Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 38(19): 4505-4520, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29650696

RESUMO

Neuropeptide Y (NPY) expression is tightly linked with the development of stress resilience in rodents and humans. Local NPY injections targeting the basolateral amygdala (BLA) produce long-term behavioral stress resilience in male rats via an unknown mechanism. Previously, we showed that activation of NPY Y1 receptors hyperpolarizes BLA principal neurons (PNs) through inhibition of the hyperpolarization-activated, depolarizing H-current, Ih The present studies tested whether NPY treatment induces stress resilience by modulating Ih NPY (10 pmol) was delivered daily for 5 d bilaterally into the BLA to induce resilience; thereafter, the electrophysiological properties of PNs and the expression of Ih in the BLA were characterized. As reported previously, increases in social interaction (SI) times persisted weeks after completion of NPY administration. In vitro intracellular recordings showed that repeated intra-BLA NPY injections resulted in hyperpolarization of BLA PNs at 2 weeks (2W) and 4 weeks (4W) after NPY treatment. At 2W, spontaneous IPSC frequencies were increased, whereas at 4W, resting Ih was markedly reduced and accompanied by decreased levels of HCN1 mRNA and protein expression in BLA. Knock-down of HCN1 channels in the BLA with targeted delivery of lentivirus containing HCN1-shRNA increased SI beginning 2W after injection and induced stress resilience. NPY treatment induced sequential, complementary changes in the inputs to BLA PNs and their postsynaptic properties that reduce excitability, a mechanism that contributes to less anxious behavior. Furthermore, HCN1 knock-down mimicked the increases in SI and stress resilience observed with NPY, indicating the importance of Ih in stress-related behavior.SIGNIFICANCE STATEMENT Resilience improves mental health outcomes in response to adverse situations. Neuropeptide Y (NPY) is associated with decreased stress responses and the expression of resilience in rodents and humans. Single or repeated injections of NPY into the basolateral amygdala (BLA) buffer negative behavioral effects of stress and induce resilience in rats, respectively. Here, we demonstrate that repeated administration of NPY into the BLA unfolds several cellular mechanisms that decrease the activity of pyramidal output neurons. One key mechanism is a reduction in levels of the excitatory ion channel HCN1. Moreover, shRNA knock-down of HCN1 expression in BLA recapitulates some of the actions of NPY and causes potent resilience to stress, indicating that this channel may be a possible target for therapy.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neuropeptídeo Y/farmacologia , Canais de Potássio/efeitos dos fármacos , Resiliência Psicológica/efeitos dos fármacos , Tonsila do Cerebelo/citologia , Animais , Ansiedade/genética , Ansiedade/psicologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Relações Interpessoais , Masculino , Microinjeções , Neuropeptídeo Y/administração & dosagem , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
2.
J Comp Neurol ; 524(12): 2418-39, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-26779765

RESUMO

Within the basolateral amygdaloid complex (BLA), neuropeptide Y (NPY) buffers against protracted anxiety and fear. Although the importance of NPY's actions in the BLA is well documented, little is known about the source(s) of NPY fibers to this region. The current studies identified sources of NPY projections to the BLA by using a combination of anatomical and neurochemical approaches. NPY innervation of the BLA was assessed in rats by examining the degree of NPY coexpression within interneurons or catecholaminergic fibers with somatostatin and tyrosine hydroxylase (TH) or dopamine ß-hydroxylase (DßH), respectively. Numerous NPY(+) /somatostatin(+) and NPY(+) /somatostatin(-) fibers were observed, suggesting at least two populations of NPY fibers within the BLA. No colocalization was noted between NPY and TH or DßH immunoreactivities. Additionally, Fluorogold (FG) retrograde tracing with immunohistochemistry was used to identify the precise origin of NPY projections to the BLA. FG(+) /NPY(+) cells were identified within the amygdalostriatal transition area (AStr) and stria terminalis and scattered throughout the bed nucleus of the stria terminalis. The subpopulation of NPY neurons in the AStr also coexpressed somatostatin. Subjecting animals to a conditioned fear paradigm increased NPY gene expression within the AStr, whereas no changes were observed within the BLA or stria terminalis. Overall, these studies identified limbic regions associated with stress circuits providing NPY input to the BLA and demonstrated that a unique NPY projection from the AStr may participate in the regulation of conditioned fear. J. Comp. Neurol. 524:2418-2439, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Complexo Nuclear Basolateral da Amígdala/metabolismo , Condicionamento Psicológico/fisiologia , Medo/fisiologia , Rede Nervosa/metabolismo , Neuropeptídeo Y/biossíntese , Animais , Complexo Nuclear Basolateral da Amígdala/química , Corpo Estriado/química , Corpo Estriado/metabolismo , Medo/psicologia , Masculino , Rede Nervosa/química , Neuropeptídeo Y/análise , Ratos , Ratos Sprague-Dawley
3.
Endocrinology ; 157(2): 820-30, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26653570

RESUMO

The neuropeptide kisspeptin (Kiss1) is integral to the advent of puberty and the generation of cyclical LH surges. Although many complex actions of Kiss1 are known, the mechanisms governing the processing/regulation of this peptide have not been unveiled. The metallo enzyme, endopeptidase 24.15 (thimet oligopeptidase), has been demonstrated to play a key role in the processing and thus the duration of action of the reproductive neuropeptide, GnRH, which signals downstream of Kiss1. Initial in silico modeling implied that Kiss1 could also be a putative substrate for EP24.15. Coincubation of Kiss1 and EP24.15 demonstrated multiple cleavages of the peptide predominantly between Arg29-Gly30 and Ser47-Phe48 (corresponding to Ser5-Phe6 in Kiss-10; Kiss-10 as a substrate had an additional cleavage between Phe6-Gly7) as determined by mass spectrometry. Vmax for the reaction was 2.37±0.09 pmol/min · ng with a Km of 19.68 ± 2.53µM, which is comparable with other known substrates of EP24.15. EP24.15 immunoreactivity, as previously demonstrated, is distributed in cell bodies, nuclei, and processes throughout the hypothalamus. Kiss1 immunoreactivity is localized primarily to cell bodies and fibers within the mediobasal and anteroventral-periventricular hypothalamus. Double-label immunohistochemistry indicated coexpression of EP24.15 and Kiss1, implicating that the regulation of Kiss1 by EP24.15 could occur in vivo. Further studies will be directed at determining the precise temporal sequence of EP24.15 effects on Kiss1 as it relates to the control of reproductive hormone secretion and treatment of fertility issues.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/enzimologia , Kisspeptinas/metabolismo , Metaloendopeptidases/metabolismo , Animais , Simulação por Computador , Escherichia coli , Feminino , Hipotálamo/metabolismo , Imuno-Histoquímica , Masculino , Espectrometria de Massas , Metestro/metabolismo , Proestro/metabolismo , Ratos
4.
Neuropeptides ; 47(4): 273-80, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23688647

RESUMO

Enhanced corticotropin releasing factor (CRF) release in the basolateral amygdala (BLA) is strongly associated with the generation of behavioral stress responses through activation of the CRF-R1 receptor subtype. Stress and anxiety-like behavior are modulated in part by the balance of peptide actions such as excitatory CRF and inhibitory neuropeptide Y (NPY) receptor activation in the BLA. While the actions of CRF are clear, little is known about the cell type influenced by CRF receptor stimulation. These studies were designed to identify the cell types within the BLA activated by intra-BLA administration of CRF using multi-label immunohistochemistry for cFos and markers for pyramidal (CaMKII-immunopositive) and interneuronal [glutamic acid decarboxylase (GAD65)] cell populations. Administration of CRF into the BLA produced a dose-dependent increase in the expression of cFos-ir. Intra-BLA injection of CRF induced significant increases in cFos-ir in the CaMKII-ir population. Although increases in cFos-ir in GAD65-ir cells were observed, this did not reach statistical significance perhaps in part due to the decreased numbers of GAD65-ir cells within the BLA after CRF treatment. These findings demonstrate that CRF, when released into the BLA, activates projection neurons and that the activity of GABAergic interneurons is also altered by CRF treatment. Decreases in the number of GAD65-ir neurons could reflect either increased or decreased activity of these cells and future studies will more directly address these possibilities. The expression of cFos is associated with longer term regulation of gene expression which may be involved in the profound long term effects of neuropeptides, such as CRF, on the activity and plasticity of BLA pyramidal neurons.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Hormônio Liberador da Corticotropina/farmacologia , Neurônios/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Glutamato Descarboxilase/metabolismo , Masculino , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley
5.
J Chem Neuroanat ; 45(1-2): 50-6, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22884996

RESUMO

Neuropeptide Y (NPY) produces potent anxiolytic effects via activation of NPY Y1 receptors (Y1r) within the basolateral amygdaloid complex (BLA). The role of NPY in the BLA was recently expanded to include the ability to produce stress resilience and long-lasting reductions in anxiety-like behavior. These persistent behavioral effects are dependent upon activity of the protein phosphatase, calcineurin (CaN), which has long been associated with shaping long-term synaptic signaling. Furthermore, NPY-induced reductions in anxiety-like behavior persist months after intra-BLA delivery, which together indicate a form of neuronal plasticity had likely occurred. To define a site of action for NPY-induced CaN signaling within the BLA, we employed multi-label immunohistochemistry to determine which cell types express CaN and if CaN colocalizes with the Y1r. We have previously reported that both major neuronal cell populations in the BLA, pyramidal projection neurons and GABAergic interneurons, express the Y1r. Therefore, this current study evaluated CaN immunoreactivity in these cell types, along with Y1r immunoreactivity. Antibodies against calcium-calmodulin kinase II (CaMKII) and GABA were used to identify pyramidal neurons and GABAergic interneurons, respectively. A large population of CaN immunoreactive cells displayed Y1r immunoreactivity (90%). Nearly all (98%) pyramidal neurons displayed CaN immunoreactivity, while only a small percentage of interneurons (10%) contained CaN immunoreactivity. Overall, these anatomical findings provide a model whereby NPY could directly regulate CaN activity in the BLA via activation of the Y1r on CaN-expressing, pyramidal neurons. Importantly, they support BLA pyramidal neurons as prime targets for neuronal plasticity associated with the long-term reductions in anxiety-like behavior produced by NPY injections into the BLA.


Assuntos
Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/ultraestrutura , Calcineurina/biossíntese , Neurônios/metabolismo , Neurônios/ultraestrutura , Receptores de Neuropeptídeo Y/biossíntese , Animais , Calcineurina/análise , Imuno-Histoquímica , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores de Neuropeptídeo Y/análise , Transdução de Sinais/fisiologia
6.
J Neurosci ; 28(4): 893-903, 2008 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18216197

RESUMO

Resilience to mental and physical stress is a key determinant for the survival and functioning of mammals. Although the importance of stress resilience has been recognized, the underlying neural mediators have not yet been identified. Neuropeptide Y (NPY) is a peptide known for its anti-anxiety-like effects mediated via the amygdala. The results of our current study demonstrate, for the first time that repeated administration of NPY directly into the basolateral nucleus of the amygdala (BLA) produces selective stress-resilient behavioral responses to an acute restraint challenge as measured in the social interaction test, but has no effect on hypothalamic-adrenal-pituitary axis activity or stress-induced hyperthermia. More importantly, the resilient behaviors observed in the NPY-treated animals were present for up to 8 weeks. Antagonizing the activity of calcineurin, a protein phosphatase involved in neuronal remodeling and present in NPY receptor containing neurons within the BLA, blocked the development of long-term, but not the acute increases in social interaction responses induced by NPY administration. This suggests that the NPY-induced long-term behavioral resilience to restraint stress may occur via mechanisms involving neuronal plasticity. These studies suggest one putative physiologic mechanism underlying stress resilience and could identify novel targets for development of therapies that can augment the ability to cope with stress.


Assuntos
Febre/fisiopatologia , Sistema Hipotálamo-Hipofisário/fisiologia , Neuropeptídeo Y/administração & dosagem , Sistema Hipófise-Suprarrenal/fisiologia , Comportamento Social , Estresse Psicológico/prevenção & controle , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , Febre/tratamento farmacológico , Febre/psicologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Masculino , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Ratos , Ratos Wistar , Estresse Psicológico/fisiopatologia , Estresse Psicológico/psicologia , Tempo
7.
Endocrinology ; 147(9): 4122-31, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16728491

RESUMO

Regulation of vasopressin (VP) and oxytocin (OT) secretion involves integration of neural signals from hypothalamic osmoreceptors, ascending catecholaminergic and peptidergic cell groups in the brain stem, and local and autoregulatory afferents. Neuropeptide Y (NPY) is one factor that stimulates the release of VP and OT from the supraoptic (SON) and paraventricular nuclei of the hypothalamus via activation of Y1 receptors (Y1R). The current studies were designed to assess the regulation and distribution of NPY Y1R expression in the SON of male rats that were either given 2% NaCl drinking water (24-72 h) or water deprived (48 h). Subjecting male rats to these conditions resulted in significant increases in both the number of cells expressing Y1R immunoreactivity (ir) and the amount of Y1R protein per cell within the SON. Y1R immunoreactivity was increased in the magnocellular but not medial parvocellular paraventricular nuclei, and Y1R mRNA levels were increased in the SON of salt-loaded rats. Subpopulations of both VP and OT cells in the hypothalamus express Y1R immunoreactivity and a greater percentage of VP-ir cells express Y1R after salt loading. To control for potential effects of dehydration-induced anorexia, a group of euhydrate animals was pair fed with animals consuming 2% NaCl. No detectable change in Y1R expression was observed in the SON of pair-fed animals, even though body weights were significantly lower than controls. These data demonstrate that NPY Y1R gene and protein expression are increased in the SON of salt-loaded and water-deprived animals and provide a mechanism whereby NPY can support VP/OT release during prolonged challenges to fluid homeostasis.


Assuntos
Desidratação/metabolismo , Hipotálamo/química , Neurônios/química , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropeptídeos/genética , Núcleo Supraóptico/química , Animais , Sangue , Peso Corporal , Expressão Gênica , Imuno-Histoquímica , Masculino , Concentração Osmolar , Ocitocina/análise , Núcleo Hipotalâmico Paraventricular/química , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/análise , Receptores de Neuropeptídeos/análise , Cloreto de Sódio/administração & dosagem , Vasopressinas/análise , Privação de Água
8.
J Pharmacol Exp Ther ; 312(3): 1257-65, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15542622

RESUMO

The effects of pretreatment with endomorphin-1 (EM-1) and endomorphin-2 (EM-2) given into the ventral periaqueductal gray (vPAG) to induce antianalgesia against the tail-flick (TF) inhibition produced by morphine given into the vPAG were studied in rats. Pretreatment with EM-1 (3.5-28 nmol) given into vPAG for 45 min dose-dependently attenuated the TF inhibition produced by morphine (9 nmol) given into vPAG. Similarly, pretreatment with EM-2 (1.7-7.0 nmol) for 45 min also attenuated the TF inhibition induced by morphine; however, a high dose of EM-2 (14 nmol) did not attenuate the morphine-produced TF inhibition. The attenuation of morphine-produced TF inhibition induced by EM-2 or EM-1 pretreatment was blocked by pretreatment with mu-opioid antagonist (-)-naloxone (55 pmol) but not nonopioid (+)-naloxone (55 pmol). However, pretreatment with a morphine-6beta-glucuronide-sensitive mu-opioid receptor antagonist 3-methoxynaltrexone (6.4 pmol) selectively blocked EM-2- but not EM-1-induced antianalgesia. Pretreatment with dynorphin A(1-17) antiserum reversed only EM-2- but not EM-1-induced antianalgesia. Pretreatment with antiserum against beta-endorphin, [Met(5)]enkephalin, [Leu(5)]enkephalin, substance P or cholecystokinin, or with delta-opioid receptor antagonist naltrindole (2.2 nmol) or kappa-opioid receptor antagonist norbinaltorphimine (6.6 nmol) did not affect EM-2-induced antianalgesia. It is concluded that EM-2 selectively releases dynorphin A(1-17) by stimulation of a novel subtype of mu-opioid receptor, tentatively designated as mu(3) in the vPAG to induce antianalgesia, whereas the antianalgesia induced by EM-1 is mediated by the stimulation of another subtype of mu(1)- or mu(2)-opioid receptor.


Assuntos
Analgesia , Oligopeptídeos/farmacologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Animais , Colecistocinina/metabolismo , Relação Dose-Resposta a Droga , Dinorfinas/metabolismo , Encefalina Leucina/metabolismo , Encefalina Metionina/metabolismo , Masculino , Morfina/antagonistas & inibidores , Naloxona/farmacologia , Ratos , Substância P/metabolismo , beta-Endorfina/metabolismo
9.
Eur J Pharmacol ; 504(3): 177-83, 2004 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-15541419

RESUMO

We previously demonstrated pretreatment with antiserum against dynorphin A1-17 attenuates endomorphin-2-induced analgesia and antianalgesia, suggesting that these endomorphin-2 effects are mediated by the release of dynorphin A1-17. Lumbar-cisternal spinal perfusion was used to measure the release of immunoreactive dynorphin A1-17 into spinal perfusates from urethane-anesthetized rats following endomorphin-2 or endomorphin-1 treatment within the perfusion solution. Treatment with endomorphin-2 (5-50 nmol) for 3 min caused a dose-dependent increase of immunoreactive dynorphin A1-17 in spinal perfusates, with a maximal increase detected between 24 and 48 min after endomorphin-2 treatment, while levels returned to baseline within 60 min. Endomorphin-2-induced release of immunoreactive dynorphin A1-17 was attenuated by pretreatment with mu-opioid receptor antagonist naloxone or 3-methoxynaltrexone. Endomorphin-1 induced a slight increase in immunoreactive dynorphin1-17 as well, but only at the highest dose used (50 nmol). Our results suggest that endomorphin-2 stimulated a specific subtype of mu-opioid receptor to induce the release of immunoreactive dynorphin A1-17 in spinal cords of rats.


Assuntos
Dinorfinas/metabolismo , Oligopeptídeos/farmacologia , Medula Espinal/metabolismo , Anestesia , Animais , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Injeções Espinhais , Masculino , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/administração & dosagem , Ratos , Receptores Opioides mu/antagonistas & inibidores , Medula Espinal/efeitos dos fármacos
10.
Neurosci Lett ; 365(3): 157-61, 2004 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-15246539

RESUMO

An unbiased conditioned place preference paradigm was used to evaluate the reward effect of selective endogenous mu-opioid ligands, endomorphin-1 and endomorphin-2, in male CD-1 mice. Pre- and post-conditioning free-movement were measured on day 1 and day 5, respectively. Conditioning sessions were conducted twice daily from day 2 through day 4 consisting of the alternate injection of conditioning drug or vehicle. Intracerebroventricular (i.c.v.) injection of endomorphin-1 (0.3-10 microg) induced place preference in a dose-dependent manner; whereas, endomorphin-2 (1-10 microg) dose-dependently induced place aversion. Both endomorphin-1-induced place preference and endomorphin-2-induced place aversion were blocked by pretreatment i.c.v. with mu-opioid receptor antagonist, beta-funaltrexamine. Selective delta-opioid receptor antagonist, naltrindole, co-administered i.c.v. with endomorphin-1 or endomorphin-2 did not affect reward effect. However, endomorphin-2-induced place aversion, but not endomorphin-1-induced place preference, was blocked by the i.c.v.-administered selective kappa-opioid receptor antagonist, WIN 44,441-3. It is concluded that endomorphin-1 produces conditioned place preference, which is mediated by the stimulation of mu-, but not delta- or kappa-opioid receptors, while endomorphin-2 produces conditioned place aversion, which is mediated by the stimulation of mu- and kappa-, but not delta-opioid receptors.


Assuntos
Condicionamento Operante/efeitos dos fármacos , Naltrexona/análogos & derivados , Oligopeptídeos/farmacologia , Animais , Azocinas/farmacologia , Injeções Intraventriculares , Ligantes , Masculino , Camundongos , Naltrexona/farmacologia , Oligopeptídeos/administração & dosagem , Oligopeptídeos/metabolismo , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo
11.
J Pharmacol Exp Ther ; 310(1): 240-6, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-14999057

RESUMO

Intrathecal (i.t.) pretreatment with a low dose (0.3 nmol) of morphine causes an attenuation of i.t. morphine-produced analgesia; the phenomenon has been defined as morphine-induced antianalgesia. The opioid-produced analgesia was measured with the tail-flick (TF) test in male CD-1 mice. Intrathecal pretreatment with low dose (0.3 nmol) of morphine time dependently attenuated i.t. morphine-produced (3.0 nmol) TF inhibition and reached a maximal effect at 45 min. Intrathecal pretreatment with morphine (0.009-0.3 nmol) for 45 min also dose dependently attenuated morphine-produced TF inhibition. The i.t. morphine-induced antianalgesia was dose dependently blocked by the nonselective mu-opioid receptor antagonist (-)-naloxone and by its nonopioid enantiomer (+)-naloxone, but not by endomorphin-2-sensitive mu-opioid receptor antagonist 3-methoxynaltrexone. Blockade of delta-opioid receptors, kappa-opioid receptors, and N-methyl-D-aspartate (NMDA) receptors by i.t. pretreatment with naltrindole, nor-binaltorphimine, and (-)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801), respectively, did not affect the i.t. morphine-induced antianalgesia. Intrathecal pretreatment with antiserum against dynorphin A(1-17), [Leu]-enkephalin, [Met]-enkephalin, beta-endorphin, cholecystokinin, or substance P also did not affect the i.t. morphine-induced antianalgesia. The i.t. morphine pretreatment also attenuated the TF inhibition produced by opioid muagonist [D-Ala2, N-Me-Phe4,Gly-ol5]-enkephalin, delta-agonist deltorphin II, and kappa-agonist U50,488H. It is concluded that low doses (0.009-0.3 nmol) of morphine given i.t. activate an antianalgesic system to attenuate opioid mu-, delta-, and kappa-agonist-produced analgesia. The morphine-induced antianalgesia is not mediated by the stimulation of opioid mu-, delta-, or kappa-receptors or NMDA receptors. Neuropeptides such as dynorphin A(1-17), [Leu]-enkephalin, [Met]-enkephalin, beta-endorphin, cholecystokinin, and substance P are not involved in this low-dose morphine-induced antianalgesia.


Assuntos
Analgesia , Morfina/administração & dosagem , Naltrexona/análogos & derivados , Dor/tratamento farmacológico , Medula Espinal/efeitos dos fármacos , Animais , Maleato de Dizocilpina/farmacologia , Interações Medicamentosas , Tolerância a Medicamentos , Dinorfinas/imunologia , Dinorfinas/metabolismo , Encefalinas/imunologia , Encefalinas/metabolismo , Masculino , Camundongos , Morfina/uso terapêutico , Naloxona/farmacologia , Naltrexona/farmacologia , Oligopeptídeos/metabolismo , Medição da Dor/efeitos dos fármacos , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Substância P/imunologia , Substância P/metabolismo , beta-Endorfina/imunologia , beta-Endorfina/metabolismo
12.
J Pharmacol Exp Ther ; 309(2): 816-24, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14755004

RESUMO

An unbiased conditioned place preference (CPP) paradigm was used to evaluate the reward effects of endogenous mu-opioid receptor ligands endomorphin-1 (EM-1) and endomorphin-2 (EM-2) from the mesolimbic posterior nucleus accumbens (Acb) shell and the ventral tegmental area (VTA) in CD rats. EM-1 (1.6-8.1 nmol) microinjected into posterior Acb shell produced CPP, whereas EM-2 (8.7-17.5 nmol) given into the same Acb shell produced conditioned place aversion (CPA). EM-1 (1.6-16.3 nmol) microinjected into the VTA produced CPP, whereas EM-2 (8.7 and 17.5 nmol) given into the same VTA site did not produce any effect, but at a high dose (35 nmol) produced CPP. EM-1 (3.3 nmol) or EM-2 (17.5 nmol) microinjected into the nigrostriatal substantia nigra was not significantly different from vehicle-injected groups. D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2) (CTOP) at 94.13 pmol or 3-methoxynaltrexone at 0.64 pmol microinjected into the posterior Acb shell blocked EM-1-induced CPP and EM-2-induced CPA. At a higher dose, CTOP (941.3 pmol) and 3-methoxynaltrexone (6.4 pmol) produced CPA and CPP, respectively. Coadministration with antiserum against dynorphin A(1-17) (Dyn) (10 microg) microinjected into the posterior Acb shell blocked EM-2-induced CPA. However, it did not affect EM-1-induced CPP. It is concluded that EM-1 and EM-2 produce site-dependent CPP and CPA, respectively, by stimulation of different subtypes of mu-opioid-receptors; stimulation of one subtype of mu-opioid-receptor at the posterior Acb shell and VTA by EM-1 induces CPP, whereas stimulation of another subtype of mu-opioid receptor at the posterior Acb shell, but not the VTA, by EM-2 induces the release of Dyn to produce CPA.


Assuntos
Condicionamento Psicológico/efeitos dos fármacos , Naltrexona/análogos & derivados , Núcleo Accumbens/efeitos dos fármacos , Oligopeptídeos/farmacologia , Somatostatina/análogos & derivados , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Dinorfinas/imunologia , Dinorfinas/farmacologia , Masculino , Microinjeções , Naltrexona/farmacologia , Núcleo Accumbens/fisiologia , Ratos , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Soro/metabolismo , Somatostatina/farmacologia , Percepção Espacial/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , Substância Negra/fisiologia , Área Tegmentar Ventral/fisiologia
13.
Neurosci Lett ; 356(2): 145-7, 2004 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-14746884

RESUMO

The G-protein activations induced by kappa-opioid receptor agonists, (-)U50,488H, U69,593 and TRK-820 in the mouse lower midbrain, striatum and limbic forebrain were determined by monitoring guanosine-5'-o-(3-[35S]thio)triphosphate ([35S]GTP gamma S) binding. All kappa-opioid receptor agonists produced approximately 40, 20 and 10% increases of [35S]GTP gamma S binding over baseline in the lower midbrain, striatum and limbic forebrain, respectively. The increases of [35S]GTP gamma S binding induced by kappa-opioid receptor agonists were completely reversed by the selective kappa-opioid receptor antagonist, norbinaltorphimine (norBNI), in all brain regions. The intrinsic activities of kappa-opioid receptor agonists for G-protein activation in brain regions observed in the present study are not correlated with densities of kappa-opioid receptor binding sites from previous reports. The present results suggest that the catalytic efficiency of kappa-opioid receptor-G-protein coupling may be variable in different brain regions.


Assuntos
Encéfalo/efeitos dos fármacos , Proteínas de Ligação ao GTP/efeitos dos fármacos , Naltrexona/análogos & derivados , Receptores Opioides kappa/agonistas , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Benzenoacetamidas/farmacologia , Encéfalo/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Masculino , Camundongos , Morfinanos/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes , Pirrolidinas/farmacologia , Receptores Opioides kappa/antagonistas & inibidores , Compostos de Espiro/farmacologia
14.
J Pharmacol Exp Ther ; 307(3): 1135-41, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14557378

RESUMO

We have previously demonstrated that both endomorphin-1 (EM-1) and endomorphin-2 (EM-2) at high doses (1.75-35 nmol) given intrathecally (i.t.) or intracerebroventricularly produce antinociception by stimulation of mu-opioid receptors. Now, we report that EM-2 at small doses (0.05-1.75 nmol), which injected alone did not produce antinociception, produces anti-analgesia against opioid agonist-induced antinociception. The tail-flick (TF) response was used to test the antinociception in male CD-1 mice. Intrathecal pretreatment with EM-2 (0.02-1.75 nmol) 45 min before i.t. morphine (3.0 nmol) injection dose dependently attenuated morphine-induced TF inhibition. On the other hand, a similar dose of EM-1 (1.64 nmol) failed to produce any antianalgesic effect. The EM-2 (1.75 nmol)-produced anti-analgesia against morphine-induced TF inhibition was blocked by i.t. pretreatment with the mu-opioid antagonist naloxone or 3-methoxynaltrexone, but not delta-opioid receptor antagonist naltrindole, kappa-opioid receptor antagonist nor-binaltorphimine, or N-methyl-d-aspartate (NMDA) receptor antagonist MK-801. The EM-2-induced antianalgesic effect against morphine-induced TF inhibition was blocked by i.t. pretreatment with antiserum against dynorphin A(1-17), but not beta-endorphin, [Met]-enkephalin, [Leu]-enkephalin, or cholecystokinin antiserum (200 microg each). The i.t. EM-2 pretreatment also attenuated the TF inhibition induced by other mu-opioid agonists, [d-Ala2,N-Me-Phe4,Gly-ol5]-enkephalin, EM-1 and EM-2, delta-opioid agonist deltorphin II, and kappa-opioid agonist (trans)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl]benzeneacetamide methane-sulfonate hydrate (U50,488H). It is concluded that EM-2 at subanalgesic doses presumably stimulates a subtype of mu-opioid receptor and subsequently induces the release of dynorphin A(1-17) to produce antianalgesic effects against mu-, delta-, or kappa-agonists-induced antinociception. The EM-2-induced antianalgesia is not mediated by the release of [Met]-enkephalin, [Leu]-enkephalin, beta-endorphin, or cholecystokinin, nor does it involve kappa- or delta-opioid or NMDA receptors in the spinal cord.


Assuntos
Analgésicos Opioides/farmacologia , Dinorfinas/fisiologia , Naltrexona/análogos & derivados , Oligopeptídeos/farmacologia , Medula Espinal/efeitos dos fármacos , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/antagonistas & inibidores , Animais , Anticorpos Bloqueadores/farmacologia , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Dinorfinas/antagonistas & inibidores , Antagonistas de Aminoácidos Excitatórios/farmacologia , Injeções Espinhais , Masculino , Camundongos , Morfina/antagonistas & inibidores , Morfina/farmacologia , Naloxona/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Receptores Opioides/agonistas
15.
J Pharmacol Exp Ther ; 303(2): 867-73, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388674

RESUMO

Intrathecal (i.t.) pretreatments with antisense oligodeoxynucleotides (AS ODNs) against exon-1, -4, or -8 of mu-opioid receptor clone (MOR-1) to knockdown different variants of MOR-1 on the antinociception induced by endomorphin-1, enomorphin-2, or [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO) given i.t. were investigated in male CD-1 mice. The antinociception was measured with the tail-flick test. AS ODNs against exon-1 (5 microg) given i.t. once daily for 3 days attenuated the antinociception induced by endomorphin-1 and endomorphin-2 with the dose-response curves shifted to the right by 4.5- and 5.3-fold, respectively. AS ODNs against exon-4 (5 microg) attenuated the antinociception induced by endomorphin-1 and endomorphin-2 with the dose-response curves shifted to the right by 2.4- and 5.3-fold, respectively. However, AS ODNs against exon-8 (5 microg) attenuated only the antinociception induced by endomorphin-1, but not endomorphin-2 with the dose-response curves shifted to the right by 3.9- and 1.3-fold, respectively. One more day of pretreatment with antisense probes failed to further reduce the antinociception. The antinociception induced by DAMGO was attenuated by i.t. pretreatment with AS ODNs directed against exon-1, and, to a lesser extent, by AS ODNs directed against exon-8. The mismatch AS ODNs against respective exon-1, -4, and -8 failed to exert significant effects. The selective actions of antisense probes directed against different exons of the MOR-1 in attenuating the antinociception induced by endomorphin-1, endomorphin-2, and DAMGO suggest that multiple splice variants of the MOR-1 exist and support the view that different subtypes of mu-opioid receptors are involved in antinociception induced by endomorphin-1, endomorphin-2, and DAMGO.


Assuntos
Analgésicos Opioides/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Medula Espinal/fisiologia , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Éxons/genética , Injeções Espinhais , Masculino , Camundongos , Oligonucleotídeos Antissenso/administração & dosagem , Oligopeptídeos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Receptores Opioides mu/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA