Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Neuropharmacology ; 180: 108307, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32941853

RESUMO

Opioid signaling controls the activity of the brain's reward system. It is involved in signaling the hedonic effects of rewards and has essential roles in reinforcement and motivational processes. Here, we focused on opioid signaling through mu and delta receptors on dopaminoceptive neurons and evaluated the role these receptors play in reward-driven behaviors. We generated a genetically modified mouse with selective double knockdown of mu and delta opioid receptors in neurons expressing dopamine receptor D1. Selective expression of the transgene was confirmed using immunostaining. Knockdown was validated by measuring the effects of selective opioid receptor agonists on neuronal membrane currents using whole-cell patch clamp recordings. We found that in the nucleus accumbens of control mice, the majority of dopamine receptor D1-expressing neurons were sensitive to a mu or delta opioid agonist. In mutant mice, the response to the delta receptor agonist was blocked, while the effects of the mu agonist were strongly attenuated. Behaviorally, the mice had no obvious impairments. The mutation did not affect the sensitivity to the rewarding effects of morphine injections or social contact and had no effect on preference for sweet taste. Knockdown had a moderate effect on motor activity in some of the tests performed, but this effect did not reach statistical significance. Thus, we found that knocking down mu and delta receptors on dopamine receptor D1-expressing cells does not appreciably affect some of the reward-driven behaviors previously attributed to opioid signaling.


Assuntos
Neurônios/metabolismo , Receptores de Dopamina D1/biossíntese , Receptores Opioides delta/deficiência , Receptores Opioides mu/deficiência , Recompensa , Analgésicos Opioides/farmacologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Receptores de Dopamina D1/genética , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
2.
Biomolecules ; 10(9)2020 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-32932811

RESUMO

Intermittent hypoxia and various pharmacological compounds protect the heart from ischemia reperfusion injury in experimental approaches, but the translation into clinical trials has largely failed. One reason may lie in species differences and the lack of suitable human in vitro models to test for ischemia/reperfusion. We aimed to develop a novel hypoxia-reoxygenation model based on three-dimensional, spontaneously beating and work performing engineered heart tissue (EHT) from rat and human cardiomyocytes. Contractile force, the most important cardiac performance parameter, served as an integrated outcome measure. EHTs from neonatal rat cardiomyocytes were subjected to 90 min of hypoxia which led to cardiomyocyte apoptosis as revealed by caspase 3-staining, increased troponin I release (time control vs. 24 h after hypoxia: cTnI 2.7 vs. 6.3 ng/mL, ** p = 0.002) and decreased contractile force (64 ± 6% of baseline) in the long-term follow-up. The detrimental effects were attenuated by preceding the long-term hypoxia with three cycles of 10 min hypoxia (i.e., hypoxic preconditioning). Similarly, [d-Ala2, d-Leu5]-enkephalin (DADLE) reduced the effect of hypoxia on force (recovery to 78 ± 5% of baseline with DADLE preconditioning vs. 57 ± 5% without, p = 0.012), apoptosis and cardiomyocyte stress. Human EHTs presented a comparable hypoxia-induced reduction in force (55 ± 5% of baseline), but DADLE failed to precondition them, likely due to the absence of δ-opioid receptors. In summary, this hypoxia-reoxygenation in vitro model displays cellular damage and the decline of contractile function after hypoxia allows the investigation of preconditioning strategies and will therefore help us to understand the discrepancy between successful conditioning in vitro experiments and its failure in clinical trials.


Assuntos
Analgésicos Opioides/farmacologia , Leucina Encefalina-2-Alanina/farmacologia , Hipóxia/tratamento farmacológico , Precondicionamento Isquêmico Miocárdico/métodos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Receptores Opioides delta/genética , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Caspase 3/genética , Caspase 3/metabolismo , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Modelos Biológicos , Contração Miocárdica/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos , Receptores Opioides delta/deficiência , Especificidade da Espécie , Engenharia Tecidual/métodos , Troponina I/metabolismo
3.
Neuropsychopharmacology ; 39(13): 2974-88, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24943644

RESUMO

The repeated cycles of cessation of consumption and relapse remain the major clinical concern in treating drug addiction. The endogenous opioid system is a crucial component of the reward circuit that participates in the adaptive changes leading to relapse in the addictive processes. We have used genetically modified mice to evaluate the involvement of µ-opioid receptor (MOR) and δ-opioid receptor (DOR) and their main endogenous ligands, the enkephalins derived from proenkephalin (PENK) and prodynorphin (PDYN), in the reinstatement of cocaine-seeking behavior. Constitutive knockout mice of MOR, DOR, PENK, and PDYN, and their wild-type littermates were trained to self-administer cocaine or to seek for palatable food, followed by a period of extinction and finally tested on a cue-induced reinstatement of seeking behavior. The four lines of knockout mice acquired operant cocaine self-administration behavior, although DOR and PENK knockout mice showed less motivation for cocaine than wild-type littermates. Moreover, cue-induced relapse was significantly decreased in MOR and DOR knockout mice. In contrast, PDYN knockout mice showed a slower extinction and increased relapse than wild-type littermates. C-Fos expression analysis revealed differential activation in brain areas related with memory and reward in these knockout mice. No differences were found in any of the four genotypes in operant responding to obtain palatable food, indicating that the changes revealed in knockout mice were not due to unspecific deficit in operant performance. Our results indicate that MOR, DOR, and PDYN have a differential role in cue-induced reinstatement of cocaine-seeking behavior.


Assuntos
Condicionamento Operante/fisiologia , Comportamento de Procura de Droga/fisiologia , Encefalinas/deficiência , Precursores de Proteínas/deficiência , Receptores Opioides mu/deficiência , Reforço Psicológico , Análise de Variância , Anestésicos Locais/administração & dosagem , Animais , Cocaína/administração & dosagem , Condicionamento Operante/efeitos dos fármacos , Sinais (Psicologia) , Encefalinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Knockout , Precursores de Proteínas/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Esquema de Reforço , Autoadministração
4.
Eur J Pharmacol ; 731: 1-7, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24657279

RESUMO

Several methodological approaches suggest that receptor heteromers exist in cell systems, but their presence in physiological tissue is widely contentious. We describe a novel method to determine if heterodimers exist in brain tissue sections using autoradiographic binding comparisons from single and double gene knockout mice, where tissues either have a full receptor complement and can form heterodimers, or are incapable of making heterodimers. We have tested this model, which we have named Knockout Subtraction Autoradiography, to determine if heterodimerisation of the kappa (KOP) and delta opioid (DOP) receptors occurs, as evidence from binding studies in cell systems suggest they are present in the brain. Using labeling of putative KOP receptor/DOP receptor heterodimers with either [(3)H]bremazocine or with [(3)H]naltrindole, two ligands which were used to provide evidence suggesting that these opioid receptor subtypes heterodimerize, we have applied a subtraction equation model based on the principle that receptor gene double knockout of either MOP receptor/KOP receptor (DOP receptor expression only) or MOP receptor/DOP receptor (KOP receptor expression only) produces tissue incapable of making the KOP receptor/DOP receptor heterodimer. We have shown in most brain regions that the labeling fits a simple additive model of monomer labeling, but that in a few brain regions opioid receptor heterodimerization does occur. The data does not support the conclusion that KOP receptor/DOP receptor heterodimerisation is widespread in the central nervous system, but does indicate that this novel methodology can detect heterodimerisation, when ligands with distinct binding affinities for monomer and heterodimer forms exist.


Assuntos
Autorradiografia/métodos , Encéfalo/metabolismo , Técnicas de Inativação de Genes , Multimerização Proteica , Receptores Opioides delta/química , Receptores Opioides kappa/química , Técnica de Subtração , Animais , Benzomorfanos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Naltrexona/análogos & derivados , Naltrexona/metabolismo , Estrutura Quaternária de Proteína , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética
5.
Philos Trans R Soc Lond B Biol Sci ; 369(1637): 20120466, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24446500

RESUMO

Mice with cytotoxic lesions of the dorsal hippocampus (DH) underestimated 15 s and 45 s target durations in a bi-peak procedure as evidenced by proportional leftward shifts of the peak functions that emerged during training as a result of decreases in both 'start' and 'stop' times. In contrast, mice with lesions of the ventral hippocampus (VH) displayed rightward shifts that were immediately present and were largely limited to increases in the 'stop' time for the 45 s target duration. Moreover, the effects of the DH lesions were congruent with the scalar property of interval timing in that the 15 s and 45 s functions superimposed when plotted on a relative timescale, whereas the effects of the VH lesions violated the scalar property. Mice with DH lesions also showed enhanced reversal learning in comparison to control and VH lesioned mice. These results are compared with the timing distortions observed in mice lacking δ-opioid receptors (Oprd1(-/-)) which were similar to mice with DH lesions. Taken together, these results suggest a balance between hippocampal-striatal interactions for interval timing and demonstrate possible functional dissociations along the septotemporal axis of the hippocampus in terms of motivation, timed response thresholds and encoding in temporal memory.


Assuntos
Comportamento Animal/fisiologia , Corpo Estriado/fisiologia , Hipocampo/fisiopatologia , Receptores Opioides delta/deficiência , Percepção do Tempo/fisiologia , Análise de Variância , Animais , Condicionamento Psicológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Neuropsychopharmacology ; 38(6): 1050-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23303070

RESUMO

Pharmacological data suggest that delta opioid receptors modulate learning and memory processes. In the present study, we investigated whether inactivation of the delta opioid receptor modifies hippocampus (HPC)- and striatum-dependent behaviors. We first assessed HPC-dependent learning in mice lacking the receptor (Oprd1(-/-) mice) or wild-type (WT) mice treated with the delta opioid antagonist naltrindole using novel object recognition, and a dual-solution cross-maze task. Second, we subjected mutant animals to memory tests addressing striatum-dependent learning using a single-solution response cross-maze task and a motor skill-learning task. Genetic and pharmacological inactivation of delta opioid receptors reduced performance in HPC-dependent object place recognition. Place learning was also altered in Oprd1(-/-) animals, whereas striatum-dependent response and procedural learning were facilitated. Third, we investigated the expression levels for a large set of genes involved in neurotransmission in both HPC and striatum of Oprd1(-/-) mice. Gene expression was modified for several key genes that may contribute to alter hippocampal and striatal functions, and bias striatal output towards striatonigral activity. To test this hypothesis, we finally examined locomotor effects of dopamine receptor agonists. We found that Oprd1(-/-) and naltrindole-treated WT mice were more sensitive to the stimulant locomotor effect of SKF-81297 (D1/D5), supporting the hypothesis of facilitated striatonigral output. These data suggest, for the first time, that delta receptor activity tonically inhibits striatal function, and demonstrate that delta opioid receptors modulate learning and memory performance by regulating the HPC/striatum balance.


Assuntos
Corpo Estriado/fisiologia , Hipocampo/fisiologia , Memória/fisiologia , Atividade Motora/fisiologia , Destreza Motora/fisiologia , Receptores Opioides delta/deficiência , Animais , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Opioides delta/genética
7.
FASEB J ; 26(8): 3483-92, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22593549

RESUMO

Pharmacological manipulation of opioid receptors alters feeding behavior. However, the individual contributions of each opioid receptor subtype on energy balance remain largely unknown. Herein, we investigated whether genetic disruption of the δ-opioid receptor (DOR) also controls energy homeostasis. Mice lacking DOR and wild-type mice were fed with standard diet and high-energy diet (HED). Mice were analyzed in vivo with the indirect calorimetry system, and tissues were analyzed by real-time PCR and Western blot analysis. DOR-knockout (KO) mice gained less weight (P<0.01) and had lower fat mass (P<0.01) when compared to WT mice fed an HED. Although DOR-KO mice were hyperphagic, they showed higher energy expenditure (P<0.05), which was the result of an increased activation of the thermogenic program in brown adipose tissue. The increased nonshivering thermogenesis involved the stimulation of uncoupling protein 1 (UCP1; P<0.01), peroxisome proliferator-activated receptor γ coactivator (PGC1α; P<0.05), and fibroblast growth factor 21 (FGF21; P<0.01). DOR deficiency also led to an attenuation of triglyceride content in the liver (P<0.05) in response to an HED. These findings reveal a novel role of DOR in the control of thermogenic markers and energy expenditure, and they provide a potential new therapeutic approach for the treatment of obesity.


Assuntos
Obesidade/etiologia , Receptores Opioides delta/deficiência , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Dieta Hiperlipídica , Metabolismo Energético/genética , Glucose/metabolismo , Homeostase/fisiologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Termogênese/fisiologia , Triglicerídeos/metabolismo
8.
J Neuroimmune Pharmacol ; 6(4): 608-16, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21938479

RESUMO

Here, we evaluated the influence of endogenous opioid activation on immune responses by examining consequences of all three opioid receptor gene (mu, delta and kappa) inactivation. In triple-opioid receptor knockout mice, splenocytes and thymocytes numbers, lymphocyte subsets as well as proliferation and cytokines induced by in vitro stimulation of T lymphocytes were measured. Compared with wild-type mice, similar lymphocyte distribution in thymus and spleen as well as comparable T lymphocyte proliferation were observed, while lower levels of IL-2 and IFNγ as well as higher levels of IL-4 and IL-10 were found in triple-opioid receptor knockout mice. Together, our results indicate a shift from TH1 to TH2 cytokines in triple-opioid receptor knockout animals, suggesting that global endogenous opioid tone drives T lymphocytes toward a TH1 profile under non-pathological conditions.


Assuntos
Peptídeos Opioides/imunologia , Receptores Opioides delta/imunologia , Receptores Opioides kappa/imunologia , Receptores Opioides mu/imunologia , Células Th1/imunologia , Animais , Citocinas/biossíntese , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Timo/citologia , Timo/imunologia
9.
Brain Res ; 1396: 54-9, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21561601

RESUMO

The false-suffocation hypothesis of panic disorder (Klein, 1993) suggested δ-opioid receptors as a possible source of the respiratory dysfunction manifested in panic attacks occurring in panic disorder (Preter and Klein, 2008). This study sought to determine if a lack of δ-opioid receptors in a mouse model affects respiratory response to elevated CO2, and whether the response is modulated by benzodiazepines, which are widely used to treat panic disorder. In a whole-body plethysmograph, respiratory responses to 5% CO2 were compared between δ-opioid receptor knockout mice and wild-type mice after saline, diazepam (1mg/kg), and alprazolam (0.3mg/kg) injections. The results show that lack of δ-opioid receptors does not affect normal response to elevated CO2, but does prevent benzodiazepines from modulating that response. Thus, in the presence of benzodiazepine agonists, respiratory responses to elevated CO2 were enhanced in δ-opioid receptor knockout mice compared to wild-type mice. This suggests an interplay between benzodiazepine receptors and δ-opioid receptors in regulating the respiratory effects of elevated CO2, which might be related to CO2 induced panic.


Assuntos
Dióxido de Carbono/toxicidade , Transtorno de Pânico/fisiopatologia , Receptores de GABA-A/fisiologia , Receptores Opioides delta/fisiologia , Insuficiência Respiratória/fisiopatologia , Animais , Benzodiazepinas/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Dióxido de Carbono/sangue , Modelos Animais de Doenças , Hipercapnia/induzido quimicamente , Hipercapnia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtorno de Pânico/tratamento farmacológico , Transtorno de Pânico/metabolismo , Pletismografia Total/métodos , Receptores de GABA-A/efeitos dos fármacos , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Insuficiência Respiratória/tratamento farmacológico , Insuficiência Respiratória/genética , Volume de Ventilação Pulmonar/fisiologia
10.
Pain ; 152(6): 1238-1248, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21295407

RESUMO

Opioid receptors are major actors in pain control and are broadly distributed throughout the nervous system. A major challenge in pain research is the identification of key opioid receptor populations within nociceptive pathways, which control physiological and pathological pain. In particular, the respective contribution of peripheral vs. central receptors remains unclear, and it has not been addressed by genetic approaches. To investigate the contribution of peripheral delta opioid receptors in pain control, we created conditional knockout mice where delta receptors are deleted specifically in peripheral Na(V)1.8-positive primary nociceptive neurons. Mutant mice showed normal pain responses to acute heat and to mechanical and formalin stimuli. In contrast, mutant animals showed a remarkable increase of mechanical allodynia under both inflammatory pain induced by complete Freund adjuvant and neuropathic pain induced by partial sciatic nerve ligation. In these 2 models, heat hyperalgesia was virtually unchanged. SNC80, a delta agonist administered either systemically (complete Freund adjuvant and sciatic nerve ligation) or into a paw (sciatic nerve ligation), reduced thermal hyperalgesia and mechanical allodynia in control mice. However, these analgesic effects were absent in conditional mutant mice. In conclusion, this study reveals the existence of delta opioid receptor-mediated mechanisms, which operate at the level of Na(V)1.8-positive nociceptive neurons. Delta receptors in these neurons tonically inhibit mechanical hypersensitivity in both inflammatory and neuropathic pain, and they are essential to mediate delta opioid analgesia under conditions of persistent pain. This delta receptor population represents a feasible therapeutic target to alleviate chronic pain while avoiding adverse central effects. The conditional knockout of delta-opioid receptor in primary afferent Na(V)1.8 neurons augmented mechanical allodynia in persistent pain models and abolished delta opioid analgesia in these models.


Assuntos
Gânglios Espinais/patologia , Nociceptores/fisiologia , Dor/genética , Dor/patologia , Receptores Opioides delta/deficiência , Analgésicos Opioides/uso terapêutico , Análise de Variância , Animais , Benzamidas/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Adjuvante de Freund/efeitos adversos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Inflamação/induzido quimicamente , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Canal de Sódio Disparado por Voltagem NAV1.8 , Nociceptores/efeitos dos fármacos , Dor/etiologia , Medição da Dor/métodos , Piperazinas/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Canais de Sódio/genética , Canais de Sódio/metabolismo , Isótopos de Enxofre/farmacocinética
11.
Biol Psychiatry ; 69(7): 700-3, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21168121

RESUMO

BACKGROUND: Converging experimental data indicate that δ opioid receptors contribute to mediate drug reinforcement processes. Whether their contribution reflects a role in the modulation of drug reward or an implication in conditioned learning, however, has not been explored. In the present study, we investigated the impact of δ receptor gene knockout on reinforced conditioned learning under several experimental paradigms. METHODS: We assessed the ability of δ receptor knockout mice to form drug-context associations with either morphine (appetitive)- or lithium (aversive)-induced Pavlovian place conditioning. We also examined the efficiency of morphine to serve as a positive reinforcer in these mice and their motivation to gain drug injections, with operant intravenous self-administration under fixed and progressive ratio schedules and at two different doses. RESULTS: Mutant mice showed impaired place conditioning in both appetitive and aversive conditions, indicating disrupted context-drug association. In contrast, mutant animals displayed intact acquisition of morphine self-administration and reached breaking-points comparable to control subjects. Thus, reinforcing effects of morphine and motivation to obtain the drug were maintained. CONCLUSION: Collectively, the data suggest that δ receptor activity is not involved in morphine reinforcement but facilitates place conditioning. This study reveals a novel aspect of δ opioid receptor function in addiction-related behaviors.


Assuntos
Analgésicos Opioides/administração & dosagem , Condicionamento Operante/fisiologia , Deficiências da Aprendizagem/genética , Morfina/administração & dosagem , Receptores Opioides delta/deficiência , Reforço Psicológico , Animais , Comportamento Apetitivo/efeitos dos fármacos , Comportamento Animal , Condicionamento Operante/efeitos dos fármacos , Discriminação Psicológica/efeitos dos fármacos , Relação Dose-Resposta a Droga , Reação de Fuga/efeitos dos fármacos , Deficiências da Aprendizagem/fisiopatologia , Camundongos , Camundongos Knockout , Motivação/efeitos dos fármacos , Autoadministração , Percepção Espacial/efeitos dos fármacos , Fatores de Tempo
12.
Neuropsychopharmacology ; 33(7): 1746-59, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17895918

RESUMO

Both mu-opioid receptors (MORs) and delta-opioid receptors (DORs) are expressed in the ventral tegmental area (VTA) and are thought to be involved in the addictive properties of opiates. However, their respective contributions to opiate reward remain unclear. We used intracranial self-administration (ICSA) to study the rewarding effects of morphine microinjections into the VTA of male and female MOR-/- and DOR-/- mice. In brains of mice tested for intra-VTA morphine self-administration, we analyzed regional Fos protein expression to investigate the neural circuitry underlying this behavior. Male and female WT and DOR-/- mice exhibited similar self-administration performances, whereas knockout of the MOR gene abolished intra-VTA morphine self-administration at all doses tested. Naloxone (4 mg/kg) disrupted this behavior in WT and DOR mutants, without triggering physical signs of withdrawal. Morphine ICSA was associated with an increase in Fos within the nucleus accumbens, striatum, limbic cortices, amygdala, hippocampus, the lateral mammillary nucleus (LM), and the ventral posteromedial thalamus (VPM). This latter structure was found to express high levels of Fos exclusively in self-administering WT and DOR-/- mice. Abolition of morphine reward in MOR-/- mice was associated with a decrease in Fos-positive neurons in the mesocorticolimbic dopamine system, amygdala, hippocampus (CA1), LM, and a complete absence within the VPM. We conclude that (i) VTA MORs, but not DORs, are critical for morphine reward and (ii) the role of VTA-thalamic projections in opiate reward deserves to be further explored.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas Oncogênicas v-fos/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Área Tegmentar Ventral/metabolismo , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Contagem de Células/métodos , Condicionamento Operante/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Entorpecentes/administração & dosagem , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Oncogênicas v-fos/genética , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Receptores Opioides delta/deficiência , Receptores Opioides mu/deficiência , Autoadministração , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/efeitos dos fármacos
13.
Eur J Neurosci ; 23(3): 830-4, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16487163

RESUMO

We have evaluated the possible involvement of delta-opioid receptor (DOR) in the development and expression of neuropathic pain. For this purpose, partial ligation of the sciatic nerve was performed in DOR knockout mice and wild-type littermates. The development of mechanical and thermal allodynia, as well as thermal hyperalgesia was evaluated by using the von Frey filament model, the cold-plate test and the plantar test, respectively. In wild-type and DOR knockout mice, sciatic nerve injury led to a neuropathic pain syndrome revealed in these nociceptive behavioural tests. However, the development of mechanical and thermal allodynia, and thermal hyperalgesia was significantly enhanced in DOR knockout mice. These results reveal the involvement of DOR in the control of neuropathic pain and suggest a new potential therapeutic use of DOR agonists.


Assuntos
Dor/etiologia , Dor/metabolismo , Receptores Opioides delta/deficiência , Neuropatia Ciática/complicações , Neuropatia Ciática/metabolismo , Análise de Variância , Animais , Comportamento Animal , Modelos Animais de Doenças , Lateralidade Funcional , Camundongos , Camundongos Knockout , Dor/genética , Medição da Dor/métodos , Tempo de Reação/fisiologia , Neuropatia Ciática/genética , Fatores de Tempo
14.
Neuropharmacology ; 48(2): 228-35, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15695161

RESUMO

Naloxone benzoylhydrazone (NalBzoH) is a ligand used to study opioid receptors. It has been suggested to act at a novel kappa3 receptor but also appears to bind to classical opioid receptors, and possibly the ORL1 receptor. We have used opioid receptor triple knockout mice, deficient in genes coding for the mu, delta and kappa-receptor, to characterise the relative contributions of opioid and ORL1 activity to the binding of this ligand, by carrying out receptor autoradiography with [3H]NalBzoH. As competing ligands we have used diprenorphine and nociceptin at 1 microM, alone or in combination, to determine the contribution of opioid and ORL1 receptor binding. At 4 nM [3H]NalBzoH showed labelling in wild-type brains indicative of broad spectrum classical opioid receptor binding. In the triple knockout brains all labelling was completely absent, suggesting that at this concentration there is no binding to ORL1 sites. However at 50 nM [3H]NalBzoH showed labelling in triple knockout brains with a distribution pattern indicative of ORL1 labelling. Quantitative analysis showed that nociceptin displaced typically 30% of the residual labelling in knockout brains whilst diprenorphine had relatively little effect. The data show that at 50 nM NalBzoH no binding was detected other than to classical opioid receptors or to ORL1 in an approximate ratio of 2:1.


Assuntos
Encéfalo/metabolismo , Naloxona/análogos & derivados , Naloxona/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Animais , Autorradiografia , Camundongos , Camundongos Knockout , Ligação Proteica/fisiologia , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética
15.
Neuroscience ; 127(2): 497-503, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15262338

RESUMO

Conventional brain microdialysis was used to assess basal and cocaine-induced dopamine (DA) levels in the nucleus accumbens of wildtype (WT) C57BL/6J mice and mice with constitutive deletion of ether mu- or delta-opioid receptors (MOR or DOR knockout [KO], respectively). Locomotor activity was assessed in these same animals. Basal locomotor activity of DOR KO was elevated relative to MOR KO, but did not differ from that of WT mice. DOR mice, but not WT or MOR KO, exhibited a significant increase in activity in response to an injection of saline. The acute administration of cocaine produced a dose-related increase in locomotor activity in the three genotypes. The locomotor activating effects of a low dose (10 mg/kg) of cocaine were enhanced in DOR KO mice whereas the locomotor activating effects of both a low and higher (20 mg/kg) dose of cocaine were reduced in MOR KO animals. Microdialysis studies revealed no difference between genotypes in basal DA levels. Acute administration of cocaine, but not saline, increased DA levels in WT and KO animals. Paradoxically, however, the magnitude of this effect was smaller in DOR KO as compared with that in either WT or MOR KO. These data indicate that constitutive deletion of either MOR or DOR results in contrasting effects upon responsiveness to cocaine, which is consistent with the distinct phenotypes previously described for these mutants.


Assuntos
Química Encefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Cocaína/farmacologia , Receptores Opioides delta/fisiologia , Receptores Opioides mu/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Química Encefálica/fisiologia , Transtornos Relacionados ao Uso de Cocaína/genética , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Genótipo , Camundongos , Camundongos Knockout , Microdiálise , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Peptídeos Opioides/metabolismo , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética
16.
J Chem Neuroanat ; 27(2): 119-28, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15121216

RESUMO

Mice lacking the mu-delta-kappa-opioid receptor (MOR/DOR/KOR) genes and their corresponding wild-type littermates have been used to quantify NPFF(1) and NPFF(2) (neuropeptide FF) receptors by in vitro autoradiography in the central nervous tissues. Adjacent coronal sections were labelled with [125I]YVP ([125I]YVPNLPQRF-NH(2)) and [125I]EYF ([125I]EYWSLAAPQRF-NH(2)) as specific radioligands for NPFF(1) and NPFF(2) receptors, respectively. NPFF(2) receptors are predominantly expressed in both genotypes, but their density increases significantly in non cortical regions of mutant mice: 64% in the amygdaloid area, 89, 308, 1214 and 49% in the nucleus of the vertical limb of the diagonal band, substantia nigra, the vestibular nucleus and the spinal cord, respectively. In contrast, the density of the NPFF(1) subtype is lower than NPFF(2) in both genotypes and significantly decreased in some brain areas of mutant mice: -99, -90 and -90% in the nucleus of the vertical limb of the diagonal band, substantia nigra and the spinal cord, respectively. This study shows that mice lacking opioid receptors have brain region-dependent increases (NPFF(2)) and decreases (NPFF(1)) in NPFF receptors densities and suggests a different functional participation of each NPFF receptor subtype in the actions of opioids.


Assuntos
Encéfalo/metabolismo , Receptores de Neuropeptídeos/metabolismo , Receptores Opioides/deficiência , Animais , Autorradiografia , Encéfalo/anatomia & histologia , Camundongos , Camundongos Knockout , Receptores Opioides/genética , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética
17.
Eur J Neurosci ; 19(8): 2239-48, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15090050

RESUMO

Delta-selective agonists have been developed to produce potent analgesic compounds with limited side-effects. DPDPE and deltorphin II are considered prototypes, but their delta-selectivity in vivo and the true ability of delta receptors to produce analgesia remain to be demonstrated. Here we have performed a parallel analysis of mu, delta and combinatorial opioid receptor knockout mice, in which we found no obvious alteration of G-protein coupling for remaining opioid receptors. We compared behavioural responses in two models of acute thermal pain following DPDPE and deltorphin II administration by intracerebroventricular route. In the tail-immersion test, both compounds were fully analgesic in delta knockout mice and totally inactive in mu knockout mice. In the hotplate test, the two compounds again produced full analgesia in delta knockout mice. In mu knockout mice, there was significant, although much lower, analgesia. Furthermore, DPDPE analgesia in the delta knockout mice was fully reversed by the mu selective antagonist CTOP in both tests. Together, this suggests that mu rather than delta receptors are recruited by the two agonists for the tail withdrawal and the hotplate responses. Finally, deltorphin II slightly prolonged jump latencies in double mu/kappa knockout mice (delta receptors only) and this response was abolished in the triple knockout mice, demonstrating that the activation of delta receptors alone can produce weak but significant mu-independent thermal antinociception.


Assuntos
D-Penicilina (2,5)-Encefalina/farmacologia , Oligopeptídeos/farmacologia , Medição da Dor/efeitos dos fármacos , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Analgesia/métodos , Animais , Relação Dose-Resposta a Droga , Feminino , Temperatura Alta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medição da Dor/métodos , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética
18.
Neuropharmacology ; 46(4): 519-30, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14975676

RESUMO

Opiate addiction is associated with abnormalities of neurofilament (NF) proteins and upregulation of cAMP signaling in the brain, which may modulate neuronal plasticity. This study investigated, using gene-targeted mice lacking mu-, delta- or kappa-opioid receptors, the role of these receptors in modulating the basal activity and the chronic effects of morphine on both intracellular targets. In WT mice, chronic treatment (5 days) with morphine (20-100 mg/kg) resulted in decreases in the immunodensity of neurofilament (NF)-L in the cerebral cortex (14-23%). In contrast, chronic morphine did not decrease NF-L in cortices of mu-, delta-, and kappa-KO mice, suggesting the involvement of the three types of opioid receptors in this effect of morphine. Also, the marked increase in phosphorylated NF-H induced by chronic morphine in WT mice (two-fold) was abolished in mu -KO mice. In cortex and/or striatum of mu-, delta- and kappa-KO mice, the basal immunodensities of Galphai1/2 proteins, the catalytic isoform (Calpha) of protein kinase A (PKA) and the total content of cAMP response element-binding protein (CREB, the nuclear target of PKA) were not different from those of WT mice. In contrast, phosphorylated CREB (the active form of this transcription factor) was reduced in cortex and/or striatum (23-26%) of mu- and delta-KO mice, but not in kappa-KO animals. These results suggest that the endogenous opioid tone acting on mu-/delta-receptors tonically stimulate CREB activation in the brain. In cortex and/or striatum of WT mice, chronic morphine did not induce upregulation of the main components of the cAMP signaling pathway. In contrast, chronic morphine treatment in mu-KO mice, but not in delta- or kappa-KO, resulted in a paradoxical upregulation of Galphai1/2 (12-19%), PKA (19-21%,) and phosphorylated CREB (21-73%), but not total CREB, in cortex and/or striatum. The induction of heterologous receptor adaptations in mu-KO mice may explain this paradoxical effect of morphine.


Assuntos
Morfina/administração & dosagem , Proteínas de Neurofilamentos/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , AMP Cíclico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Neurofilamentos/genética , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética
19.
Eur J Neurosci ; 17(1): 155-9, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12534979

RESUMO

Several studies have shown a functional relationship between the endogenous cannabinoid and opioid systems. However, acute effects of Delta9-tetrahydrocannabinol (THC) and physical dependence were not modified in knockout mice with single deletion of mu (MOR), delta (DOR) or kappa (KOR) opioid receptors. To further investigate the neurobiological basis of cannabinoid dependence, we have evaluated acute pharmacological responses, rewarding effects, tolerance and dependence to THC in double MOR/DOR knockout mice. Antinociception and hypolocomotion induced by acute THC administration remained unaffected, whereas the hypothermic effect was slightly attenuated in these double knockout mice. During chronic THC treatment, knockout mice developed slower tolerance to the hypothermic effect, but the development of tolerance to antinociceptive and hypolocomotor effects was unchanged. The rewarding properties of THC, measured in the conditioned place preference paradigm, were reduced in knockout mice. Interestingly, the somatic manifestations of THC withdrawal were also significantly attenuated in mutant mice, suggesting that a cooperative action of MOR and DOR is required for the entire expression of THC dependence.


Assuntos
Analgésicos não Narcóticos/farmacologia , Dronabinol/efeitos adversos , Psicotrópicos/efeitos adversos , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Animais , Canabinoides/efeitos adversos , Tolerância a Medicamentos , Euforia/efeitos dos fármacos , Camundongos , Camundongos Knockout , Receptores Opioides delta/deficiência , Receptores Opioides mu/deficiência , Transtornos Relacionados ao Uso de Substâncias
20.
J Neurosci ; 22(3): 1146-54, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11826143

RESUMO

Repeated THC administration produces motivational and somatic adaptive changes leading to dependence in rodents. To investigate the molecular basis for cannabinoid dependence and its possible relationship with the endogenous opioid system, we explored delta9-tetrahydrocannabinol (THC) activity in mice lacking mu-, delta- or kappa-opioid receptor genes. Acute THC-induced hypothermia, antinociception, and hypolocomotion remained unaffected in these mice, whereas THC tolerance and withdrawal were minimally modified in mutant animals. In contrast, profound phenotypic changes are observed in several place conditioning protocols that reveal both THC rewarding and aversive properties. Absence of microreceptors abolishes THC place preference. Deletion of kappa receptors ablates THC place aversion and furthermore unmasks THC place preference. Thus, an opposing activity of mu- and kappa-opioid receptors in modulating reward pathways forms the basis for the dual euphoric-dysphoric activity of THC.


Assuntos
Canabinoides/farmacologia , Abuso de Maconha/fisiopatologia , Motivação , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Canabinoides/antagonistas & inibidores , Cruzamentos Genéticos , Dronabinol/antagonistas & inibidores , Dronabinol/farmacologia , Tolerância a Medicamentos/genética , Hipotermia/induzido quimicamente , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Piperidinas/farmacologia , Psicotrópicos/antagonistas & inibidores , Psicotrópicos/farmacologia , Pirazóis/farmacologia , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética , Recompensa , Rimonabanto , Comportamento Espacial/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA