Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Life Sci ; 93(25-26): 1010-6, 2013 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-24084045

RESUMO

UNLABELLED: Approximately one third of the adult U.S. population suffers from some type of on-going, chronic pain annually, and many more will have some type of acute pain associated with trauma or surgery. First-line therapies for moderate to severe pain include prescriptions for common mu opioid receptor agonists such as morphine and its various derivatives. The epidemic use, misuse and diversion of prescription opioids have highlighted just one of the adverse effects of mu opioid analgesics. Alternative approaches include novel opioids that target delta or kappa opioid receptors, or compounds that interact with two or more of the opioid receptors. AIMS: Here we report the pharmacology of a newly synthesized bifunctional opioid agonist (RV-Jim-C3) derived from combined structures of fentanyl and enkephalin in rodents. RV-Jim-C3 has high affinity binding to both mu and delta opioid receptors. MAIN METHODS: Mice and rats were used to test RV-Jim-C3 in a tailflick test with and without opioid selective antagonist for antinociception. RV-Jim-C3 was tested for anti-inflammatory and antihypersensitivity effects in a model of formalin-induced flinching and spinal nerve ligation. To rule out motor impairment, rotarod was tested in rats. KEY FINDINGS: RV-Jim-C3 demonstrates potent-efficacious activity in several in vivo pain models including inflammatory pain, antihyperalgesia and antiallodynic with no significant motor impairment. SIGNIFICANCE: This is the first report of a fentanyl-based structure with delta and mu opioid receptor activity that exhibits outstanding antinociceptive efficacy in neuropathic pain, reducing the propensity of unwanted side effects driven by current therapies that are unifunctional mu opioid agonists.


Assuntos
Dor Aguda/tratamento farmacológico , Analgésicos Opioides/farmacologia , Dor Crônica/tratamento farmacológico , Encefalinas/farmacologia , Fentanila/análogos & derivados , Fentanila/química , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Analgésicos Opioides/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Encefalinas/química , Fentanila/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo
2.
J Pharmacol Exp Ther ; 339(2): 499-508, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21856861

RESUMO

Approximately 795,000 people experience a new or recurrent stroke in the United States annually. The purpose of this study was to assess the protective effect of a nonselective opioid receptor agonist, biphalin, in brain edema and infarct damage by using both in vitro and in vivo models of stroke. In an in vivo model of ischemia, biphalin significantly decreased edema (66.6 and 58.3%) and infarct (52.2 and 56.4%) ratios in mouse transient (60-min occlusion/24-h reperfusion) and permanent (6 h) middle cerebral artery occlusion models, respectively. Biphalin administration also showed decreased neurodegeneration in hippocampal, cortical, and striatal brain tissue after ischemia, evidenced by reduced Fluoro-Jade C staining. In addition, biphalin improved neurological function after stroke injury evidenced by neurological score and locomotor activity evaluation. Biphalin significantly decreased penumbral expression of Na(+), K(+), 2Cl(-) cotransporter (NKCC) and the translocation of the conventional isoforms of protein kinase C (PKC). It also reversed the activation of PKC-induced cell volume increase during ischemia in primary neuronal cell cultures exposed to 1 h of oxygen glucose deprivation. These data suggest that opioid receptor activation provides neuroprotection during stroke, and a possible explanation of this mechanism could be the inhibition of NKCC function via the regulation of PKC-dependent cell signaling.


Assuntos
Edema Encefálico/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Encefalinas/farmacologia , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Receptores Opioides/agonistas , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Edema Encefálico/patologia , Edema Encefálico/fisiopatologia , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos , Terapia de Alvo Molecular , Atividade Motora/efeitos dos fármacos , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Proteína Quinase C/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
3.
Indian J Physiol Pharmacol ; 47(3): 311-7, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-14723317

RESUMO

The present study was carried out in five cats which did not attack the rats spontaneously. Predatory attack on an anaesthetized rat was elicited by electrical stimulation of lateral hypothalamus at a mean current strength of 650 microA. The attack was accompanied by minimal affective display and culminated in neck biting. Microinfusions of DAME (delta-alanine methionine enkephaline) in 500 ng dose in substantia nigra facilitated the predatory attack and there was a significant reduction in the threshold current strength for affective display as well as somatomotor components. Microinfusions of naloxone, an opioid antagonist in 1.0 microg dose when DAME effect was at its peak reversed the facilitatory effects and the threshold returned to the control levels within 10 minutes of naloxone infusion at the same locus. Microinfusions of naloxone alone in similar dosage completely blocked the predatory attack response as indicated by an increase in the threshold current strength for somatomotor as well as affective display components. The somatomotor were completely inhibited and could not be elicited even when the current strength was increased to 1000 microA. Control injections of saline in similar volumes (0.5 microl) failed to produce any response Microinfusions of naloxone in lower dose (250 ng) failed to produce any blocking effect. These findings indicate that hypothalamically elicited predatory attack is facilitated by enkephalinergic mechanisms operating at the midbrain level.


Assuntos
Encefalinas/farmacologia , Hipotálamo/fisiologia , Comportamento Predatório/fisiologia , Substância Negra/fisiologia , Agressão/efeitos dos fármacos , Animais , Gatos , Relação Dose-Resposta a Droga , Estimulação Elétrica , Eletrodos Implantados , Encefalina Metionina/administração & dosagem , Encefalina Metionina/antagonistas & inibidores , Encefalina Metionina/farmacologia , Encefalinas/administração & dosagem , Encefalinas/antagonistas & inibidores , Feminino , Hipotálamo/anatomia & histologia , Masculino , Microinjeções , Naloxona/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Substância Negra/anatomia & histologia
4.
J Neurophysiol ; 84(2): 1050-61, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10938327

RESUMO

Previously, it was determined that microinjection of morphine into the caudal portion of subnucleus caudalis mimicked the facilitatory effects of intravenous morphine on cornea-responsive neurons recorded at the subnucleus interpolaris/caudalis (Vi/Vc) transition region. The aim of the present study was to determine the opioid receptor subtype(s) that mediate modulation of corneal units and to determine whether opioid drugs affected unique classes of units. Pulses of CO(2) gas applied to the cornea were used to excite neurons at the Vi/Vc ("rostral" neurons) and the caudalis/upper cervical spinal cord transition region (Vc/C1, "caudal" neurons) in barbiturate-anesthetized male rats. Microinjection of morphine sulfate (2.9-4.8 nmol) or the selective mu receptor agonist D-Ala, N-Me-Phe, Gly-ol-enkephalin (DAMGO; 1.8-15.0 pmol) into the caudal transition region enhanced the response in 7 of 27 (26%) rostral units to CO(2) pulses and depressed that of 10 units (37%). Microinjection of a selective delta ([D-Pen(2,5)] (DPDPE); 24-30 pmol) or kappa receptor agonist (U50488; 1.8-30.0 pmol) into the caudal transition region did not affect the CO(2)-evoked responses of rostral units. Caudal units were inhibited by local DAMGO or DPDPE but were not affected by U50,488H. The effects of DAMGO and DPDPE were reversed by naloxone (0.2 mg/kg iv). Intravenous morphine altered the CO(2)-evoked activity in a direction opposite to that of local DAMGO in 3 of 15 units, in the same direction as local DAMGO but with greater magnitude in 4 units, and in the same direction with equal magnitude as local DAMGO in 8 units. CO(2)-responsive rostral and caudal units projected to either the thalamic posterior nucleus/zona incerta region (PO/ZI) or the superior salivatory/facial nucleus region (SSN/VII). However, rostral units not responsive to CO(2) pulses projected only to SSN/VII and caudal units not responsive to CO(2) projected only to PO/ZI. It was concluded that the circuitry for opioid analgesia in corneal pain involves multiple sites of action: inhibition of neurons at the caudal transition region, by intersubnuclear connections to modulate rostral units, and by supraspinal sites. Local administration of opioid agonists modulated all classes of corneal units. Corneal stimulus modality was predictive of efferent projection status for rostral and caudal units to sensory thalamus and reflex areas of the brain stem.


Assuntos
Analgésicos Opioides/farmacologia , Córnea/inervação , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Bulbo/citologia , Células do Corno Posterior/fisiologia , Tálamo/citologia , Núcleo Inferior Caudal do Nervo Trigêmeo/citologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos não Narcóticos/farmacologia , Animais , Piscadela/efeitos dos fármacos , Piscadela/fisiologia , Dióxido de Carbono/farmacologia , Vias Eferentes/fisiologia , Eletrofisiologia , D-Penicilina (2,5)-Encefalina/farmacologia , Encefalinas/farmacologia , Nervo Facial/citologia , Masculino , Bulbo/efeitos dos fármacos , Bulbo/fisiologia , Microinjeções , Morfina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Estimulação Química , Núcleo Inferior Caudal do Nervo Trigêmeo/efeitos dos fármacos , Núcleo Inferior Caudal do Nervo Trigêmeo/fisiologia
5.
Neuroscience ; 93(3): 1025-31, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10473267

RESUMO

delta-Opioid receptors belong to the superfamily of G protein-coupled receptors, characterized by seven putative transmembrane domains, and have been shown to interact with a host of effector systems. It has been suggested that the charge on the conserved aspartic acid residue at position 128 in transmembrane domain 3 of the delta-opioid receptor contributes to both the conformation of the receptor binding pocket and the molecular rearrangements which accompany the establishment of high-affinity states of the receptor. In light of this, we used site-directed mutagenesis to determine whether this residue participates in the transmission of signals to adenylyl cyclase, the effector with which opioid receptors have been classically associated. Substitution of this aspartic acid (D128) for the neutral amino acid alanine, or the protonated amino acids lysine and histidine, constitutively couples the receptor to adenylyl cyclase, as evidenced by a curtailed response to forskolin stimulation in transfected cells. In addition, this constitutive activity can be blocked by pretreatment of the transfected cells with pertussis toxin. Interestingly, naloxone blocks this effect in cells expressing the D128A mutant, but acts as an agonist at the D128K mutant. Our findings support the hypothesis that the interaction between agonist and receptor promotes conformational changes that may be mimicked, at least in part, by mutation of the aspartate residue at position 128. Furthermore, these changes appear to be involved not only in receptor activation, but also in the functional discrimination between agonists and antagonists.


Assuntos
Adenilil Ciclases/metabolismo , Mutação Puntual , Receptores Opioides delta/genética , Sistemas do Segundo Mensageiro/fisiologia , Toxina Adenilato Ciclase , Inibidores de Adenilil Ciclases , Substituição de Aminoácidos , Animais , Ácido Aspártico/química , Ligação Competitiva , Células COS , Chlorocebus aethiops , Códon/genética , Colforsina/farmacologia , AMP Cíclico/biossíntese , DNA Complementar/genética , Diprenorfina/metabolismo , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Ligantes , Camundongos , Mutagênese Sítio-Dirigida , Naloxona/metabolismo , Toxina Pertussis , Ligação Proteica/efeitos dos fármacos , Receptores Opioides delta/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Transfecção , Fatores de Virulência de Bordetella/farmacologia
6.
Eur J Endocrinol ; 139(5): 546-51, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9849821

RESUMO

We have investigated the inter-relationship between the opioid and aminergic systems in the control of secretion of the pro-oestrous LH surge and the involvement of delta-opioid receptor subtypes in this process. Conscious female rats bearing a cannula in the femoral artery were injected i.p. with a selective delta-opioid receptor agonist (DPDPE) either alone or with the opioid antagonist (naloxone) at 1300 h on the day of pro-oestrus. Blood samples were collected hourly between 1500 h and 1900 h, and plasma LH levels were measured by RIA. At the end of this period (1900 h), the animals were autopsied and the concentrations of the amines (noradrenaline (NA), dopamine (DA), 5-hydroxytryptamine (5HT)) and their metabolites (dihydroxyphenolglycol (DHPG) and 5-hydroxyindoleacetic acid (5HIAA), metabolites of NA and 5HT respectively) were determined by HPLC with electrochemical detection in the medial preoptic area, suprachiasmatic nucleus, median eminence and arcuate nucleus. DPDPE abolished the LH surge and concomitantly decreased hypothalamic NA and DHPG concentrations in all the areas examined. The levels of DA, 5HT and 5HIAA were also reduced in all hypothalamic regions studied, except DA and 5HIAA in the suprachiasmatic nucleus. Naloxone reversed these inhibitory effects of the delta-agonist. We conclude that activation of delta-opioid receptors may exert an inhibitory effect on LH release. The effect is probably an indirect one mediated by the monoaminergic systems, as they are suppressed by DPDPE in nearly all the hypothalamic regions studied.


Assuntos
Monoaminas Biogênicas/metabolismo , Encefalinas/farmacologia , Fase Folicular/fisiologia , Hipotálamo/metabolismo , Hormônio Luteinizante/sangue , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides delta/agonistas , Animais , D-Penicilina (2,5)-Encefalina , Feminino , Hipotálamo/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Artigo em Inglês | MEDLINE | ID: mdl-9829295

RESUMO

1. The effects of opioids have been characterized in portions of the neural circuitry proposed to underly the development and maintenance of addiction. One possible mechanism is modulation of function of endogenous transmitters. 2. Cells in the prefrontal cortex, a brain area involved in cognitive function and processes relevant to addiction, are described that exhibit morphine-associated attenuation of activation response to glutamate but not acetylcholine. 3. The predominantly excitatory response of prefrontal cortical cells to local application of glutamate and acetylcholine were differentially modified by systemic and local application of opioids. 4. Local mu opioid effects mimic those of systemic morphine to a more limited degree. 5. Morphine attenuates the response of prefrontal cortical cells to activation of excitatory afferents from the mediodorsal thalamus, and to a lesser degree, from the basolateral amygdala and the hippocampus. 6. Morphine modulation of prefrontal excitatory activation is naloxone-reversible.


Assuntos
Analgésicos Opioides/farmacologia , Encefalinas/farmacologia , Potenciais Evocados/efeitos dos fármacos , Morfina/farmacologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Acetilcolina/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , Estimulação Elétrica , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Ácido Glutâmico/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Masculino , Naloxona/farmacologia , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/fisiologia , Tálamo/efeitos dos fármacos , Tálamo/fisiologia
8.
Neurosci Lett ; 252(1): 13-6, 1998 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-9756347

RESUMO

In vitro receptor binding and quantitative autoradiography were used to determine whether cannabinoid receptors in rat lumbar spinal cord are localized to the central terminals of nociceptive primary afferents. Rats were treated as neonates with capsaicin to destroy sensory C-fibers. The densities of cannabinoid and mu opioid receptors in the spinal cord of the adult rats were compared with age-matched vehicle controls. Neonatal capsaicin produced a moderate but reliable suppression (16%) of [3H]CP55,940 binding to cannabinoid receptors. By contrast, the binding of [3H][D-Ala2-MePhe4,Gly-ol5]enkephalin (DAMGO) to mu receptors was depleted by approximately 60% in near adjacent sections. These data suggest that only a subpopulation of cannabinoid receptors is situated on the central terminals of primary afferent C-fibers. The present data provide anatomical evidence for a dissociation between cannabinoid and mu opioid modulation of sensory transmission at the level of the primary afferent inputs to the spinal cord.


Assuntos
Capsaicina/farmacologia , Receptores de Droga/análise , Receptores Opioides mu/análise , Medula Espinal/química , Analgésicos Opioides/farmacologia , Animais , Animais Recém-Nascidos , Autorradiografia , Cicloexanóis/farmacologia , Regulação para Baixo/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Feminino , Imunossupressores/farmacologia , Fibras Nervosas/química , Nociceptores/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Canabinoides , Receptores de Droga/agonistas , Receptores Opioides mu/agonistas , Trítio
9.
J Pharmacol Exp Ther ; 285(3): 1266-73, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9618432

RESUMO

The present study examined protein kinase A (PKA) and protein kinase C (PKC) involvement in the maintenance of cellular tolerance to mu opioid receptor agonists resulting from chronic opiate exposure in neurosecretory cells of the hypothalamic arcuate nucleus (ARC). The possibility that the diminution of mu opioid receptor/effector coupling produced by acute 17beta-estradiol or chronic opiate exposures is mediated by a common kinase pathway also was investigated. Intracellular recordings were made in hypothalamic slices prepared from ovariectomized female guinea pigs. The mu opioid receptor agonist D-Ala2, N-Me-Phe4, Gly-ol5-enkephalin (DAMGO) produced dose-dependent hyperpolarizations of ARC neurons. Chronic morphine treatment for 4 days reduced DAMGO potency 2.5-fold with no change in the maximal response. This effect was mimicked by a 20-min bath application of the PKA activator cAMP, Sp-isomer, or the PKC activator phorbol-12,13-dibutyrate. A 30-min bath application of the broad-spectrum protein kinase inhibitor staurosporine completely abolished the reduced DAMGO potency seen in morphine-tolerant neurosecretory cells, including those immunopositive for gonadotropin-releasing hormone. The effect of staurosporine was mimicked by the PKA inhibitor cAMP, Rp-isomer, but not by the PKC inhibitor calphostin C. Finally, a 20-min bath application of 17beta-estradiol did not further reduce DAMGO potency in morphine-tolerant ARC neurons. Therefore, increased PKA activity maintains cellular tolerance to mu opioid receptor agonists in ARC neurosecretory cells caused by chronic morphine treatment. Furthermore, acute 17beta-estradiol and chronic opiate treatments attenuate mu opioid receptor-mediated responses via a common PKA pathway.


Assuntos
Analgésicos Opioides/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Morfina/farmacologia , Sistemas Neurossecretores/efeitos dos fármacos , Receptores Opioides mu/agonistas , Animais , Tolerância a Medicamentos , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Estrogênios/metabolismo , Feminino , Cobaias , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Sistemas Neurossecretores/fisiologia , Proteína Quinase C/metabolismo
10.
Brain Res ; 791(1-2): 341-6, 1998 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-9593984

RESUMO

mu-Opioid receptors play a critical role in the regulation of the female reproductive cycle, and estrogen modulates the coupling of mu-opioid receptors to a potassium channel in the basal hypothalamus (BH) of the female guinea pig. Therefore, we ascertained the distribution of mu-opioid receptors in the BH with autoradiography using the mu-opioid selective agonist [3H]DAMGO. In addition, we investigated the effects of estrogen on DAMGO- or the GABAB receptor agonist baclofen-stimulated [35S]GTPgammaS binding in the BH. Based on the high density of mu-opioid receptors, but the lack of effects of estrogen on [35S]GTPgammaS binding, we conclude that mu-opioid receptor interaction with its G-protein is not the target of estrogen's actions.


Assuntos
Encefalinas/farmacologia , Estrogênios/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hipotálamo/efeitos dos fármacos , Receptores Opioides mu/agonistas , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/metabolismo , Feminino , Cobaias , Hipotálamo/metabolismo , Sistemas Neurossecretores/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Ensaio Radioligante , Estimulação Química , Radioisótopos de Enxofre
11.
Brain Res ; 786(1-2): 133-42, 1998 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-9554984

RESUMO

Extracellular recordings were made from 161 warm-sensitive, six cold-sensitive and 153 temperature-insensitive neurons in slices of the preoptic area/anterior hypothalamus (PO/AH) of rats, to investigate the effects of the kappa-receptor opioid agonist dynorphin A1-17 and the delta-receptor opioid agonist DPDPE on neuronal response characteristics. While 61% of the neurons exhibited kappa-receptors, delta-receptors were only present in 37% of the neurons. No co-localization was observed between kappa- and delta-receptors, whereas mu-receptors could be co-localized with kappa- as well as delta-receptors. Antagonistic effects on tonic activity were induced by different concentrations of the kappa-agonist dynorphin A1-17. At 0.5 nM, the excitatory effect was predominant, while 50% of the neurons were already inhibited at 5 nM and inhibition was the major effect at 100 nM. A significant increase in temperature sensitivity was observed in warm-sensitive neurons during administration of 0.5 nM dynorphin A1-17; in contrast, the temperature sensitivity was significantly decreased at the high dose of 100 nM. In most of the neurons responding to the delta-receptor agonist DPDPE (0.5-100 nM) the firing rate was decreased. The temperature sensitivity was only affected in warm-sensitive neurons, and was increased in the majority of neurons at 0.5 and 5 nM, but predominantly decreased at higher concentrations. The effects of low concentrations of dynorphin A1-17 and DPDPE were prevented by pre- and co-perfusion of the appropriate antagonists. The present results suggest that changes of the temperature sensitivity of warm-sensitive PO/AH neurons are an important mechanism for the effect of low doses of opioids on body temperature.


Assuntos
Hipotálamo/fisiologia , Neurônios/fisiologia , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Sensação Térmica/fisiologia , Animais , Dinorfinas/farmacologia , Eletrofisiologia , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo Anterior/efeitos dos fármacos , Técnicas In Vitro , Masculino , Neurônios/efeitos dos fármacos , Área Pré-Óptica/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores Opioides mu/agonistas , Sensação Térmica/efeitos dos fármacos
12.
J Pharmacol Exp Ther ; 285(2): 496-505, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9580589

RESUMO

This study examined the signal transduction correlates of mu opioid agonist efficacy in two systems: mu receptor-transfected mMOR-CHO cell and rat thalamic membranes. The potency and maximal stimulation of [35S]GTP gamma S binding by various agonists was measured in the presence of excess GDP and compared with receptor binding affinity under identical assay conditions. Results showed that the relative maximal stimulation produced by these agonists was greater in mMOR-CHO cell than in rat thalamic membranes; some drugs that were full agonists in mMOR-CHO cells were partial agonists in the thalamus, and some partial agonists in the transfected cells were full antagonists in the thalamus. Furthermore, there was receptor reserve for G-protein activation by some agonists in mMOR-CHO cell membranes, but no receptor reserve was detected in rat thalamic membranes. Saturation analysis of agonist-stimulated [35S]GTP gamma S binding revealed that full agonists produced both a higher Bmax and apparent affinity of [35S]GTP gamma S binding than partial agonists. Correlation of the Bmax and KD of agonist-stimulated [35S]GTP gamma S binding with agonist intrinsic efficacy revealed only a moderate correlation with either parameter alone, but a highly significant correlation (r > 0.9) with a combination of the two parameters (Bmax/KD). These results suggest that the intrinsic efficacy of agonists at G-protein-coupled receptors is determined primarily by the ability of the agonist-occupied receptor to promote high-affinity GTP binding to the G-protein and to catalytically activate a maximal number G-proteins.


Assuntos
Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia , Transdução de Sinais , Tálamo/metabolismo , Animais , Células CHO , Cricetinae , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Proteínas de Ligação ao GTP/fisiologia , Camundongos , Ratos , Receptores Opioides mu/genética
13.
J Pharmacol Exp Ther ; 285(2): 820-7, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9580632

RESUMO

To analyze the selectivity of delta receptor subtypes to regulate different classes of G proteins, the expression of the alpha-subunits of Gi2, Gi3, Go1, Go2, Gq and G11 transducer proteins was reduced by administration of oligodeoxynucleotides (ODNs) complementary to sequences in their respective mRNAs. Mice receiving antisense ODNs to Gi2 alpha, Gi3 alpha, Go2 alpha and G11 alpha subunits showed an impaired antinociceptive response to all the delta agonists evaluated. An ODN to Go1 alpha specifically blocked the antinociceptive effect of the agonist of delta-1 receptors, [D-Pen2,5]enkephalin (DPDPE), without altering the activity of [D-Ala2]deltorphin II or [D-Ser2]-Leu-enkephalin-Thr (DSLET). In mice treated with an ODN to Gq alpha, the effects of the agonists of delta-2-opioid receptors were reduced, but not those of DPDPE. Thus, Go1 proteins are selectively linked to delta-1-mediated analgesia, and Gq proteins are related to delta-2-evoked antinociception. After impairing the synthesis of Go1 alpha subunits, DPDPE exhibited an antagonistic activity on the antinociception produced by [D-Ala2]deltorphin II. After treatment with ODNs complementary to sequences in Gq alpha or PLC-beta 1 mRNAs, the analgesic capacity of [D-Ala2]deltorphin II was diminished. However, the delta-2-agonist did not alter the antinociceptive activity of DPDPE. An ODN complementary to nucleotides 7 to 26 of the murine delta receptor reduced the analgesic potency of [D-Ala2]deltorphin II, but not that observed for DPDPE. In these mice, [D-Ala2]deltorphin II did not antagonize the effect of DPDPE. These results suggest the existence of different molecular forms of the delta opioid receptor, and the involvement of inositol-signaling pathways in the supraspinal antinociceptive effects of delta agonists.


Assuntos
Analgesia , Proteínas de Ligação ao GTP/análise , Inositol/metabolismo , Receptores Opioides delta/fisiologia , Analgésicos Opioides/farmacologia , Animais , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Proteínas de Ligação ao GTP/imunologia , Masculino , Camundongos , Oligonucleotídeos Antissenso/farmacologia , Receptores Opioides delta/classificação , Fosfolipases Tipo C/fisiologia
14.
J Neurosci ; 18(5): 1671-8, 1998 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9464992

RESUMO

Opioidergic inhibition of neurons in the centrolateral nucleus of the thalamus was investigated using an in vitro thalamic slice preparation from young rats. The mu-opioid receptor agonist D-Ala2,N-Me-Phe4,glycinol5-enkephalin (DAMGO) evoked a hyperpolarization and decrease in input resistance that was reversible, concentration-dependent, and persisted in the presence of tetrodotoxin. Application of the specific mu-receptor antagonist Cys2,Tyr3,Orn5,Pen7-amide blocked this response. The respective delta- and kappa-opioid receptor agonists, (D-Pen2, D-Pen5)-enkephalin and (+/-)-trans-U-50488 methanesulfonate had no effect. Voltage-clamp experiments showed that DAMGO activated an inwardly rectifying potassium conductance (GKIR) characterized by rectification at hyperpolarized potentials that increased in elevated extracellular potassium concentrations, a complete block by Ba2+ (1 mM), and a voltage-dependent block by Cs+. The extent of mu-opioid inhibition in other thalamic nuclei was then investigated. Widespread inhibition similar to that seen in the centrolateral nucleus was observed in a number of sensory, motor, intralaminar, and midline nuclei. Our results suggest that the net action of opioids would depend on their source: exogenous (systemically administered) opiates inhibiting the entire thalamus and favoring the shift of cell firing from tonic to bursting mode; and endogenously released opioids acting on specific thalamic nuclei, their release depending on the origin of the presynaptic input.


Assuntos
Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Peptídeos Opioides/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Animais , Membrana Celular/fisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Feminino , Técnicas In Vitro , Masculino , Inibição Neural/fisiologia , Neurônios/fisiologia , Técnicas de Patch-Clamp , Potássio/fisiologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores Opioides mu/agonistas , Núcleos Talâmicos/citologia , Núcleos Talâmicos/efeitos dos fármacos , Núcleos Talâmicos/fisiologia , Tálamo/citologia , Tálamo/fisiologia
15.
Brain Res ; 782(1-2): 18-23, 1998 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-9519245

RESUMO

The paraventricular nucleus of the hypothalamus (PVN) and the central nucleus of the amygdala (CNA) are two forebrain structures which are important in regulation of ingestive behavior. DAMGO is one of the most reliable and potent mu-selective opioid ligands that increases feeding in both of these brain nuclei. Administration of naloxone, an opioid antagonist, into the CNA prior to DAMGO blocks DAMGO-induced increases in food intake. The effect of this drug combination on food intake has not been evaluated in the PVN. However, intra-PVN injection of naloxone decreases deprivation and NPY-induced feeding. It has been suggested that CNA may modulate activity of midbrain and caudal brainstem centers via the hypothalamus. Based on these data, we evaluated whether an opioid-opioid interaction is present between the CNA and PVN which might affect feeding behavior. To test this, rats were doubly cannulated with 1 cannula placed in the PVN and 1 cannula in the CNA, allowing for co-administration of the opioid agonist into the PVN and the opioid antagonist into the CNA, and vice versa. CNA DAMGO increased feeding more than two-fold as compared to the vehicle-injected rats. When doses of 10, 12.5 and 25 micrograms of naltrexone (NTX) were injected into the PVN, CNA DAMGO no longer increased food intake above control levels. In the reverse situation, PVN DAMGO also increased food intake above control levels. However, when NTX was administrated unilaterally into the CNA at a relatively high dose (25 micrograms) or bilaterally (12.5 micrograms), PVN DAMGO-induced feeding was not altered. This suggests that an opioid-opioid signaling pathway exists from the CNA to the PVN which influences feeding via mu opioid receptors, whereas such a pathway from the PVN to the CNA does not seem to exist.


Assuntos
Tonsila do Cerebelo/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Encefalinas/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Receptores Opioides mu/agonistas , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Injeções , Masculino , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
16.
Eur J Pharmacol ; 326(2-3): 251-5, 1997 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-9196278

RESUMO

The effect of [Met5]enkephalin, [Leu5]enkephalin, proenkephalin, dynorphin-(1-17) or beta-endorphin on the cytotoxic (51Cr release assay) activity of natural killer cells and macrophages/monocytes was studied in mice. It was found that a single i.p. injection of [Met5]enkephalin, [Leu5]enkephalin, beta-endorphin, dynorphin or proenkephalin as well as repeated treatment with both enkephalins increased natural killer cell activity. In vitro only [Met5]enkephalin stimulated natural killer cells. Opioid peptides did not affect the cytotoxic activity of macrophages/monocytes. In addition to functional alterations, both enkephalins and beta-endorphin increased the percentage of cells with natural killer phenotype. The results of this study suggest that the increase in natural killer cytotoxicity after opioid peptides injected once or for 14 days may result from an increased number of natural killer cells in the spleen.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Endorfinas/farmacologia , Encefalinas/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Animais , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Feminino , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia
17.
Neuroreport ; 8(4): 937-40, 1997 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-9141068

RESUMO

The neuronal-specific G protein Gz is known to interact with a large variety of receptors for neurotransmitters and hormones. Fatty acylations on the N-terminus of the alpha subunit of Gz (alpha z) provide anchorage to the plasma membrane. Fatty acylation-deficient mutants of alpha z have previously been shown to exhibit altered signaling properties. Since the N-terminus of alpha z is likely to play a critical role in beta gamma binding, we examined the ability of these mutants to interact with beta gamma subunits by means of receptor-mediated stimulation of beta gamma-sensitive type II adenylyl cyclase. Our results indicate that lack of myristoylation, but not lack of palmitoylation, impaired the ability of alpha z to mediate receptor-induced release of beta gamma subunits.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Mutagênese Sítio-Dirigida , Receptores Opioides delta/fisiologia , Acilação , Toxina Adenilato Ciclase , Adenilil Ciclases/metabolismo , Animais , Células COS , AMP Cíclico/metabolismo , DNA Complementar , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Proteínas de Ligação ao GTP/biossíntese , Proteínas de Ligação ao GTP/genética , Substâncias Macromoleculares , Ácido Mirístico , Ácidos Mirísticos/metabolismo , Neurônios/metabolismo , Ácido Palmítico/metabolismo , Mutação Puntual , Receptores Opioides delta/biossíntese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Transfecção , Fatores de Virulência de Bordetella/farmacologia
18.
J Pharmacol Exp Ther ; 280(2): 1057-64, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9023324

RESUMO

The present study examined the potential for cross-tolerance development between mu-opioid and gamma-aminobutyric acidB receptor agonists, in hypothalamic arcuate neurons, resulting from chronic morphine treatment. Intracellular recordings were made in hypothalamic slices prepared from ovariectomized female guinea pigs. The mu-opioid receptor agonist D-Ala2,N-Me-Phe4,Gly-ol5-enkephalin and the gamma-aminobutyric acidB receptor agonist baclofen produced dose-dependent membrane hyperpolarizations of arcuate neurons. The reversal potential for both agonist-induced hyperpolarizations was near -95 mV, indicative of the activation of an underlying K+ conductance. Coadministration of maximally effective concentrations of D-Ala2,N-Me-Phe4,Gly-ol5-enkephalin and baclofen produced a response that was not additive, indicating a convergence onto a common K+ channel. In arcuate neurons, including a subset that was immunopositive for tyrosine hydroxylase, chronic morphine treatment for 4 to 7 days produced a 3.2-fold reduction in the potency, with no change in the efficacy, of D-Ala2,N-Me-Phe4,Gly-ol5-enkephalin. In contrast, it affected neither the potency nor the efficacy of baclofen. Therefore, chronic morphine exposure does not produce cross-tolerance between mu-opioid and gamma-aminobutyric acidB receptor agonists in A12 dopamine neurons, suggesting that convergence upon a common effector is not a sufficient criterion for the development of cross-tolerance between receptor systems.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Baclofeno/farmacologia , Encefalinas/farmacologia , Agonistas dos Receptores de GABA-B , Hipotálamo/fisiologia , Dependência de Morfina/fisiopatologia , Morfina/farmacologia , Neurônios/fisiologia , Receptores Opioides mu/agonistas , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Tolerância a Medicamentos , Eletrofisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Feminino , Cobaias , Hipotálamo/efeitos dos fármacos , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Morfina/sangue , Neurônios/efeitos dos fármacos , Tetrodotoxina/farmacologia , Tirosina 3-Mono-Oxigenase/análise
19.
Mol Pharmacol ; 51(1): 87-96, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9016350

RESUMO

G protein activation by different mu-selective opioid agonists was examined in rat thalamus, SK-N-SH cells, and mu-opioid receptor-transfected mMOR-CHO cells using agonist-stimulated guanosine-5'-O-(gamma-thio)-triphosphate ([35S]GTP gamma S) binding to membranes in the presence of excess GDP. [D-Ala2, N-MePhe4, Gly5-ol]Enkephalin (DAMGO) was the most efficacious agonist in rat thalamus and SK-N-SH cells, followed by (in rank order) fentanyl = morphine > > buprenorphine. In mMOR-CHO cells expressing a high density of mu receptors, no differences were observed among DAMGO, morphine or fentanyl, but these agonists were more efficacious than buprenorphine, which was more efficacious than levallorphan. In all three systems, efficacy differences were magnified by increasing GDP concentrations, indicating that the activity state of G proteins can affect agonist efficacy. Scatchard analysis of net agon stimulated [35S]GTP gamma S binding revealed two major components responsible for agonist efficacy differences. First, differences in the KD values of agonist-stimulated [35S]GTP gamma S binding between high efficacy agonists (DAMGO, fentanyl, and morphine) and classic partial agonists (buprenorphine and levallorphan) were observed in all three systems. Second, differences in the Bmax value of agonist-stimulated [35S]GTP gamma S binding were observed between DAMGO and morphine or fentanyl in rat thalamus and SK-N-SH cells and between the high efficacy agonists and buprenorphine or levallorphan in all three systems. These results suggest that mu-opioid agonist efficacy is determined by the magnitude of the receptor-mediated affinity shift in the binding of GTP (or[35S]GTP gamma S) versus GDP to the G protein and by the number of G proteins activated per occupied receptor.


Assuntos
Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Receptores Opioides mu/agonistas , Transdução de Sinais , Tálamo/metabolismo , Animais , Autorradiografia , Células CHO , Linhagem Celular , Cricetinae , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Encefalinas/farmacologia , Guanosina Difosfato/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/fisiologia
20.
Eur J Pharmacol ; 339(1): 27-31, 1997 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-9450613

RESUMO

The effects of pretreatment with a highly selective protein kinase C inhibitor, calphostin C, on the antinociception induced by the intracerebroventricular (i.c.v.) administration of the mu-opioid receptor agonist [D-Ala2,NMePhe4,Gly-ol5]enkephalin (DAMGO) were studied in diabetic and non-diabetic mice. The antinociceptive potency of i.c.v. DAMGO in diabetic mice was lower than that in non-diabetic mice. I.c.v. pretreatment with phorbol 12,13-dibutyrate (50 pmol), a protein kinase C activator, for 60 min attenuated the antinociception induced by i.c.v. DAMGO in non-diabetic mice, but not in diabetic mice. I.c.v. pretreatment with calphostin C (3 pmol) for 60, 120 and 240 min, but not 10 min, increased the antinociceptive effect of DAMGO (10 ng) in diabetic mice, but not in non-diabetic mice. The dose-response curve for DAMGO-induced antinociception in diabetic mice, but not in non-diabetic mice, was shifted to the left by i.c.v. pretreatment with calphostin C (3 pmol) for 60 min. In non-diabetic mice, i.c.v. pretreatment with a high dose of calphostin C (10 pmol) for 60 and 120 min potentiated DAMGO-induced antinociception. These results indicate that protein kinase C may be involved in DAMGO-induced antinociception in mice. Furthermore, these results suggest that the attenuation of DAMGO-induced antinociception in diabetic mice may be due in part to increased protein kinase C activity.


Assuntos
Analgésicos Opioides/farmacologia , Diabetes Mellitus Experimental/enzimologia , Encefalinas/farmacologia , Nociceptores/efeitos dos fármacos , Proteína Quinase C/fisiologia , Animais , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Inibidores Enzimáticos/farmacologia , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos ICR , Naftalenos/farmacologia , Dibutirato de 12,13-Forbol/farmacologia , Proteína Quinase C/antagonistas & inibidores , Valores de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA