Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Brain ; 147(1): 26-38, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-37633259

RESUMO

Restless legs syndrome (RLS) is responsive to opioid, dopaminergic and iron-based treatments. Receptor blocker studies in RLS patients suggest that the therapeutic efficacy of opioids is specific to the opioid receptor and mediated indirectly through the dopaminergic system. An RLS autopsy study reveals decreases in endogenous opioids, ß-endorphin and perhaps Met-enkephalin in the thalamus of RLS patients. A total opioid receptor knock-out (mu, delta and kappa) and a mu-opioid receptor knock-out mouse model of RLS show circadian motor changes akin to RLS and, although both models show sensory changes, the mu-opioid receptor knock mouse shows circadian sensory changes closest to those seen in idiopathic RLS. Both models show changes in striatal dopamine, anaemia and low serum iron. However, only in the total receptor knock-out mouse do we see the decreases in serum ferritin that are normally found in RLS. There are also decreases in serum iron when wild-type mice are administered a mu-opioid receptor blocker. In addition, the mu-opioid receptor knock-out mouse also shows increases in striatal zinc paralleling similar changes in RLS. Adrenocorticotropic hormone and α-melanocyte stimulating hormone are derived from pro-opiomelanocortin as is ß-endorphin. However, they cause RLS-like symptoms and periodic limb movements when injected intraventricularly into rats. These results collectively suggest that an endogenous opioid deficiency is pathogenetic to RLS and that an altered melanocortin system may be causal to RLS as well.


Assuntos
Analgésicos Opioides , Síndrome das Pernas Inquietas , Humanos , Ratos , Camundongos , Animais , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Síndrome das Pernas Inquietas/diagnóstico , Síndrome das Pernas Inquietas/tratamento farmacológico , Melanocortinas/uso terapêutico , beta-Endorfina/uso terapêutico , Ferro , Dopamina
2.
Allergy ; 78(6): 1585-1594, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37129453

RESUMO

BACKGROUND: Pruritus is identified as an adverse drug reaction to arsenic trioxide, but the association of arsenic exposure with pruritus has not been investigated. METHODS: A cross-sectional study was conducted in Shimen, China. A Mendelian randomization analysis was conducted to confirm the causal relationship between genetically predicted percentages of monomethylated arsenic (MMA%) and dimethylated arsenic (DMA%) in urine with chronic pruritus in UK Biobank. A case-control study was then conducted to determine the biomarker for pruritus. Arsenite-treated mice were used to confirm the biomarker, and von Frey test was used to induce scratching bouts. Last, a randomized, double-blind, placebo-controlled trial was conducted to test the efficacy of naloxone in arsenic-exposed patients with pruritus in Shimen. RESULTS: Hair arsenic (µg/g) showed a dose-response relationship with the intensity of itch in 1079 participants, with odds ratios (OR) of 1.11 for moderate-to-severe itch (p = 0.012). The Mendelian randomization analysis confirmed the causal relationship, with ORs of 1.043 for MMA% (p = 0.029) and 0.904 for DMA% (p = 0.077) above versus under median. Serum ß-endorphin was identified as a significant biomarker for the intensity of itch (p < 0.001). Consistently, treatment with arsenite upregulated the level of ß-endorphin (p = 0.002) and induced scratching bouts (p < 0.001) in mice. The randomized controlled trial in 126 participants showed that treatment with sublingual naloxone significantly relieved the intensity of itch in arsenic-exposed participants in 2 weeks (ß = -0.98, p = 0.04). CONCLUSION: Arsenic exposure is associated with pruritus, and ß-endorphin serves as a biomarker of pruritus. Naloxone relieves pruritus in patients with arseniasis.


Assuntos
Arsênio , Arsenitos , Animais , Camundongos , Arsênio/toxicidade , Arsenitos/uso terapêutico , beta-Endorfina/uso terapêutico , Biomarcadores , Estudos de Casos e Controles , Estudos Transversais , Análise da Randomização Mendeliana , Naloxona/uso terapêutico , Prurido/tratamento farmacológico , Prurido/etiologia , Humanos
3.
Environ Toxicol Pharmacol ; 95: 103979, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36174909

RESUMO

The study aimed to examine the genetic contribution to buprenorphine (BUP) treatment in individuals with opioid use disorder (OUD), with a specific focus on BDNF and OPRM1 genes. A total of 113 controls and 111 OUD patients receiving sublingual BUP/naloxone were enrolled. OPRM1 A118G and BDNF Val66Met polymorphisms were investigated by PCR-FRLP. Plasma BDNF and beta-endorphin levels were assessed by ELISA kits in both groups. Blood BUP levels were measured by LC-MS/MS and normalized with daily BUP dose (BUP/D). OPRM1 A118G and BDNF Val66Met polymorphisms didn't have an effect on plasma beta-endorphin and BDNF levels in OUD patients, respectively. Interestingly, OUD patients had significantly higher plasma BDNF and lower beta-endorphin levels compared to the controls (p < 0.001). A negative and significant correlation between plasma BUP/D and BDNF levels was found. Age onset of first use was associated with OPRM1 A118G polymorphism. The findings indicated that sublingual BUP/naloxone may increase plasma BDNF levels, but may decrease beta-endorphin levels in individuals with OUD. Plasma BDNF level seemed to be decreased in a BUP/D concentration-dependent manner.


Assuntos
Buprenorfina , Transtornos Relacionados ao Uso de Opioides , Fator Neurotrófico Derivado do Encéfalo/genética , Buprenorfina/uso terapêutico , Combinação Buprenorfina e Naloxona/uso terapêutico , Cromatografia Líquida , Humanos , Transtornos Relacionados ao Uso de Opioides/complicações , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/genética , Receptores Opioides mu/genética , Receptores Opioides mu/uso terapêutico , Espectrometria de Massas em Tandem , beta-Endorfina/genética , beta-Endorfina/uso terapêutico
4.
Pain Med ; 20(1): 28-36, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30256990

RESUMO

Background: Chronic low back pain (CLBP) is usually quantified using the visual analog scale (VAS). However, the VAS is a subjective measure and prone to reporting bias, therefore making it difficult to differentiate patients with true pain from those seeking to obtain secondary gain. This study aimed to evaluate the feasibility of using plasma ß-endorphin as an objective biomarker for CLBP. Methods: We searched PubMed, Embase, and the Cochrane Library for randomized trials that compared treatment vs sham procedures for patients with CLBP. Changes in VAS and ß-endorphin levels between baseline and final evaluations were assessed for the treatment and control groups. A meta-regression model was developed to evaluate the association between the ß-endorphin level and VAS. Results: We included data from seven trials involving 375 patients. There was no significant difference in VAS scores and ß-endorphin levels between both groups at baseline. At final evaluation, the treatment group demonstrated significantly greater improvements in VAS scores and an increased ß-endorphin level compared with the control group. The change in the plasma ß-endorphin level may be a surrogate marker of treatment response for patients with CLBP (explanatory power: 80%). The plasma ß-endorphin level might be rarely affected by sham procedures. For patients with CLBP, the baseline ß-endorphin level may reflect the intensity of CLBP (explanatory power: 66%). Conclusions: A change in plasma ß-endorphin level may be a surrogate marker of the treatment response for patients with CLBP. Advancements in ß-endorphin measurements may help us better quantify pain intensity.


Assuntos
Dor Crônica/tratamento farmacológico , Dor Lombar/terapia , Medição da Dor , beta-Endorfina/uso terapêutico , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
5.
Alcohol Clin Exp Res ; 39(1): 146-57, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25623413

RESUMO

BACKGROUND: Alcohol exposure has adverse effects on stress physiology and behavioral reactivity. This is suggested to be due, in part, to the effect of alcohol on ß-endorphin (ß-EP)-producing neurons in the hypothalamus. In response to stress, ß-EP normally provides negative feedback to the hypothalamic-pituitary-adrenal axis and interacts with other neurotransmitter systems in the amygdala to regulate behavior. We examined whether ß-EP neuronal function in the hypothalamus reduces the corticosterone response to acute stress, attenuates anxiety-like behaviors, and modulates alcohol drinking in rats. METHODS: To determine whether ß-EP neuronal transplants modulate the stress response, anxiety behavior, and alcohol drinking, we implanted differentiated ß-EP neurons into the paraventricular nucleus (PVN) of the hypothalamus of normal, prenatal alcohol-exposed, and alcohol-preferring (P) and alcohol-non-preferring (NP) rats. We then assessed corticosterone levels in response to acute restraint stress and other markers of stress response in the brain and anxiety-like behaviors in the elevated plus maze and open-field assays. RESULTS: We showed that ß-EP neuronal transplants into the PVN reduced the peripheral corticosterone response to acute stress and attenuated anxiety-like behaviors. Similar transplants completely reduced the hypercorticosterone response and elevated anxiety behaviors in prenatal alcohol-exposed adult rats. Moreover, we showed that ß-EP reduced anxiety behavior in P rats with minimal effects on alcohol drinking during and following restraint stress. CONCLUSIONS: These data further establish a role of ß-EP neurons in the hypothalamus for regulating physiological stress response and anxiety behavior and resemble a potential novel therapy for treating stress-related psychiatric disorders in prenatal alcohol-exposed children and those genetically predisposed to increased alcohol consumption.


Assuntos
Consumo de Bebidas Alcoólicas/terapia , Ansiedade/terapia , Neurônios/transplante , Núcleo Hipotalâmico Paraventricular/cirurgia , Efeitos Tardios da Exposição Pré-Natal/terapia , beta-Endorfina/uso terapêutico , Tonsila do Cerebelo/metabolismo , Animais , Corticosterona/sangue , Hormônio Liberador da Corticotropina/biossíntese , Feminino , Masculino , Aprendizagem em Labirinto , Camundongos Endogâmicos , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Receptores de Hormônio Liberador da Corticotropina/biossíntese , Restrição Física , beta-Endorfina/metabolismo
6.
Endocr Rev ; 33(4): 623-51, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22736674

RESUMO

Proopiomelanocortin (POMC)-derived peptides such as melanocortins and ß-endorphin (ß-ED) exert their pleiotropic effects via binding to melanocortin receptors (MCR) and opioid receptors (OR). There is now compelling evidence for the existence of a functional POMC system within the osteoarticular system. Accordingly, distinct cell types of the synovial tissue and bone have been identified to generate POMC-derived peptides like ß-ED, ACTH, or α-MSH. MCR subtypes, especially MC1R, MC2R (the ACTH receptor), MC3R, and MC4R, but also the µ-OR and δ-OR, have been detected in various cells of the synovium, cartilage, and bone. The respective ligands of these POMC-derived peptide receptors mediate an increasing number of newly recognized biological effects in the osteoarticular system. These include bone mineralization and longitudinal growth, cell proliferation and differentiation, extracellular matrix synthesis, osteoprotection, and immunomodulation. Importantly, bone formation is also regulated by the central melanocortin system via a complex hormonal interplay with other organs and tissues involved in energy metabolism. Among the POMC-derived peptides examined in cell culture systems from osteoarticular tissue and in animal models of experimentally induced arthritis, α-MSH, ACTH, and MC3R-specific agonists appear to have the most promising antiinflammatory actions. The effects of these melanocortin peptides may be exploited in future for the treatment of patients with inflammatory and degenerative joint diseases.


Assuntos
Artrite Reumatoide/metabolismo , Osso e Ossos/metabolismo , Cartilagem/metabolismo , Articulações/metabolismo , Osteoartrite/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/etiologia , Osso e Ossos/efeitos dos fármacos , Cartilagem/efeitos dos fármacos , Humanos , Articulações/efeitos dos fármacos , Melanocortinas/farmacologia , Melanocortinas/uso terapêutico , Osteoartrite/tratamento farmacológico , Osteoartrite/etiologia , Pró-Opiomelanocortina/genética , Transdução de Sinais , beta-Endorfina/farmacologia , beta-Endorfina/uso terapêutico
7.
Clin J Pain ; 23(9): 750-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18075400

RESUMO

INTRODUCTION: Increasing evidence suggests the existence of sex differences in pain perception. Adenosine, an early messenger for myocardial ischemia induces angina pectorislike symptoms in healthy volunteers and in patients with ischemic heart disease. AIMS: To study whether sex influences adenosine-provoked chest pain and the analgesic effect of the opioid receptor agonist beta-endorphin. MATERIALS AND METHODS: Twenty patients (10 male and 10 female) with significant coronary artery disease and 20 healthy volunteers (10 male and 10 female) were studied. Both the hand algometer and Borg CR-10 scale were used to estimate chest pain. Chest pain was provoked double-blind by injections of placebo, 1/3, 2/3, 3/3 of maximal tolerable dose of adenosine twice in randomized order. This procedure was repeated after bolus injection of beta-endorphin followed by infusion and repeated a third time after bolus injection of naloxone 0.8 mg. Central chest pain and physiologic responses were quantified using hemodynamic and psychophysical methods. RESULTS: Pain estimate by hand algometer and the Borg CR-10 scale was correlated (r=0.77, P<0.001). Both sexes reported a dose-dependent increase of adenosine-provoked chest pain with no differences for maximum tolerable dose of adenosine per kilogram. beta-Endorphin administration lowered adenosine-provoked pain in both male patients and male healthy volunteers (P=0.02) but not in women. Naloxone tended to increase the pain perception in male patients (P=0.052) and male healthy volunteers (P=0.054), but did not have any significant effect on pain modalities in female. CONCLUSIONS: In conclusion, women were resistant to beta-endorphin modulation of adenosine-provoked chest pain. In male patients, beta-endorphin induced analgesia.


Assuntos
Adenosina , Dor no Peito/tratamento farmacológico , Dor no Peito/etiologia , Isquemia Miocárdica/complicações , Caracteres Sexuais , beta-Endorfina/uso terapêutico , Adulto , Análise de Variância , Relação Dose-Resposta a Droga , Método Duplo-Cego , Interações Medicamentosas , Eletrocardiografia/métodos , Feminino , Frequência Cardíaca/efeitos dos fármacos , Humanos , Masculino , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor
8.
J Clin Endocrinol Metab ; 90(9): 5167-74, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15956086

RESUMO

CONTEXT: The opioid system is involved in blood pressure regulation in both normal humans and patients with essential hypertension. OBJECTIVE: The objective of the study was to investigate the effects of a high-dose infusion of beta-endorphin, an opioid peptide, on blood pressure and on the hormonal profile in healthy subjects and in hypertensive patients and the mediation played by opioid receptor agonism. DESIGN, SETTING, AND PARTICIPANTS: According to a randomized double-blind design, 11 healthy subjects (controls) and 12 hypertensive inpatients (mean age, 38.9 and 40.4 yr, respectively) received 1-h iv infusion of beta-endorphin (250 mug/h) and, on another occasion, the same infusion protocol preceded by the opioid antagonist naloxone (8 mg). MAIN OUTCOME MEASURES: Hemodynamic and hormonal measurements were performed at established times during the infusion protocols. RESULTS: At baseline, circulating beta-endorphin, norepinephrine, and endothelin-1 in hypertensive patients were significantly (P < 0.05) higher than in controls. In controls, beta-endorphin reduced blood pressure (P < 0.01) and circulating norepinephrine (P < 0.02) and increased plasma atrial natriuretic factor (P < 0.003) and GH (P < 0.0001). In hypertensive patients, beta-endorphin decreased systemic vascular resistance (P < 0.0001), blood pressure (P < 0.0001), and plasma norepinephrine (P < 0.0001) and endothelin-1 (P < 0.0001) and raised circulating atrial natriuretic factor (P < 0.0001), GH (P < 0.0001), and IGF-I (P < 0.0001). These hemodynamic and hormonal responses to beta-endorphin in hypertensive patients were significantly (P < 0.0001) greater than in controls but were annulled in all individuals when naloxone preceded beta-endorphin infusion. CONCLUSIONS: High doses of beta-endorphin induce hypotensive and beneficial hormonal effects in humans, which are enhanced in essential hypertension and are mediated by opioid receptors.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Hormônios/sangue , Hipertensão/tratamento farmacológico , Hipertensão/fisiopatologia , Receptores Opioides/agonistas , beta-Endorfina/administração & dosagem , Adulto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Hemodinâmica/efeitos dos fármacos , Humanos , Hipertensão/sangue , Masculino , Pessoa de Meia-Idade , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , beta-Endorfina/uso terapêutico
9.
Muscle Nerve ; 31(4): 481-6, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15704144

RESUMO

beta-Endorphin and a C-terminal analogue have been shown to decrease muscle fatigue and increase glucose uptake in muscles of normal mice. In order to provide evidence whether these peptides might be useful in muscle-wasting conditions and whether the two actions of the peptides are interdependent, the effect of beta-endorphin on muscle fatigue and glucose uptake was studied using isolated hemidiaphragm preparations of dystrophic mice as well as normal mice. Muscle contractions were elicited by high-frequency stimulation of the phrenic nerve. Glucose uptake was measured using (nonmetabolizable) 2-deoxy-D-[1-(3)H]glucose. beta-Endorphin and the C-terminal analogue reduced fatigue in normal muscles of males but not females. Insulin had no effect in either sex. The peptides increased 2-deoxyglucose uptake in contracting and noncontracting muscles of normal males and females. beta-Endorphin reduced fatigue and increased deoxyglucose uptake in dystrophic muscles. The effect on fatigue was not due to increased glucose uptake, as the energy substrate present was pyruvate. Nerve stimulation released beta-endorphin immunoreactivity from intramuscular nerves of dystrophic mice. It is hypothesized that beta-endorphin released from motor nerves as well as from the pituitary could be responsible for improving muscle function during exercise. beta-Endorphin or analogues could have therapeutic use in muscle-wasting disease.


Assuntos
Glucose/metabolismo , Fadiga Muscular/efeitos dos fármacos , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular Animal/metabolismo , beta-Endorfina/uso terapêutico , Animais , Diafragma/efeitos dos fármacos , Diafragma/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fadiga Muscular/fisiologia , beta-Endorfina/farmacologia
10.
Am Heart J ; 148(3): E13, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15389249

RESUMO

BACKGROUND: Cardiomyocytes produce opioid peptides and receptors. beta-Endorphin is increased in the plasma of patients with congestive heart failure (CHF). We evaluated whether an intravenous infusion of beta-endorphin exerted any effect on cardiovascular function and on the neurohormonal milieu in patients with mild to moderate CHF. METHODS: According to a double-blind, placebo-controlled design, 10 patients (5 men, age 46.9 +/- 8.2 years [mean +/- SD]) with CHF and New York Heart Association functional class II to III received, in random order, 1-hour intravenous infusion of beta-endorphin (500 microg/h) and, on a separate occasion, received placebo and underwent echocardiographic and laboratory measurements at baseline and during infusions. RESULTS: beta-Endorphin significantly increased left ventricular ejection fraction (LVEF) (P =.0001) and stroke volume (P =.0001), and reduced systemic vascular resistance (P =.031) in patients with CHF. These changes were paralleled by a significant increase in plasma levels of glucagon (P =.0001), GH (P =.0001), and IGF-1 (P =.0001), and a significant decrease in plasma levels of endothelin (P =.0001) and catecholamines (P =.01). No hemodynamic and neurohormonal changes were observed during the placebo study in any patient. CONCLUSIONS: We conclude that a short-term, high dose infusion of beta-endorphin improves LVEF, reduces systemic vascular resistance, blunts the neurohormonal activation, and stimulates the GH/IGF-1 axis in patients with mild to moderate CHF.


Assuntos
Cardiomiopatia Dilatada/fisiopatologia , beta-Endorfina/farmacologia , Adulto , Cardiomiopatia Dilatada/sangue , Cardiomiopatia Dilatada/tratamento farmacológico , Catecolaminas/sangue , Doença Crônica , Ecocardiografia , Teste de Esforço , Tolerância ao Exercício/efeitos dos fármacos , Feminino , Hemodinâmica/efeitos dos fármacos , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Volume Sistólico/efeitos dos fármacos , beta-Endorfina/sangue , beta-Endorfina/uso terapêutico
11.
Pain ; 104(1-2): 55-63, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12855314

RESUMO

Although exogenous administration of beta-endorphin to the arcuate nucleus of hypothalamus (ARC) had been shown to produce antinociception, the role of endogenous beta-endorphin of the ARC in nociceptive processing has not been studied directly. The aim of the present study was to investigate the effect of endogenous beta-endorphin in the ARC on nociception in rats with carrageenan-induced inflammation. The hindpaw withdrawal latency (HWL) to noxious thermal and mechanical stimulation was assessed by the hot-plate test and the Randall Selitto Test. Intra-ARC injection of naloxone had no significant influence on the HWL to thermal and mechanical stimulation in intact rats. The HWL decreased significantly after intra-ARC injection of 1 or 10 microg of naloxone in rats with inflammation, but not with 0.1 microg of naloxone. Furthermore, intra-ARC administration of the selective mu-opioid receptor antagonist beta-funaltrexamine (beta-FNA) decreased the nociceptive response latencies to both stimulation in a dose-dependent manner in rats with inflammation, while intra-ARC administration of the selective delta-opioid receptor antagonist naltrindole or the selective kappa-opioid receptor antagonist nor-binaltorphimine (nor-BNI) showed no influences on the nociceptive response latency. The antiserum against beta-endorphin, administered to the ARC, also dose-dependently reduced the HWL in rats with inflammation. The results indicate that endogenous beta-endorphin in the ARC plays an important role in the endogenous antinociceptive system in rats with inflammation, and that its effect is predominantly mediated by the mu-opioid receptor.


Assuntos
Analgésicos/uso terapêutico , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Inflamação/tratamento farmacológico , beta-Endorfina/antagonistas & inibidores , beta-Endorfina/uso terapêutico , Analgésicos/antagonistas & inibidores , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Soros Imunes/farmacologia , Inflamação/induzido quimicamente , Masculino , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Wistar , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/fisiologia , beta-Endorfina/fisiologia
12.
J Pharmacol Exp Ther ; 306(1): 394-400, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12721333

RESUMO

Antagonistic properties of buprenorphine for epsilon- and micro -opioid receptors were characterized in beta-endorphin- and [d-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin (DAMGO)-induced antinociception, respectively, with the tail-flick test in male ICR mice. epsilon-Opioid receptor agonist beta-endorphin (0.1-1 micro g), micro -opioid receptor agonist DAMGO (0.5-20 ng), or buprenorphine (0.1-20 micro g) administered i.c.v. dose dependently produced antinociception. The antinociception induced by 10 micro g of buprenorphine given i.c.v. was completely blocked by the pretreatment with beta-funaltrexamine (beta-FNA) (0.3 micro g i.c.v.), indicating that the buprenophine-induced antinociception is mediated by the stimulation of the micro -opioid receptor. The antinociceptive effects induced by beta-endorphin (1 micro g i.c.v.) and DAMGO (16 ng i.c.v.) were dose dependently blocked by pretreatment with smaller doses of buprenorphine (0.001-1 micro g i.c.v.), but not by a higher dose of buprenorphine (10 micro g i.c.v.). beta-FNA at a dose (0.3 micro g i.c.v.) that strongly attenuated DAMGO-induced antinociception had no effect on the antinociception produced by beta-endorphin (1 micro g i.c.v.). However, pretreatment with buprenorphine (0.1-10 micro g) in mice pretreated with this same dose of beta-FNA was effective in blocking beta-endorphin-induced antinociception. beta-FNA was 226-fold more effective at antagonizing the antinociception induced by DAMGO (16 ng i.c.v.) than by beta-endorphin (1 micro g i.c.v.). The antinociception induced by delta-opioid receptor agonist [d-Ala2]deltorphin II (10 micro g i.c.v.) or kappa1-opioid receptor agonist trans-3,4-dichloro-N-methyl-N-(2-[1-pyrrolidinyl]cyclohexyl)benzeneacetamine methanesulfonate salt [(-)-U50,488H] (75 micro g i.c.v.) was not affected by pretreatment with buprenorphine (0.1-1.0 micro g i.c.v.). It is concluded that buprenorphine, at small doses, blocks epsilon-opioid receptor-mediated beta-endorphin-induced antinociception and micro -opioid receptor-mediated DAMGO-induced antinociception, and at high doses produces a micro -opioid receptor-mediated antinociception.


Assuntos
Analgésicos Opioides/uso terapêutico , Analgésicos/uso terapêutico , Buprenorfina/uso terapêutico , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes , Dor/tratamento farmacológico , Receptores Opioides mu/antagonistas & inibidores , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/uso terapêutico , Analgésicos/farmacologia , Analgésicos não Narcóticos/uso terapêutico , Animais , Buprenorfina/farmacologia , Modelos Animais de Doenças , Ala(2)-MePhe(4)-Gly(5)-Encefalina/uso terapêutico , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos ICR , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Oligopeptídeos/uso terapêutico , Receptores Opioides , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Fatores de Tempo , beta-Endorfina/uso terapêutico
13.
Artigo em Inglês | MEDLINE | ID: mdl-11817510

RESUMO

The effect exerted by two beta-endorphin fragments (DTgammaE and DEgammaE) was investigated on the acute opioid dependence induced by mu, kappa and delta receptor agonists in vitro. After a 4-min in vitro exposure to morphine (less selective mu agonist), DAGO (highly selective mu agonist), U50-488H (highly selective kappa agonist) and beta-endorphin (selective mu-delta agonist), a strong contracture of guinea pig isolated ileum was observed after the addition of naloxone. This effect was also observed when rabbit isolated jejunum was pretreated with deltorphin (highly selective delta agonist). DTgammaE or DEgammaE injection treatment before or after morphine, DAGO, U50-488H, beta-endorphin or deltorphin were able to both prevent and reverse the naloxone-induced contracture after exposure to the opioid agonists in a concentration-dependent fashion. Our results indicate that both DTgammaE or DEgammaE are able to reduce significantly opioid dependence in vitro, suggesting an important functional interaction between beta-endorphin fragments and opioid dependence induced by mu, kappa and delta receptors.


Assuntos
Endorfinas/farmacologia , Endorfinas/uso terapêutico , Íleo/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , beta-Endorfina/farmacologia , beta-Endorfina/uso terapêutico , Analgésicos Opioides/efeitos adversos , Animais , Antidepressivos/uso terapêutico , Relação Dose-Resposta a Droga , Cobaias , Íleo/fisiologia , Masculino , Morfina/efeitos adversos , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico
14.
Pharmacol Toxicol ; 91(6): 398-403, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12688385

RESUMO

It is well established that cholecystokinin (CCK) reduces the antinociceptive effect of opioids. The level of CCK and CCK receptors, as well as CKK release, exhibits considerable plasticity after nerve injury and inflammation, conditions known to be associated with chronic pain. Such altered CCK release coupled in some situation with changes in CCK receptor levels may underlie the clinical phenomenon of varying opioid sensitivity in different clinical pain conditions. In particular, neuropathic pain after injury to the peripheral and central nervous system does not respond well to opioids, which is likely to be caused by increased activity in the endogenous CCK system. CCK receptor antagonists may thus be useful as analgesics in combination with opioids to treat neuropathic pain.


Assuntos
Analgesia , Colecistocinina , Inflamação/tratamento farmacológico , Morfina/antagonistas & inibidores , Dor/etiologia , Receptores da Colecistocinina , beta-Endorfina/antagonistas & inibidores , Animais , Colecistocinina/efeitos adversos , Colecistocinina/líquido cefalorraquidiano , Colecistocinina/fisiologia , Doença Crônica , Humanos , Morfina/uso terapêutico , Ratos , Receptores da Colecistocinina/antagonistas & inibidores , Receptores da Colecistocinina/efeitos dos fármacos , Receptores da Colecistocinina/fisiologia , Traumatismos da Medula Espinal/metabolismo , beta-Endorfina/uso terapêutico
15.
Vestn Ross Akad Med Nauk ; (4): 27-30, 2001.
Artigo em Russo | MEDLINE | ID: mdl-11496754

RESUMO

Among various approaches to long-term correction of the highest functions of the brain there are two methods that are particularly promising. These included: 1) induction of autoantibodies against the enzymes involved in the metabolism of neuroregulators by means of immunization of respective heterologous enzymes; 2) immunization by covalent conjugates of monomolecular neurotropic compounds and neuropeptides with antigen carriers. The investigations of both methods are reviewed and illustrated in experiments on albino rats during alcoholization and some other processes of pathological behavior formation. Evidence is provided for that behavior can be corrected for several months or longer.


Assuntos
Alcoolismo/terapia , Encéfalo/fisiologia , Atividade Nervosa Superior , Imunização , Alcoolismo/fisiopatologia , Animais , Barreira Hematoencefálica , Feminino , Hormônio do Crescimento/imunologia , Hormônio do Crescimento/uso terapêutico , Humanos , Oligopeptídeos/imunologia , Oligopeptídeos/uso terapêutico , Ratos , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/terapia , Fatores de Tempo , beta-Endorfina/imunologia , beta-Endorfina/uso terapêutico
16.
Neuropeptides ; 30(5): 479-84, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8923511

RESUMO

The effect of nicotine administered supraspinally on antinociception induced by supraspinally administered opioids was examined in ICR mice. The intracerebroventricular (i.c.v.) injection of nicotine alone at doses from 1 to 12 micrograms produced only a minimal inhibition of the tail-flick response. Morphine (0.2 micrograms), beta-endorphin (0.1 microgram), D-Pen2.5-enkephalin (DPDPE; 0.5 microgram), trans-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl) cyclohexyl] benzeocetamide (U50, 488H; 6 micrograms) caused only slight inhibition of the tail-flick response. Nicotine dose dependently enhanced inhibition of the tail-flick response induced by i.c.v. administered morphine (0.2 microgram) or beta-endorphin (0.1 microgram). The degree of enhancing effect of nicotine toward beta-endorphin-induced inhibition of the tail-flick response was greater than toward morphine-induced inhibition of the tail-flick response. However, i.c.v. administered nicotine at the same doses was not effective in enhancing the inhibition of the tail-flick response induced by DPDPE (0.5 microgram) or U50, 488H (6 micrograms) administered i.c.v. Our results suggest that stimulation of supraspinal nicotinic receptors may enhance antinociception induced by morphine (a mu-opioid receptor agonist) and beta-endorphin (an epsilon-opioid receptor agonist) administered supraspinally. However, the activation of nicotinic receptors at supraspinal sites may not be involved in enhancing the antinociception induced by DPDPE (a delta-opioid receptor agonist) or U50, 488H (a kappa-opioid receptor agonist) administered supraspinally.


Assuntos
Analgésicos/farmacologia , Morfina/farmacologia , Nicotina/farmacologia , Nociceptores/efeitos dos fármacos , Dor/tratamento farmacológico , beta-Endorfina/farmacologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida , Analgésicos/administração & dosagem , Analgésicos/uso terapêutico , Animais , Sinergismo Farmacológico , D-Penicilina (2,5)-Encefalina , Encefalinas/farmacologia , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfina/administração & dosagem , Morfina/uso terapêutico , Nicotina/administração & dosagem , Nicotina/uso terapêutico , Dor/fisiopatologia , Medição da Dor , Pirrolidinas/administração & dosagem , Pirrolidinas/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides kappa/antagonistas & inibidores , beta-Endorfina/administração & dosagem , beta-Endorfina/uso terapêutico
17.
Neuropeptides ; 30(5): 485-90, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8923512

RESUMO

The present study was designed to investigate the modulatory effects of blockade of spinal histamine receptors on antinociception induced by spinally administered morphine, beta-endorphin and U50, 488H. The effects of intrathecal (i.t.) injections with cyproheptadine (a histamine-1 (H1) receptor antagonist), ranitidine (an H2 receptor antagonist), or thioperamide (an H3 receptor antagonist) injected i.t., on the antinociception induced by morphine, beta-endorphin or trans-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl) cyclohexyl] benzeocetamide (U50, 488H) injected intrathecally (i.t.) were studied. The antinociception was assayed using the tail-flick test. The i.t. injection of cyproheptadine (20 micrograms), ranitidine (20 micrograms), or thioperamide (20 micrograms) alone did not produce any antinociceptive effect. i.t. pretreatment with cyproheptadine attenuated the inhibition of the tail-flick response induced by i.t. administered morphine or beta-endorphin, but not U50, 488H. In addition, i.t. pretreatment with ranitidine attenuated the inhibition of the tail-flick response induced by i.t. administered morphine, beta-endorphin, or U50, 488H. Furthermore, the i.t. pretreatment with thioperamide attenuated the inhibition of the tail-flick response induced by beta-endorphin or U50, 488H, but not morphine, administered i.t. Our results indicate that spinal H1 receptors may be involved in the production of antinociception induced by spinally applied morphine or beta-endorphin- but not U50, 488H. Spinal H2 receptors appear to be involved in spinally administered morphine-, beta-endorphin- and U50, 488H-induced antinociception. Supraspinal histamine H3 receptors may be involved in the production of antinociception induced by supraspinally applied beta-endorphin or U50, 488H, but not morphine.


Assuntos
Analgésicos/antagonistas & inibidores , Ciproeptadina/farmacologia , Antagonistas dos Receptores Histamínicos H1/farmacologia , Morfina/farmacologia , Dor/tratamento farmacológico , Piperidinas/farmacologia , Pirrolidinas/farmacologia , Ranitidina/farmacologia , Medula Espinal/fisiopatologia , beta-Endorfina/farmacologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida , Analgésicos/administração & dosagem , Analgésicos/uso terapêutico , Animais , Ciproeptadina/administração & dosagem , Antagonistas dos Receptores Histamínicos H1/administração & dosagem , Antagonistas dos Receptores H2 da Histamina/administração & dosagem , Antagonistas dos Receptores H2 da Histamina/farmacologia , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfina/administração & dosagem , Morfina/uso terapêutico , Dor/fisiopatologia , Medição da Dor , Piperidinas/administração & dosagem , Pirrolidinas/administração & dosagem , Pirrolidinas/uso terapêutico , Ranitidina/administração & dosagem , Receptores Histamínicos H1/efeitos dos fármacos , Receptores Histamínicos H1/fisiologia , Receptores Histamínicos H2/efeitos dos fármacos , Receptores Histamínicos H2/fisiologia , Receptores Histamínicos H3/efeitos dos fármacos , Receptores Histamínicos H3/fisiologia , beta-Endorfina/administração & dosagem , beta-Endorfina/uso terapêutico
18.
Neuroreport ; 7(12): 1982-4, 1996 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-8905708

RESUMO

Whether i.v. infusion of beta-endorphin or metenkephalin can modify adenosine-provoked angina pectoris-like pain was investigated in healthy volunteers with a double-blind controlled design. All seven volunteers experienced dose-related adenosine-provoked chest pain. Metenkephalin did not modulate the dose-effect curve for adenosine while beta-endorphin counteracted (p < 0.01) the development of pain. The results suggest that peripheral p.subtype opioid receptors are involved in the modulation and may play a role in the genesis of silent and painful myocardial ischaemia.


Assuntos
Angina Pectoris/induzido quimicamente , Encefalinas/uso terapêutico , Dor/tratamento farmacológico , beta-Endorfina/uso terapêutico , Adulto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Humanos , Masculino , Pessoa de Meia-Idade
19.
J Neurol Sci ; 129 Suppl: 127-30, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7595603

RESUMO

The pro-opiomelanocortin-derived peptides beta-endorphin (beta-EP) and alpha-melanocortin (alpha-MSH) were administered to normal and dystrophic C57BL6J mice. All groups of normal and dystrophic mice which had been treated with the two peptides gained significant body weight, as did the normal and dystrophic saline-treated male controls, but the normal and dystrophic female controls did not. The plasma activity of creatine phosphokinase (CPK) was lower in normal mice and dystrophic males which had been treated with the two peptides compared to the corresponding controls. There was no significant difference between the plasma LDH activity in any of the peptide-treated and the corresponding control groups. The activity of CPK was significantly higher in the extensor digitorum longus (EDL) muscles, but not the soleus muscles, of the peptide-treated dystrophic mice compared to the corresponding controls. Administration of alpha-MSH alone or beta-EP alone had no significant effect on the body weight or plasma CPK activity of dystrophic mice compared to the controls. However the activity of CPK was significantly higher in the EDL muscles of the alpha-MSH-treated mice than in the corresponding controls. It is possible that beta-EP and alpha-MSH act synergistically on the neuromuscular system to protect the muscles from damage.


Assuntos
Distrofia Muscular Animal/tratamento farmacológico , Fármacos Neuromusculares/uso terapêutico , alfa-MSH/uso terapêutico , beta-Endorfina/uso terapêutico , Animais , Peso Corporal/efeitos dos fármacos , Creatina Quinase/metabolismo , Feminino , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Distrofia Muscular Animal/patologia , Tamanho do Órgão/efeitos dos fármacos
20.
J Neuroimmunol ; 48(1): 81-90, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8227310

RESUMO

The neurotropic murine coronavirus, MHV-JHM (JHMV) causes encephalitis and paralytic-demyelinating disease in susceptible strains of mice and rats, serving as a model for human demyelinating diseases such as multiple sclerosis. In this communication, we report that a single intracerebral administration of the naturally occurring neuropeptide, beta-endorphin, reduced the incidence of JHMV-induced paralytic-demyelinating disease 40-50% in C57Bl/6 mice. Protection from disease was accompanied by significantly reduced virus replication in the brain as early as 3 days post-infection and did not occur in irradiated, or immunoincompetent mice. The data suggest that beta-endorphin engages immune mechanisms of host resistance to JHMV infection to protect the mice from disease.


Assuntos
Infecções por Coronavirus/imunologia , Doenças Desmielinizantes/imunologia , Vírus da Hepatite Murina , Paralisia/prevenção & controle , beta-Endorfina/uso terapêutico , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/sangue , Encéfalo/microbiologia , Infecções por Coronavirus/microbiologia , Doenças Desmielinizantes/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Vírus da Hepatite Murina/imunologia , Vírus da Hepatite Murina/isolamento & purificação , Vírus da Hepatite Murina/fisiologia , Relação Estrutura-Atividade , Replicação Viral , Irradiação Corporal Total , beta-Endorfina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA