Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 141(3): 1489-501, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16750886

RESUMO

Examples of spontaneous oscillating neural activity contributing to both pathological and physiological states are abundant throughout the CNS. Here we report a spontaneous oscillating intermittent synaptic current located in lamina I of the neonatal rat spinal cord dorsal horn. The spontaneous oscillating intermittent synaptic current is characterized by its large amplitude, slow decay time, and low-frequency. We demonstrate that post-synaptic N-methyl-D-aspartate receptors (NMDARs) mediate the spontaneous oscillating intermittent synaptic current, as it is inhibited by magnesium, bath-applied d-2-amino-5-phosphonovalerate (APV), or intracellular MK-801. The NR2B subunit of the NMDAR appears important to this phenomenon, as the NR2B subunit selective NMDAR antagonist, alpha-(4-hydroxphenyl)-beta-methyl-4-benzyl-1-piperidineethanol tartrate (ifenprodil), also partially inhibited the spontaneous oscillating intermittent synaptic current. Inhibition of spontaneous glutamate release by the AMPA/kainate receptor antagonist 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) or the mu-opioid receptor agonist [D-Ala2, N-Me-Phe4, Gly5] enkephalin-ol (DAMGO) inhibited the spontaneous oscillating intermittent synaptic current frequency. Marked inhibition of spontaneous oscillating intermittent synaptic current frequency by tetrodotoxin (TTX), but not post-synaptic N-(2,6-dimethylphenylcarbamoylmethyl)triethylammonium bromide (QX-314), suggests that the glutamate release important to the spontaneous oscillating intermittent synaptic current is dependent on active neural processes. Conversely, increasing dorsal horn synaptic glutamate release by GABAA or glycine inhibition increased spontaneous oscillating intermittent synaptic current frequency. Moreover, inhibiting glutamate transporters with threo-beta-benzyloxyaspartic acid (DL-TBOA) increased spontaneous oscillating intermittent synaptic current frequency and decay time. A possible functional role of this spontaneous NMDAR-mediated excitatory postsynaptic current in modulating nociceptive transmission within the spinal cord is discussed.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Células do Corno Posterior/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Medula Espinal/citologia , Antagonistas Adrenérgicos alfa/farmacologia , Analgésicos Opioides/farmacologia , Animais , Animais Recém-Nascidos , Ácido Aspártico/farmacologia , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Técnicas In Vitro , Lidocaína/análogos & derivados , Lidocaína/farmacologia , Magnésio/farmacologia , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Inibição Neural/efeitos da radiação , Técnicas de Patch-Clamp/métodos , Piperidinas/farmacologia , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/efeitos da radiação , Ratos , Ratos Sprague-Dawley , Tetrodotoxina/farmacologia
2.
J Neurophysiol ; 85(2): 485-94, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11160487

RESUMO

Long-term potentiation (LTP) involves a prolonged increase in neuronal excitability following repeated afferent input. This phenomenon has been extensively studied in the hippocampus as a model of learning and memory. Similar long-term increases in neuronal responses have been reported in the dorsal horn of the spinal cord following intense primary afferent stimulation. In these studies, we utilized the spinal cord slice preparation to examine effects of the potently antinociceptive mu opioids in modulating primary afferent/dorsal horn neurotransmission as well as LTP of such transmission. Transverse slices were made from the lumbar spinal cord of 10- to 17-day-old rats, placed in a recording chamber, and perfused with artificial cerebrospinal fluid also containing bicuculline (10 microM) and strychnine (1 microM). Primary afferent activation was achieved in the spinal slice by electrical stimulation of the dorsal root (DR) or the tract of Lissauer (LT) which is known to contain a high percentage of small diameter fibers likely to transmit nociception. Consistent with this anatomy, response latencies of LT-evoked field potentials in the dorsal horn were considerably slower than the response latencies of DR-evoked potentials. Only LT-evoked field potentials were found to be reliably inhibited by the mu opioid receptor agonist [D-Ala(2), N-Me-Phe(4), Gly(5)] enkephalin-ol (DAMGO, 1 microM), although evoked potentials from both DR and LT were blocked by the AMPA/kainate glutamate receptor antagonist 6-cyano-7-nitroquinoxalene-2,3-dione. Moreover repeated stimulation of LT produced LTP of LT- but not DR-evoked potentials. In contrast, repeated stimulation of DR showed no reliable LTP. LTP of LT-evoked potentials depended on N-methyl-D-aspartate (NMDA) receptor activity, in that it was attenuated by the NMDA antagonist APV. Moreover, such LTP was inhibited by DAMGO interfering with LTP induction mechanisms. Finally, in whole cell voltage-clamp studies of Lamina I neurons, DAMGO inhibited excitatory postsynaptic current (EPSC) response amplitudes from LT stimulation-evoked excitatory amino acid release but not from glutamate puffed onto the cell and increased paired-pulse facilitation of EPSCs evoked by LT stimulation. These studies suggest that mu opioids exert their inhibitory effects presynaptically, likely through the inhibition of glutamate release from primary afferent terminals, and thereby inhibit the induction of LTP in the spinal dorsal horn.


Assuntos
Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Receptores Opioides mu/agonistas , Medula Espinal/fisiologia , Animais , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Gânglios Espinais/fisiologia , Técnicas In Vitro , Inibição Neural/fisiologia , Terminações Pré-Sinápticas/fisiologia , Ratos
4.
J Biol Chem ; 275(33): 25082-8, 2000 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-10833508

RESUMO

G protein-activated inwardly rectifying potassium channels (Kir3) are widely expressed throughout the brain, and regulation of their activity modifies neuronal excitability and synaptic transmission. In this study, we show that the neurotrophin brain-derived neurotrophic factor (BDNF), through activation of TrkB receptors, strongly inhibited the basal activity of Kir3. This inhibition was subunit dependent as functional homomeric channels of either Kir3.1 or Kir3.4 were significantly inhibited, whereas homomeric channels composed of Kir3.2 were insensitive. The general tyrosine kinase inhibitors genistein, Gö 6976, and K252a but not the serine/threonine kinase inhibitor staurosporine blocked the BDNF-induced inhibition of the channel. BDNF was also found to directly stimulate channel phosphorylation because Kir3.1 immunoprecipitated from BDNF-stimulated cells showed enhanced labeling by anti-phosphotyrosine-specific antibodies. The BDNF effect required specific tyrosine residues in the amino terminus of Kir3.1 and Kir3.4 channels. Mutations of either Tyr-12, Tyr-67, or both in Kir3.1 or mutation of either Tyr-32, Tyr-53, or both of Kir3. 4 channels to phenylalanine significantly blocked the BDNF-induced inhibition. The insensitive Kir3.2 was made sensitive to BDNF by adding a tyrosine (D41Y) and a lysine (P32K) upstream to generate a phosphorylation site motif analogous to that present in Kir3.4. These results suggest that neurotrophin activation of TrkB receptors may physiologically control neuronal excitability by direct tyrosine phosphorylation of the Kir3.1 and Kir3.4 subunits of G protein-gated inwardly rectifying potassium channels.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/metabolismo , Receptor trkB/metabolismo , Tirosina/metabolismo , Sequência de Aminoácidos , Animais , Carbazóis/farmacologia , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Eletrofisiologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Genisteína/farmacologia , Alcaloides Indólicos , Indóis/farmacologia , Lisina/química , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Fosforilação , Canais de Potássio/química , Testes de Precipitina , RNA Mensageiro/metabolismo , Estaurosporina/farmacologia , Xenopus
5.
J Neurosci ; 20(12): 4379-88, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10844006

RESUMO

kappa opioid receptor activation inhibits granule cell-mediated excitatory neurotransmission in the hippocampal formation via a decrease in glutamate release from both perforant path and mossy fiber terminals. We now report a third, anatomically and pharmacologically distinct site of such kappa opioid inhibition within the hippocampus. Granule cell population responses to selective stimulation of an excitatory hilar pathway were decreased by the kappa(1) opioid receptor agonist U69,593, an effect blocked by the kappa(1) antagonist norbinaltorphimine. U69,593 also inhibited hilar path induced long-term potentiation (LTP) of granule cell responses. LTP in this pathway was also blocked by the NMDA receptor antagonist d-2-amino-5-phosphonovalerate, unlike granule cell mossy fiber LTP in CA3. The kappa opioid peptide dynorphin is present in hilar mossy fiber collaterals. Ultrastructural analysis of these collaterals demonstrated dynorphin-containing vesicles in asymmetric synapses formed between axon terminals and granule cell dendrites, suggesting direct granule cell-granule cell connections. Evoked release of endogenous dynorphin within the hilus was effective in reducing hilar excitation of granule cells, although this release, in contrast to the release of dynorphin in the dentate molecular layer, was not dependent on L-type calcium channels. No hilar path excitation was observed in the absence of bicuculline, suggesting a strong GABA(A)-mediated inhibition of this pathway. However, hilar path activity could be seen after LTP, with or without bicuculline. Thus, kappa opioids can inhibit granule cell recurrent excitation, likely via effects on excitatory mossy fiber collaterals. Such collaterals are thought to be important in mediating temporal lobe epilepsy.


Assuntos
Benzenoacetamidas , Giro Denteado/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Neurônios/fisiologia , Pirrolidinas/farmacologia , Sinapses/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , Analgésicos/farmacologia , Animais , Axônios/efeitos dos fármacos , Axônios/fisiologia , Dendritos/efeitos dos fármacos , Dendritos/fisiologia , Giro Denteado/efeitos dos fármacos , Dinorfinas/análise , Dinorfinas/fisiologia , Cobaias , Técnicas In Vitro , Masculino , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Receptores de GABA-A/fisiologia , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inibidores , Sinapses/efeitos dos fármacos
7.
J Pharmacol Exp Ther ; 284(3): 1147-55, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9495877

RESUMO

The effects of kappa opioids on seizures and seizure-induced histopathology were investigated with the pilocarpine model of temporal lobe epilepsy. Rats treated with the kappa opioid receptor agonist U50488h before pilocarpine showed: 1) increased seizure latency; 2) decreased seizure duration; 3) decreased mossy fiber sprouting; and 4) increased hilar neuron survival when compared with rats pretreated with saline. Behavioral effects of U50488h were blocked by the kappa opioid receptor antagonist norbinaltorphimine (nBNI), whereas the changes caused by U50488h in the histological response to pilocarpine were not blocked by nBNI. Rats treated with nBNI before pilocarpine exhibited: 1) increased incidence of seizures; 2) increased mossy fiber sprouting; and 3) increased hilar neuron loss when compared with rats treated with pilocarpine alone. These changes suggest a protective role of endogenously released kappa opioids in this seizure model. The location of functional kappa opioid receptors in the rat dentate gyrus was documented electrophysiologically to enable correlation with kappa opioid effects on histopathology. The kappa selective agonist, U69593, reversibly decreased the amplitude of excitatory postsynaptic potentials in the middle molecular layer of the dentate gyrus from the ventral but not the more dorsal portion of the hippocampal formation. Thus, kappa opioids decreased the severity and incidence of behavioral seizures and secondarily decreased seizure-induced histopathology via the decreased incidence of seizures.


Assuntos
(trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Anticonvulsivantes/farmacologia , Pilocarpina/farmacologia , Receptores Opioides kappa/fisiologia , Convulsões/prevenção & controle , Animais , Comportamento Animal/efeitos dos fármacos , Giro Denteado/efeitos dos fármacos , Giro Denteado/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Opioides kappa/agonistas , Convulsões/etiologia , Convulsões/patologia
8.
J Neurophysiol ; 78(4): 1860-8, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9325355

RESUMO

Temporal lobe epilepsy is associated with a characteristic pattern of synaptic reorganization in the hippocampal formation, consisting of neuronal loss and aberrant growth of mossy fiber collaterals into the dentate gyrus inner molecular layer. We have used the rat pilocarpine model of temporal lobe epilepsy to study the functional consequences of mossy fiber sprouting on excitatory activity and kappa-opioid receptor-mediated inhibition. Using the whole cell voltage-clamp technique, we found that abnormal excitatory activity was evident in granule cells of the dentate gyrus from pilocarpine-treated rats. The frequency of spontaneous excitatory postsynaptic currents (EPSCs) was increased greatly in cells from tissue in which significant mossy fiber sprouting had developed. In the presence of bicuculline, giant spontaneous EPSCs, with large amplitudes and long durations, were seen only in association with mossy fiber sprouting. Giant EPSCs also could be evoked by low-intensity stimulation of the perforant path. Mossy fibers release not only excitatory amino acids, but also opioid peptides. kappa-Opioid receptor-mediated inhibition in normal Sprague-Dawley rats was seen only in hippocampal sections from the ventral pole. In pilocarpine-treated rats, however, kappa receptor-mediated effects were seen in both ventral and more dorsal sections. Thus in this model of temporal lobe epilepsy, several types of abnormal excitatory activity were observed, thereby supporting the idea that mossy fiber sprouting leads to recurrent excitatory connections. At the same time, inhibition of excitatory activity by kappa-opioid receptors was increased, perhaps representing an endogenous anticonvulsant mechanism.


Assuntos
Giro Denteado/efeitos dos fármacos , Epilepsia do Lobo Temporal/induzido quimicamente , Pilocarpina/farmacologia , Receptores Opioides kappa/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley
9.
J Pharmacol Exp Ther ; 281(1): 123-8, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9103488

RESUMO

We investigated whether chronic, in vivo administration of U50,488H, a kappa-1 opioid agonist, caused the development of tolerance to both the electrophysiological effects of applied kappa opioids and endogenously released dynorphins. In hippocampal slices from drug-naive guinea pigs, application of U69,593, a kappa-1 agonist, produced a concentration-dependent inhibition (EC50 = 20 nM) of the amplitude of the granule cell population response in the dentate gyrus. In slices from chronically U50,488H-treated animals, the concentration-response curve for U69,593 was shifted 3-fold to the right (EC50 = 59 nM), with a significant decrease in the maximal effect of U69,593. We also found that the effects of endogenously released dynorphins were significantly attenuated by chronic U50,488H treatment. There was no cross-tolerance between kappa and mu opioid receptor agonists as measured with the in vitro electrophysiological assay, and the noncompetitive N-methyl-D-aspartate receptor antagonist MK801 did not prevent the development of tolerance to either the electrophysiological effects or the hypothermic effects of kappa opioids. Our study demonstrates that receptor-selective tolerance to the kappa opioid actions in the guinea pig hippocampus does develop after chronic U50,488H treatment; but, unlike the mechanisms reported to underlie tolerance to kappa opioid analgesia, the inhibitory effects in the hippocampus did not depend on activation of N-methyl-D-aspartate receptors.


Assuntos
Benzenoacetamidas , Hipocampo/efeitos dos fármacos , Receptores Opioides kappa/efeitos dos fármacos , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida , Animais , Maleato de Dizocilpina/farmacologia , Tolerância a Medicamentos , Cobaias , Hipocampo/fisiologia , Técnicas In Vitro , Masculino , Pirrolidinas/farmacologia , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores Opioides mu/efeitos dos fármacos
10.
Anesth Analg ; 82(5): 1043-8, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8610865

RESUMO

Although patient-controlled analgesia (PCA) pumps have been in use for more than a decade, the optimal PCA analgesic has yet to be identified. Many drugs are used; however, morphine remains the "gold standard" of opioid analgesics worldwide. The present study evaluated morphine and hydromorphone (Dilaudid) PCA with respect to analgesic efficacy, side effects, mood, and cognitive function. Sixty-one opioid naive patients undergoing lower abdominal surgery participated in the double-blind protocol. Verbal rating scores, use of medication, and side effects for the two medications were recorded. Cognitive functioning was assessed by computation of Digit Symbol and Trails Making B Tests. Self-reported affective state (mood) was measured by Profile of Mood States (POMS) inventory. Both medications provided adequate analgesia without a difference in side effects. Cognitive performance was poorer in the hydromorphone group (P < 0.05). Patients receiving hydromorphone reported less anger/hostility (P < 0.01) and generally better mood elevations on the other subscales than those receiving morphine. A similar incidence of side effects and dose medication can be anticipated with morphine and hydromorphone. When considering cognitive effects, morphine had less adverse consequences, while hydromorphone appeared to result in improved mood. We conclude that hydromorphone may provide a suitable alternative to morphine.


Assuntos
Analgesia Controlada pelo Paciente , Analgésicos Opioides/administração & dosagem , Hidromorfona/administração & dosagem , Morfina/administração & dosagem , Abdome/cirurgia , Adolescente , Adulto , Afeto/efeitos dos fármacos , Idoso , Analgesia Controlada pelo Paciente/instrumentação , Analgésicos Opioides/efeitos adversos , Ira , Atenção/efeitos dos fármacos , Cognição/efeitos dos fármacos , Método Duplo-Cego , Feminino , Hostilidade , Humanos , Hidromorfona/efeitos adversos , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Pessoa de Meia-Idade , Morfina/efeitos adversos , Destreza Motora/efeitos dos fármacos , Dor Pós-Operatória/prevenção & controle , Desempenho Psicomotor/efeitos dos fármacos
11.
Neuron ; 14(6): 1265-72, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7605635

RESUMO

Granule cells in the guinea pig dentate gyrus release kappa opioid neuropeptides, dynorphins, from dendrites as well as from axon terminals. We have found that both L- and N-type calcium channel antagonists inhibited dendritic dynorphin release. In contrast, N-type but not L-type calcium channel antagonists inhibited axonal dynorphin release. Neither L- nor N-type channel antagonists directly altered the effects of kappa opioid receptor activation. By inhibiting dynorphin release, L-type channel antagonists also facilitated the induction of long-term potentiation of the perforant path-granule cell synapse. These studies establish that a single cell type can release a transmitter from two different cellular domains and provide new distinction between axonal and dendritic transmitter release mechanisms.


Assuntos
Axônios/metabolismo , Canais de Cálcio/fisiologia , Dendritos/metabolismo , Dinorfinas/metabolismo , Hipocampo/ultraestrutura , Animais , Axônios/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Dendritos/efeitos dos fármacos , Cobaias , Hipocampo/fisiologia , Isradipino/farmacologia , Potenciação de Longa Duração , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Nifedipino/farmacologia , Peptídeos/farmacologia , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/fisiologia , ômega-Conotoxina GVIA
12.
J Neurophysiol ; 72(4): 1697-705, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7823095

RESUMO

1. Activation of kappa 1-opioid receptors inhibits excitatory transmission in the hippocampal dentate gyrus of the guinea pig. The present studies used both anatomic and physiological approaches to distinguish between a pre- and postsynaptic localization of these receptors. 2. The entorhinal cortex was lesioned unilaterally to cause degeneration of perforant path afferents to the dentate molecular layer, and kappa 1-opioid binding sites were measured by labeling with the selective agonist, [3H]-U69593. Binding density was reduced significantly in the dentate gyrus molecular layer ipsilateral to the lesion compared with the contralateral molecular layer and with sham-lesioned controls. 3. Paired-pulse facilitation is a neurophysiologic paradigm that has been used to differentiate pre- and postsynaptic sites of action for agents that inhibit excitatory neurotransmission. U69593 reduced the amplitude of single population spikes and increased the degree of paired pulse facilitation. The potentiation of paired-pulse facilitation was maintained when the stimulation intensity was increased to compensate for the inhibition of excitatory transmission. These effects of kappa 1-receptor activation were similar to those seen after presynaptic inhibition of excitatory neurotransmitter release and support the hypothesis that U69593 presynaptically inhibits excitatory amino acid release in the dentate gyrus. 4. Local application of glutamate by pressure ejection in the dentate molecular layer evoked field excitatory postsynaptic potentials that mimicked those evoked by electrical stimulation of the perforant path. Both responses were sensitive to the non-N-methyl-D-aspartate glutamate receptor antagonist, 6-cyano-7-nitroquinoxaline-2,3-dione. U69593 inhibited responses evoked by perforant path stimulation but had no effect on responses evoked by glutamate application.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Benzenoacetamidas , Ácido Glutâmico/metabolismo , Hipocampo/fisiologia , Inibição Neural/fisiologia , Receptores Opioides kappa/fisiologia , Transmissão Sináptica/fisiologia , Vias Aferentes/fisiologia , Analgésicos/farmacocinética , Animais , Autorradiografia , Técnicas de Cultura , Dominância Cerebral/fisiologia , Cobaias , Masculino , Pirrolidinas/farmacocinética
13.
J Neurosci ; 14(8): 4740-7, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7913954

RESUMO

NMDA receptor-mediated long-term potentiation (LTP) of dentate granule cell responses to perforant path stimulation was inhibited by the kappa 1 opioid receptor agonist U69,593. This inhibition was reversed stereospecifically by naloxone and blocked by the selective kappa 1 antagonist norbinaltorphimine (NBNI). NBNI, by itself, had no effect on LTP induced by threshold stimulation but significantly enhanced LTP from more prolonged stimulation. This effect of NBNI suggests that endogenous opioids can regulate LTP in the dentate gyrus. In support of this hypothesis, stimulation of dynorphin-containing fibers also blocked LTP production in an NBNI-sensitive manner. Finally, dynorphin-mediated inhibition of LTP acts primarily on mechanisms of induction rather than maintenance or expression, since dynorphin released immediately before, but not immediately after, perforant path stimulation blocked LTP. Thus, exogenous and endogenous kappa opioids can inhibit induction of long-term potentiation at the perforant path-granule cell synapse and may therefore regulate plastic changes in synaptic transmission in a brain region thought to play an important role in processes of both learning and memory and epileptogenesis.


Assuntos
Benzenoacetamidas , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Receptores Opioides kappa/fisiologia , Animais , Dinorfinas/farmacologia , Estimulação Elétrica , Cobaias , Hipocampo/efeitos dos fármacos , Masculino , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Pirrolidinas/farmacologia , Receptores Opioides kappa/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos
14.
J Neurosci ; 14(6): 3736-50, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7911518

RESUMO

The granule cell population response to perforant path stimulation decreased significantly within seconds following release of endogenous dynorphin from dentate granule cells. The depression was blocked by the opioid receptor antagonists naloxone and norbinaltorphimine, suggesting that the effect was mediated by dynorphin activation of kappa 1 type opioid receptors. Pharmacological application of dynorphin B in the molecular layer was effective at reducing excitatory synaptic transmission from the perforant path, but application in the hilus had no significant effect. These results suggest that endogenous dynorphin peptides may be released from a local source within the dentate molecular layer. By light microscopy, dynorphin-like immunoreactivity (dynorphin-LI) was primarily found in granule cell axons in the hilus and stratum lucidum with only a few scattered fibers evident in the molecular layer. At the extreme ventral pole of the hippocampus, a diffuse band of varicose processes was also seen in the molecular layer, but this band was not present in more dorsal sections similar to those used for the electrophysiological studies. Electron microscopic analysis of the molecular layer midway along the septotemporal axis revealed that dynorphin-LI was present in dense-core vesicles in both spiny dendrites and unmyelinated axons with the majority (74%) of the dynorphin-LI-containing dense-core vesicles found in dendrites. Neuronal processes containing dynorphin-LI were observed throughout the molecular layer. The results suggest that dynorphin release from granule cell processes in the molecular layer regulates excitatory inputs entering the hippocampus from cerebral cortex, thus potentially counteracting such excitation-induced phenomena as epileptogenesis or long-term potentiation.


Assuntos
Dinorfinas/fisiologia , Endorfinas/fisiologia , Granulócitos/metabolismo , Hipocampo/metabolismo , Inibição Neural/fisiologia , Neurotransmissores/fisiologia , Animais , Dinorfinas/metabolismo , Endorfinas/metabolismo , Cobaias , Hipocampo/citologia , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Fatores de Tempo , Distribuição Tecidual
15.
Nature ; 363(6428): 451-4, 1993 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-8099201

RESUMO

Although anatomical and neurochemical studies suggest that endogenous opioids act as neurotransmitters, their roles in normal and pathophysiological regulation of synaptic transmission are not defined. Here we examine the actions of prodynorphin-derived opioid peptides in the guinea-pig hippocampus and show that physiological stimulation of the dynorphin-containing dentate granule cells can release endogenous dynorphins, which then activate kappa 1 opioid receptors present in the molecular layer of the dentate gyrus. Activation of kappa 1 receptors by either pharmacologically applied agonist or endogenously released peptide reduces excitatory transmission in the dentate gyrus, as shown by a reduction in the excitatory postsynaptic currents evoked by stimulation of the perforant path, a principal excitatory afferent. In addition, released dynorphin peptides were found to block the induction of long-term potentiation (LTP) at the granule cell-perforant path synapse. The results indicate that endogenous dynorphins function in this hippocampal circuit as retrograde, inhibitory neurotransmitters.


Assuntos
Benzenoacetamidas , Dinorfinas/fisiologia , Hipocampo/fisiologia , Neurotransmissores/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Glutamatos/metabolismo , Ácido Glutâmico , Cobaias , Técnicas In Vitro , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Inibição Neural/fisiologia , Pirrolidinas/farmacologia , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Fatores de Tempo
16.
Clin J Pain ; 8(3): 255-9, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1421740

RESUMO

OBJECTIVE: We report the case of a woman with presumed cancer pain treated with escalating doses of opiates despite no evident improvement in her pain and several deleterious side effects. PATIENT: A 62-year-old woman with cervical myelopathy and a diagnosis of a spinal cord tumor was referred to the University of Washington Medical Center complaining of chest tightness, multiple joint pains, nausea, constipation, seizures and a deteriorating memory. At the time of admission she was confined to her bed with a full-time attendant and was receiving 240 milligrams of intravenous morphine per hour for her pain. INTERVENTION: Diagnostic studies failed to find any evidence of neoplasm and revealed only an old hemorrhage within the cervical spinal cord. A program of increasing physical and occupational therapy and decreasing opiate intake was initiated. RESULTS: Within a month the patient's pain complaints decreased, as did the rest of her presenting complaints. Her activities of daily living greatly increased making attendant care no longer necessary. CONCLUSIONS: This case report illustrates some of the hazards of opioid therapy in the management of patients with chronic pain. Our patient's opiate therapy was expensive, gave her undesirable side effects, and did not reduce her pain complaints or improve her function. In the treatment of chronic pain, of noncancerous or cancerous origin, a) systemic opioids may not be effective in reducing pain complaints in every patient, b) treatment efficacy evaluation should always include functional endpoints, and c) nonefficacious treatments should not be continued indefinitely.


Assuntos
Entorpecentes/uso terapêutico , Neoplasias/complicações , Dor/tratamento farmacológico , Doença Crônica , Feminino , Hemorragia/complicações , Humanos , Pessoa de Meia-Idade , Entorpecentes/efeitos adversos , Dor/complicações , Manejo da Dor , Medula Espinal/patologia
17.
Brain Behav Immun ; 1(4): 318-28, 1987 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-3453207

RESUMO

We previously reported that daily exposure for 4 days to an inescapable form of footshock stress, known to cause opioid-mediated analgesia, suppressed the cytotoxic activity of splenic natural killer (NK) cells in rats. Similarly, daily injection of high doses of morphine (greater than or equal to 30 mg/kg) for 4 days also suppressed splenic NK cell activity. We now report that a single exposure to the opioid form of footshock stress or a single high dose of morphine induces suppression of splenic NK cell cytotoxicity. This effect is evident 3 h after treatment, returning to normal by 24 h. Morphine-induced NK suppression is evident in both male and female rats, is blocked by the opiate antagonist naltrexone, and develops tolerance. Morphine-induced NK suppression is seen in cells derived simultaneously from the spleen, bone marrow, and peripheral blood, suggesting that this suppression does not result from a selective egress of NK cells from the spleen.


Assuntos
Células Matadoras Naturais/efeitos dos fármacos , Morfina/farmacologia , Estresse Fisiológico/imunologia , Animais , Citotoxicidade Imunológica/efeitos dos fármacos , Eletrochoque , Feminino , Terapia de Imunossupressão , Células Matadoras Naturais/imunologia , Naltrexona/farmacologia , Ratos , Ratos Endogâmicos F344 , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia
18.
Neuropharmacology ; 26(11): 1589-93, 1987 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2829049

RESUMO

Male rats injected with pentobarbital (50.0 mg/kg i.p.) showed increased sleeping time after the acute administration of morphine (5.0 or 10.0 mg/kg s.c.). This effect was antagonized by naltrexone (2.0 mg/kg s.c.). The enhancement displayed stereoselectivity as levorphanol greatly lengthened the sleeping time induced by pentobarbital while dextrophan only produced a slight increase. Rats implanted for 3 days with pellets of morphine base (75.0 mg) were tolerant to the analgesic effects of morphine (2.5 mg/kg s.c.) but showed an even greater increase in pentobarbital-induced sleeping time than rats treated acutely. No potentiation was observed in subjects that were also implanted with a pellet of naltrexone (30.0 mg). It is concluded that the potentiation of pentobarbital-induced sleeping time produced by opiates is mediated by opiate receptors, but fails to show the development of tolerance.


Assuntos
Entorpecentes/farmacologia , Pentobarbital/farmacologia , Receptores Opioides/efeitos dos fármacos , Sono/efeitos dos fármacos , Animais , Dextrorfano/farmacologia , Sinergismo Farmacológico , Levorfanol/farmacologia , Masculino , Naltrexona/farmacologia , Ratos , Ratos Endogâmicos , Estereoisomerismo , Fatores de Tempo
20.
Proc Natl Acad Sci U S A ; 83(18): 7114-7, 1986 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3018757

RESUMO

We previously reported that a single systemic injection of a high dose of morphine (greater than or equal to 20 mg/kg) transiently suppresses splenic natural killer cell cytotoxicity in rats. The present study examined the possibility that the immune-suppressive effect of morphine is mediated by opiate receptors in the brain. Supporting this hypothesis, we found that morphine (20 or 40 micrograms) injected into the lateral ventricle suppressed natural killer cell activity to the same degree as a systemic dose higher by three orders of magnitude. This effect was blocked by an opiate antagonist, naltrexone. Natural killer cell activity was unaffected by systemic administration of N-methyl morphine, a morphine analogue that does not cross the blood-brain barrier. These data implicate opiate receptors in the brain in morphine-induced suppression of natural killer cell cytotoxicity.


Assuntos
Encéfalo/efeitos dos fármacos , Imunossupressores/farmacologia , Morfina/farmacologia , Receptores Opioides/efeitos dos fármacos , Animais , Citotoxicidade Imunológica/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Masculino , Naltrexona/farmacologia , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...