Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 222
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Behav Brain Res ; 203(2): 232-9, 2009 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-19450625

RESUMO

The potential neuroleptic-like effect of ampullosporin A, a new peptaibol, isolated from the fungus Sepedonium ampullosporum HKI-0053, was characterized using specific behavioural models and methods. Ampullosporin A (amp) disrupted the retrieval of a well-trained conditioned reaction and normalized the behavioural effects of subchronic ketamine treatment in the social interaction test in a dose which showed only inconsiderable side effects. The experiments demonstrated that the substance did not antagonize the apomorphine (apo) induced hyperactivity. On the other hand, the locomotor stimulation induced by the NMDA receptor antagonist MK-801 was nearly completely suppressed by ampullosporin A, supposing interactions with the glutamatergic system. Binding studies demonstrated no interaction with dopaminergic D(1) and D(2) receptors. However, amp can alter the activity of glutamate receptors. The results resemble characteristics of an atypical neuroleptic drug. But further experiments are necessary to validate the suggested neuroleptic-like activity.


Assuntos
Peptídeos/farmacologia , Animais , Antipsicóticos/efeitos adversos , Antipsicóticos/farmacologia , Ansiedade/induzido quimicamente , Apomorfina/farmacologia , Aprendizagem da Esquiva/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Encéfalo/metabolismo , Catalepsia/induzido quimicamente , Maleato de Dizocilpina/farmacologia , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Hipercinese/induzido quimicamente , Hipercinese/metabolismo , Masculino , Atividade Motora/efeitos dos fármacos , Peptaibols , Peptídeos/efeitos adversos , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Teste de Desempenho do Rota-Rod , Comportamento Social
2.
Neuropharmacology ; 56(4): 768-78, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19162047

RESUMO

Recent evidence suggests that opioid analgesia and tolerance can be modulated by metabotropic glutamate receptors. Therefore, we studied the functional coupling and desensitization of the micro-opioid receptor (MOR) in human embryonic kidney (HEK) 293 cells which co-express metabotropic glutamate receptor 5 (mGluR5). As demonstrated by the D-Ala2,N-MePhe4,Gl-ol5-enkephalin (DAMGO)-induced inhibition of intracellular cAMP level and by binding studies, the co-expression of mGluR5 had no substantial effect on the agonist binding sites and functional coupling of the MOR. However, in MOR/ mGluR5 co-expressing cells, the non-competitive mGluR5 antagonist MPEP (2-methyl-6-(phenyl-ethynyl)-pyridine) decreases the DAMGO-induced MOR phosphorylation, internalization, and desensitization, whereas non-selective competitive mGluR antagonists or agonists had no effects. These findings indicate that an allosteric modulation of mGluR5 can affect the agonist-induced MOR signalling and regulation. As a mechanistic basis for the observed effects we suggested an interaction/heterodimerization of MOR and mGluR5, which is supported by the DAMGO-induced co-internalization of MOR and mGluR5 and by the increase of MPEP binding sites (Bmax) and a change of the binding affinity (K(D)) of mGluR5 receptors after the co-expression of MOR. In addition, co-immunoprecipitation experiments revealed evidence for an interaction between MOR and mGluR5 which is facilitated by MPEP treatment.


Assuntos
Analgésicos Opioides/farmacologia , Receptores de Ácido Caínico/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Analgésicos Opioides/metabolismo , Western Blotting , Linhagem Celular , Células Cultivadas , AMP Cíclico/metabolismo , Endocitose/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Ligantes , Microscopia Confocal , Neurônios/efeitos dos fármacos , Fosforilação , Piridinas/farmacologia , Ensaio Radioligante , Receptores de Ácido Caínico/química , Transfecção
3.
Neuropharmacology ; 49(8): 1160-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16098996

RESUMO

The influence of opioid antagonists and of morphine on rat hippocampal slices in a model of reversible hypoxia/hypoglycemia was investigated by assessment of evoked field potentials (population spike amplitude). In control slices, a brief hypoxia/hypoglycemia led to a loss of field potentials followed by an impaired recovery (40-50% of baseline) during reperfusion. In contrast, restoration was significantly improved when the opioid receptor antagonists funaltrexamine (mu) or naltrindole (delta) were administered prior to and during hypoxia/hypoglycemia. In addition, recovery was improved in brain slices derived from mu-opioid receptor-deficient mice as compared to wild-type mice, indicating a deleterious role of endogenous opioids in hypoxia/hypoglycemia. Exogenous opiate exposure with morphine (0.1, 1.0, 10 microM) prior to hypoxia/hypoglycemia caused a slight concentration dependent increase of evoked field potentials. When morphine exposure was terminated after 1h and immediately followed by hypoxia/hypoglycemia, an impaired recovery of population spike amplitude was obtained, dependent on morphine concentration during preincubation. These results demonstrate that morphine aggravates neurotoxic effects of hypoxia/hypoglycemia. Conversely, when onset of hypoxia/hypoglycemia was delayed for 3h after morphine termination, a significantly improved recovery was observed. Similarly, in vivo administration of morphine 12h prior to slice preparation resulted in a dose dependent improved recovery of field potentials after hypoxia/hypoglycemia. These results provide evidence that preconditioning with morphine is able to induce neuroprotective effects.


Assuntos
Analgésicos Opioides/farmacologia , Hipocampo/fisiologia , Hipoglicemia/fisiopatologia , Hipóxia Encefálica/fisiopatologia , Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Animais , Química Encefálica/efeitos dos fármacos , Potenciais Evocados/efeitos dos fármacos , Técnicas In Vitro , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Fármacos Neuroprotetores , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/psicologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/patologia
4.
Brain Res ; 1043(1-2): 225-30, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15862537

RESUMO

Immunohistochemistry for delta-opioid receptor (DOR) was performed on the rat cranial sensory ganglia. The immunoreactivity was detected in 16%, 19% and 11% of neurons in the trigeminal, jugular and petrosal ganglia, respectively. The nodose ganglion was devoid of such neurons. DOR-immunoreactive (IR) neurons were mostly small to medium-sized (trigeminal, range = 62-851 microm(2), mean +/- SD = 359 +/- 175 microm(2); jugular, range = 120-854 microm(2), mean +/- SD = 409 +/- 196 microm(2); petrosal, range = 167-1146 microm(2), mean +/- SD = 423 +/- 233 microm(2)). Double immunofluorescence method revealed that all DOR-IR neurons were also immunoreactive for calcitonin gene-related peptide. The cutaneous and mucosal epithelia in the oro-facial region, tooth pulp, taste bud and carotid body were innervated by DOR-IR nerve fibers. In the brainstem, IR nerve terminals were located in the superficial medullary dorsal horn and dorsomedial part of the subnucleus oralis as well as the solitary tract nucleus. The present study suggests that DOR-IR neurons may be associated with nociceptive and/or chemoreceptive function in the cranial sensory ganglia.


Assuntos
Neurônios/metabolismo , Receptores Opioides delta/metabolismo , Gânglio Trigeminal/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Células Quimiorreceptoras/metabolismo , Imuno-Histoquímica , Masculino , Nociceptores/metabolismo , Gânglio Nodoso/citologia , Gânglio Nodoso/metabolismo , Ratos , Ratos Sprague-Dawley , Gânglio Trigeminal/citologia
5.
Neurosci Res ; 51(4): 445-51, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15740807

RESUMO

Immunohistochemistry for substance P, somatostatin and vanilloid receptor subtype 1 as well as receptors for somatostatin and opioids was performed on the trigeminal ganglion in wild-type and Brn-3a knockout mice at postnatal day 0. In wild-type mice, the trigeminal ganglion contained abundant substance P-, vanilloid receptor subtype 1-, sst2A receptor- and delta-opioid receptor-immunoreactive neurons, while the ganglion had only a few mu-opioid receptor-immunoreactive neurons. The Brn-3a deficiency had an effect on the cell size but not the number of substance P-immunoreactive neurons. In knockout mice, the proportion of small immunoreactive neurons markedly increased and that of medium- to large-sized immunoreactive ones correspondingly decreased (mean +/- S.D. = 54.7 +/- 29.1 microm2, range = 10.9-220.8 microm2) compared to wild-type mice (mean +/- S.D. = 116.6 +/- 58.6 microm2, range = 27.3-400.7 microm2). As for vanilloid receptor subtype 1-immunoreactive neurons, the number and cell size was barely affected by the deficiency. On the other hand, the loss of Brn-3a caused a decrease in the number of sst2A receptor- or delta-opioid receptor-immunoreactive neurons (more than 95% reduction) and an increase in the number of mu-opioid receptor-immunoreactive neurons (9.3-fold increase). Somatostatin-immunoreactive neurons were not detected in the trigeminal ganglion of wild-type or mutant mice at postnatal day 0. The present study suggests that Brn-3a deficiency may have effects on the survival of trigeminal nociceptors and their expression of some neurochemical substances.


Assuntos
Proteínas de Ligação a DNA/deficiência , Neurônios/metabolismo , Nociceptores/metabolismo , Fatores de Transcrição/deficiência , Gânglio Trigeminal/metabolismo , Animais , Contagem de Células , Tamanho Celular , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Receptores de Droga/metabolismo , Receptores da Neurocinina-1/metabolismo , Receptores Opioides/metabolismo , Receptores de Somatostatina/metabolismo , Canais de Cátion TRPV , Fator de Transcrição Brn-3 , Fator de Transcrição Brn-3A
6.
Naunyn Schmiedebergs Arch Pharmacol ; 369(3): 287-93, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14963640

RESUMO

The relevance of mu-opioid systems for central excitability and kindling related disturbed learning performance was underlined by investigations using mu-opioid receptor knockout mice. Mice lacking mu-opioid receptors showed an accelerated kindling development induced by the convulsant drug pentylenetetrazol. Blockade of delta-opioid receptors by naltrindole suppressing kindling development in wild-type animals led to a further acceleration of kindled seizure development in the knockout mice. Mice lacking mu-opioid receptors showed such a low learning performance in the shuttle box, that the kindling induced learning deficit as seen in wild-type mice was not detected. The results were discussed on the basis of receptor binding studies with regard to subtypes of glutamatergic receptors, delta-opioid and somatostatin receptors. An increase in glutamate and somatostatin binding could contribute to the enhanced excitability in the-mu-opioid receptor knockout mice.


Assuntos
Excitação Neurológica/metabolismo , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Ácido Glutâmico/metabolismo , Excitação Neurológica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Pentilenotetrazol/toxicidade , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Receptores Opioides mu/fisiologia
7.
Neuroscience ; 120(3): 807-13, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12895520

RESUMO

Immunohistochemistry for the somatostatin sst2A receptor was performed on the rat trigeminal ganglion to know its function in the trigeminal nervous system. The immunoreactivity was detected in 9.4% of primary sensory neurons in the ganglion. These neurons were small to medium-sized (range=106.5-1123.2 microm(2); mean+/-S.D.=506.3+/-213.2 microm(2)) and predominantly located in the rostromedial part of the ophthalmo-maxillary division. They were also immunoreactive for calcitonin gene-related peptide and the vanilloid receptor subtype 1. In addition, 13.7% of trigeminal neurons which were retrogradely traced with fluorogold from the nasal mucosa exhibited sst2A receptor-immmunoreactivity. Trigeminal neurons which innervated the facial skin and tooth pulp were devoid of the immunoreactivity. In the brainstem trigeminal sensory nuclear complex, both the neuronal cell body and the neuropil exhibited sst2A receptor-immunoreactivity in the superficial medullary dorsal horn.The present study indicates that sst2A receptor-immunoreactive trigeminal nociceptors innervate the nasal mucosa. They may project to the superficial laminae of the medullary dorsal horn.


Assuntos
Receptores de Somatostatina/análise , Gânglio Trigeminal/química , Animais , Tronco Encefálico/química , Imuno-Histoquímica , Masculino , Neurônios/química , Neurópilo/química , Ratos , Ratos Sprague-Dawley , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/análise , Receptores de Droga/análise
8.
Brain Res Mol Brain Res ; 104(2): 143-7, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12225868

RESUMO

Behavioral experiments demonstrate that stress alters the individual's attitude towards opiates. In search for the underlying neuronal mechanisms we investigated the influence of stress on morphine-induced c-fos expression in the brain, and, vice versa, the influence of morphine application on the brain's c-fos response to stress. In our experiments, mild stress was induced either by brief immobilization (1 min) or by exposing the rats to a noisy and unfamiliar environment. These kinds of stress, unlike severe stress, did not elicit c-fos expression in the paraventricular nucleus of the hypothalamus. However, c-fos expression was observed in the lateral septum, medial striatum, claustrum and in the cingulate and piriform cortices under these conditions. The stress-induced c-fos induction was markedly decreased by a moderate (10 mg/kg) dose of morphine. On the other hand, morphine alone (50 mg/kg) caused only a weak c-fos expression in nai;ve animals despite of the rather high dose. If, however, this morphine dose was applied in the presence of a stressful stimulus, a pronounced c-fos expression in the dorsal striatum resulted. This c-fos signal was comparable with the signal seen in morphine-sensitized animals. Thus, distressing conditions seem to alter the brain's response to morphine at the level of gene expression, and this could be important for initiating voluntary opiate intake.


Assuntos
Encéfalo/efeitos dos fármacos , Tolerância a Medicamentos/fisiologia , Morfina/farmacologia , Transtornos Relacionados ao Uso de Opioides/metabolismo , Estresse Fisiológico/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Ambiente Controlado , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ruído/efeitos adversos , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Restrição Física , Estresse Fisiológico/fisiopatologia , Síndrome de Abstinência a Substâncias/fisiopatologia
9.
Gut ; 50(1): 52-60, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11772967

RESUMO

BACKGROUND AND AIMS: Recently, novel somatostatin receptor (sstr) subtype specific ligand analogues have been developed for medical treatment of neuroendocrine tumours expressing different sstrs (sstr1-5). At present, individual expression patterns of sstr subtypes are based on methods such as in situ hybridisation and polymerase chain reaction at the transcriptional level. Therefore, we generated subtype specific antibodies against sstr1, 2A, 3, and 5 and analysed their presence, cellular localisation, distribution, and expression pattern in 33 gastrinomas, 36 insulinomas, and 35 tumours associated with a carcinoid syndrome by immunohistochemistry at the translational level. METHODS: Western blotting experiments were performed in the normal human pancreas used as a reference organ and in tumour tissues; at the cellular level, sstrs were localised by immunohistochemistry in tissue paraffin sections. RESULTS: In western blot analyses, the antibodies identified the respective receptors in their correct molecular range in extracts of the pancreas and neuroendocrine tumours. Using immunohistochemistry and immunofluorescence, the antibodies specifically detected the receptors in islet cells of the normal pancreas. Immunohistochemistry in the tumours revealed that all investigated sstr subtypes were highly expressed in the different tumour types. The frequency and expression pattern of the individual sstr subtypes varied considerably not only between the different tumour types but also in each patient. CONCLUSIONS: We conclude that immunohistochemistry with subtype specific antibodies can be used in clinical routine work to analyse sstr expression patterns for each patient before treatment and to facilitate well directed individual medical therapy by administering subtype specific somatostatin analogues.


Assuntos
Tumores Neuroendócrinos/imunologia , Receptores de Somatostatina/imunologia , Especificidade de Anticorpos , Western Blotting , Cromatografia Líquida de Alta Pressão , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Imunização
10.
Pharmacol Ther ; 91(3): 167-77, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11744066

RESUMO

People with a genetic predisposition for substance abuse have defects in genes for the opioid peptides and receptors. A high number of polymorphisms have been detected in the mu-opioid receptor, some of which result in pharmacological alterations. The opioid peptide proopiomelanocortin proved extraordinarily rich in mutations that often lead to severe phenotypical consequences. Prodynorphin displays a polymorphic regulation of transcription. Variants of the mu- and the delta-opioid receptor showed positive associations with opiate and/or alcohol addiction in some studies. However, these associations were weak, indicating a small contribution of the opioid system to these disorders.


Assuntos
Predisposição Genética para Doença , Polimorfismo Genético , Receptores Opioides delta/genética , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Transtornos Relacionados ao Uso de Substâncias/genética , Alelos , Encefalinas/genética , Encefalinas/farmacologia , Humanos , Precursores de Proteínas/genética , Precursores de Proteínas/farmacologia , Receptores Opioides delta/fisiologia , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/fisiologia , beta-Endorfina/genética , beta-Endorfina/farmacologia
11.
Brain Res Mol Brain Res ; 95(1-2): 129-37, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11687284

RESUMO

There is limited knowledge about the distribution of the different G-protein-coupled receptor kinases (GRKs) in the rat brain, especially for the recently cloned isoforms GRK5 and GRK6. In this work an overview will be given of the mRNA expression patterns of four G-protein-coupled receptor kinases, GRK2 (betaARK1), GRK3 (betaARK2), GRK5 and GRK6 in the rat brain. As now shown by us and recently by others GRK2 and GRK3 are widely distributed in rat brain with nearly the same expression pattern. But GRK3, in general, appeared to be weaker expressed than GRK2 in most brain areas. Exceptions were the islands of Calleja, the compact part of the substantia nigra and the locus coeruleus. GRK3 mRNA was very low expressed or absent in the striatum and in some hypothalamic and thalamic nuclei. The expression pattern of GRK6 was also similar to GRK2. In the caudate putamen GRK6 yielded the strongest hybridization signal of all GRK types. GRK5 took a special position. The message for this form was not expressed ubiquitously in the brain but was mainly localized in limbic brain regions with a very prominent expression in the lateral septal area. GRK5 may therefore be involved in reward and addiction. Accordingly, a higher expression level of GRK5 mRNA was found in the lateral septum of cocaine-sensitized rats as compared to controls.


Assuntos
Encéfalo/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Encéfalo/anatomia & histologia , Cocaína/farmacologia , Quinase 3 de Receptor Acoplado a Proteína G , Quinase 5 de Receptor Acoplado a Proteína G , Quinases de Receptores Acoplados a Proteína G , Hibridização In Situ , Masculino , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Quinases de Receptores Adrenérgicos beta
12.
J Biol Chem ; 276(47): 43901-8, 2001 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-11572871

RESUMO

Morphine and the endogenous opioid peptide beta-endorphin exert neuromodulatory as well as immunomodulatory effects, which are transduced by mu-opioid receptors. In this report we show that stimulation with interleukin-4 induces mu-opioid receptor transcripts in human primary blood cells (T cells and polymorphonuclear leukocytes), immune cell lines (Raji, U-937, and HMEC-1), and dendritic cells. In nonstimulated immune cells this gene is silent. In addition, mu receptor transcription is up-regulated by interleukin-4 in cultures of primary rat neurons. Transient transfection experiments in Raji and SH SY5Y neuronal cells with human and rat reporter gene constructs linked the interleukin-4 effect directly to cis-active mu receptor promoter elements located at nucleotide -997 on the human gene and nucleotide -727 on the rat gene. The interleukin-4 response elements function orientation independently. They bind STAT6 transcription factors as shown by electrophoretic mobility shift assays. In the human gene, a single nucleotide polymorphism within the interleukin-4 response element reduces the trans-activating potential of this element by 50%, which may affect the phenotype of persons carrying this variation. These findings provide a molecular basis for understanding bidirectional interactions between the opioid system and the immune system.


Assuntos
Alelos , Regulação da Expressão Gênica/efeitos dos fármacos , Variação Genética , Interleucina-4/farmacologia , Receptores Opioides mu/genética , Transativadores/genética , Transcrição Gênica/efeitos dos fármacos , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Primers do DNA , Humanos , Neurônios/metabolismo , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Fator de Transcrição STAT6 , Transativadores/metabolismo
13.
J Biol Chem ; 276(43): 39772-8, 2001 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-11514577

RESUMO

In this study we report that human phosphatidylethanolamine-binding protein (hPBP) facilitates heterotrimeric G protein-coupled signaling. In Xenopus laevis oocytes, coexpression of hPBP with human mu opioid receptor, human delta opioid receptor, or human somatostatin receptor 2 evoked an agonist-induced increase in potassium conductance of G protein-activated inwardly rectifying potassium channels. This activation of heterotrimeric G protein signaling in oocytes could also be elicited by injection of bacterially overexpressed and purified hPBP. Stimulatory effect was pertussis toxin-sensitive and present even in the absence of coexpressed receptors. Additionally, an increase in G protein-mediated inhibition of adenylate cyclase activity, measured by the inhibition of forskolin-mediated cAMP accumulation, could be detected in HEK293 and NIH3T3 cells after expression of hPBP and in Xenopus oocytes after injection of hPBP. As [(35)S]guanosine 5'-3-O-(thio)triphosphate (GTPgammaS) binding to membranes prepared from hPBP-expressing cells was significantly elevated and recombinant hPBP dose-dependently stimulated [(35)S]GTPgammaS binding to native membranes, the results presented provide strong evidence that hPBP-induced effects are G protein-dependent. These data suggest a novel function of hPBP in regulating G protein and G protein-coupled receptor signaling in vivo.


Assuntos
Proteína de Ligação a Androgênios , Proteínas de Transporte/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Cloretos/metabolismo , Sequência Conservada , AMP Cíclico/metabolismo , Condutividade Elétrica , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Oócitos , Proteína de Ligação a Fosfatidiletanolamina , Proteínas de Transferência de Fosfolipídeos , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/agonistas , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores Opioides delta , Receptores Opioides mu/metabolismo , Receptores de Somatostatina/metabolismo , Transdução de Sinais , Xenopus
14.
Naunyn Schmiedebergs Arch Pharmacol ; 363(5): 562-8, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11383718

RESUMO

Mice lacking D1 receptors were used to study the role of these receptors in morphine-induced antinociception and locomotor sensitisation. In the hot-plate test D1 receptor deficient (-/-) and wild-type (+/+) mice showed similar reaction times under basal conditions. A single injection of 1.25 mg/kg and 2.5 mg/kg morphine resulted in a stronger antinociceptive response in D1 receptor deficient mice than in wild-type animals. Tolerance to the analgesic effect did not develop in both groups of animals when 12.5 mg/kg morphine was chronically applied twice daily for 13 days. There was no change in basal locomotor activity between saline-injected wild-type and D1 receptor deficient mice. After chronic treatment wild-type mice showed a continuous increase in locomotor activity, indicating the development of sensitisation. In contrast, a subchronic administration of morphine did not change locomotor activity in mutant mice. The lack of the development of locomotor sensitisation in D1 deficient mice was associated with reduced levels of immunoreactive mu opioid receptors in dorsal striatal patches as compared to wild-type mice. In contrast, no change in the distribution of immunoreactive mu receptors could be detected in areas related to pain pathways such as the spinal cord. Taken together, these results suggest an involvement of D1 receptors in morphine-induced locomotor activity and analgesia.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Medição da Dor/efeitos dos fármacos , Receptores de Dopamina D1/deficiência , Receptores de Dopamina D1/genética , Animais , Corpo Estriado/metabolismo , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/fisiologia , Células do Corno Posterior/metabolismo , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Receptores de Dopamina D1/metabolismo
15.
J Biol Chem ; 276(33): 31408-14, 2001 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-11359768

RESUMO

The main analgesic effects of the opioid alkaloid morphine are mediated by the mu-opioid receptor. In contrast to endogenous opioid peptides, morphine activates the mu-opioid receptor without causing its rapid endocytosis. Recently, three novel C-terminal splice variants (MOR1C, MOR1D, and MOR1E) of the mouse mu-opioid receptor (MOR1) have been identified. In the present study, we show that these receptors differ substantially in their agonist-selective membrane trafficking. MOR1 and MOR1C stably expressed in human embryonic kidney 293 cells exhibited phosphorylation, internalization, and down-regulation in the presence of the opioid peptide [d-Ala(2),Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO) but not in response to morphine. In contrast, MOR1D and MOR1E exhibited robust phosphorylation, internalization, and down-regulation in response to both DAMGO and morphine. DAMGO elicited a similar desensitization (during an 8-h exposure) and resensitization (during a 50-min drug-free interval) of all four mu-receptor splice variants. After morphine treatment, however, MOR1 and MOR1C showed a faster desensitization and no resensitization as compared with MOR1D and MOR1E. These results strongly reinforce the hypothesis that receptor phosphorylation and internalization are required for opioid receptor reactivation thus counteracting agonist-induced desensitization. Our findings also suggest a mechanism by which cell- and tissue-specific C-terminal splicing of the mu-opioid receptor may significantly modulate the development of tolerance to the various effects of morphine.


Assuntos
Morfina/farmacologia , Receptores Opioides mu/metabolismo , Processamento Alternativo , Sequência de Aminoácidos , Animais , Western Blotting , Regulação para Baixo , Endocitose , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Humanos , Camundongos , Dados de Sequência Molecular , Fosforilação , Isoformas de Proteínas/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/química
16.
J Biol Chem ; 276(17): 14027-36, 2001 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-11134004

RESUMO

Several recent studies suggest that G protein-coupled receptors can assemble as heterodimers or hetero-oligomers with enhanced functional activity. However, inactivation of a fully functional receptor by heterodimerization has not been documented. Here we show that the somatostatin receptor (sst) subtypes sst(2A) and sst(3) exist as homodimers at the plasma membrane when expressed in human embryonic kidney 293 cells. Moreover, in coimmunoprecipitation studies using differentially epitope-tagged receptors, we provide direct evidence for heterodimerization of sst(2A) and sst(3). The sst(2A)-sst(3) heterodimer exhibited high affinity binding to somatostatin-14 and the sst(2)-selective ligand L-779,976 but not to the sst(3)-selective ligand L-796,778. Like the sst(2A) homodimer, the sst(2A)-sst(3) heterodimer stimulated guanosine 5'-3-O-(thio)triphosphate (GTPgammaS) binding, inhibition of adenylyl cyclase, and activation of extracellular signal-regulated kinases after exposure to the sst(2)-selective ligand L-779,976. However, unlike the sst(3) homodimer, the sst(2A)-sst(3) heterodimer did not promote GTPgammaS binding, adenylyl cyclase inhibition, or extracellular signal-regulated kinase activation in the presence of the sst(3)-selective ligand L-796,778. Interestingly, during prolonged somatostatin-14 exposure, the sst(2A)-sst(3) heterodimer desensitized at a slower rate than the sst(2A) and sst(3) homodimers. Both sst(2A) and sst(3) homodimers underwent agonist-induced endocytosis in the presence of somatostatin-14. In contrast, the sst(2A)-sst(3) heterodimer separated at the plasma membrane, and only sst(2A) but not sst(3) underwent agonist-induced endocytosis after exposure to somatostatin-14. Together, heterodimerization of sst(2A) and sst(3) results in a new receptor with a pharmacological and functional profile resembling that of the sst(2A) receptor, however with a greater resistance to agonist-induced desensitization. Thus, inactivation of sst(3) receptor function by heterodimerization with sst(2A) or possibly other G protein-coupled receptors may explain some of the difficulties in detecting sst(3)-specific binding and signaling in mammalian tissues.


Assuntos
Amidas/química , Indóis , Ligantes , Receptores de Somatostatina/química , Adenilil Ciclases/metabolismo , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , Dimerização , Relação Dose-Resposta a Droga , Endocitose , Ativação Enzimática , Epitopos/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Imuno-Histoquímica , Concentração Inibidora 50 , Cinética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Testes de Precipitina , Ligação Proteica , Transdução de Sinais , Somatostatina/química , Somatostatina/metabolismo , Transfecção
17.
J Physiol Paris ; 94(3-4): 199-204, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11087997

RESUMO

The diverse biological effects of the hormone somatostatin are mediated by five genetic different receptor subtypes (sst1-sst5), which belong to the superfamily of G-protein coupled receptors with seven transmembrane domains. The sst2 subtype is unique among the somatostatin receptors in its structure, since it is expressed in two protein variants which differ within their carboxy-terminal ends, generated by alternative splicing. Within the 5' untranslated region of the gene two introns separate three transcriptional units with distinct promoters. Due to the latter feature, the sst2 gene is also unique among all somatostatin receptor genes regarding its transcriptional regulation. The three alternative promoters are tissue specifically active and show alternative responsiveness to extracellular signals. The second sst2 promoter is important for expression of the gene in tissues where somatostatin has essential physiological functions, such as brain, pituitary and gastrointestinal tissues. Furthermore, it contains cis-acting regulatory elements involved in the transcriptional response to elevated cyclic AMP levels and glucocorticoids.


Assuntos
Regulação da Expressão Gênica , Regiões Promotoras Genéticas/fisiologia , Receptores de Somatostatina/genética , Animais , Sequência de Bases , AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Glucocorticoides/metabolismo , Camundongos , Sítios de Splice de RNA/genética , Receptores de Somatostatina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional/fisiologia
18.
J Physiol Paris ; 94(3-4): 259-64, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11088003

RESUMO

The cloning of five members of the somatostatin receptor family, sst1-sst5, as well as two isoforms of the somatostatin receptor 2, sst2A and sst2B, enabled us to generate specific anti-peptide antisera against unique sequences in the carboxyl-terminal tail of each somatostatin receptor subtype. We used these antibodies in multicolor immunofluorescent studies aimed to examine the regional and subcellular distribution of somatostatin receptors in adult rat brain. Several findings are notable: The cloned sst1 receptor is primarily localized to axons, and therefore most likely functions in a presynaptic manner. The cloned sst2 receptor isoforms exhibit strikingly different distributions, however, both sst2A and sst2B are confined to the plasma membrane of neuronal somata and dendrites, and therefore most likely function in a postsynaptic manner. The cloned sst3 receptor appears to be excluded from 'classical' pre- or postsynaptic sites but is selectively targeted to neuronal cilia. The cloned sst4 receptor is preferentially distributed to distal dendrites, and therefore most likely functions postsynaptically. The cloned sst5 receptor was not detectable in the adult rat brain, however, prominent sst5 expression was found in the pituitary. Furthermore, sst1-containing axons either co-contained somatostatin or were closely apposed by somatostatin-positive terminals in a regional-specific manner. Neuronal somata and dendrites containing either sst2A, sst2B or sst4 were found to exist in close proximity, although not necessarily synaptically linked, to somatostatin-positive terminals. Together, in the central nervous system the effects of somatostatin are mediated by several different receptor proteins which are distributed with considerable regional overlap. However, there appears to be a high degree of specialization among somatostatin receptor subtypes with regard to their subcellular targeting. This subtype-selective targeting may be the underlying principal of organization that allows somatostatinergic modulation of neuronal activity via both pre- and postsynaptic mechanisms.


Assuntos
Especificidade de Anticorpos , Química Encefálica , Receptores de Somatostatina/análise , Receptores de Somatostatina/imunologia , Animais , Proteínas de Membrana , Ratos
19.
FEBS Lett ; 480(2-3): 156-60, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11034319

RESUMO

delta Opioid receptors were identified in human melanomas by RT-PCR and radioligand binding. In all tumors an additional PCR amplificate was detected in which 144 bp within the third exon were deleted. This fragment corresponded to the third cytoplasmic domain of the receptor protein. The short variant resulted from atypical mRNA processing. There were no common splice recognition sequences around the deleted fragment; instead its excision resembled the removal of a transposon. The deletion was not detected in normal human melanocytes nor in human or rat brain. However, it was present in a human neuroblastoma cell line (SH-SY5Y). Thus, it appears that the occurrence of the short delta opioid receptor is correlated to malignancy.


Assuntos
Neoplasias/metabolismo , Processamento Pós-Transcricional do RNA , Receptores Opioides delta/genética , Animais , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Citoplasma/metabolismo , DNA Complementar , Humanos , Melanócitos/citologia , Melanoma , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/metabolismo , Pigmentos Biológicos , RNA Mensageiro , Ratos , Receptores Opioides delta/metabolismo , Células Tumorais Cultivadas
20.
Naunyn Schmiedebergs Arch Pharmacol ; 361(6): 584-9, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10882032

RESUMO

Morphine-induced place preference was demonstrated recently in wild-type mice, whereas this conditioned behaviour was not observed in mu-opioid receptor-deficient mice. In the present study, we investigated locomotor effects of subcutaneously (s.c.) injected morphine as well as intracerebroventricular (i.c.v.) morphine self-administration in mu-opioid receptor-knockout mice. After s.c. morphine injection, locomotor activity significantly increased in wild-type animals. As expected, in the self-administration test the rate of self-administration constantly increased in wild-type mice reflecting reward effects of morphine. This increase was independent of locomotor/motor activity. In contrast, self-administration rates and locomotor/motor activity significantly decreased in the receptor-deficient animals. It was shown that this aversive effect might partly be due to kappa-opioid receptor interaction.


Assuntos
Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Análise de Variância , Animais , Autorradiografia , Corpo Estriado/metabolismo , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Comportamento Exploratório/efeitos dos fármacos , Hipocampo/metabolismo , Injeções Intraventriculares , Camundongos , Camundongos Knockout , Morfina/administração & dosagem , Ensaio Radioligante , Distribuição Aleatória , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/genética , Autoadministração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...