male Sprague-Dawley rats were administered naltrexone, 7-8 mg/kg per day, or saline continuously for seven days by osmotic minipumps, after which time their brains were processed for immunohistochemistry and receptor autoradiography on adjacent fresh frozen tissue sections. Semiquantitative immunohistochemistry was performed using a radiolabelled secondary antibody for autoradiographic determination and a set of radioactive standards. Results demonstrate an overall concordance between the distribution of mu opioid receptors as measured by the two different methods with a few exceptions. Following naltrexone administration, mu receptor immunoreactivity was significantly higher in the amygdala, thalamus, hippocampus, and interpeduncular nucleus as compared with the saline-treated control animals. [3H]D-Ala2,N-Me-Phe4,Gly-ol5-enkephalin binding to mu opioid receptors was significantly higher in the globus pallidus, amygdala, thalamus, hypothalamus, hippocampus, substantia nigra, ventral tegmental area, central gray, and interpeduncular nucleus of the naltrexone-treated rats. These findings indicate that in some brain regions chronic naltrexone exposure increases the total number of mu opioid receptors, while in other regions there is an increase in the percent of active receptors without an observable change in the total number of receptors. Quantitative receptor immunodetection together with ligand autoradiography provides a new approach for investigating the regulation of mu opioid receptors on tissue sections.


Assuntos
Química Encefálica/efeitos dos fármacos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/análise , Regulação para Cima/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Animais , Autorradiografia , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Masculino , Naltrexona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Trítio
20.
Neuropsychopharmacology ; 19(2): 146-53, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9629568

RESUMO

In our previous positron emission tomography studies striatal binding for both [11C]SCH23390 and [11C]N-methylspiperone (NMSP) were decreased in the rat brain on the last day of chronic (14 days) binge cocaine administration. We have found that [11C]SCH23390 binding to dopamine D1 receptors returns to saline control levels within ten days withdrawal from chronic binge cocaine and remains at control levels after 21 days withdrawal. An 18% decrease in [11C]NMSP binding to dopamine D2 receptors was observed after ten days withdrawal. However, importantly, after 21 days withdrawal [11C]NMSP binding was at saline control levels. Changes of in vivo [11C]NMSP binding required a longer abstinence period for normalization than [11C]SCH23390 binding. The apparent recovery of dopamine D2 receptors after prolonged abstinence from chronic cocaine and the different rates of normalization for dopamine D1 versus D2 receptors may be critical information for development of pharmacotherapies for cocaine dependent patients.


Assuntos
Benzazepinas/metabolismo , Encéfalo/metabolismo , Cocaína/farmacologia , Receptores de Dopamina D2/metabolismo , Espiperona/análogos & derivados , Síndrome de Abstinência a Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Análise de Variância , Animais , Encéfalo/diagnóstico por imagem , Radioisótopos de Carbono , Masculino , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Espiperona/farmacocinética , Fatores de Tempo , Tomografia Computadorizada de Emissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA