Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 358
Filtrar
1.
Peptides ; 87: 20-27, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27840228

RESUMO

Brain region-specific expression of proteolytic enzymes can control the biological activity of endogenous neuropeptides and has recently been targeted for the development of novel drugs, for neuropathic pain, cancer, and Parkinson's disease. Rapid and sensitive analytical methods to profile modulators of enzymatic activity are important for finding effective inhibitors with high therapeutic value. Combination of in situ enzyme histochemistry with MALDI imaging mass spectrometry allowed developing a highly sensitive method for analysis of brain-area specific neuropeptide conversion of synthetic and endogenous neuropeptides, and for selection of peptidase inhibitors that differentially target conversion enzymes at specific anatomical sites. Conversion and degradation products of Dynorphin B as model neuropeptide and effects of peptidase inhibitors applied to native brain tissue sections were analyzed at different brain locations. Synthetic dynorphin B (2pmol) was found to be converted to the N-terminal fragments on brain sections whereas fewer C-terminal fragments were detected. N-ethylmaleimide (NEM), a non-selective inhibitor of cysteine peptidases, almost completely blocked the conversion of dynorphin B to dynorphin B(1-6; Leu-Enk-Arg), (1-9), (2-13), and (7-13). Proteinase inhibitor cocktail, and also incubation with acetic acid displayed similar results. Bioconversion of synthetic dynorphin B was region-specific producing dynorphin B(1-7) in the cortex and dynorphin B (2-13) in the striatum. Enzyme inhibitors showed region- and enzyme-specific inhibition of dynorphin bioconversion. Both phosphoramidon (inhibitor of the known dynorphin converting enzyme neprilysin) and opiorphin (inhibitor of neprilysin and aminopeptidase N) blocked cortical bioconversion to dynorphin B(1-7), wheras only opiorphin blocked striatal bioconversion to dynorphin B(2-13). This method may impact the development of novel therapies with aim to strengthen the effects of endogenous neuropeptides under pathological conditions such as chronic pain. Combining histochemistry and MALDI imaging MS is a powerful and sensitive tool for the study of inhibition of enzyme activity directly in native tissue sections.


Assuntos
Encéfalo/metabolismo , Dinorfinas/metabolismo , Endorfinas/metabolismo , Neuropeptídeos/metabolismo , Animais , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Dinorfinas/antagonistas & inibidores , Dinorfinas/química , Dinorfinas/isolamento & purificação , Endorfinas/antagonistas & inibidores , Endorfinas/química , Endorfinas/isolamento & purificação , Glicopeptídeos/administração & dosagem , Humanos , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/química , Neuropeptídeos/isolamento & purificação , Oligopeptídeos/administração & dosagem , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Inibidores de Proteases/administração & dosagem , Ratos , Proteínas e Peptídeos Salivares/administração & dosagem , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
2.
Can J Physiol Pharmacol ; 94(9): 955-60, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27285300

RESUMO

The present study focused on the interactive pain regulation of endokinin A/B (EKA/B, the common C-terminal decapeptide in EKA and EKB) or endokinin C/D (EKC/D, the common C-terminal duodecapeptide in EKC and EKD) on chimeric peptide MCRT (YPFPFRTic-NH2, based on YPFP-NH2 and PFRTic-NH2) at the supraspinal level in mice. Results demonstrated that the co-injection of nanomolar EKA/B and MCRT showed moderate regulation, whereas 30 pmol EKA/B had no effect on MCRT. The combination of EKC/D and MCRT produced enhanced antinociception, which was nearly equal to the sum of the mathematical values of single EKC/D and MCRT. Mechanism studies revealed that pre-injected naloxone attenuated the combination significantly compared with the equivalent analgesic effects of EKC/D alone, suggesting that EKC/D and MCRT might act on two totally independent pathways. Moreover, based on the above results and previous reports, we made two reasonable hypotheses to explain the cocktail-induced analgesia, which may potentially pave the way to explore the respective regulatory mechanisms of EKA/B, EKC/D, and MCRT and to better understand the complicated pain regulation of NK1 and µ opioid receptors, as follows: (1) MCRT and endomorphin-1 possibly activated different µ subtypes; and (2) picomolar EKA/B might motivate the endogenous NPFF system after NK1 activation.


Assuntos
Endorfinas/farmacologia , Medição da Dor/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/farmacologia , Taquicininas/farmacologia , Analgésicos/administração & dosagem , Analgésicos/farmacologia , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Endorfinas/administração & dosagem , Endorfinas/antagonistas & inibidores , Infusões Intraventriculares , Masculino , Camundongos , Naloxona/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Precursores de Proteínas/administração & dosagem , Precursores de Proteínas/antagonistas & inibidores , Taquicininas/administração & dosagem , Taquicininas/antagonistas & inibidores
3.
Regul Pept ; 179(1-3): 23-8, 2012 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-22960407

RESUMO

A chimeric opioid peptide (MCRT, YPFPFRTic-NH(2)) was here designed and synthesized. This peptide was based on morphiceptin (YPFP-NH(2)) and a neuropeptide FF (NPFF) derivative (PFRTic-NH(2)) sharing one proline. This peptide is intended to produce potent analgesia. MCRT was found to induce analgesic activity in a dose- and time-dependent manner, as indicated by a tail flick latency test in mice to which it had been intracerebroventricularly administered (5-60 min, 0.025-2.5 nmol/kg (0.5-50 pmol per mouse), ED(50)=1.49 nmol/kg). At 2.5nmol/kg, MCRT showed significantly higher levels of analgesic activity than morphiceptin or PFR(Tic)amide at 2500 nmol/kg. Naltrindole and cyprodime were found to partially but significantly inhibit this analgesic activity, but naloxone blocked it completely. The kappa opioid receptor antagonist nor-BNI was found to slightly inhibit MCRT and morphiceptin. Pre-injection of BIBP3226 and co-administration of NPFF and MCRT showed that NPFF receptors were involved in the analgesia of MCRT. BIBP3226 was found to weaken the analgesic effects of MCRT, but BIBP3226 could not block the analgesic effects of PFR(Tic)amide. Overall, MCRT was found to have stronger analgesic activity than morphiceptin or PFR(Tic)amide when interacting with mixed µ/δ opioid receptor interactions. MCRT also showed partial interaction with NPFF receptors.


Assuntos
Analgésicos Opioides/farmacologia , Endorfinas/farmacologia , Neuropeptídeos/farmacologia , Peptídeos Opioides/farmacologia , Receptores de Neuropeptídeos/metabolismo , Tetra-Hidroisoquinolinas/farmacologia , Analgesia/métodos , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/síntese química , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Relação Dose-Resposta a Droga , Endorfinas/administração & dosagem , Endorfinas/antagonistas & inibidores , Cobaias , Masculino , Camundongos , Morfinanos/farmacologia , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Neuropeptídeos/administração & dosagem , Neuropeptídeos/metabolismo , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/síntese química , Prolina/metabolismo , Receptores de Neuropeptídeos/antagonistas & inibidores , Tetra-Hidroisoquinolinas/administração & dosagem , Tetra-Hidroisoquinolinas/metabolismo , Fatores de Tempo
4.
Pharmacol Biochem Behav ; 93(3): 291-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19258022

RESUMO

Previously, we found that distinct brain areas predict individual selection bias in decisions between small immediate ("Now") and larger delayed rewards ("Later"). Furthermore, such selection bias can be manipulated by endogenous opioid blockade. To test whether blocking endogenous opioids with naltrexone (NTX) alters brain activity during decision-making in areas predicting individual bias, we compared fMRI BOLD signal correlated with Now versus Later decision-making after acute administration of NTX (50 mg) or placebo. We tested abstinent alcoholics and control subjects in a double-blind two-session design. We defined regions of interest (ROIs) centered on activation peaks predicting Now versus Later selection bias. NTX administration significantly increased BOLD signal during decision-making in the right lateral orbital gyrus ROI, an area where enhanced activity during decision-making predicts Later bias. Exploratory analyses identified additional loci where BOLD signal during decision-making was enhanced (left orbitofrontal cortex, left inferior temporal gyrus, and cerebellum) or reduced (right superior temporal pole) by NTX. Additional analyses identified sites, including the right lateral orbital gyrus, in which NTX effects on BOLD signal predicted NTX effects on selection bias. These data agree with opioid receptor expression in human frontal and temporal cortices, and suggest possible mechanisms of NTX's therapeutic effects.


Assuntos
Tomada de Decisões/efeitos dos fármacos , Endorfinas/antagonistas & inibidores , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Adulto , Alcoolismo/patologia , Alcoolismo/psicologia , Mapeamento Encefálico , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Comportamento de Escolha/efeitos dos fármacos , DNA/genética , Endorfinas/genética , Feminino , Genótipo , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Rede Nervosa/anatomia & histologia , Oxigênio/sangue , Polimorfismo de Nucleotídeo Único , Escalas de Graduação Psiquiátrica , Receptores Opioides mu/genética , Adulto Jovem
5.
Curr Pharm Des ; 12(8): 1001-12, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16533167

RESUMO

Beta-endorphin were detected in the endocrine pancreas and seem able to influence insulin and glucagon release. Hence, endogenous opioids could have a role in glucoregulation and in the pathogenesis of obesity beyond the previously detected effects on appetite. Metabolic abnormalities, such as hyperinsulinemia, insulin-resistance and obesity, are common features of polycystic ovary syndrome (PCOS), and seem to have a pathogenetic role in this disorder. A link between opioids and PCOS-related hyperinsulinism is suggested by the finding of altered central opioid tone and elevated beta-endorphins levels, directly correlated with body weight, in these patients. Furthermore, naloxone and naltrexone significantly reduce the insulin response to glucose load only in hyperinsulinemic PCOS patients. This effect is obtained chiefly through an improvement of insulin clearance. Naltrexone is also able to ameliorate the abnormal gonadotrophins secretion and to improve the ovarian responsiveness in obese PCOS women undergoing ovulation induction with exogenous GnRH. Such effects are believed to be obtained through an amelioration of hyperinsulinemia. Gonadal steroids modulate the opioid system both centrally and in peripheral districts. Nevertheless, the decline of ovarian function does not abolish the opioidergic control of glucoregulation. Post-menopausal period is characterised by a high prevalence of hyperinsulinemia and insulin-resistance. In particular, an association between hyperinsulinemia and increased opioid activity was found in postmenopausal women showing a central body fat distribution. Both naloxone and naltrexone ameliorate the metabolic imbalance also when it appears in the climacteric period, and mainly by increasing insulin clearance. The benefits of naltrexone may represent in the future a useful tool for the treatment of women with hyperinsulinism in the clinical practice.


Assuntos
Endorfinas/antagonistas & inibidores , Hiperinsulinismo/tratamento farmacológico , Resistência à Insulina , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Síndrome do Ovário Policístico/tratamento farmacológico , Ensaios Clínicos como Assunto , Endorfinas/metabolismo , Feminino , Humanos , Hiperinsulinismo/metabolismo , Menopausa/metabolismo , Naloxona/uso terapêutico , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Síndrome do Ovário Policístico/metabolismo
6.
Psychosom Med ; 66(2): 198-206, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15039504

RESUMO

OBJECTIVE: Sex differences in pain sensitivity and stress reactivity have been well documented. Little is known about the role of the endogenous opioid system in these differences. This study was conducted to compare adrenocortical, pain sensitivity, and blood pressure responses to opioid blockade using naltrexone in men and women. METHODS: Twenty-six participants completed 2 sessions during which placebo or 50 mg of naltrexone was administered, using a double-blind, counterbalanced design. Thermal pain threshold and heat tolerance were assessed. Participants also rated pain during a 90-second cold pressor test (CPT) and completed the McGill Pain Questionnaire (MPQ) after each pain challenge. Blood and saliva samples and cardiovascular and mood measures were obtained throughout the sessions. RESULTS: Plasma cortisol, adrenocorticotropin, beta endorphin, prolactin, and salivary cortisol levels increased similarly in men and women after naltrexone administration compared with placebo. Women reported more pain during both pain procedures and had lower thermal pain tolerance. In response to naltrexone, women exhibited reduced blood pressure responses and reduced MPQ pain ratings after CPT. No effects of naltrexone on these measures were found in men. CONCLUSIONS: Although men and women exhibited similar hormonal responses to opioid receptor blockade, women reported less pain and showed smaller blood pressure responses during CPT. Results suggest differential effects of the endogenous opioid system on pain perception and blood pressure in men and women.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Endorfinas/antagonistas & inibidores , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Hormônio Adrenocorticotrópico/sangue , Pressão Sanguínea/efeitos da radiação , Método Duplo-Cego , Endorfinas/fisiologia , Feminino , Temperatura Alta , Humanos , Hidrocortisona/análise , Hidrocortisona/sangue , Masculino , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Placebos , Prolactina/sangue , Saliva/química , Fatores Sexuais , beta-Endorfina/sangue
7.
Biosci Biotechnol Biochem ; 67(11): 2501-4, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14646221

RESUMO

The effects of intracerebroventricular (i.c.v.) injection of bovine beta-casomorphin-5 (beta-CM-5: Tyr-Pro-Phe-Pro-Gly), a micro-opioid agonist derived from milk beta-casein, on step-down type passive avoidance tasks were investigated in mice. Intracerebroventricular administration of a high dose (10 microg) of beta-CM-5 produced a significant decrease in step-down latency. beta-Funaltrexamine (5 microg, i.c.v.) almost completely reversed the beta-CM-5-induced shortening of step-down latency, although neither naltrindole (5 ng, i.c.v.) nor nor-binaltorphimine (5 microg, i.c.v.) had any significant influence on the effect of beta-CM-5. Meanwhile, a low dose (0.5 microg, i.c.v.) of beta-CM-5 inhibited scopolamine (1 mg/kg)-induced impairment of passive avoidance response. These results indicated that a high dose of beta-CM-5 induces amnesia, whereas a low dose ameliorates scopolamine-induced amnesia.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Endorfinas/farmacologia , Naltrexona/análogos & derivados , Fragmentos de Peptídeos/farmacologia , Tempo de Reação/efeitos dos fármacos , Analgésicos/farmacologia , Animais , Ventrículos Cerebrais/efeitos dos fármacos , Ventrículos Cerebrais/fisiologia , Endorfinas/administração & dosagem , Endorfinas/antagonistas & inibidores , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/antagonistas & inibidores , Escopolamina/farmacologia
8.
J Neuroimmunol ; 141(1-2): 30-9, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12965251

RESUMO

Neuroimmune interactions control pain through activation of opioid receptors on sensory nerves by immune-derived opioid peptides. Here we evaluate mechanisms of intrinsic pain inhibition at different stages of Freund's adjuvant-induced inflammation of the rat paw. We use immunohistochemistry and paw pressure testing. Our data show that in early (6 h) inflammation leukocyte-derived beta-endorphin, met-enkephalin and dynorphin A activate peripheral mu-, delta- and kappa-receptors to inhibit nociception. In addition, central opioid mechanisms seem to contribute significantly to this effect. At later stages (4 days), antinociception is exclusively produced by leukocyte-derived beta-endorphin acting at peripheral mu and delta receptors. Corticotropin-releasing hormone (CRH) is an endogenous trigger of these effects at both stages. These findings indicate that peripheral opioid mechanisms of pain inhibition gain functional relevance with the chronicity of inflammation.


Assuntos
Dor/patologia , Dor/prevenção & controle , Animais , Hormônio Liberador da Corticotropina/administração & dosagem , Dinorfinas/antagonistas & inibidores , Dinorfinas/biossíntese , Dinorfinas/fisiologia , Edema/imunologia , Edema/metabolismo , Edema/fisiopatologia , Endorfinas/antagonistas & inibidores , Endorfinas/biossíntese , Endorfinas/fisiologia , Encefalina Metionina/antagonistas & inibidores , Encefalina Metionina/biossíntese , Encefalina Metionina/fisiologia , Adjuvante de Freund/administração & dosagem , Membro Posterior , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Injeções Subcutâneas , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Leucócitos/fisiologia , Masculino , Naloxona/administração & dosagem , Dor/imunologia , Limiar da Dor , Ratos , Ratos Wistar , Estresse Fisiológico/imunologia , Estresse Fisiológico/metabolismo , Estresse Fisiológico/fisiopatologia , Fatores de Tempo
9.
Life Sci ; 73(20): 2591-602, 2003 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-12967683

RESUMO

This study was conducted to more clearly delineate the possible role of endogenous opioid receptors and opioid peptides in general anesthesia-associated hypotension in rats. Exposure to 2% isoflurane in oxygen produced a triphasic change in mean arterial pressure (MAP), including an early phase in which MAP fell by -28.4 +/- 2.2%. The magnitude of this early-phase hypotension was attenuated in rats pretreated with intravenous (i.v.) mu-subtype-selective doses of either naloxone or methylnaloxone but not central doses of the selective mu-opioid antagonist beta-funaltrexamine. This early hypotensive phase was also reduced following i.v. pretreatment with antiserum against methionine-enkephalin but not beta-endorphin. These findings suggest that early-phase isoflurane-induced hypotension may be due to activation of peripheral mu-opioid receptors by an endogenous opioid peptide, possibly related to methionine-enkephalin.


Assuntos
Anestésicos Inalatórios/farmacologia , Endorfinas/fisiologia , Hipotensão/induzido quimicamente , Isoflurano/farmacologia , Naltrexona/análogos & derivados , Receptores Opioides/fisiologia , Anestésicos Intravenosos/farmacologia , Animais , Anticorpos Bloqueadores/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Endorfinas/antagonistas & inibidores , Encefalina Metionina/farmacologia , Temperatura Alta , Injeções Intraventriculares , Masculino , Naloxona/administração & dosagem , Naloxona/farmacologia , Naltrexona/efeitos adversos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Uretana/farmacologia , beta-Endorfina/farmacologia
10.
Br J Sports Med ; 35(4): 223-30, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11477013

RESUMO

OBJECTIVES: The immune changes induced by a bout of prolonged and vigorous exercise have been suggested to be a useful experimental model of sepsis and the inflammatory response. Available literature was reviewed to evaluate this hypothesis. METHODS: Literature describing the immune response to various patterns of exercise was compared with data on the immune changes observed during sepsis and inflammation. RESULTS: Although there are qualitative similarities between the immune responses to exercise and sepsis, the magnitude of the changes induced by most forms of exercise remains much smaller than in a typical inflammatory response. Indeed, the exercise induced changes in some key elements such as plasma cytokine concentrations are too small to be detected reliably by current technology. CONCLUSIONS: If exercise is to provide a valid model of sepsis and the inflammatory response, it will be necessary to focus on subjects who are willing to exercise extremely hard, to use the pattern of exercise that has the greatest effect on the immune system, and to combine this stimulus with other psychological, environmental, or nutritional stressors.


Assuntos
Exercício Físico/fisiologia , Inflamação/imunologia , Modelos Imunológicos , Sepse/imunologia , Anti-Inflamatórios/farmacologia , Citocinas/sangue , Citocinas/metabolismo , Endorfinas/antagonistas & inibidores , Humanos , Células Matadoras Naturais/imunologia , Leucócitos/imunologia , Fisiologia Comparada , Estudos Prospectivos , RNA Mensageiro/imunologia , Estresse Fisiológico/imunologia
11.
J Physiol Pharmacol ; 51(2): 359-68, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10898106

RESUMO

The aim of the present studies was to investigate (1) the presence of LH receptor (LHR) in porcine separated small (SLCs) and large (LLCs) luteal cells taken from mid-luteal corpora lutea and (2) the influence of opioid agonist, FK 33-824 (FK) on LHR gene expression in these cells. Immunocytochemistry revealed intense staining for LHR in both SLCs and LLCs. Reverse transcription-polymerase chain reaction (RT-PCR) and Southern hybridization were used to check the effect of FK and hCG on LHR gene expression. The LHR gene expression was observed in non-stimulated LLCs and in both types of cells after treatment with FK or hCG. FK stimulated LHR gene expression in SLCs and inhibited the gene expression in LLCs. Moreover, FK inhibited and potentiated stimulatory influence of hCG on the gene expression in LLCs and SLCs, respectively. These results suggest that LHR gene expression in porcine luteal cells can be regulated by opioid peptides.


Assuntos
Corpo Lúteo/metabolismo , Endorfinas/fisiologia , Expressão Gênica , Receptores do LH/genética , Receptores do LH/metabolismo , Animais , Tamanho Celular , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Corpo Lúteo/citologia , Corpo Lúteo/efeitos dos fármacos , D-Ala(2),MePhe(4),Met(0)-ol-encefalina/farmacologia , Endorfinas/antagonistas & inibidores , Feminino , Expressão Gênica/efeitos dos fármacos , Suínos , Distribuição Tecidual
12.
Peptides ; 21(5): 717-22, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10876055

RESUMO

Intraplantar injection of the enzymatically stable, N-methylated kyotorphin analog Tyr(NMe)-Arg-OH produced marked and sharp nociceptive flexor responses in a dose-dependent manner. A significant response was observed with this compound at a dose of 0. 01 amol (6000 molecules). Tyr(NMe)-Arg-OH-nociception was completely blocked by the kyotorphin antagonist leucyl-arginine and its enzymatically stable, N-methylated analog, as well as by CP-99994, a specific neurokinin 1 antagonist. These findings suggest that the nociceptive effect produced by Tyr(NMe)-Arg-OH in subattomol doses occurs via specific interaction with the kyotorphin receptor and that the extraordinary potency observed may result from amplification through local substance P release.


Assuntos
Endorfinas/química , Endorfinas/farmacologia , Dor/induzido quimicamente , Analgésicos/farmacologia , Animais , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Endorfinas/antagonistas & inibidores , Masculino , Camundongos , Antagonistas dos Receptores de Neurocinina-1
14.
Hypertension ; 29(1 Pt 1): 105-10, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9039088

RESUMO

To investigate the involvement of endogenous opioids in acute increases in blood pressure and their functional relationship with atrial natriuretic factor and endothelin-1, we assessed plasma levels of beta-endorphin, met-enkephalin, dynorphin B, catecholamines, atrial natriuretic factor, and endothelin-1 before and after administration of the opioid antagonist naloxone hydrochloride (8 mg i.v.) in 28 hypertensive patients with a stress-induced acute increase in blood pressure. Ten patients with established mild or moderate essential hypertension and 10 normotensive subjects served as control groups. Opioids, atrial natriuretic factor, and endothelin-I were radioimmunoassayed after chromatographic preextraction; catecholamines were determined by high-performance liquid chromatography with electrochemical detection. Patients with an acute increase in blood pressure (systolic, 203.2 +/- 2.2 mm Hg; diastolic, 108.4 +/- 1.3) had plasma opioid, catecholamine, and atrial natriuretic factor levels significantly (P < .01) higher than hypertensive control patients (systolic pressure, 176.4 +/- 1.0 mm Hg; diastolic, 100.0 +/- 1.4), who had a hormonal pattern similar to that of normotensive subjects (systolic pressure, 123.2 +/- 1.5 mm Hg; diastolic, 75.0 +/- 2.0). Endothelin-1 did not differ in any group. In patients with an acute increase in blood pressure, naloxone significantly (P < .01) reduced blood pressure, heart rate, opioids, catecholamines, and atrial natriuretic factor 10 minutes after administration. Naloxone effects on blood pressure, heart rate, opioids, and catecholamines wore off within 20 minutes. In control groups, naloxone failed to modify any of the considered parameters. Our findings suggest that pressor effects of opioid peptides mediated by the autonomic nervous system during stress-induced acute episodes of blood pressure increase in hypertensive patients.


Assuntos
Pressão Sanguínea/fisiologia , Hipertensão/fisiopatologia , Peptídeos Opioides/fisiologia , Fator Natriurético Atrial/antagonistas & inibidores , Fator Natriurético Atrial/sangue , Fator Natriurético Atrial/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Dinorfinas/antagonistas & inibidores , Dinorfinas/sangue , Dinorfinas/fisiologia , Endorfinas/antagonistas & inibidores , Endorfinas/sangue , Endorfinas/fisiologia , Endotelina-1/antagonistas & inibidores , Endotelina-1/sangue , Endotelina-1/fisiologia , Encefalina Metionina/antagonistas & inibidores , Encefalina Metionina/sangue , Encefalina Metionina/fisiologia , Feminino , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Humanos , Hipertensão/sangue , Hipertensão/etiologia , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Naloxona/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Norepinefrina/antagonistas & inibidores , Norepinefrina/sangue , Norepinefrina/fisiologia , Peptídeos Opioides/antagonistas & inibidores , Peptídeos Opioides/sangue , Estresse Fisiológico/complicações , Estresse Psicológico/complicações , beta-Endorfina/antagonistas & inibidores , beta-Endorfina/sangue , beta-Endorfina/fisiologia
16.
Brain Res ; 743(1-2): 86-92, 1996 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-9017234

RESUMO

The aim of the present study was to evaluate the action of the descending modulation system from the locus coeruleus (LC) in a rat model of unilateral hyperalgesic inflammation. Unilateral hindlimb inflammation was produced by a subcutaneous injection of carrageenan (6 mg in 0.15 ml saline). One week before testing, rats received bilateral lesions of the LC using anodal current. Nociception was assessed by measuring withdrawal of the paw from a noxious thermal stimulus. Four hours after carrageenan injection, paw withdrawal latencies (PWLs) in the inflamed paw of the LC-lesioned rats were significantly shorter than those of the sham-operated rats. This difference in PWL between the two groups was not observed at 7 days, whereas edema and hyperalgesia still remained in the inflamed paw. At 4 h, systemic naloxone produced a further decrease of the PWL in the LC-lesioned rats but not in the sham-operated rats. These results suggest that inflammation-induced activation of the descending modulation system from the LC occurs in only the acute phase of inflammation and that a decrease in the extent of the development of hyperalgesia in the acute phase of inflammation might depend on the interaction between the descending modulation system from the LC and the opioid inhibitory system.


Assuntos
Endorfinas/fisiologia , Hiperalgesia/fisiopatologia , Inflamação/fisiopatologia , Locus Cerúleo/fisiologia , Naloxona/farmacologia , Dor/fisiopatologia , Análise de Variância , Animais , Carragenina , Modelos Animais de Doenças , Vias Eferentes/efeitos dos fármacos , Vias Eferentes/fisiologia , Endorfinas/antagonistas & inibidores , Membro Posterior , Hiperalgesia/induzido quimicamente , Inflamação/induzido quimicamente , Locus Cerúleo/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
17.
J Neuroendocrinol ; 8(8): 647-52, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8866253

RESUMO

The present study was undertaken to evaluate the role and possible interaction of the endogenous opioid peptide (EOP) and corticotropin-releasing factor (CRF) in the acute stress-induced suppression of gonadotropin secretion in ovariectomized estrogen-primed rats. An intravenous (i.v.) injection of naloxone (10 or 20 mg/kg), an EOP antagonist, significantly elevated serum luteinizing hormone (LH) levels within 10 min in non-stressed animals. The naloxone-induced LH release was completely eliminated when tested 30 min after the onset of acute immobilization. In a subsequent study, it was found that suppression of the naloxone-induced LH release occurred as early as 5 min after the stress onset, and was still evident 60 min after the end of a 30-min period of immobilization. The effect of naloxone was restored 3 h after liberation of the animal from the 30-min immobilization. An intraventricular (i.c.v.) injection of CRF (1 or 5 micrograms) also significantly suppressed, in a dose-related manner, the effect of a subsequent i.v. injection of naloxone. However, an i.c.v. injection of alpha-helical CRF(9-41) (25 or 50 micrograms), a CRF antagonist, prior to immobilization, could not interfere with the suppressive effect of stress on naloxone-induced LH release. These results suggest that both acute immobilization stress and CRF can inhibit the LH secretory activity without mediation by EOP neurons. However, the stress-related suppression may involve non-CRF mechanism(s).


Assuntos
Hormônio Liberador da Corticotropina/farmacologia , Endorfinas/antagonistas & inibidores , Hormônio Luteinizante/metabolismo , Naloxona/antagonistas & inibidores , Ovário/fisiologia , Estresse Fisiológico/fisiopatologia , Análise de Variância , Animais , Depressão Química , Estrogênios/farmacologia , Feminino , Injeções Intraventriculares , Ovariectomia , Ratos , Ratos Wistar , Restrição Física , Taxa Secretória/efeitos dos fármacos
18.
Neurosci Lett ; 212(1): 1-4, 1996 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-8823748

RESUMO

L-Arginine (L-Arg) is an endogenous substrate for nitric oxide synthase (NOS). In the present study, we examined whether L-tyrosyl-L-Arg (kyotorphin), an endogenous analgesic neuropeptide, might be a substrate for inducible NOS (iNOS) in the brain. Both kyotorphin and L-Arg caused an accumulation of nitrites in lipopolysaccharide (LPS)-treated glial cells cultured from infant rat brains. However, such accumulation of nitrites was not induced by NG-nitro-L-Arg (a NOS inhibitor), L-tyrosyl-D-Arg (D-kyotorphin) or D-Arg. L-Leucyl-L-Arg (an antagonist for kyotorphin receptors) or bestatin (an inhibitor for kyotorphin-hydrolyzing peptidase) did not inhibit the kyotorphin-induced accumulation of nitrites in LPS-treated cells. On the contrary, L-Leucyl-L-Arg caused an accumulation of nitrites in a concentration-dependent manner. The results indicate that nitric oxide (NO) is produced in LPS-treated glial cells directly from kyotorphin through the catalytic action of iNOS.


Assuntos
Analgésicos/metabolismo , Endorfinas/metabolismo , Neuroglia/enzimologia , Óxido Nítrico Sintase/metabolismo , Análise de Variância , Animais , Arginina/metabolismo , Dipeptídeos/farmacologia , Eletroforese em Gel de Poliacrilamida , Endorfinas/antagonistas & inibidores , Lipopolissacarídeos , Ratos , Ratos Wistar , Especificidade por Substrato , Tirosina/metabolismo
20.
Psychopharmacology (Berl) ; 122(4): 330-5, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8657829

RESUMO

Morphiceptin, a selective mu opioid agonist, injected into the medial preoptic area (MPOA), delayed the onset of copulation in male rats, but did not affect genital reflexes, sexual motivation or general motor activity. In a dose-dependent manner, morphiceptin (100 ng and 1000 ng) injected into the MPOA increased mount and intromission latencies. Similar injections of morphiceptin into the ventromedial hypothalamus had no effect on any parameter of copulation. The increase in copulatory latencies following the injection of the highest dose of morphiceptin was blocked by pretreatment with the opioid antagonist naloxone. In the X-maze task, morphiceptin had no effect on sexual motivation, as measured by the percentage of trials on which the male chose the female's chamber, but it increased the number of trials in which the subject did not select a chamber within 60 s and the latency to the female the first time he chose her chamber. Similar to the copulation task, the mount and intromission latencies were also increased in the X-maze, after the male reached the female. Morphiceptin in the MPOA had no effect on ex copula genital reflexes, tested in restrained supine males, or on motor activity, tested in a grid box. These results suggest that morphiceptin disrupts either the specific copulatory somatomotor pattern or a more general motivational component.


Assuntos
Analgésicos/farmacologia , Copulação/efeitos dos fármacos , Endorfinas/farmacologia , Área Pré-Óptica/efeitos dos fármacos , Analgésicos/antagonistas & inibidores , Animais , Endorfinas/antagonistas & inibidores , Feminino , Hipotálamo Médio/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...