Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 278: 11-9, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25086317

RESUMO

Alterations in hippocampal neurogenesis affect spatial learning, though, the relative contributions of cell proliferation and cell survival on this process are poorly understood. The current study utilized mu opioid receptor (MOR-1) knockout (KO) mice on two background strains, C57BL/6 and 129S6, to assess cell survival as well as determine the impact on spatial learning using the Morris water maze. These experiments were designed to extend prior work showing that both C57BL/6 and 129S6 MOR-1 KO mice have an increased number of proliferating cells in the dentate gyrus (DG) when compared to wild-type (WT) mice. The current study indicates that newly born neurons in the DG of C57BL/6 MOR-1 KO mice exhibit enhanced survival when compared to WT mice, while new neurons in the DG of 129S6 MOR-1 KO mice do not. In addition, C57BL/6 MOR-1 KO mice have a lower number of apoptotic cells in the DG compared to WT mice while, in contrast, 129S6 MOR-1 KO mice have a higher number of apoptotic cells in this region. These alterations collectively contribute to an increase in the granule cell number in the DG of C57BL/6 MOR-1 KO mice, while the total number of granule cells in 129S6 MOR-1 KO mice is unchanged. Thus, although C57BL/6 and 129S6 MOR-1 KO mice both exhibit increased cell proliferation in the DG, the impact of the MOR-1 mutation on cell survival differs between strains. Furthermore, the decrease in DG cell survival displayed by 129S6 MOR-1 KO mice is correlated with functional deficits in spatial learning, suggesting that MOR-1-dependent alterations in the survival of new neurons in the DG, and not MOR-1-dependent changes in proliferation of progenitor cells in the DG, is important for spatial learning.


Assuntos
Giro Denteado/fisiologia , Neurogênese , Receptores Opioides mu/fisiologia , Aprendizagem Espacial/fisiologia , Animais , Morte Celular , Sobrevivência Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Opioides mu/genética , Especificidade da Espécie
2.
Neuroscience ; 206: 49-59, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22280973

RESUMO

The endogenous opioid system is involved in various physiological processes, including neurogenesis in the dentate gyrus (DG) of the hippocampus. In the current study, we investigated the role of the mu opioid receptor (MOR-1) on DG neurogenesis and measured glucocorticoid levels following several injection paradigms to supplement the neurogenesis experiments. MOR-1 knockout (KO) mice on C57BL/6 and 129S6 backgrounds were injected with bromodeoxyuridine (BrdU) using either a single injection or two different repeated injection protocols and then sacrificed at different time points. The total number of BrdU and proliferating cell nuclear antigen (PCNA) positive cells in the DG is significantly increased in MOR-1 KO mice compared with wild type (WT) on both strains after repeated injection, but not after a single injection. Plasma corticosterone (CORT) levels increased similarly in MOR-1 KO and WT mice following both single and repeated injection, indicating that the stress response is activated following any injection protocol, but that the mechanism responsible for the increase in BrdU labeling in MOR-1 KO mice is CORT-level independent. Finally, WT 129S6 mice, independent of genotype, showed higher levels of plasma CORT compared with WT C57BL/6 mice in both noninjected controls and following injection at two separate time points; these levels were inversely correlated with low numbers of BrdU cells in the DG in 129S6 mice compared with C57BL/6 mice. In summary, these data demonstrate that loss of MOR-1 increases BrdU labeling in the DG independent of CORT levels, but only following a repeated injection, illustrating the capability of injection paradigms to influence cell-proliferative responses in a genotype-dependent manner.


Assuntos
Bromodesoxiuridina/administração & dosagem , Giro Denteado/metabolismo , Neurogênese/fisiologia , Receptores Opioides mu/metabolismo , Coloração e Rotulagem/métodos , Animais , Cortisona/sangue , Imunofluorescência , Imuno-Histoquímica , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Radioimunoensaio , Receptores Opioides mu/genética
3.
Endocrinology ; 150(4): 1739-47, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19022883

RESUMO

Amidated peptides are critically involved in many physiological functions. Genetic deletion of peptidylglycine alpha-amidating monooxygenase (PAM), the only enzyme that can synthesize these peptides, is embryonically lethal. The goal of the present study was the identification of physiological functions impaired by haploinsufficiency of PAM. Regulation of the hypothalamic-pituitary-thyroid axis and body temperature, functions requiring contributions from multiple amidated peptides, were selected for evaluation. Based on serum T(4) and pituitary TSH-beta mRNA levels, mice heterozygous for PAM (PAM(+/-)) were euthyroid at baseline. Feedback within the hypothalamic-pituitary-thyroid axis was impaired in PAM(+/-) mice made hypothyroid using a low iodine/propylthiouracil diet. Despite their normal endocrine response to cold, PAM(+/-) mice were unable to maintain body temperature as well as wild-type littermates when kept in a 4 C environment. When provided with additional dietary copper, PAM(+/-) mice maintained body temperature as well as wild-type mice. Pharmacological activation of vasoconstriction or shivering also allowed PAM(+/-) mice to maintain body temperature. Cold-induced vasoconstriction was deficient in PAM(+/-) mice. This deficit was eliminated in PAM(+/-) mice receiving a diet with supplemental copper. These results suggest that dietary deficiency of copper, coupled with genetic deficits in PAM, could result in physiological deficits in humans.


Assuntos
Cobre/farmacologia , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/genética , Complexos Multienzimáticos/deficiência , Complexos Multienzimáticos/genética , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/genética , Temperatura Baixa , Cobre/administração & dosagem , Suplementos Nutricionais , Feminino , Genótipo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Oxigenases de Função Mista/fisiologia , Complexos Multienzimáticos/fisiologia , Fenilefrina/farmacologia , Piperazinas/farmacologia , Piridinas/farmacologia , Radioimunoensaio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Reologia , Proteína Desacopladora 1 , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
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 ; 147(2): 439-44, 2007 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-17544222

RESUMO

Opioids are extensively used analgesics yet can paradoxically increase pain sensitivity in humans and rodents. This hyperalgesia is extensively conceptualized to be a consequence of opioid receptor activity, perhaps providing an adaptive response to analgesia, and to utilize N-methyl-D-aspartate (NMDA) receptors. These assumptions were tested here in opioid receptor triple knock-out (KO) mice lacking all three genes encoding opioid receptors (mu, delta, and kappa) by comparing their thermal nociceptive responses to the opioids morphine and oxymorphone with those of B6129F(1) controls. Injecting acute opioid bolus doses in controls caused maximal analgesia that was completely abolished in KO mice, confirming the functional consequence of the KO mouse opioid receptor deficiency. Continuous opioid infusion by osmotic pump in control mice also initially caused several consecutive days of analgesia that was shortly thereafter followed by several consecutive days of hyperalgesia. In contrast, continuously infusing KO mice with opioids caused no detectable analgesic response, but only immediate and steady declines in nociceptive thresholds culminating in several days of unremitting hyperalgesia. Finally, injecting the non-competitive NMDA receptor antagonist MK-801 during opioid infusion markedly reversed hyperalgesia in control but not KO mice. These data demonstrate that sustained morphine and oxymorphone delivery causes hyperalgesia independently of prior or concurrent opioid or NMDA receptor activity or opioid analgesia, indicating the contribution of mechanisms outside of current conceptions, and are inconsistent with proposals of hyperalgesia as a causative factor of opioid analgesic tolerance.


Assuntos
Analgésicos Opioides/farmacologia , Dor/genética , Dor/fisiopatologia , Receptores Opioides/genética , Receptores Opioides/fisiologia , Analgésicos Opioides/administração & dosagem , Animais , Interpretação Estatística de Dados , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/genética , Hiperalgesia/fisiopatologia , Camundongos , Camundongos Knockout , Morfina/farmacologia , Oximorfona/farmacologia , Medição da Dor/efeitos dos fármacos , Tempo de Reação/fisiologia , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos
6.
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
7.
Neuropeptides ; 39(6): 559-67, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16289278

RESUMO

Agonist stimulation of opioid receptors increases feeding in rodents, while opioid antagonists inhibit food intake. The pan-opioid antagonist, LY255582, produces a sustained reduction in food intake and body weight in rodent models of obesity. However, the specific receptor subtype(s) responsible for this activity is unknown. To better characterize the pharmacology of LY255582, we examined the binding of a radiolabeled version of the molecule, [(3)H]-LY255582, in mouse brain using autoradiography. In mouse brain homogenates, the K(d) and B(max) for [(3)H]-LY255582 were 0.156 +/- 0.07 nM and 249 +/- 14 fmol/mg protein, respectively. [(3)H]-LY255582 bound to slide mounted sections of mouse brain with high affinity and low non-specific binding. High levels of binding were seen in areas consistent with the known localization of opioid receptors. These areas included the caudate putamen, nucleus accumbens, claustrum, medial habenula, dorsal endopiriform nucleus, basolateral nucleus of the amygdala, hypothalamus, thalamus and ventral tegmental area. We compared the binding distribution of [(3)H]-LY255582 to the opioid receptor antagonist radioligands [(3)H]-naloxone (mu), [(3)H]-naltrindole (delta) and [(3)H]-norBNI (kappa). The overall distribution of [(3)H]-LY255582 binding sites was similar to that of the other ligands. No specific [(3)H]-LY255582 binding was noted in sections of mu-, delta- and kappa-receptor combinatorial knockout mice. Therefore, it is likely that LY255582 produces its effects on feeding and body weight gain through a combination of mu-, delta- and kappa-receptor activity.


Assuntos
Encéfalo/metabolismo , Cicloexanos/metabolismo , Piperidinas/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Animais , Autorradiografia , Sítios de Ligação , Encéfalo/anatomia & histologia , Cicloexanos/química , Camundongos , Camundongos Knockout , Estrutura Molecular , Naloxona/metabolismo , Naltrexona/análogos & derivados , Naltrexona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Piperidinas/química , Receptores Opioides delta/genética , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Trítio/química , Trítio/metabolismo
8.
Neuroscience ; 130(2): 359-67, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15664692

RESUMO

There is general agreement that dopaminergic neurons projecting from the ventral tegmental area (VTA) to the nucleus accumbens and prefrontal cortex play a key role in drug reinforcement. The activity of these neurons is strongly modulated by the inhibitory and excitatory input they receive. Activation of mu-opioid receptors, located on GABAergic neurons in the VTA, causes hyperpolarization of these GABAergic neurons, thereby causing a disinhibition of VTA dopaminergic neurons. This effect of mu-opioid receptors upon GABA neurotransmission is a likely mechanism for mu-opioid receptor modulation of drug reinforcement. We studied mu-opioid receptor signaling in relation to cocaine reinforcement in wild-type and mu-opioid receptor knockout mice using a cocaine self-administration paradigm and in vitro electrophysiology. Cocaine self-administration was reduced in mu-opioid receptor knockout mice, suggesting a critical role of mu-opioid receptors in cocaine reinforcement. The frequency of spontaneous inhibitory post-synaptic currents onto dopaminergic neurons in the ventral tegmental area was increased in mu-opioid receptor knockout mice compared with wild-type controls, while the frequency of spontaneous excitatory post-synaptic currents was unaltered. The reduced cocaine self-administration and increased GABAergic input to VTA dopaminergic neurons in mu-opioid receptor knockout mice supports the notion that suppression of GABAergic input onto dopaminergic neurons in the VTA contributes to mu-opioid receptor modulation of cocaine reinforcement.


Assuntos
Cocaína/farmacologia , Neurônios/metabolismo , Receptores Opioides mu/genética , Reforço Psicológico , Área Tegmentar Ventral/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/metabolismo , Vias Aferentes/fisiopatologia , Animais , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Modelos Animais de Doenças , Dopamina/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Neurônios/efeitos dos fármacos , Autoadministração , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/fisiopatologia
9.
Neuroscience ; 125(1): 211-20, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15051160

RESUMO

Cocaine-induced behavioral sensitization is a complex phenomenon involving a number of neuromodulator and neurotransmitter systems. To specifically investigate the role of the micro opioid receptor (MOR) in cocaine-induced behavioral sensitization in mice, both genetic and pharmacological approaches were undertaken. MOR-1 deficient mice of varying backgrounds (C57BL/6J, 129S6, F1 hybrid 129S6xC57BL/6J and 129S6xC57BL/6J) and wild-type C57BL/6J mice exposed continuously to naltrexone, an opioid receptor antagonist, received single daily injections of saline or cocaine for 10 days. All mice received a single cocaine challenge 7 days following the last saline or cocaine injection to test for the expression of sensitization. The locomotor-stimulating and sensitizing effects of cocaine observed in MOR-1 wild-type mice were absent in MOR-1 knockout mice maintained on the mixed 129S6xC57BL/6J background. In contrast, MOR-1 deficient mice developed on a C57BL/6J background showed an accentuated sensitivity to cocaine-induced locomotion. Cocaine's psychomotor activating effects were more pronounced in the MOR-1 C57BL/6J knockouts injected daily with cocaine than in the MOR-1 wild-type mice. Similar locomotor-stimulating and sensitizing effects were found in both F1 hybrid 129S6xC57BL/6J MOR-1 wild-type and MOR-1 knockout mice, while the 129S6 strain showed an overall indifference to cocaine. That is, both the locomotor-stimulating and sensitizing effects of cocaine were absent in both MOR-1 wild-type and MOR-1 knockout mice maintained on the 129S6 background. Lastly, the locomotor-stimulating and sensitizing effects of cocaine were attenuated in C57BL/6J wild-type mice exposed continuously to naltrexone. Collectively, these data support a role for opioidergic involvement in cocaine-influenced behavior in mice. Moreover, MORs appear to differentially modulate a sensitized response to cocaine in different strains of mice as delineated by MOR-1 gene deletion and pharmacological antagonism.


Assuntos
Adaptação Fisiológica/fisiologia , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Receptores Opioides mu/genética , Animais , Comportamento Animal , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/deficiência
10.
J Endocrinol ; 179(2): 227-35, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14596674

RESUMO

Analysis of knockout mice suggests that the neurotropin-inducible secreted polypeptide VGF (non-acronymic) plays an important role in the regulation of energy balance. VGF is synthesized by neurons in the central and peripheral nervous systems (CNS, PNS), as well as in the adult pituitary, adrenal medulla, endocrine cells of the stomach and pancreatic beta cells. Thus VGF, like cholecystokinin, leptin, ghrelin and other peptide hormones that have been shown to regulate feeding and energy expenditure, is synthesized in both the gut and the brain. Although detailed developmental studies of VGF localization in the CNS and PNS have been completed, little is known about the ontogeny of VGF expression in endocrine and neuroendocrine tIssues. Here, we report that VGF mRNA is detectable as early as embryonic day 15.5 in the developing rat gastrointestinal and esophageal lumen, pancreas, adrenal, and pituitary, and we further demonstrate that VGF mRNA is synthesized in the gravid rat uterus, together supporting possible functional roles for this polypeptide outside the nervous system and in the enteric plexus.


Assuntos
Sistemas Neurossecretores/química , Sistemas Neurossecretores/embriologia , Proteínas/genética , RNA Mensageiro/análise , Glândulas Suprarrenais/química , Glândulas Suprarrenais/embriologia , Animais , Proteínas de Ligação a DNA/genética , Sistema Endócrino/química , Sistema Endócrino/embriologia , Feminino , Fatores de Transcrição Fushi Tarazu , Fator de Transcrição GATA2 , Proteínas de Homeodomínio/genética , Humanos , Hibridização In Situ/métodos , Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/embriologia , Neuropeptídeos , Fatores de Transcrição Box Pareados , Hipófise/química , Hipófise/embriologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares , Homologia de Sequência , Fator Esteroidogênico 1 , Fatores de Transcrição/genética , Útero/química
11.
Neuroscience ; 117(1): 157-68, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12605902

RESUMO

The opioid receptor-like 1 receptor is a novel member of the opioid receptor family and its endogenous peptide ligand has been termed nociceptin and orphanin FQ. Activation of the opioid receptor-like 1 receptor by nociceptin/orphanin FQ in vivo produces hyperalgesia when this peptide is given supraspinally but analgesia at the spinal level. Nociceptin/orphanin FQ also reverses stress-induced analgesia, suggesting that the peptide has anti-opioid properties. Nociceptin/orphanin FQ knockout mice show alterations in pain sensitivity and stress responses and display increased morphine dependence, suggesting an interaction of the nociceptin/orphanin FQ system with classical opioid receptor function. To determine if the behavioural phenotype of nociceptin/orphanin FQ knockout mice reflects changes in either opioid receptor-like 1 or classical opioid receptor expression, we have carried out quantitative autoradiography of the opioid receptor-like 1, mu-, delta- and kappa-opioid receptors in the brains of these animals. Receptor density was measured on coronal sections from wild-type, heterozygous and homozygous mice using [(3)H]nociceptin, [(3)H][D-Ala(2)-N-methyl-Phe(4)-Gly(5) ol] enkephalin, [(3)H]deltorphin-I, or [(3)H](-)-N-methyl-N-[7-(1-pyrrodinyl)-1-oxospiro[4,5]dec-8-yl]-4-benzofuranacetamide to label opioid receptor-like 1, mu-, delta- and kappa-receptors, respectively. A region-specific up-regulation of the opioid receptor-like 1 receptor (up to 135%) was seen in brains from homozygous mice. Mu-Receptors also showed significant differences between genotypes whilst changes in delta- and kappa- receptors were minor. In conclusion the region-specific up-regulation of the opioid receptor-like 1 receptor indicates a tonic role for nociceptin/orphanin FQ in some brain structures and may suggest the peptide regulates the receptor expression in these regions. The changes in the opioid receptor-like 1 receptor may relate to the anxiogenic phenotype of these animals but the observed change in mu-receptors does not correlate with altered morphine responses.


Assuntos
Encéfalo/metabolismo , Receptores Opioides/biossíntese , Receptores Opioides/deficiência , Regulação para Cima/fisiologia , Animais , Química Encefálica/fisiologia , Regulação da Expressão Gênica/fisiologia , Camundongos , Camundongos Knockout , Receptores Opioides/análise , Receptores Opioides/genética , Receptor de Nociceptina
12.
Proc Natl Acad Sci U S A ; 99(5): 3087-92, 2002 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-11854475

RESUMO

The serine protease prohormone convertase 2 (PC2), principally involved in the processing of polypeptide hormone precursors in neuroendocrine tissues, requires interaction with the neuroendocrine protein 7B2 to generate an enzymatically active form. 7B2 null mice express no PC2 activity and release large quantities of uncleaved ACTH, resulting in a lethal endocrine condition that resembles pituitary Cushing's (Westphal, C. H., Muller, L., Zhou, A., Bonner-Weir, S., Schambelan, M., Steiner, D. F., Lindberg, I. & Leder, P. (1999) Cell 96, 689). Here, we have compared the 7B2 and PC2 null mouse models to determine why the 7B2 null, but not the PC2 null, exhibits a lethal disease state. Both 7B2 and PC2 nulls contained highly elevated pituitary adrenocorticotropic hormone (ACTH); the neurointermediate lobe content of ACTH in 7B2 nulls was 13-fold higher than in WT mice; that of the PC2 null was 65-fold higher. However, circulating ACTH levels were much higher in the 7B2 null than in the PC2 null. Because hypothalamic inhibitory dopaminergic control represents the major influence on intermediate lobe proopiomelanocortin-derived peptide secretion, dopamine levels were measured, and they revealed that 7B2 null pituitaries contained only one-fourth of WT pituitary dopamine. Adrenalectomized 7B2 null animals survived past the usual time of death at 5 weeks; a month after adrenalectomy, they exhibited normal levels of pituitary dopamine, circulating ACTH, and corticosterone. Elevated corticosterone, therefore, seems to play a central role in the lethal phenotype of the 7B2 null, whereas a 7B2-mediated dopaminergic deficiency state may be involved in the actual ACTH hypersecretion phenomenon. Interestingly, adrenalectomized 7B2 nulls also developed unexpectedly severe obesity.


Assuntos
Glândulas Suprarrenais/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Síndrome de Cushing/metabolismo , Dopamina/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Hormônios Hipofisários/fisiologia , Adrenalectomia , Hormônio Adrenocorticotrópico/genética , Animais , Síndrome de Cushing/etiologia , Síndrome de Cushing/mortalidade , Humanos , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteína Secretora Neuroendócrina 7B2 , Fenótipo , Hipófise/metabolismo , Adeno-Hipófise/metabolismo , Hormônios Hipofisários/genética , Pró-Opiomelanocortina/genética , Pró-Proteína Convertase 2 , RNA Mensageiro , Subtilisinas/genética , Subtilisinas/fisiologia
13.
J Neurosci ; 21(19): 7788-92, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11567069

RESUMO

Dopamine systems are intimately involved with opioid actions. Pharmacological studies suggest an important modulatory effect of dopamine and its receptors on opioid analgesia. We have now examined these interactions in a knock-out model in which the dopamine(2) (D(2)) receptor has been disrupted. Loss of D(2) receptors enhances, in a dose-dependent manner, the analgesic actions of the mu analgesic morphine, the kappa(1) agonist U50,488H and the kappa(3) analgesic naloxone benzoylhydrazone. The responses to the delta opioid analgesic [d-Pen(2),d-Pen(5)]enkephalin were unaffected in the knock-out animals. Loss of D(2) receptors also potentiated spinal orphanin FQ/nociceptin analgesia. Antisense studies using a probe targeting the D(2) receptor revealed results similar to those observed in the knock-out model. The modulatory actions of D(2) receptors were independent of final sigma receptor systems because the final sigma agonist (+)-pentazocine lowered opioid analgesia in all mice, including the D(2) knock-out group. Thus, dopamine D(2) receptors represent an additional, significant modulatory system that inhibits analgesic responses to mu and kappa opioids.


Assuntos
Analgesia , Analgésicos Opioides/farmacologia , Receptores de Dopamina D2/deficiência , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Antagonistas de Dopamina/farmacologia , Antagonistas dos Receptores de Dopamina D2 , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , D-Penicilina (2,5)-Encefalina/farmacologia , Heterozigoto , Camundongos , Camundongos Knockout , Morfina/farmacologia , Naloxona/análogos & derivados , Naloxona/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Peptídeos Opioides/farmacologia , Medição da Dor/efeitos dos fármacos , Pentazocina/farmacologia , Receptores de Dopamina D2/genética , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Receptores sigma/agonistas , Sulpirida/farmacologia , Nociceptina
14.
J Neurosci ; 21(16): 5864-70, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11487609

RESUMO

The bioactivity of neuropeptides can be regulated by a variety of post-translational modifications, including proteolytic processing. Here, gene-targeted mice producing defective prohormone convertase 2 (PC2) were used to examine the post-translational processing of two neuroendocrine prohormones, pro-opiomelanocortin (POMC) and pro-orphanin FQ (pOFQ)/nociceptin (N), in the brain. Reversed-phase HPLC and gel-exclusion chromatography were combined with specific radioimmunoassays to analyze the processing patterns of these two prohormones in the hypothalamus and the amygdala. In the case of POMC, the lack of PC2 activity completely prevented carboxy-shortening of beta-endorphins and greatly diminished conversion of beta-lipotropin to gamma-lipotropin and beta-endorphin. Although conversion of beta-lipotropin to beta-endorphin decreased, the lack of PC2 activity caused an increase in beta-lipotropin and beta-endorphin levels in the mutant animals, but no increases in POMC or biosynthetic intermediates were seen. The extent of OFQ/N production was significantly lower in PC2-deficient mice and there was an accumulation of relatively large amounts of pOFQ/N and biosynthetic intermediates. These results demonstrate that PC2 is directly involved in the biogenesis of two brain neuropeptides in vivo and suggest that the specific prohormone and cellular context influences neuropeptide processing by PCs.


Assuntos
Encéfalo/metabolismo , Pró-Opiomelanocortina/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Receptores Opioides/metabolismo , Subtilisinas/biossíntese , Tonsila do Cerebelo/química , Tonsila do Cerebelo/metabolismo , Animais , Química Encefálica , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Marcação de Genes , Heterozigoto , Homozigoto , Hipotálamo/química , Hipotálamo/metabolismo , Camundongos , Camundongos Knockout , Pró-Proteína Convertase 2 , Radioimunoensaio , Subtilisinas/genética , beta-Endorfina/biossíntese , beta-Lipotropina/biossíntese , beta-Lipotropina/metabolismo
15.
Neuroscience ; 104(1): 217-22, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11311544

RESUMO

It has been hypothesized that morphine tolerance and dependence in mice following chronic exposure may reflect increased compensatory activity of antiopioid systems. The endogenous peptide nociceptin/orphanin FQ has been shown to have anti-opioid effects, for example antagonizing morphine analgesia. Moreover, chronic morphine administration increases synthesis of the peptide, and morphine tolerance and dependence can be attenuated or reversed by antagonists and agonists of the nociceptin/orphanin FQ receptor, respectively. The present study seeks to confirm a role for nociceptin/orphanin FQ in opioid tolerance and dependence by comparing morphine ED(50) values and naloxone-precipitated withdrawal jumping in mice homozygous (knock-out) and heterozygous for a null mutation of the Npnc1 gene encoding the nociceptin/orphanin FQ propeptide, and their wild type littermates, following chronic morphine exposure. Relative to morphine-naive control mice, significant rightward shifts in the morphine dose-response curve, resulting in increased morphine ED(50) values (approximately two to three-fold), was observed for all genotypes following three days of repeated systemic morphine injections. However, no differences between genotypes in the magnitude of tolerance were observed. In contrast, knock-out mice displayed significantly increased naloxone-precipitated withdrawal jumping relative to heterozygous and wild-type mice following implantation with a morphine pellet (25mg) for 72h. Use of nociception/orphaninFQ transgenic knock-out mice thus demonstrate the differential involvement of nociceptin/orphanin FQ in morphine tolerance and dependence.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Tolerância a Medicamentos/genética , Camundongos Knockout/metabolismo , Dependência de Morfina/genética , Peptídeos Opioides/deficiência , Analgésicos Opioides/farmacologia , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiopatologia , Masculino , Camundongos , Camundongos Knockout/genética , Morfina/farmacologia , Dependência de Morfina/metabolismo , Dependência de Morfina/fisiopatologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Peptídeos Opioides/genética , Nociceptina
16.
J Neuroimmunol ; 114(1-2): 173-80, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11240029

RESUMO

We have examined the chemotactic responsiveness of thymocytes to selective mu-, kappa-, and delta-opioid agonists. Our results show that developing T cells migrate in response to mu-, but not kappa- or delta-opioids. The mu-opioid response appears to be dependent on the classical mu-opioid receptor (MOR-1) since the chemotactic response is blocked by a selective mu-opioid antagonist, and is absent in thymocytes from MOR-1-deficient mice. Flow cytometric analysis of the mu-opioid responsive cells shows that these cells consist predominantly of highly immature CD4- CD8- T cells. These results represent the first demonstration of the functional expression of mu-opioid receptors by developing T cells.


Assuntos
Receptores Opioides mu/genética , Receptores Opioides mu/imunologia , Linfócitos T/fisiologia , Timo/citologia , Timo/crescimento & desenvolvimento , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos não Narcóticos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Diferenciação Celular/imunologia , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Expressão Gênica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organismos Livres de Patógenos Específicos , Linfócitos T/citologia , Timo/imunologia
17.
Brain Res ; 880(1-2): 102-8, 2000 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-11032994

RESUMO

Improgan is an analog of the H(2) antagonist cimetidine that does not act on known histamine receptors, but induces highly effective analgesia in rodents following intracerebroventricular (icv) administration. Since the mechanism of action of this compound remains unknown, improgan analgesia was characterized presently with the tail immersion nociceptive test in mutant mice lacking either the mu (exon 1 of MOR-1), delta (exon 2 of DOR-1) or kappa (exon 3 of KOR-1) opioid receptor. Improgan (30 microg, icv) induced reversible, maximal analgesia in both sexes of all three genotypes (+/+, +/- and -/-) of MOR-1 mutant mice 10 and 20 min after administration, whereas morphine analgesia was reduced (+/-) or abolished (-/-) in these subjects. In DOR-1 mutant mice, improgan was equally effective in all three genotypes, despite the reduction (+/-) or complete loss (-/-) of delta opioid receptor (3H-[D-Pen(2), D-Pen(5)]enkephalin, DPDPE) binding. Similarly, improgan analgesia was equivalent in all three genotypes of KOR-1 mutant mice, whereas kappa-mediated analgesia (U50,488) and kappa opioid (3H-U69,593) binding were abolished in the homozygous (-/-) mice. These studies demonstrate that improgan analgesia does not require intact MOR-1, DOR-1, or KOR-1 genes, and support the hypothesis that improgan-like analgesics act in the CNS by non-opioid mechanisms.


Assuntos
Analgésicos não Narcóticos/farmacologia , Ventrículos Cerebrais/fisiologia , Cimetidina/análogos & derivados , Receptores Opioides mu/genética , Receptores Opioides/fisiologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgesia , Animais , Ventrículos Cerebrais/efeitos dos fármacos , Cimetidina/administração & dosagem , Cimetidina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Éxons , Feminino , Regulação da Expressão Gênica , Genótipo , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Knockout , Receptores Opioides/deficiência , Receptores Opioides/genética , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides delta/fisiologia , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/deficiência , Receptores Opioides mu/fisiologia
18.
J Neurochem ; 75(4): 1763-70, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10987860

RESUMO

Prodynorphin, a multifunctional precursor of several important opioid peptides, is expressed widely in the CNS. It is processed at specific single and paired basic sites to generate various biologically active products. Among the prohormone convertases (PCs), PC1 and PC2 are expressed widely in neuroendocrine tissues and have been proposed to be the major convertases involved in the biosynthesis of hormonal and neural peptides. In this study we have examined the physiological involvement of PC2 in the generation of dynorphin (Dyn) peptides in mice lacking active PC2 as a result of gene disruption. Enzymological and immunological assays were used to confirm the absence of active PC2 in these mice. The processing profiles of Dyn peptides extracted from brains of these mice reveal a complete lack of Dyn A-8 and a substantial reduction in the levels of Dyn A-17 and Dyn B-13. Thus, PC2 appears to be involved in monobasic processing, leading to the generation of Dyn A-8, Dyn A-17, and Dyn B-13 from prodynorphin under physiological conditions. Brains of heterozygous mice exhibit only half the PC2 activity of wild-type mice; however, the levels of Dyn peptides in these mice are similar to those of wild-type mice, suggesting that a 50% reduction in PC2 activity is not sufficient to significantly reduce prodynorphin processing. The disruption of the PC2 gene does not lead to compensatory up-regulation in the levels of other convertases with similar substrate specificity because we find no significant changes in the levels of PC1, PC5/PC6, or furin in these mice as compared with wild-type mice. Taken together, these results support a critical role for PC2 in the generation of Dyn peptides.


Assuntos
Encéfalo/metabolismo , Encefalinas/metabolismo , Pró-Proteína Convertase 1 , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Subtilisinas/deficiência , Animais , Ácido Aspártico Endopeptidases/metabolismo , Western Blotting , Química Encefálica , Cromatografia em Gel , Fluorometria , Furina , Heterozigoto , Camundongos , Camundongos Knockout , Neuropeptídeos/metabolismo , Fragmentos de Peptídeos/análise , Pró-Proteína Convertase 2 , Pró-Proteína Convertase 5 , Pró-Proteína Convertases , Radioimunoensaio , Serina Endopeptidases/metabolismo , Subtilisinas/genética , Subtilisinas/metabolismo
19.
Mol Endocrinol ; 14(9): 1472-82, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10976924

RESUMO

Insulin-like growth factor binding protein 2 (IGFBP-2) is one member of the family of IGF binding proteins believed to have both endocrine functions elicited by modulating serum IGF half-life and transport as well as autocrine/paracrine functions that result from blocking or enhancing the availability of IGFs to bind cell surface receptors. To clarify the in vivo role of IGFBP-2, we have used gene targeting to introduce a null IGFBP-2 allele into the mouse genome. Animals homozygous for the altered allele are viable and fertile, contain no IGFBP-2 mRNA, and have no detectable IGFBP-2 in the adult circulation. Heterozygous and homozygous animals showed no significant differences in prenatal or postnatal body growth. Analyses of organ weights in adult males, however, revealed that spleen weight was reduced and liver weight was increased in the absence of IGFBP-2. In addition, ligand blot analyses of sera from adult IGFBP-2 null males showed that IGFBP-1, IGFBP-3, and IGFBP-4 levels were increased relative to wild-type mice. These results demonstrate that up-regulation of multiple IGFBPs accompanies the absence of IGFBP-2 and that IGFBP-2 has a critical role, either directly or indirectly, in modulating spleen and liver size.


Assuntos
Crescimento/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Deleção de Sequência , Animais , Peso Corporal , Fertilidade/genética , Coração/anatomia & histologia , Heterozigoto , Homozigoto , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/deficiência , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Rim/anatomia & histologia , Fígado/anatomia & histologia , Pulmão/anatomia & histologia , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão , RNA Mensageiro/genética , Mapeamento por Restrição , Baço/anatomia & histologia
20.
Dev Biol ; 219(2): 197-213, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10694416

RESUMO

The superior cervical ganglion (SCG) is a well-characterized model of neural development, in which several regulatory signals have been identified. Vasoactive intestinal peptide (VIP) has been found to regulate diverse ontogenetic processes in sympathetics, though functional requirements for high peptide concentrations suggest that other ligands are involved. We now describe expression and functions of pituitary adenylate cyclase-activating polypeptide (PACAP) during SCG ontogeny, suggesting that the peptide plays critical roles in neurogenesis. PACAP and PACAP receptor (PAC(1)) mRNA's were detected at embryonic days 14.5 (E14.5) through E17.5 in vivo and virtually all precursors exhibited ligand and receptor, indicating that the system is expressed as neuroblasts proliferate. Exposure of cultured precursors to PACAP peptides, containing 27 or 38 residues, increased mitogenic activity 4-fold. Significantly, PACAP was 1000-fold more potent than VIP and a highly potent and selective antagonist entirely blocked effects of micromolar VIP, consistent with both peptides acting via PAC(1) receptors. Moreover, PACAP potently enhanced precursor survival more than 2-fold, suggesting that previously defined VIP effects were mediated via PAC(1) receptors and that PACAP is the more significant developmental signal. In addition to neurogenesis, PACAP promoted neuronal differentiation, increasing neurite outgrowth 4-fold and enhancing expression of neurotrophin receptors trkC and trkA. Since PACAP potently activated cAMP and PI pathways and increased intracellular Ca(2+), the peptide may interact with other developmental signals. PACAP stimulation of precursor mitosis, survival, and trk receptor expression suggests that the signaling system plays a critical autocrine role during sympathetic neurogenesis.


Assuntos
Neuropeptídeos/metabolismo , Receptores do Hormônio Hipofisário/metabolismo , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , AMP Cíclico/metabolismo , DNA/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Ligantes , Mitose/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptor trkA/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores do Hormônio Hipofisário/genética , Sistemas do Segundo Mensageiro , Gânglio Cervical Superior/efeitos dos fármacos , Gânglio Cervical Superior/embriologia , Gânglio Cervical Superior/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...