Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-38187718

RESUMO

Increases in drug consumption over time, also known as escalation, is a key behavioral component of substance use disorder (SUD) that is related to potential harm to users, such as overdose. Studying escalation also allows researchers to investigate the transition from casual drug use to more SUD-like drug use. Understanding the neurobiological systems that drive this transition will inform therapeutic treatments in the aim to prevent increases in drug use and the development of SUD. The kappa opioid receptor (KOR) system is typically known for its role in negative affect, which is commonly found in SUD as well. Furthermore, the KOR system has also been implicated in drug use and importantly, modulating the negative effects of drug use. However, the specific neuronal subpopulation expressing KOR involved has not been identified. Here, we first demonstrated that pharmacologically inhibiting KOR in the nucleus accumbens core (NAcC), as a whole, blocks cocaine escalation under long-access self-administration conditions. We then demonstrated that KOR expressed on ventral tegmental area (VTA) neurons but not NAcC neurons is sufficient for blocking cocaine escalation by utilizing a novel virally-mediated CRISPR-SaCas9 knock-out of the oprk1 gene. Together, this suggests that activation of KOR on VTA terminals in the NAcC drives the transition to the SUD-like phenotype of escalation of cocaine consumption.

2.
Brain Res ; 1314: 44-55, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-19716811

RESUMO

Stress is a complex experience that carries both aversive and motivating properties. Chronic stress causes an increase in the risk of depression, is well known to increase relapse of drug seeking behavior, and can adversely impact health. Several brain systems have been demonstrated to be critical in mediating the negative affect associated with stress, and recent evidence directly links the actions of the endogenous opioid neuropeptide dynorphin in modulating mood and increasing the rewarding effects of abused drugs. These results suggest that activation of the dynorphin/kappa opioid receptor (KOR) system is likely to play a major role in the pro-addictive effects of stress. This review explores the relationship between dynorphin and corticotropin-releasing factor (CRF) in the induction of dysphoria, the potentiation of drug seeking, and stress-induced reinstatement. We also provide an overview of the signal transduction events responsible for CRF and dynorphin/KOR-dependent behaviors. Understanding the recent work linking activation of CRF and dynorphin/KOR systems and their specific roles in brain stress systems and behavioral models of addiction provides novel insight to neuropeptide systems that regulate affective state.


Assuntos
Encéfalo/fisiopatologia , Dinorfinas/fisiologia , Receptores Opioides kappa/fisiologia , Estresse Psicológico/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Animais , Encéfalo/metabolismo , Comorbidade , Hormônio Liberador da Corticotropina/fisiologia , Humanos , Transtornos do Humor/induzido quimicamente , Transtornos do Humor/metabolismo , Transtornos do Humor/fisiopatologia , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Recidiva , Estresse Psicológico/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo
3.
Neuroscience ; 161(2): 381-91, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19328839

RESUMO

In the present study, we asked whether multiple intrathecal injections of deltorphin II, a selective delta opioid receptor (DOPR) agonist, induced DOPR tolerance in three behavioral assays. Unilateral inflammation caused by complete Freund's adjuvant (CFA) injection into the rat or mouse hind paw (CFA model) induced thermal hyperalgesic response that was transiently and dose-dependently reduced by intrathecal administration of deltorphin II or morphine. In both rodent species, the effect of deltorphin II was not modified by a single prior administration of deltorphin II, suggesting an absence of acute tolerance in this paradigm. Repeated administration of intrathecal deltorphin II or s.c. SB-235863 (five consecutive injections over 60 h) also failed to impair the antihyperalgesic response to delta opioid receptor agonist, whereas repeated intrathecal or s.c. injections of morphine induced a significant decrease in the subsequent thermal antihyperalgesic response to morphine. In mice, deltorphin II also induced a rapid, transient motor incoordination/ataxia-like behavior as tested with the accelerating rotarod. In contrast to the antihyperalgesic responses, tolerance to the motoric effect of deltorphin II was evident in mice previously exposed to multiple intrathecal agonist injections, but not multiple saline administrations. Using the tail flick antinociceptive test, we found that DOPR-mediated analgesia was significantly reduced by repeated exposure to deltorphin II. Altogether, these observations suggest that repeated injections of DOPR agonists induce differential tolerance effects on antihyperalgesic, antinociceptive, and motor incoordination/ataxia-like behaviors related to DOPR activation by deltorphin II.


Assuntos
Tolerância a Medicamentos , Atividade Motora/efeitos dos fármacos , Dor/tratamento farmacológico , Receptores Opioides delta/agonistas , Administração Cutânea , Animais , Artrite Experimental/fisiopatologia , Relação Dose-Resposta a Droga , Adjuvante de Freund , Temperatura Alta , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Morfina/efeitos adversos , Derivados da Morfina/administração & dosagem , Derivados da Morfina/efeitos adversos , Oligopeptídeos/administração & dosagem , Oligopeptídeos/efeitos adversos , Dor/fisiopatologia , Ratos , Ratos Sprague-Dawley
4.
Neuroscience ; 150(4): 807-17, 2007 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-17997230

RESUMO

Analgesic effects of delta opioid receptor (DOR) -selective agonists are enhanced during persistent inflammation and arthritis. Although the underlying mechanisms are still unknown, membrane density of DOR was shown to be increased 72 h after induction of inflammation, an effect abolished in mu opioid receptor (MOR) -knockout (KO) mice [Morinville A, Cahill CM, Kieffer B, Collier B, Beaudet A (2004b) Mu-opioid receptor knockout prevents changes in delta-opioid receptor trafficking induced by chronic inflammatory pain. Pain 109:266-273]. In this study, we demonstrated a crucial role of MOR in DOR-mediated antihyperalgesia. Intrathecal administration of the DOR selective agonist deltorphin II failed to induce antihyperalgesic effects in MOR-KO mice, whereas it dose-dependently reversed thermal hyperalgesia in wild-type mice. The antihyperalgesic effects of deltorphin II were blocked by naltrindole but not d-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2) (CTOP) suggesting that this agonist was mainly acting through DOR. SNC80-induced antihyperalgesic effects in MOR-KO mice were also attenuated as compared with littermate controls. In contrast, kappa opioid receptor knockout did not affect deltorphin II-induced antihyperalgesia. As evaluated using mice lacking endogenous opioid peptides, the regulation of DOR's effects was also independent of beta-endorphin, enkephalins, or dynorphin opioids known to be released during persistent inflammation. We therefore conclude that DOR-mediated antihyperalgesia is dependent on MOR expression but that activation of MOR by endogenous opioids is probably not required.


Assuntos
Hiperalgesia/tratamento farmacológico , Receptores Opioides delta/metabolismo , Receptores Opioides mu/fisiologia , Animais , Relação Dose-Resposta a Droga , Dinorfinas/deficiência , Encefalinas/deficiência , Adjuvante de Freund , Hiperalgesia/etiologia , Inflamação/induzido quimicamente , Inflamação/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Naltrexona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Oligopeptídeos/administração & dosagem , Medição da Dor , Precursores de Proteínas/deficiência , Tempo de Reação/efeitos dos fármacos , Receptores Opioides kappa/deficiência , Receptores Opioides mu/deficiência , Somatostatina/administração & dosagem , Somatostatina/análogos & derivados , beta-Endorfina/deficiência
5.
Neuroscience ; 146(4): 1795-807, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17467916

RESUMO

Phosphorylation of specific sites in the second intracellular loop and in the C-terminal domain have previously been suggested to cause desensitization and internalization of the mu-opioid receptor (MOP-R). To assess sites of MOP-R phosphorylation in vivo, affinity-purified, phosphoselective antibodies were raised against either phosphothreonine-180 in the second intracellular loop (MOR-P1) or the C-terminal domain of MOP-R containing phosphothreonine-370 and phosphoserine-375 (MOR-P2). We found that MOR-P2-immunoreactivity (IR) was significantly increased within the striatum of wild-type C57BL/6 mice after injection of the agonist fentanyl. Pretreatment with the antagonist naloxone blocked the fentanyl-induced increase. Furthermore, mutant mice lacking MOP-R showed only non-specific nuclear MOR-P2-IR before or after fentanyl treatment, confirming the specificity of the MOR-P2 antibodies. To assess whether MOP-R phosphorylation occurs following endogenous opioid release, we induced chronic neuropathic pain by partial sciatic nerve ligation (pSNL), which caused a significant increase in MOR-P2-IR in the striatum. pSNL also induced signs of mu opioid receptor tolerance demonstrated by a rightward shift in the morphine dose response in the tail withdrawal assay and by a reduction in morphine conditioned place preference (CPP). Mutant mice selectively lacking all forms of the beta-endorphin peptides derived from the proopiomelanocortin (Pomc) gene did not show increased MOR-P2-IR, decreased morphine antinociception, or reduced morphine CPP following pSNL. In contrast gene deletion of either proenkephalin or prodynorphin opioids did not block the effects of pSNL. These results suggest that neuropathic pain caused by pSNL in wild-type mice activates the release of the endogenous opioid beta-endorphin, which subsequently induces MOP-R phosphorylation and opiate tolerance.


Assuntos
Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Ciática/metabolismo , Analgésicos Opioides/farmacologia , Análise de Variância , Animais , Comportamento Animal , Linhagem Celular Transformada , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Interações Medicamentosas , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Proteínas de Fluorescência Verde/biossíntese , Humanos , Hiperalgesia/etiologia , Camundongos , Camundongos Knockout , Mutagênese/fisiologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Fosforilação/efeitos dos fármacos , Fosfotreonina/imunologia , Fosfotreonina/metabolismo , Receptores Opioides mu/química , Ciática/complicações , Ciática/patologia , Transfecção , beta-Endorfina/deficiência , beta-Endorfina/metabolismo
6.
Synapse ; 42(3): 185-92, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11746715

RESUMO

The behavioral effects of psychostimulants, which are produced at least in part through inhibition of the dopamine transporter (DAT), are modulated by kappa-opioid receptors (KOR) in the nucleus accumbens (Acb). Using electron microscopic immunocytochemistry, we reveal that in the Acb KOR labeling is mainly, and DAT immunoreactivity is exclusively, presynaptic. From 400 KOR-labeled presynaptic structures, including axon terminals, intervaricosities, and small axons, 51% expressed DAT and 29% contacted another population of terminals exclusively labeled for DAT. Within axonal profiles that contained both antigens, DAT and KOR were prominently localized to plasma membrane segments that showed overlapping distributions of the respective immunogold-silver and immunoperoxidase markers. KOR labeling was also localized to membranes of small synaptic vesicles in terminals with or without DAT immunoreactivity. In addition, from 24 KOR-immunoreactive dendritic spines 42% received convergent input from DAT-containing varicosities and unlabeled terminals forming asymmetric, excitatory-type synapses. Our results provide the first ultrastructural evidence that in the Acb, KOR is localized to strategic sites for involvement in the direct presynaptic release and/or reuptake of dopamine. These data also suggest a role for KOR in the presynaptic modulation of other neurotransmitters and in the postsynaptic excitatory responses of single spiny neurons in the Acb. Dual actions on dopamine terminals and their targets in the Acb may account for KOR-mediated attenuation of drug reinforcement and sensitization.


Assuntos
Axônios/química , Glicoproteínas de Membrana , Proteínas de Membrana Transportadoras/análise , Proteínas do Tecido Nervoso , Núcleo Accumbens/química , Receptores Opioides kappa/análise , Animais , Axônios/ultraestrutura , Membrana Celular/química , Membrana Celular/ultraestrutura , Dendritos/química , Dendritos/ultraestrutura , Dopamina/fisiologia , Proteínas da Membrana Plasmática de Transporte de Dopamina , Imuno-Histoquímica , Proteínas de Membrana Transportadoras/biossíntese , Proteínas de Membrana Transportadoras/imunologia , Microscopia Imunoeletrônica , Núcleo Accumbens/citologia , Ratos , Receptores Opioides kappa/biossíntese , Receptores Opioides kappa/imunologia
8.
Mol Pharmacol ; 59(6): 1360-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11353794

RESUMO

The mu-opioid receptor (MOR) contains four highly conserved cytoplasmic tyrosine residues that may serve to regulate receptor activity. For Xenopus laevis oocytes coexpressing the rat MOR and the heteromultimeric potassium channel, K(IR)3.1/3.2, pretreatment with insulin produced both a 40% suppression in the basal channel conductance and potentiation of response to the mu-opioid agonist [D-Ala(2),methyl-Phe(4),Gly(5)-ol]enkephalin (DAMGO) to 155% of matched, untreated control cells. Insulin-induced potentiation of the DAMGO response was concentration-dependent and reversed after 1 h. Insulin pretreatment increased the maximal effect of DAMGO, but did not change its EC(50) value. Potentiation of the DAMGO response did not result from a recruitment of MOR to the cell surface, as measured by specific binding of the opioid peptide antagonist [(3)H]d-Phe((3)H)-Cys-Tyr-d-Trp-Arg-Thr-Pen-Thr-NH(2) (cyclic) to whole-oocytes, but instead the potentiation was probably caused by an increase in intrinsic efficacy of G protein coupling. The involvement of tyrosine residues on the putative intracellular loops of the MOR was demonstrated with four point-mutated receptors, replacing tyrosine with phenylalanine to create MOR(Y96F), MOR(Y106F), MOR(Y166F), and MOR(Y336F). None of these mutations significantly altered the EC(50) value for DAMGO compared with wild-type MOR, and insulin pretreatment still potentiated the effect of 1 microM DAMGO in oocytes containing either MOR(Y96F) or MOR(Y336F) to 137 +/- 10 and 124 +/- 8%, respectively. However, insulin did not significantly potentiate the DAMGO response with oocytes containing either MOR(Y106F) or MOR(Y166F), suggesting that these two sites were responsible for the insulin-induced opioid potentiation. The tyrosine-kinase inhibitors genistein (100 microM) or K-252a (20 microM) did not block the insulin-induced potentiation of the DAMGO response, but coincubation of insulin with either the MAP kinase inhibitor PD98,059 (20 microM) or phosphatase inhibitor orthovanadate (30 microM) completely blocked the potentiation. The results suggest the hypothesis that the potentiation was caused by dephosphorylation of the two tyrosines in MOR. To test this hypothesis, we measured the recovery rates after insulin treatment. As predicted, tyrosine kinase inhibition by K-252a significantly slowed the reversal and phosphatase inhibition by orthovanadate significantly accelerated the recovery. These findings support a rapid modulatory role for insulin on opioid signal transduction, possibly through the dephosphorylation of the MOR at tyrosines 106 and 166 by an insulin-activated MAP kinase/protein tyrosine phosphatase cascade. We conclude that tyrosine phosphorylation of the mu-opioid receptor regulates receptor-G protein coupling efficacy.


Assuntos
Insulina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/metabolismo , Receptores Opioides mu/metabolismo , Tirosina/metabolismo , Animais , Eletrofisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Hipoglicemiantes/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutagênese Sítio-Dirigida , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/fisiologia , Ratos , Receptores Opioides mu/genética , Receptores Opioides mu/fisiologia , Transdução de Sinais , Transfecção , Xenopus laevis
9.
J Biol Chem ; 276(18): 14855-60, 2001 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-11278615

RESUMO

We previously showed that activation of the human endothelin A receptor (HETAR) by endothelin-1 (Et-1) selectively inhibits the response to mu opioid receptor (MOR) activation of the G-protein-gated inwardly rectifying potassium channel (Kir3). The Et-1 effect resulted from PLA2 production of an eicosanoid that inhibited Kir3. In this study, we show that Kir3 inhibition by eicosanoids is channel subunit-specific, and we identify the site within the channel required for arachidonic acid sensitivity. Activation of the G-protein-coupled MOR by the selective opioid agonist D-Ala(2)Glyol, enkephalin, released Gbetagamma that activated Kir3. The response to MOR activation was significantly inhibited by Et-1 activation of HETAR in homomeric channels composed of either Kir3.2 or Kir3.4. In contrast, homomeric channels of Kir3.1 were substantially less sensitive. Domain deletion and channel chimera studies suggested that the sites within the channel required for Et-1-induced inhibition were within the region responsible for channel gating. Mutation of a single amino acid in the homomeric Kir3.1 to produce Kir3.1(F137S)(N217D) dramatically increased the channel sensitivity to arachidonic acid and Et-1 treatment. Complementary mutation of the equivalent amino acid in Kir3.4 to produce Kir3.4(S143T)(D223N) significantly reduced the sensitivity of the channel to arachidonic acid- and Et-1-induced inhibition. The critical aspartate residue required for eicosanoid sensitivity is the same residue required for Na(+) regulation of PIP(2) gating. The results suggest a model of Kir3 gating that incorporates a series of regulatory steps, including Gbetagamma, PIP(2), Na(+), and arachidonic acid binding to the channel gating domain.


Assuntos
Eicosanoides/farmacologia , Proteínas de Ligação ao GTP/fisiologia , Ativação do Canal Iônico , Fosfatidilinositol 4,5-Difosfato/metabolismo , Bloqueadores dos Canais de Potássio , Sódio/metabolismo , Animais , Humanos , Mutagênese Sítio-Dirigida , Canais de Potássio/genética , Ratos
10.
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
11.
J Biol Chem ; 276(7): 4894-900, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11060299

RESUMO

To determine the sites in the mu-opioid receptor (MOR) critical for agonist-dependent desensitization, we constructed and coexpressed MORs lacking potential phosphorylation sites along with G-protein activated inwardly rectifying potassium channels composed of K(ir)3.1 and K(ir)3.4 subunits in Xenopus oocytes. Activation of MOR by the stable enkephalin analogue, [d-Ala(2),MePhe(4),Glyol(5)]enkephalin, led to homologous MOR desensitization in oocytes coexpressing both G-protein-coupled receptor kinase 3 (GRK3) and beta-arrestin 2 (arr3). Coexpression with either GRK3 or arr3 individually did not significantly enhance desensitization of responses evoked by wild type MOR activation. Mutation of serine or threonine residues to alanines in the putative third cytoplasmic loop and truncation of the C-terminal tail did not block GRK/arr3-mediated desensitization of MOR. Instead, alanine substitution of a single threonine in the second cytoplasmic loop to produce MOR(T180A) was sufficient to block homologous desensitization. The insensitivity of MOR(T180A) might have resulted either from a block of arrestin activation or arrestin binding to MOR. To distinguish between these alternatives, we expressed a dominant positive arrestin, arr2(R169E), that desensitizes G protein-coupled receptors in an agonist-dependent but phosphorylation-independent manner. arr2(R169E) produced robust desensitization of MOR and MOR(T180A) in the absence of GRK3 coexpression. These results demonstrate that the T180A mutation probably blocks GRK3- and arr3-mediated desensitization of MOR by preventing a critical agonist-dependent receptor phosphorylation and suggest a novel GRK3 site of regulation not yet described for other G-protein-coupled receptors.


Assuntos
Arrestinas/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização , Proteínas Serina-Treonina Quinases/fisiologia , Receptores Opioides mu/metabolismo , Animais , Arrestinas/genética , Relação Dose-Resposta a Droga , Regulação para Baixo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Quinase 3 de Receptor Acoplado a Proteína G , Mutagênese Sítio-Dirigida , Oócitos/metabolismo , Fosfotreonina/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/genética , Transfecção , Xenopus , beta-Arrestinas
14.
J Biol Chem ; 275(49): 38281-5, 2000 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-10995763

RESUMO

To explore the role of highly conserved tyrosine residues in the putative cytoplasmic domains of the seven-transmembrane G protein-coupled opioid receptors, we expressed the rat kappa-opioid receptor (KOR) in Xenopus oocytes and then activated the intrinsic insulin receptor tyrosine kinase. KOR activation by the agonist produced a strong increase in potassium current through coexpressed G protein-gated inwardly rectifying potassium channels (K(IR)3). Brief pretreatment with insulin caused a 60% potentiation of the KOR-activated response. The insulin-induced increase in kappa-opioid response was blocked by the tyrosine kinase inhibitor genistein. In contrast, insulin had no effect on the basal activity of K(IR)3, suggesting that KOR is the target of the tyrosine kinase cascade. Mutation of tyrosine residues to phenylalanines in either the first or second intracellular loop of KOR to produce KOR(Y87F) and KOR(Y157F) had no effect on either the potency or maximal effect of. However, neither KOR(Y87F)- nor KOR(Y157F)-mediated responses were potentiated by insulin treatment. Insulin pretreatment shifted the dose-response curve for activation of KOR by increasing the maximal response without changing the EC(50) value for. These results suggest that insulin increases the efficacy of KOR activation by phosphorylating two tyrosine residues in the first and second intracellular loops of the receptor. Thus, tyrosine phosphorylation may provide an important mechanism for modulation of G protein-coupled receptor signaling.


Assuntos
Benzenoacetamidas , Fosfotirosina/metabolismo , Pirrolidinas/farmacologia , Receptores Opioides kappa/agonistas , Sequência de Aminoácidos , Analgésicos/farmacologia , Animais , Células Cultivadas , Feminino , Genisteína/farmacologia , Insulina/farmacologia , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Fosforilação , Estrutura Secundária de Proteína , Ratos , Receptores Opioides kappa/química , Receptores Opioides kappa/genética , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/química , Xenopus laevis
15.
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
16.
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
17.
Proc Natl Acad Sci U S A ; 96(26): 15268-73, 1999 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-10611374

RESUMO

Synaptic vesicle protein 2 (SV2) is a membrane glycoprotein common to all synaptic and endocrine vesicles. Unlike many proteins involved in synaptic exocytosis, SV2 has no homolog in yeast, indicating that it performs a function unique to secretion in higher eukaryotes. Although the structure and protein interactions of SV2 suggest multiple possible functions, its role in synaptic events remains unknown. To explore the function of SV2 in an in vivo context, we generated mice that do not express the primary SV2 isoform, SV2A, by using targeted gene disruption. Animals homozygous for the SV2A gene disruption appear normal at birth. However, they fail to grow, experience severe seizures, and die within 3 weeks, suggesting multiple neural and endocrine deficits. Electrophysiological studies of spontaneous inhibitory neurotransmission in the CA3 region of the hippocampus revealed that loss of SV2A leads to a reduction in action potential-dependent gamma-aminobutyric acid (GABA)ergic neurotransmission. In contrast, action potential-independent neurotransmission was normal. Analyses of synapse ultrastructure suggest that altered neurotransmission is not caused by changes in synapse density or morphology. These findings demonstrate that SV2A is an essential protein and implicate it in the control of exocytosis.


Assuntos
Hipocampo/fisiologia , Glicoproteínas de Membrana/deficiência , Proteínas do Tecido Nervoso/deficiência , Transmissão Sináptica/fisiologia , Animais , Encéfalo/anatomia & histologia , Sistema Endócrino/anormalidades , Genes Letais , Homozigoto , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout/crescimento & desenvolvimento , Mutagênese , Proteínas do Tecido Nervoso/genética , Malformações do Sistema Nervoso , Isoformas de Proteínas , Convulsões/genética , Sinapses/ultraestrutura , Ácido gama-Aminobutírico/metabolismo
18.
Neuron ; 23(4): 787-98, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10482244

RESUMO

It is hypothesized that Ca2+ stimulation of calmodulin (CaM)-activated adenylyl cyclases (AC1 or AC8) generates cAMP signals critical for late phase LTP (L-LTP) and long-term memory (LTM). However, mice lacking either AC1 or AC8 exhibit normal L-LTP and LTM. Here, we report that mice lacking both enzymes (DKO) do not exhibit L-LTP or LTM. To determine if these defects are due to a loss of cAMP increases in the hippocampus, DKO mice were unilaterally cannulated to deliver forskolin. Administration of forskolin to area CA1 before training restored normal LTM. We conclude that Ca2+-stimulated adenylyl cyclase activity is essential for L-LTP and LTM and that AC1 or AC8 can produce the necessary cAMP signal.


Assuntos
Adenilil Ciclases/metabolismo , Cálcio/fisiologia , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Animais , Aprendizagem da Esquiva/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Calmodulina/fisiologia , Colforsina/farmacologia , Sinais (Psicologia) , Eletrofisiologia , Medo/fisiologia , Medo/psicologia , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Imuno-Histoquímica , Potenciação de Longa Duração/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Camundongos Knockout , Microscopia Confocal
19.
J Biol Chem ; 274(34): 23802-7, 1999 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-10446141

RESUMO

We used the Xenopus oocyte expression system to examine the regulation of rat kappa opioid receptor (rKOR) function by G protein receptor kinases (GRKs). kappa agonists increased the conductance of G protein-activated inwardly rectifying potassium channels in oocytes co-expressing KOR with Kir3.1 and Kir3.4. In the absence of added GRK and beta-arrestin 2, desensitization of the kappa agonist-induced potassium current was modest. Co-expression of either GRK3 or GRK5 along with beta-arrestin 2 significantly increased the rate of desensitization, whereas addition of either beta-arrestin 2, GRK3, or GRK5 alone had no effect on the KOR desensitization rate. The desensitization was homologous as co-expressed delta opioid receptor-evoked responses were not affected by KOR desensitization. The rate of GRK3/beta-arrestin 2-dependent desensitization was reduced by truncation of the C-terminal 26 amino acids, KOR(Q355Delta). In contrast, substitution of Ala for Ser within the third intracellular loop [KOR(S255A,S260A, S262A)] did not reduce the desensitization rate. Within the C-terminal region, KOR(S369A) substitution significantly attenuated desensitization, whereas the KOR(T363A) and KOR(S356A,T357A) point mutations did not. These results suggest that co-expression of GRK3 or GRK5 and beta-arrestin 2 produced homologous, agonist-induced desensitization of the kappa opioid receptor by a mechanism requiring the phosphorylation of the serine 369 of rKOR.


Assuntos
Arrestinas/fisiologia , Benzenoacetamidas , Proteínas Serina-Treonina Quinases , Receptores Proteína Tirosina Quinases/fisiologia , Receptores Opioides kappa/fisiologia , Sequência de Aminoácidos , Animais , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Feminino , Quinase 3 de Receptor Acoplado a Proteína G , Dados de Sequência Molecular , Pirrolidinas/farmacologia , Ratos , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/química , Relação Estrutura-Atividade , beta-Arrestina 2 , beta-Arrestinas
20.
J Neurosci ; 19(10): 3773-80, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10234009

RESUMO

Desensitization of cannabinoid receptor signaling by a G-protein coupled receptor kinase (GRK) was examined using the Xenopus oocyte expression system. Application of a CB1 agonist, WIN 55,212-2, evoked a concentration-dependent increase in K+ conductance (Kir3) in oocytes coexpressing rat CB1 with the G-protein-gated, inwardly rectifying K+ channels Kir3.1 and Kir3.4. Desensitization was slight during continuous agonist application in the absence of GRK and arrestin. However, coexpression of GRK3 and beta-arrestin 2 (beta-arr2) caused profound homologous CB1 receptor desensitization, supporting the hypothesis that GRK3 and beta-arr2 effectively produce CB1 receptor desensitization. To identify the regions of the CB1 receptor responsible for GRK3- and beta-arr2-mediated desensitization, we constructed several CB1 receptor mutants. Truncation of the C-terminal tail of CB1 receptor at residue 418 (Delta418) almost completely abolished desensitization but did not affect agonist activation of Kir3. In contrast, truncation at residues 439 and 460 did not significantly affect GRK3- and beta-arr2-dependent desensitization. A deletion mutant (Delta418-439) did not desensitize, indicating that residues within this region are important for GRK3- and beta-arr2-mediated desensitization. Phosphorylation in this region was likely involved in desensitization, because mutation of either of two putative phosphorylation sites (S426A or S430A) significantly attenuated desensitization. CB1 receptors rapidly internalize after activation by agonist. Phosphorylation of S426 or S430 was not necessary for internalization, because the S426A/S430A CB1 mutant internalized when stably expressed in AtT20 cells. These studies establish that CB1 desensitization can be regulated by a GRK and that different receptor domains are involved in GRK- and beta-arrestin-dependent desensitization and CB1 internalization.


Assuntos
Estrutura Terciária de Proteína , Receptores de Droga/fisiologia , Sequência de Aminoácidos , Animais , Benzoxazinas , Testes Genéticos , Dados de Sequência Molecular , Morfolinas/farmacologia , Mutação , Naftalenos/farmacologia , Oócitos , Fosforilação , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Receptores de Canabinoides , Receptores de Droga/química , Receptores de Droga/genética , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA