Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.597
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 631(8021): 686-693, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38961287

RESUMO

The µ-opioid receptor (µOR) is a well-established target for analgesia1, yet conventional opioid receptor agonists cause serious adverse effects, notably addiction and respiratory depression. These factors have contributed to the current opioid overdose epidemic driven by fentanyl2, a highly potent synthetic opioid. µOR negative allosteric modulators (NAMs) may serve as useful tools in preventing opioid overdose deaths, but promising chemical scaffolds remain elusive. Here we screened a large DNA-encoded chemical library against inactive µOR, counter-screening with active, G-protein and agonist-bound receptor to 'steer' hits towards conformationally selective modulators. We discovered a NAM compound with high and selective enrichment to inactive µOR that enhances the affinity of the key opioid overdose reversal molecule, naloxone. The NAM works cooperatively with naloxone to potently block opioid agonist signalling. Using cryogenic electron microscopy, we demonstrate that the NAM accomplishes this effect by binding a site on the extracellular vestibule in direct contact with naloxone while stabilizing a distinct inactive conformation of the extracellular portions of the second and seventh transmembrane helices. The NAM alters orthosteric ligand kinetics in therapeutically desirable ways and works cooperatively with low doses of naloxone to effectively inhibit various morphine-induced and fentanyl-induced behavioural effects in vivo while minimizing withdrawal behaviours. Our results provide detailed structural insights into the mechanism of negative allosteric modulation of the µOR and demonstrate how this can be exploited in vivo.


Assuntos
Analgésicos Opioides , Avaliação Pré-Clínica de Medicamentos , Naloxona , Receptores Opioides mu , Bibliotecas de Moléculas Pequenas , Animais , Humanos , Masculino , Camundongos , Regulação Alostérica/efeitos dos fármacos , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Sítios de Ligação/efeitos dos fármacos , Microscopia Crioeletrônica , Fentanila/antagonistas & inibidores , Fentanila/farmacologia , Cinética , Ligantes , Modelos Moleculares , Morfina/antagonistas & inibidores , Morfina/farmacologia , Naloxona/administração & dosagem , Naloxona/química , Naloxona/metabolismo , Naloxona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/metabolismo , Antagonistas de Entorpecentes/farmacologia , Overdose de Opiáceos/tratamento farmacológico , Conformação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Células Sf9 , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Camundongos Endogâmicos C57BL
2.
Bull Exp Biol Med ; 170(1): 30-34, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33222084

RESUMO

Activity of compound GSB-106, a low-molecular mimetic of loop 4 of the brain neurotrophic factor (BDNF), was studied in experimental morphine withdrawal syndrome simulated in outbred rats. Single and subchronic (5 intraperitoneal injections) administration of GSB-106 in a dose of 0.1 mg/kg significantly reduced the total index of morphine withdrawal syndrome by 55.2 and 45.6%, respectively. GSB-106 reduced the severity of some behavioral signs (piloerection, gnashing of teeth, wet-dog shaking, and runaway attempts), but had no effect on mechanical allodynia formed in the rats with dependence. Subchronic treatment with GSB-106 prevented the increase in the content of ΔFosB (product of early response gene) in the striatum induced by morphine withdrawal. The results confirmed the concept on the involvement of neurotrophins, specifically BDNF and its analogs, in the mechanisms associated with the formation of opiate dependence.


Assuntos
Dipeptídeos/farmacologia , Dependência de Morfina/tratamento farmacológico , Morfina/antagonistas & inibidores , Antagonistas de Entorpecentes/farmacologia , Peptidomiméticos/farmacologia , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Animais , Animais não Endogâmicos , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Expressão Gênica , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Injeções Intraperitoneais , Masculino , Morfina/efeitos adversos , Dependência de Morfina/genética , Dependência de Morfina/metabolismo , Dependência de Morfina/fisiopatologia , Entorpecentes/efeitos adversos , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Síndrome de Abstinência a Substâncias/genética , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/fisiopatologia
3.
Planta Med ; 84(4): 221-224, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28793355

RESUMO

This study sought to determine whether the cannabis constituent cannabidiol attenuates the development of morphine reward in the conditioned place preference paradigm. Separate groups of mice received either saline or morphine in combination with one of four doses of cannabidiol using three sets of drug/no-drug conditioning trials. After drug-place conditioning, morphine mice displayed robust place preference that was attenuated by 10 mg/kg cannabidiol. Further, when administered alone, this dose of cannabidiol was void of rewarding and aversive properties. The finding that cannabidiol blocks opioid reward suggests that this compound may be useful in addiction treatment settings.


Assuntos
Canabidiol/farmacologia , Morfina/farmacologia , Comportamento Espacial/efeitos dos fármacos , Animais , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/antagonistas & inibidores , Recompensa
4.
Vet Anaesth Analg ; 45(2): 190-194, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29409803

RESUMO

OBJECTIVE: To evaluate whether subcutaneous (SC) metoclopramide (0.2 mg kg-1) administered 30 minutes prior to (T30) or simultaneously with (T0) intramuscular (IM) morphine (0.2 mg kg-1) and dexmedetomidine (0.003 mg kg-1) reduces the incidence of nausea and emesis in healthy dogs. STUDY DESIGN: Prospective, randomized and blinded study. ANIMALS: A total of 45 dogs scheduled for elective procedures. METHODS: Dogs were assigned randomly to three groups to be administered SC metoclopramide (0.2 mg kg-1) 30 minutes before (group M30) or simultaneously (group M0) to IM morphine (0.2 mg kg-1) and dexmedetomidine (0.003 mg kg-1). Dogs in the control group (group C) were administered SC saline at T30 and T0. Dogs were observed for 30 minutes after premedication to evaluate signs of nausea (continuous lip-licking and sialorrhoea) and emesis. Signs of pain or discomfort caused by SC injections were also recorded. RESULTS: There were no statistical differences amongst groups for age, body weight and sex. More dogs developed continuous lip-licking in group C (12/15, 80.0%) compared to dogs in group M30 (1/15, 6.7%) and dogs in group M0 (5/15, 33.3%; p = 0.0001 and p = 0.01, respectively). More dogs developed sialorrhoea in group M0 (8/15, 53.3%) and in group C (10/15, 66.7%) compared to dogs in group M30 (2/15, 13.3%; p = 0.03 and p = 0.004, respectively). More dogs vomited in group M0 (4/15, 26.7%) and in group C (9/15, 60.0%) compared to dogs in group M30 (0/15, 0.0%; p = 0.05 and p = 0.0003, respectively). None of the dogs demonstrated signs of pain or discomfort during SC metoclopramide injection. CONCLUSIONS AND CLINICAL RELEVANCE: Subcutaneous metoclopramide at 0.2 mg kg-1 may reduce IM morphine and dexmedetomidine-induced nausea and emesis if administered 30 minutes in advance. It is effective in reducing lip-licking even when administered concurrently with IM morphine-dexmedetomidine.


Assuntos
Analgésicos Opioides/efeitos adversos , Antieméticos/uso terapêutico , Cães , Metoclopramida/uso terapêutico , Morfina/efeitos adversos , Náusea/veterinária , Vômito/veterinária , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/antagonistas & inibidores , Animais , Dexmedetomidina/administração & dosagem , Antagonistas de Dopamina/uso terapêutico , Feminino , Masculino , Morfina/administração & dosagem , Morfina/antagonistas & inibidores , Náusea/induzido quimicamente , Pré-Medicação/veterinária , Método Simples-Cego , Vômito/induzido quimicamente
5.
J Pharmacol Sci ; 133(1): 9-17, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28034513

RESUMO

We investigated the mechanisms underlying the suppression of the rewarding effects of opioids using the femur bone cancer (FBC) mouse model. The rewarding and antinociceptive effects of subcutaneously administered morphine and oxycodone in the FBC model mice were assessed using the conditioned place preference test and the von-Frey test. In FBC mice, antinociceptive doses of morphine (30 mg/kg) and oxycodone (5 mg/kg) did not produce the rewarding effects but excessive doses of morphine (300 mg/kg) and oxycodone (100 mg/kg) did. Western blot analyses revealed a transient and significant increase in phosphorylated-extracellular regulated kinase (p-ERK) levels in ventral tegmental area (VTA) 5 min after the administration of morphine in sham-group. Interestingly, in FBC group, a regular dose of morphine did not increase p-ERK levels but a high dose of morphine caused an increase in p-ERK level 5 min after administration. The rewarding effects of a regular dose of and a high dose of morphine in the sham-operation and FBC model, respectively, were significantly inhibited by the MEK inhibitor. The suppression of p-ERK might result in resistance to these rewarding effects under the conditions of bone cancer.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Morfina/farmacologia , Oxicodona/farmacologia , Receptores Opioides mu/agonistas , Recompensa , Regulação para Cima/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/enzimologia , Analgésicos/farmacologia , Animais , Butadienos/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Camundongos , Morfina/antagonistas & inibidores , Nitrilas/farmacologia , Oxicodona/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Ensaio Radioligante , Receptores Opioides mu/metabolismo
6.
Biol Pharm Bull ; 40(3): 278-283, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-27980244

RESUMO

There have been few detailed reports on respiratory depression due to overdoses of opioids in terminal cancer patients. We investigated the situation of treatment with opioid antagonists for respiratory depression that occurred after administration of opioid at optimal doses in terminal cancer patients, to clarify pathological changes as well as causative factors. In 2443 terminal cancer patients receiving opioids, 7 patients (0.3%) received opioid antagonists: 6, morphine (hydrochloride, 5; sulfate, 1); 1, oxycodone. The median dosage of opioids was 13.3 mg/d, as converted to morphine injection. Respiratory depression occurred on this daily dose in 4 patients and after changed dose and route in 3 patients. Opioids were given through the vein in 6 patients and by the enteral route in 1 patient. Concomitant drugs included nonsteroidal anti-inflammatory drugs in 3 patients and zoledronic acid in 2 patients. In morphine-receiving patients, renal functions were significantly worsened at the time of administration of an opioid antagonist than the day before the start of opioid administration. These findings indicate that the proper use of opioids was safe and acceptable in almost all terminal cancer patients. In rare cases, however, a risk toward respiratory depression onset is indicated because morphine and morphine-6-glucuronide become relatively excessive owing to systemic debility due to disease progression, especially respiratory and renal dysfunctions. At the onset of respiratory depression, appropriate administration of an opioid antagonist mitigated the symptoms. Thereafter, opioid switching or continuous administration at reduced dosages of the same opioids prevented the occurrence of serious adverse events.


Assuntos
Analgésicos Opioides/administração & dosagem , Morfina/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Neoplasias/complicações , Dor/tratamento farmacológico , Insuficiência Respiratória/prevenção & controle , Idoso , Idoso de 80 Anos ou mais , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/uso terapêutico , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Morfina/efeitos adversos , Morfina/antagonistas & inibidores , Morfina/uso terapêutico , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Neoplasias/tratamento farmacológico , Dor/etiologia , Insuficiência Respiratória/etiologia , Estudos Retrospectivos , Assistência Terminal
7.
Am J Drug Alcohol Abuse ; 43(1): 78-86, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27712113

RESUMO

BACKGROUND: Treatments for reducing opioid withdrawal are limited and prone to problematic side effects. Laboratory studies, clinical observations, and limited human trial data suggest 5-HT3-receptor antagonists and antihistamines may be effective. OBJECTIVES: This double-blind, crossover, placebo-controlled study employing an acute physical dependence model evaluated whether (i) treatment with a 5-HT3-receptor antagonist (palonosetron) would reduce opioid withdrawal symptoms, and (ii) co-administration of an antihistamine (hydroxyzine) would enhance any treatment effect. METHODS: At timepoint T = 0, healthy (non-opioid dependent, non-substance abuser) male volunteers (N = 10) were pre-treated with either a) placebo, b) palonosetron IV (0.75 mg), or c) palonosetron IV (0.75 mg) and hydroxyzine PO (100 mg) in a crossover study design. This was followed at T = 30 by intravenous morphine (10 mg/70kg). At T = 165, 10 mg/70kg naloxone IV was given to precipitate opioid withdrawal. The objective opioid withdrawal score (OOWS) and subjective opioid withdrawal score (SOWS) were determined 5 and 15 minutes after naloxone administration (T = 170, 180, respectively). Baseline measurements were recorded at T = -30 and T = -15. RESULTS: Comparison of average baseline OOWS scores with OOWS scores obtained 15 minutes after naloxone was significant (p = 0.0001). Scores from 15 minutes post-naloxone infusion showed significant differences in OOWS scores between treatment groups: placebo, 3.7 ± 2.4; palonosetron, 1.5 ± 0.97; and palonosetron with hydroxyzine, 0.2 ± 0.1333. CONCLUSIONS: Pretreatment with palonosetron significantly reduced many signs of experimentally-induced opioid withdrawal. Co-administration with hydroxyzine further reduced opioid withdrawal severity. These results suggest that 5-HT3 receptor antagonists, alone or in combination with an antihistamine, may be useful in the treatment of opioid withdrawal.


Assuntos
Hidroxizina/uso terapêutico , Isoquinolinas/uso terapêutico , Quinuclidinas/uso terapêutico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Estudos Cross-Over , Método Duplo-Cego , Sinergismo Farmacológico , Voluntários Saudáveis , Humanos , Masculino , Morfina/efeitos adversos , Morfina/antagonistas & inibidores , Naloxona/farmacologia , Palonossetrom , Adulto Jovem
8.
Cell Physiol Biochem ; 39(5): 1930-1940, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27771708

RESUMO

BACKGROUND/AIMS: The purpose of this study was to investigate the implications of protein kinase C-epsilon (PKCε), Extracellular Signal-regulated Kinase 1/2 (ERK1/2) and mitochondrial permeability transition pore (mPTP) in myocardial protection induced by morphine postconditioning (MpostC). METHODS: The isolated rat hearts were randomly assigned into one of eight groups. Hearts in time control (TC) group were constantly perfused for 105min. Hearts in ischemia-reperfusion (I/R) group were subjected to 45 min of ischemia followed by 1 h of reperfusion. MpostC was induced by 10 min of morphine administration at the onset of reperfusion. εV1-2 (an inhibitor of PKCε) and PD (an inhibitor of ERK1/2) was administered with or without morphine during the first 10 min of reperfusion following the 45 min of ischemia. I/R injury was assessed by functional parameters, creatine kinase-MB (CK-MB) release and infarct size (IS/AAR). Additional hearts were excised at 20 min following reperfusion to detect the membrane-specific translocation of PKCε, ERK1/2 phosphorylation, mitochondrial permeability transition (MPT) and cytochrome C (Cyt-c) release. RESULTS: MpostC markedly reduced infarct size (IS/AAR), CK-MB release, and improved cardiac function recovery. However, these protective effects were partly abolished in the presence of εV1-2 or PD. Compared to TC group, the membrane translocation of PKCε, ERK1/2 phosphorylation, mPTP opening, and Cyt-c release were significantly increased in I/R hearts. MpostC further increased the membrane translocation of PKCε and ERK1/2 phosphorylation, and significantly inhibited mPTP opening and Cyt-c release. However, those protective effects induced by MpostC were abolished by εV1-2 or PD, which, used alone, showed no influence on reperfusion injury. CONCLUSIONS: These findings suggest that MpostC protects isolated rat hearts against ischemia-reperfusion injury via activating PKCε-ERK1/2 pathway and inhibiting mPTP opening.


Assuntos
Proteínas de Transporte da Membrana Mitocondrial/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Morfina/farmacologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteína Quinase C-épsilon/genética , Animais , Creatina Quinase Forma MB/metabolismo , Citocromos c/metabolismo , Regulação da Expressão Gênica , Pós-Condicionamento Isquêmico/métodos , Masculino , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Morfina/antagonistas & inibidores , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Técnicas de Cultura de Órgãos , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C-épsilon/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
9.
J Pharmacol Exp Ther ; 358(2): 209-15, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27278234

RESUMO

Toll-like receptor 4 (TLR4) signaling is implied in opioid reinforcement, reward, and withdrawal. Here, we explored whether TLR4 signaling is involved in the acute psychomotor-stimulating effects of heroin, 6-acetylmorphine (6-AM), and morphine as well as whether there are differences between the three opioids regarding TLR4 signaling. To address this, we examined how pretreatment with (+)-naloxone, a TLR4 active but opioid receptor (OR) inactive antagonist, affected the acute increase in locomotor activity induced by heroin, 6-AM, or morphine in mice. We also assessed the effect of pretreatment with (-)-naloxone, a TLR4 and OR active antagonist, as well as the pharmacokinetic profiles of (+) and (-)-naloxone in the blood and brain. We found that (-)-naloxone reduced acute opioid-induced locomotor activity in a dose-dependent manner. By contrast, (+)-naloxone, administered in doses assumed to antagonize TLR4 but not ORs, did not affect acute locomotor activity induced by heroin, 6-AM, or morphine. Both naloxone isomers exhibited similar concentration versus time profiles in the blood and brain, but the brain concentrations of (-)-naloxone reached higher levels than those of (+)-naloxone. However, the discrepancies in their pharmacokinetic properties did not explain the marked difference between the two isomers' ability to affect opioid-induced locomotor activity. Our results underpin the importance of OR activation and do not indicate an apparent role of TLR4 signaling in acute opioid-induced psychomotor stimulation in mice. Furthermore, there were no marked differences between heroin, 6-AM, and morphine regarding involvement of OR or TLR4 signaling.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Heroína/farmacologia , Derivados da Morfina/farmacologia , Morfina/farmacologia , Naloxona/química , Naloxona/farmacologia , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiologia , Heroína/antagonistas & inibidores , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Morfina/antagonistas & inibidores , Derivados da Morfina/antagonistas & inibidores , Naloxona/sangue , Naloxona/farmacocinética , Transdução de Sinais/efeitos dos fármacos , Estereoisomerismo , Relação Estrutura-Atividade , Receptor 4 Toll-Like/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-26390873

RESUMO

BACKGROUND: Opiates such as morphine are the most powerful analgesics, but their protracted use is restrained by the development of tolerance to analgesic effects. Recent works suggest that tolerance to morphine might be due to its inability to promote mu opioid receptor endocytosis, and the co-injection of morphine with a mu opioid receptor internalizing agonist like [D-Ala(2),N-Me-Phe(4),Gly-ol(5)]enkephalin reduces tolerance to morphine. So far, no studies have been conducted to evaluate the ability of methadone to reduce morphine tolerance in morphine-pretreated animals, a treatment sequence that could be encountered in opiate rotation protocol. We investigated the ability of methadone (a mu opioid receptor internalizing agonist used in therapy) to reverse morphine tolerance and the associated cellular mechanisms in the periaqueductal gray matter, a region involved in pain control. METHODS: We measured analgesic response following a challenge dose of morphine in the hot plate test and investigated regulation of mu opioid receptor (coupling and endocytosis) and some cellular mechanisms involved in tolerance such as adenylate cyclase superactivation and changes in N-methyl-d-aspartate receptor subunits expression and phosphorylation state. RESULTS: A chronic treatment with morphine promoted tolerance to its analgesic effects and was associated with a lack of mu opioid receptor endocytosis, adenylate cyclase overshoot, NR2A and NR2B downregulation, and phosphorylation of NR1. We reported that a methadone treatment in morphine-treated mice reversed morphine tolerance to analgesia by promoting mu opioid receptor endocytosis and blocking cellular mechanisms of tolerance. CONCLUSIONS: Our data might lead to rational strategies to tackle opiate tolerance in the frame of opiate rotation.


Assuntos
Tolerância a Medicamentos , Metadona/farmacologia , Morfina/antagonistas & inibidores , Morfina/farmacologia , Adenilil Ciclases/metabolismo , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Endocitose , Masculino , Camundongos , Medição da Dor/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/biossíntese , Receptores Opioides mu/metabolismo
11.
Ann Behav Med ; 50(4): 497-505, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26809850

RESUMO

BACKGROUND: Recent studies suggest that participant expectations influence pain ratings during conditioned pain modulation testing. The present study extends this work by examining expectancy effects among individuals with and without chronic back pain after administration of placebo, naloxone, or morphine. PURPOSE: This study aims to identify the influence of individual differences in expectancy on changes in heat pain ratings obtained before, during, and after a forearm ischemic pain stimulus. METHODS: Participants with chronic low back pain (n = 88) and healthy controls (n = 100) rated heat pain experience (i.e., "test stimulus") before, during, and after exposure to ischemic pain (i.e., "conditioning stimulus"). Prior to testing, participants indicated whether they anticipated that their heat pain would increase, decrease, or remain unchanged during ischemic pain. RESULTS: Analysis of the effects of expectancy (pain increase, decrease, or no change), drug (placebo, naloxone, or morphine), and group (back pain, healthy) on changes in heat pain revealed a significant main effect of expectancy (p = 0.001), but no other significant main effects or interactions. Follow-up analyses revealed that individuals who expected lower pain during ischemia reported significantly larger decreases in heat pain as compared with those who expected either no change (p = 0.004) or increased pain (p = 0.001). CONCLUSIONS: The present findings confirm that expectancy is an important contributor to conditioned pain modulation effects, and therefore significant caution is needed when interpreting findings that do not account for this individual difference. Opioid mechanisms do not appear to be involved in these expectancy effects.


Assuntos
Antecipação Psicológica , Dor Crônica/psicologia , Condicionamento Psicológico/efeitos dos fármacos , Dor Lombar/psicologia , Morfina/farmacologia , Naloxona/farmacologia , Medição da Dor/psicologia , Adulto , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Feminino , Humanos , Masculino , Morfina/antagonistas & inibidores , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Adulto Jovem
12.
J Neurosci ; 34(38): 12850-64, 2014 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-25232120

RESUMO

Synaptodendritic injury is thought to underlie HIV-associated neurocognitive disorders and contributes to exaggerated inflammation and cognitive impairment seen in opioid abusers with HIV-1. To examine events triggering combined transactivator of transcription (Tat)- and morphine-induced synaptodendritic injury systematically, striatal neuron imaging studies were conducted in vitro. These studies demonstrated nearly identical pathologic increases in dendritic varicosities as seen in Tat transgenic mice in vivo. Tat caused significant focal increases in intracellular sodium ([Na(+)]i) and calcium ([Ca(2+)]i) in dendrites that were accompanied by the emergence of dendritic varicosities. These effects were largely, but not entirely, attenuated by the NMDA and AMPA receptor antagonists MK-801 and CNQX, respectively. Concurrent morphine treatment accelerated Tat-induced focal varicosities, which were accompanied by localized increases in [Ca(2+)]i and exaggerated instability in mitochondrial inner membrane potential. Importantly, morphine's effects were prevented by the µ-opioid receptor antagonist CTAP and were not observed in neurons cultured from µ-opioid receptor knock-out mice. Combined Tat- and morphine-induced initial losses in ion homeostasis and increases in [Ca(2+)]i were attenuated by the ryanodine receptor inhibitor ryanodine, as well as pyruvate. In summary, Tat induced increases in [Na(+)]i, mitochondrial instability, excessive Ca(2+) influx through glutamatergic receptors, and swelling along dendrites. Morphine, acting via µ-opioid receptors, exacerbates these excitotoxic Tat effects at the same subcellular locations by mobilizing additional [Ca(2+)]i and by further disrupting [Ca(2+)]i homeostasis. We hypothesize that the spatiotemporal relationship of µ-opioid and aberrant AMPA/NMDA glutamate receptor signaling is critical in defining the location and degree to which opiates exacerbate the synaptodendritic injury commonly observed in neuroAIDS.


Assuntos
Cálcio/metabolismo , Dendritos/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mitocôndrias/efeitos dos fármacos , Morfina/farmacologia , Sódio/metabolismo , Sinapses/efeitos dos fármacos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Nefropatia Associada a AIDS/induzido quimicamente , Nefropatia Associada a AIDS/metabolismo , Nefropatia Associada a AIDS/patologia , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dendritos/metabolismo , Dendritos/patologia , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/metabolismo , Morfina/antagonistas & inibidores , N-Metilaspartato/farmacologia , Cultura Primária de Células , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Rianodina/farmacologia , Sinapses/metabolismo , Sinapses/patologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/antagonistas & inibidores , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
13.
BMC Anesthesiol ; 15: 12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25745356

RESUMO

BACKGROUND: Morphine-induced hyperalgesia and tolerance significantly limits its clinical use in relieving acute and chronic pain. Melatonin, a pineal gland neurohormone, has been shown to participate in certain neuropsychopharmacological actions. The present study investigated the effect of melatonin on morphine-induced hyperalgesia and tolerance and possible involvement of protein kinase C (PKC)/N-methyl-D-aspartate (NMDA) pathway in melatonin-mediated. METHODS: Experiments were performed on adult, male Sprague-Dawley rats. Melatonin (10 mg/kg, intraperitoneal, i.p.) or saline was administrated 10 min after morphine injection (10 mg/kg, subcutaneous, s.c.) each day for consecutive 14 days. Withdrawal threshold of the hindpaw to mechanical and thermal stimulation was measured before any drug administration and one hour after melatonin or saline on each designated test day. On the 15(th) day, thermal withdrawal was measured after s.c. morphine (20 mg/kg), but not melatonin, and morphine tolerance was measured and expressed by MPAE% (percent of maximal possible anti-nociceptive effect) of morphine. Levels of expression of protein kinase C gamma (PKCγ) and NMDA receptor subtype NR1 in spinal cord were detected by Western blotting. RESULTS: The mechanical withdrawal threshold and thermal withdrawal latency decreased and shortened significantly (i.e., threshold decreased) in rats that received morphine treatment for two weeks compared with that in rats receiving saline. This morphine-induced mechanical and thermal hyperalgesia were greatly attenuated by co-administration of morphine with melatonin. The MPAE% representing morphine analgesic effect was reduced approximately 60% in rats that received morphine treatment. However, following the treatment of morphine with melatonin, the MPAE% was reduced only about 30%, comparing with those that received saline treatment as control. Administration of morphine alone resulted in significantly increased expression of PKCγ and NR1 proteins in the spinal cord. These increased levels of expression of PKCγ and NR1 were significantly inhibited by co-administration of morphine with melatonin. CONCLUSIONS: Our findings demonstrate that melatonin have potential to attenuate repetitive morphine-induced hyperalgesia and tolerance, possibly by inhibiting PKCγ and NR1 activities in the spinal cord.


Assuntos
Tolerância a Medicamentos/fisiologia , Hiperalgesia/prevenção & controle , Hiperalgesia/fisiopatologia , Melatonina/farmacologia , Morfina/antagonistas & inibidores , Proteína Quinase C/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Hiperalgesia/induzido quimicamente , Masculino , Morfina/farmacologia , Medição da Dor/efeitos dos fármacos , Proteína Quinase C/biossíntese , Ratos , Receptores de N-Metil-D-Aspartato/biossíntese , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
14.
Int J Neuropsychopharmacol ; 17(1): 127-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24103337

RESUMO

The maladaptive drug memory developed between the drug-rewarding effect and environmental cues contributes to difficulty in preventing drug relapse. Established reward memories can be disrupted by pharmacologic interventions following their reactivation. Rapamycin, an inhibitor of mammalian target of rapamycin (mTOR) kinase, has been proved to be involved in various memory consolidation. However, it is less well characterized in drug memory reconsolidation. Using a conditioned place preference (CPP) procedure, we examined the effects of systemically administered rapamycin on reconsolidation of drug memory in rats. We found that systemically administered rapamycin (0.1 or 10 mg/kg, i.p.) after re-exposure to drug-paired environment, dose dependently decreased the expression of CPP 1 d later, and the effect lasted for up to 14 d and could not be reversed by a priming injection of morphine. The effect of rapamycin on morphine-associated memory was specific to drug-paired context, and rapamycin had no effect on subsequent CPP expression when rats were exposed to saline-paired context or homecage. These results indicated that systemic administration of rapamycin after memory reactivation can persistently inhibit the drug seeking behaviour via disruption of morphine memory reconsolidation in rats. Additionally, the effect of rapamycin on memory reconsolidation was reproduced in cocaine CPP and alcohol CPP. Furthermore, rapamycin did not induce conditioned place aversion and had no effect on locomotor activity and anxiety behaviour. These findings suggest that rapamycin could erase the acquired drug CPP in rats, and that mTOR activity plays an important role in drug reconsolidation and is required for drug relapse.


Assuntos
Comportamento de Procura de Droga/efeitos dos fármacos , Memória/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Recompensa , Sirolimo/farmacologia , Animais , Cocaína/antagonistas & inibidores , Cocaína/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Sinais (Psicologia) , Etanol/antagonistas & inibidores , Etanol/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Morfina/antagonistas & inibidores , Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Ratos
15.
Artigo em Russo | MEDLINE | ID: mdl-25713873

RESUMO

The article concerns study of effects of polyclonal antibodies to serotonin-modulating anticonsolidation protein (SMAP) being in direct dependence on serotonin level and providing intracellular transduction of serotonergic signal, on positive reinforcement effect of morphine in rats. The task was formed in Wistar male rats in the model of morphine self-administration as a result of pressing of one of two levers attached to the wall, joined to the pump delivering each time 100 µg of morphine directly into the vena jugularis. In the 1st series of studies brain cingulate cortex and hypothalamus were taken from the rats achieved stable level of morphine intake and SMAP level was measured with indirect immune-enzyme assay. It was shown that in the morphine-self-injected rats SMAP level in the cingulate cortex is significantly upregulated (p = 0.01), while in the hypothalamus it was left unchanged. In the 2nd series of studies the rats with stable level of morphine intake were administered intraperitoneally with anti-SMAP rabbit polyclonal antibodies (experimental group) or non-immune γ-globulins (control group). Soon after antibodies administration the animals of the experimental group demonstrated manifold decrease of morphine intake lasted for 8 days (p < 0.008), whereas it did not change in the controls. SMAP upregulation in the brain cingulate cortex in the rats with stable morphine intake, obviously, indicates to its engagement in positive reinforcement effect of morphine. Blockade of SMAP activity with anti-SMAP antibodies in the nerve cells induced sharp decrease of morphine intake due to disturbances of transduction through intracellular serotonin's signal channels.


Assuntos
Anticorpos/farmacologia , Giro do Cíngulo/efeitos dos fármacos , Dependência de Morfina/tratamento farmacológico , Morfina/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Animais , Anticorpos/isolamento & purificação , Transporte Biológico/efeitos dos fármacos , Condicionamento Operante , Regulação da Expressão Gênica/efeitos dos fármacos , Giro do Cíngulo/metabolismo , Giro do Cíngulo/fisiopatologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Injeções Intraperitoneais , Masculino , Morfina/administração & dosagem , Dependência de Morfina/metabolismo , Dependência de Morfina/fisiopatologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Coelhos , Ratos , Ratos Wistar , Autoadministração , Transdução de Sinais
16.
Antibiot Khimioter ; 59(7-8): 16-24, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25975103

RESUMO

UNLABELLED: Rengalin liquid formulation on the basis of antibodies to bradikinin histamine and morphine was specially designed for the treatment of cough in children. The three-component combination in therapeutically active against both dry and wet cough due to effect on diverse pathogenetic aspects of the cough reflex. The aim of the multicenter, comparative, randomized clinical trial was to estimate the efficacy and safety of rengalin in the treatment of cough in patients with acute respiratory infection (ARI) of the upper respiratory tract. METHODS: One hundred forty six patients at the age of 3 to 17 years (the average age of 8.2 ± 3.6 years) from 14 medical centres of Russia were observed. The patients suffered from dry/nonproductive, frequent, sore cough preventing from day-time activity and/or night sleep (≥ 4 by the Cough Severity Scale). The cough duration ranged from 12 hours to 3 days. For 3 days the patients of group 1 (n = 71) and group 2 (n = 75) were treated with rengalin and sinekod (butamirate) respectively. For the following 4 days the patients (in case of viscid expectoration were treated with ambroxole in the age doses. The results of the Per Protokol Analysis (n = 67 rengalin group and n = 73 sinekod group) with an account of the Non-Infectiority Design are presented. RESULTS: In 3 days the number of the group 1 patients with significant improvement/recovery by the day and night estimates amounted to 90% and 88% respectively (vs. 81% and 88% in the group 2 patients, no night opisodes of cough after 3-days rengalin use being recorded in 52% of the patients vs. 34% in the sinekod group patients (p = 0.0003). On the 7th day of the treatment with rengalin the number of the children with significant improvement of or recovery from day-time cought amounted to 99%and that of the patients with significant improvement of or recovery from night-time cough amounted to 93%, in 90% of them no night-time cough being recorded (p = 0.0008). As for the patients of the reference group, the respective values were 93% and 90%, no night-time cough being recorded in 81% of the patients. The time required for development of productive/moist cough during the 3-day treatment course in the patients of both the group was the same (2.9 ± 0.3 days in the patients of group 1 and 2.9 ± 0.4 days in the group 2 patients. Moreover, in 34% of the rengalin dry cough became residual (as rare episode of tussiculation with scantly exudation). After 3-day course of the rengalin therapy, 66% of the patients was treated with ambroxole (versus 95% in sinecod group (p < 0.0001) based on comparative analysis and χ2 = 17.7, p > 0.0001 by the results of the frequency analysis). The total duration of cough in the patients of groups 1 and 2 was 6.5 ± 0.8 and 6.7 ± 0.7 days respectively (the comparability truth, p = 0.0001). The severity of the day-time cough by the area under the curve estimates for 7 days of the treatment in the rengalin group patients was equel to 14.3 ± 5.6 numbers--days and that of the patients of the sinekod? group was equal to 15.9?6.1 numbers - days. The severity of the night-time cough was equal to 4.2 ± 2.7 number--days respectively. In 2 patients (3%) treated with sinekod signs of ARI generalization was observed after the 3-day treatment (p > 0.0001). The research physicians-investigators (CGI-EL Scale) the combination of the anti- and protussive activities in one drug to be efficient and absolutely safe for the chilgren. The therapeutic efficacy in the patients of the rengalin group was higher in 3 days (2.1 ± 0.5 numbers) and even in 7 days (2.7 ± 0.5 numbers). The results value in the patients of the sinekod group being 1.8 ± 0.4 and 2.5 ± 0.6 numbers (one-wayANOVA for repeated estimates ANOVA: Visit - F(1/138) = 146, p < 0.0001, TREATMENT--F(1/138) = 9.0, p = 0.003). The factor of the side effects in the patients of the rengalin group was zero (no side effects due to the treatment were recorded in the patients), whereas in the patients treated with sinekod for 3 days the respective value was 0.1 ± 0.3 (true superiority of rengalin by the ANOVA data. TREATMENT--F(1/138) = 4.7, p = 0.03). The efficacy factor of the rengalin was also in its favour (ANOVA: Visit--F(1/138) = 182, p < 0.0001, TREATMENT--F(1y138) = 7.3, p = 0.008). In the patients treated with rengalin there were defected no deviations in the biochemical and general clinical analyses of blood and urine, no adverse reactions characteristic of antitussive drugs of the action. 100-percent adherence to the therapy was stated. CONCLUSION: He antitussive effect of rengalin in the treatment of frequent dry day-time and night-time cough was observed earlier and proved to be comparable with that of butamirate (sinekod). Rengalin prevented significant exudation and viscid expectoration in many patients, promoted rapid residual in the patients with dry cough and the patients recovery. The use of rengalin for 3 days significantly lowered the percentage of the patients requiring treatment with mucolytics at the subsequent stages of ARI.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Antitussígenos/uso terapêutico , Tosse/tratamento farmacológico , Antagonistas dos Receptores Histamínicos/uso terapêutico , Fenilbutiratos/uso terapêutico , Infecções Respiratórias/tratamento farmacológico , Administração Oral , Adolescente , Ambroxol/uso terapêutico , Anticorpos Neutralizantes/biossíntese , Antitussígenos/metabolismo , Bradicinina/antagonistas & inibidores , Bradicinina/imunologia , Criança , Pré-Escolar , Tosse/fisiopatologia , Expectorantes/uso terapêutico , Feminino , Histamina/imunologia , Antagonistas dos Receptores Histamínicos/metabolismo , Humanos , Masculino , Morfina/antagonistas & inibidores , Morfina/imunologia , Infecções Respiratórias/fisiopatologia , Federação Russa
17.
J Pharmacol Exp Ther ; 345(3): 512-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23528610

RESUMO

The chronic use of opioids in humans, accompanied by the development of tolerance, is a dangerous phenomenon in its own right. However, chronic opioid use is often made more dangerous by the coconsumption of other substances. It has been observed that the blood level of opioids in postmortem analyses of addicts, who consumed ethanol along with the opioid, was much less than that observed in individuals who died from opioids alone. This relationship between ethanol and opioids led us to investigate the hypothesis that ethanol alters tolerance to opioids. In the present study, we report that ethanol significantly and dose-dependently reduced the antinociceptive tolerance produced by morphine and the cross-tolerance between [D-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin (DAMGO) and morphine in the mouse tail-flick test. The reversal of morphine tolerance was partially blocked by both the gamma receptor blocker bicuculline and by the γ-aminobutyric acid (GABA)(B) receptor blocker phaclofen and the administration of both inhibitors completely reversed the effects of ethanol on morphine tolerance. Diazepam, like ethanol, decreased morphine tolerance. However, this inhibition was reversed by the GABA(A) antagonist bicuculline but not by the GABA(B) antagonist phaclofen. These findings have important implications for individuals who abuse opioids and ethanol as well as suggest a mechanism to reduce the amount of opioid needed in chronic pain treatment.


Assuntos
Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Morfina/antagonistas & inibidores , Morfina/farmacologia , Animais , Baclofeno/análogos & derivados , Baclofeno/farmacologia , Bicuculina/farmacologia , Diazepam/farmacologia , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Antagonistas GABAérgicos/farmacologia , Hipnóticos e Sedativos/farmacologia , Imersão , Injeções Intraventriculares , Masculino , Camundongos , Medição da Dor/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-B/efeitos dos fármacos
18.
Neurochem Res ; 38(1): 153-61, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23070470

RESUMO

Uridine, a pyrimidine nucleoside, has been proposed to be a potential signaling molecule in the central nervous system. The understanding of uridine release in the brain is therefore of fundamental importance. The present study was performed to determine the characteristics of basal and morphine-induced uridine release in the striatum of freely moving mice by using the microdialysis technique. To ascertain whether extracellular uridine was derived from neuronal release, the following criteria were applied: sensitivity to (a) K(+) depolarization, (b) Na(+) channel blockade and (c) removal of extracellular Ca(2+). Uridine levels were not greatly affected by infusion of tetrodotoxin (TTX) and were unaffected by either Ca(2+)-free medium or in the presence of EGTA (a calcium chelator), suggesting that basal extracellular uridine levels were maintained mainly by non-vesicular release mechanisms. In addition, both systemic and local application of morphine increased striatal uridine release. The morphine-induced release was reversed by naloxone pretreatment, but was unaffected by TTX or EGTA infusion. Moreover, co-administration of morphine and nitrobenzylthioinosine (NBTI, an inhibitor of nucleotide transporter) produced increases of uridine levels similar to that produced by NBTI or morphine alone, suggesting a nucleotide transporter mechanism involved. Taken together, these findings suggest that morphine produces a µ-opioid receptor-mediated uridine release via nucleoside transporters in a TTX- and calcium-independent manner.


Assuntos
Morfina/farmacologia , Entorpecentes/farmacologia , Neostriado/metabolismo , Uridina/metabolismo , Animais , Western Blotting , Cálcio/fisiologia , Quelantes/farmacologia , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Ácido Egtázico/farmacologia , Camundongos , Microdiálise , Morfina/antagonistas & inibidores , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neostriado/efeitos dos fármacos , Proteínas de Transporte de Nucleotídeos/biossíntese , Proteínas de Transporte de Nucleotídeos/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia
19.
Eur Rev Med Pharmacol Sci ; 17(13): 1748-51, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23852898

RESUMO

AIM: This study investigated the antinociceptive effects of aqueous extract of Murraya koenigii (AEMK) leaves (200, 400 and 800 mg/kg, orally) on animal models of acute and persistent pain and its modulation by naloxone. MATERIALS AND METHODS: Antinociceptive effects were assessed using tail-flick, hot plate and formalin tests in mice. To differentiate between central and peripheral antinociceptive effect of AEMK, naloxone (2 mg/kg) was administered along with the 800 mg/kg dose of extract. Morphine was used as a standard drug. RESULTS: AEMK and morphine significantly increased the tail-flick latency (tfl) and paw licking/jumping latency in tail-flick and hot plate tests, respectively, in comparison to control. Also, in both the tests AEMK and morphine significantly increased the AUC0-120 min. In formalin test, AEMK (400 mg/kg and 800 mg/kg) and morphine significantly reduced licking time in both early and late phases in comparison to control. CONCLUSIONS: Thus, in all three pain models AEMK showed antinociceptive effect, which was blocked by naloxone suggesting the involvement of opioidergic central mechanism.


Assuntos
Analgésicos/antagonistas & inibidores , Analgésicos/farmacologia , Murraya/química , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Formaldeído , Temperatura Alta , Masculino , Camundongos , Morfina/antagonistas & inibidores , Morfina/farmacologia , Medição da Dor , Extratos Vegetais/farmacologia , Folhas de Planta/química , Tempo de Reação/efeitos dos fármacos
20.
Am J Physiol Cell Physiol ; 303(6): C607-15, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22763121

RESUMO

Opiates have been reported to induce T cell loss. We evaluated the role of vitamin D receptor (VDR) and the activation of the renin-angiotensin system (RAS) in morphine-induced T cell loss. Morphine-treated human T cells displayed downregulation of VDR and the activation of the RAS. On the other hand, a VDR agonist (EB1089) enhanced T cell VDR expression both under basal and morphine-stimulated states. Since T cells with silenced VDR displayed the activation of the RAS, whereas activation of the VDR was associated with downregulation of the RAS, it appears that morphine-induced T cell RAS activation was dependent on the VDR status. Morphine enhanced reactive oxygen species (ROS) generation in a dose-dependent manner. Naltrexone (an opiate receptor antagonist) inhibited morphine-induced ROS generation and thus, suggested the role of opiate receptors in T cell ROS generation. The activation of VDR as well as blockade of ANG II (by losartan, an AT(1) receptor blocker) also inhibited morphine-induced T cell ROS generation. Morphine not only induced double-strand breaks (DSBs) in T cells but also attenuated DNA repair response, whereas activation of VDR not only inhibited morphine-induced DSBs but also enhanced DNA repair. Morphine promoted T cell apoptosis; however, this effect of morphine was inhibited by blockade of opiate receptors, activation of the VDR, and blockade of the RAS. These findings indicate that morphine-induced T cell apoptosis is mediated through ROS generation in response to morphine-induced downregulation of VDR and associated activation of the RAS.


Assuntos
Apoptose/fisiologia , Regulação para Baixo/fisiologia , Morfina/farmacologia , Receptores de Calcitriol/metabolismo , Sistema Renina-Angiotensina/fisiologia , Linfócitos T/fisiologia , Apoptose/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Células Jurkat , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/fisiologia , Morfina/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA