Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
China Pharmacy ; (12): 2299-2304, 2023.
Article in Chinese | WPRIM | ID: wpr-988795

ABSTRACT

Opioid receptor agonist-antagonists are a class of drugs which have both agonistic and antagonistic effects on opioid receptors. These drugs already on the market mainly include pentazocine, butorphanol, nalbuphine, buprenorphine, dezocine and so on. Compared with pure opioid receptor agonists such as morphine and fentanyl, these drugs have strong analgesic effects, less addictive, and less side effects such as cough, itching and respiratory depression. Due to the different tendentious effects of opioid receptor agonists-antagonists among different endogenous opioid receptors (μ, κ, δ, etc.), different receptors of subtypes can exhibit different or even opposite effects in terms of affecting emotions and drug dependence. Therefore, the rational use of these drugs can effectively reduce the occurrence of adverse reactions and drug abuse caused by opioid drugs. With the deepening of research on various endogenous opioid receptor subtypes and related drugs in the academic community, opioid receptor agonists- antagonists have broad application space and prospects in improving adverse reactions to opioid drugs and enhancing patient drug compliance.

2.
Neuroscience Bulletin ; (6): 403-416, 2022.
Article in English | WPRIM | ID: wpr-929123

ABSTRACT

Spinal cord stimulation (SCS)-induced analgesia was characterized, and its underlying mechanisms were examined in a spared nerve injury model of neuropathic pain in rats. The analgesic effect of SCS with moderate mechanical hypersensitivity was increased with increasing stimulation intensity between the 20% and 80% motor thresholds. Various frequencies (2, 15, 50, 100, 10000 Hz, and 2/100 Hz dense-dispersed) of SCS were similarly effective. SCS-induced analgesia was maintained without tolerance within 24 h of continuous stimulation. SCS at 2 Hz significantly increased methionine enkephalin content in the cerebrospinal fluid. The analgesic effect of 2 Hz was abolished by μ or κ opioid receptor antagonist. The effect of 100 Hz was prevented by a κ antagonist, and that of 10 kHz was blocked by any of the μ, δ, or κ receptor antagonists, suggesting that the analgesic effect of SCS at different frequencies is mediated by different endorphins and opioid receptors.


Subject(s)
Animals , Rats , Analgesics , Narcotic Antagonists/pharmacology , Neuralgia/therapy , Opioid Peptides , Receptors, Opioid/physiology , Receptors, Opioid, kappa , Spinal Cord , Spinal Cord Stimulation
3.
Chinese Journal of Clinical Pharmacology and Therapeutics ; (12): 373-379, 2020.
Article in Chinese | WPRIM | ID: wpr-855856

ABSTRACT

AIM: To investigate the effect of opioid μ receptor on testosterone and its peripheral mechanism. METHODS: Levels of testosterone and testosterone synthetase mRNA in testis plasma and testis homogenate, IGF1 protein and mRNA expression in testis homogenate were detected by intratesticular morphine injection in vivo animal model and treatment of specific μ receptor antagonists and agonists in vitro cultured testis. RESULTS:Serum testosterone level was decreased after testis was injected with morphine [(722.11±121.02) vs. (346.91±75.31) pg/mL; t=7.898, P=0.001]. Testis homogenate testosterone level [(133.61±16.82) vs. (66.56±14.96) pg/mg protein; t=8.941, P=0.001] and expression of testosterone synthetase were decreased. Testis IGF1 level [(7.93±2.13) vs. (2.54±0.74) ng/mg protein, t=7.155, P=0.001] and IGF1 mRNA [(3.22±1.12) vs. (1.66±0.55), t=3.751, P=0.001] expression were decreased in intratesticular morphine injection model. Testosterone level in culture media, IGF1 and expression of testosterone synthetase were decreased after testis was injected with DAMGO in vitro model. And testosterone level, IGF1 and expression of testosterone synthetase were increased after testis was injected with CTOP.CONCLUSION: Morphine acts on opioid μ receptor on the surface of testicular Sertoli cells and inhibits the synthesis of IGF1. It decreases expression of testosterone synthetase Scarb1, Star, 3βHsd, Cyp17α1 and 17βHsd from testicular Leydig cells. which leads to a decrease in testosterone synthesis.

4.
Journal of Integrative Medicine ; (12): 396-403, 2018.
Article in English | WPRIM | ID: wpr-691052

ABSTRACT

<p><b>OBJECTIVE</b>The present study aimed to evaluate the analgesic and anti-inflammatory effects of far infrared-emitting ceramics (cFIRs) in a model of persistent inflammatory hyperalgesia and to elucidate the possible mechanisms of these effects.</p><p><b>METHODS</b>Mice were injected with complete Freund's adjuvant (CFA) and treated with cFIRs via placement on a pad impregnated with cFIRs on the bottom of the housing unit for different periods of time. Mice underwent mechanical hyperalgesia and edema assessments, and tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and IL-10 levels were measured. Twenty-four hours after CFA injection and 30 min before cFIR treatment, mice were pretreated with a nonselective adenosinergic antagonist, caffeine, the selective adenosine receptor A antagonist, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX), the selective cannabinoid receptor type 1 antagonist, AM281, the selective cannabinoid receptor type 2 antagonist, AM630, or the nonselective opioid receptor antagonist, naloxone, and mechanical hyperalgesia was assessed.</p><p><b>RESULTS</b>cFIRs statistically (P < 0.05) decreased CFA-induced mechanical hyperalgesia ((82.86 ± 5.21)% in control group vs (56.67 ± 9.54)% in cFIR group) and edema ((1699.0 ± 77.8) μm in control group vs (988.7 ± 107.6) μm in cFIR group). cFIRs statistically (P < 0.05) reduced TNF-α ((0.478 ± 0.072) pg/mg of protein in control group vs (0.273 ± 0.055) pg/mg of protein in cFIR group) and IL-1β ((95.81 ± 3.95) pg/mg of protein in control group vs (80.61 ± 4.71) pg/mg of protein in cFIR group) levels and statistically (P < 0.05) increased IL-10 ((18.32 ± 0.78) pg/mg of protein in control group vs (25.89 ± 1.23) pg/mg of protein in cFIR group) levels in post-CFA-injected paws. Peripheral pre-administration of inhibitory neuroreceptor antagonists (caffeine, DPCPX, AM281, AM630 and naloxone) prevented the analgesic effects of cFIRs (P < 0.05).</p><p><b>CONCLUSION</b>These data provide additional support for the use of cFIRs in the treatment of painful inflammatory conditions and contribute to our understanding of the neurobiological mechanisms of the therapeutic effects of cFIRs.</p>

5.
Braz. J. Pharm. Sci. (Online) ; 54(1): e17217, 2018. tab
Article in English | LILACS | ID: biblio-951905

ABSTRACT

ABSTRACT O sistema opioidérgico envolve a regulação do sono e da vigília. É possível, portanto, que os polimorfismos genéticos no OPRM1 influenciem na qualidade do sono. Este estudo investigou a associação de polimorfismos do OPRM1 com a qualidade subjetiva do sono entre indivíduos sem tratamento prévio com opióides. Este estudo observacional de corte transversal envolveu 161 homens que nunca haviam se tornado opióides (média de idade = 27,74 anos; variação: 18 a 63 anos). A qualidade subjetiva do sono foi avaliada com a versão traduzida e validada em malaio do Índice de Qualidade do Sono de Pittsburgh (PSQI). O DNA foi extraído do sangue total e submetido à reação em cadeia da polimerase (PCR) para dois polimorfismos OPRM1 (118A> G e IVS2 + 691G> C). Sujeitos combinados com 118A e IVS2 + 691Galelos (haplótipo AC) apresentaram escores significativamente mais baixos do PSQI [média (DP) = 4,29 (1,76)] em comparação àqueles sem o haplótipo [4,99 (2,50)] (p = 0,004). Por outro lado, os indivíduos com genótipo heterozigótico combinado (GC / AG diplotipo) apresentaram escores significativamente mais altos do PSQI em comparação àqueles sem o diplótipo [6,04 (2,48) vs 4,54 (2,22), p = 0,004]. Em indivíduos sem tratamento prévio com opiáceos, o haplótipo AC e o diplótipo GC / AG para os polimorfismos 118A> G e IVS2 + 691G> C do OPRM1 estão associados a uma melhor e pior qualidade do sono, respectivamente.


Subject(s)
Humans , Male , Adolescent , Adult , Middle Aged , Sleep/genetics , Sleep Wake Disorders , Receptors, Opioid, mu/analysis , Polymorphism, Genetic/genetics , Receptors, Opioid/analysis
6.
Basic & Clinical Medicine ; (12): 422-426, 2017.
Article in Chinese | WPRIM | ID: wpr-510499

ABSTRACT

Respiratory depression is a common adverse effect of opioids .Currently, the mechanisms and manage-ment of opioid-induced respiratory depression ( OIRD) are one of the research foci .OIRD arises from stimulation ofμ-opioid receptors in the pre-Botzinger complex and the Kolliker-Fuse neurons .Adenylyl cyclase , calcium chan-nels, and G-protein-gated inwardly rectifying potassium (GIRK) channels may be the key cellular signaling mecha-nisms of OIRD .

7.
Asian Pacific Journal of Tropical Biomedicine ; (12): 198-201, 2016.
Article in Chinese | WPRIM | ID: wpr-950842

ABSTRACT

Objective: To investigate the analgesic and anti-inflammatory property and possible involvement of opioid receptors of ethyl acetate extract from aerial parts of Daphne mucronata (D. mucronata) in mice by formalin test. Methods: Single doses of 2.5, 5.0 and 10.0 mg/kg of body weight of ethyl acetate extract of D. mucronata were intraperitoneally administered to the mice 30 min before analgesic test. The anti-nociceptive effect of preparations was evaluated based on the formalin in mice. Results: The results indicated that the extract (2.5, 5.0 and 10.0 mg/kg) increased the pain threshold of mice and induced analgesia in both phases of formalin test. Like morphine sulfate (5.0 mg/kg, i.p.), the extract also showed more effective analgesic effect on the late phase of formalin test. Pre-treatment of animals with naloxone (5.0 mg/kg i.p.) did not inhibit the effects of the extract. Conclusions: Our findings suggest that D. mucronata contains potential analgesic and anti-inflammatory compounds which support its traditional use. Moreover, it seems that the analgesic and anti-inflammatory effects of the extract is mediated by non-opioid mechanisms. Further pharmacological studies are required to determine whether the analgesic mechanisms are actually responsible for such properties.

8.
Journal of Breast Cancer ; : 206-209, 2016.
Article in English | WPRIM | ID: wpr-166631

ABSTRACT

Distribution of A118G single nucleotide polymorphism (SNP) in the mu-opioid receptor 1 gene (OPRM1) differs with ethnicity. We assessed the distribution of this SNP in Korean women with breast cancer and compared it with that in women of other ethnicities with breast cancer. Distribution of SNP genotypes was as follows: 49.8% for AG genotype, 40.6% for AA genotype, and 9.6% for GG genotype. Logistic regression analysis showed a negative association between the presence of the G allele at position 118 of OPRM1 and breast cancer in the studied population (odds ratios [OR], 0.635; p=0.002). However, the AG and GG genotypes were not associated with breast cancer in the studied population (OR, 0.719; p=0.130). The proportions of the AG and GG genotypes of the OPRM1 SNP were higher in Korean women with breast cancer than in those of other ethnicities.


Subject(s)
Adult , Female , Humans , Alleles , Breast Neoplasms , Breast , Genotype , Logistic Models , Polymorphism, Single Nucleotide , Receptors, Opioid, mu , Retrospective Studies
9.
Rev. bras. anestesiol ; 65(3): 186-190, May-Jun/2015. graf
Article in English | LILACS | ID: lil-748921

ABSTRACT

BACKGROUND AND OBJECTIVES: Tramadol is known as a central acting analgesic drug, used for the treatment of moderate to severe pain. Local analgesic effect has been demonstrated, in part due to local anesthetic-like effect, but other mechanisms remain unclear. The role of peripheral opioid receptors in the local analgesic effect is not known. In this study, we examined role of peripheral opioid receptors in the local analgesic effect of tramadol in the plantar incision model. METHODS: Young male Wistar rats were divided into seven groups: control, intraplantar tramadol, intravenous tramadol, intravenous naloxone-intraplantar tramadol, intraplantar naloxone-intraplantar tramadol, intravenous naloxone-intravenous tramadol, and intravenous naloxone. After receiving the assigned drugs (tramadol 5 mg, naloxone 200 µg or 0.9% NaCl), rats were submitted to plantar incision, and withdrawal thresholds after mechanical stimuli with von Frey filaments were assessed at baseline, 10, 15, 30, 45 and 60 min after incision. RESULTS: Plantar incision led to marked mechanical hyperalgesia during the whole period of observation in the control group, no mechanical hyperalgesia were observed in intraplantar tramadol group, intraplantar naloxone-intraplantar tramadol group and intravenous naloxone-intraplantar tramadol. In the intravenous tramadol group a late increase in withdrawal thresholds (after 45 min) was observed, the intravenous naloxone-intravenous tramadol group and intravenous naloxone remained hyperalgesic during the whole period. CONCLUSIONS: Tramadol presented an early local analgesic effect decreasing mechanical hyperalgesia induced by plantar incision. This analgesic effect was not mediated by peripheral opioid receptors. .


JUSTIFICATIVA E OBJETIVOS: Tramadol é conhecido como um fármaco analgésico de ação central, usado para o tratamento de dor moderada a grave. O efeito analgésico local foi demonstrado, em parte devido ao efeito semelhante ao anestésico local, mas outros mecanismos permanecem obscuros. O papel dos receptores opioides periféricos no efeito analgésico local não é conhecido. Neste estudo, examinamos o papel dos receptores opioides periféricos no efeito analgésico local de tramadol em modelo de incisão plantar. MÉTODOS: Ratos Wistar, jovens e machos, foram divididos em sete grupos: controle, tramadol intraplantar, tramadol intravenoso, tramadol intraplantar-naloxona intravenosa, tramadol intraplantar-naloxona intraplantar, tramadol intravenoso-naloxona intravenosa e naloxona intravenosa. Após receber os medicamentos designados (5 mg de tramadol, 200 mg de naloxona ou NaCl a 0,9%, os ratos foram submetidos à incisão plantar e os limiares de retirada após estímulos mecânicos com filamentos de von Frey foram avaliados no início do estudo e nos minutos 10, 15, 30, 45 e 60 após a incisão. RESULTADOS: A incisão plantar levou à hiperalgesia mecânica acentuada durante todo o período de observação no grupo controle; hiperalgesia mecânica não foi observada nos grupos tramadol intraplantar, tramadol intraplantar-naloxona intraplantar e tramadol intraplantar-naloxona intravenosa. No grupo tramadol intravenoso, um aumento tardio do limiar de retirada (após 45 minutos) foi observado. Os grupos tramadol intravenoso-naloxona intravenosa e naloxona intravenosa permaneceram hiperalgésicos durante todo o período. CONCLUSÕES: Tramadol apresentou efeito analgésico local inicial e diminuiu a hiperalgesia mecânica induzida pela incisão plantar. Esse efeito analgésico não foi mediado por receptores opioides periféricos. .


JUSTIFICACIÓN Y OBJETIVOS: Al tramadol se le conoce como un medicamento analgésico de acción central usado para el tratamiento del dolor moderado a intenso. El efecto analgésico local quedó demostrado, en parte, a causa del efecto similar al del anestésico local, pero otros mecanismos permanecen sin clarificar. El rol de los receptores opiáceos periféricos en el efecto analgésico local no se conoce. En este estudio, examinamos el papel de los receptores opiáceos periféricos en el efecto analgésico local del tramadol en un modelo de incisión plantar. MÉTODOS: Ratones Wistar, jóvenes y machos, fueron divididos en 7 grupos: control, tramadol intraplantar, tramadol intravenoso, tramadol intraplantar-naloxona intravenosa, tramadol intraplantar-naloxona intraplantar, tramadol intravenoso-naloxona intravenosa, y naloxona intravenosa. Después de recibir los medicamentos designados (5 mg de tramadol, 200 µg de naloxona o NaCl al 0,9%), los ratones fueron sometidos a la incisión plantar, y los umbrales de retirada de la pata posteriores a los estímulos mecánicos con filamentos de von Frey fueron evaluados al inicio del estudio y en los minutos 10, 15, 30, 45 y 60 después de la incisión. RESULTADOS: La incisión plantar conllevó hiperalgesia mecánica acentuada durante todo el período de observación en el grupo control; la hiperalgesia mecánica no fue observada en los grupos tramadol intraplantar, tramadol intraplantar-naloxona intraplantar, y tramadol intraplantar-naloxona intravenosa. En el grupo tramadol intravenoso, fue observado un aumento tardío del umbral de retirada (después de 45 min); los grupos tramadol intravenoso-naloxona intravenosa y naloxona intravenosa permanecieron hiperalgésicos durante todo el período. CONCLUSIONES: El tramadol presentó un efecto analgésico local inicial, disminuyendo la hiperalgesia mecánica inducida por la incisión plantar. Ese efecto analgésico no fue mediado por receptores opiáceos periféricos. .


Subject(s)
Animals , Male , Rats , Pain, Postoperative/drug therapy , Tramadol/pharmacology , Hyperalgesia/drug therapy , Analgesics, Opioid/pharmacology , Time Factors , Tramadol/administration & dosage , Rats, Wistar , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Disease Models, Animal , Analgesics, Opioid/administration & dosage , Injections , Injections, Intravenous , Naloxone/administration & dosage , Naloxone/pharmacology
10.
Annals of Rehabilitation Medicine ; : 331-339, 2015.
Article in English | WPRIM | ID: wpr-153689

ABSTRACT

OBJECTIVE: To investigate the effects of aerobic exercise on neuropathic pain and verify whether regular treadmill exercise alters opioid receptor expression in the rostral ventral medulla (RVM) in a neuropathic pain rat model. METHODS: Thirty-two male Sprague-Dawley rats were used in the study. All rats were divided into 3 groups, i.e., group A, sham group (n=10); group B, chronic constriction injury (CCI) group (n=11); and group C, CCI+exercise group (n=11). Regular treadmill exercise was performed for 30 minutes a day, 5 days a week, for 4 weeks at the speed of 8 m/min for 5 minutes, 11 m/min for 5 minutes, and 22 m/min for 20 minutes. Withdrawal threshold and withdrawal latency were measured before and after the regular exercise program. Immunohistochemistry and Western blots analyses were performed using antibodies against micro-opioid receptor (MOR). RESULTS: Body weight of group C was the lowest among all groups. Withdrawal thresholds and withdrawal latencies were increased with time in groups B and C. There were significant differences of withdrawal thresholds between group B and group C at 1st, 2nd, 3rd, and 4th weeks after exercise. There were significant differences of withdrawal latencies between group B and group C at 3rd and 4th weeks after exercise. MOR expression of group C was significantly decreased, as compared to that of group B in the RVM and spinal cord. CONCLUSION: In neuropathic pain, exercise induced analgesia could be mediated by desensitization of central MOR by endogenous opioids, leading to the shift of RVM circuitry balance to pain inhibition.


Subject(s)
Animals , Humans , Male , Rats , Analgesia , Analgesics, Opioid , Antibodies , Blotting, Western , Body Weight , Constriction , Exercise , Immunohistochemistry , Medulla Oblongata , Models, Animal , Neuralgia , Rats, Sprague-Dawley , Receptors, Opioid , Spinal Cord
11.
Journal of International Pharmaceutical Research ; (6): 200-205, 2014.
Article in Chinese | WPRIM | ID: wpr-845780

ABSTRACT

Objective To stably express rat mu-opioid receptor (rMOR) in PC12 cells with characteristics of neurons. Methods After the lentiviral vector pBPLV-rMOR-eGFP was constructed, the lentivirus was packaged and used to infect the PC12 cells. PC12 cells stably expressing rMOR was screened by the flow cytometry and the limiting dilution assay. The affinity and quantity of rMOR protein expressed in the PC12 were verified by radio-ligand binding assay. The function of rMOR was analyzed by cAMP overshooting. Results The eGFP protein in PC12-rMOR cells infected by the lentivirus could be clearly shown by the fluorescence microscope. Cell lines grew normally after every clone was enlargedly cultured. The affinity (Kd) and quantity (Bmax) values of rMOR were (0.51 ± 0.07) nmol/L and (1.58 ± 0.15)pmol/mg protein respectively in 3H-diprenorphine binding assay. The cAMP content increased (255 ±25.2) % after naloxone precipitated in chronic morphine-treated cells. Exogenous agmatine could dose-dependently inhibit the overshooting of cAMP by naloxone precipitated. Conclusion We have successfully established the PC12 cell model co-expressing stably rMOR and I1 style imidazoline receptor(I1R) without α2 adrenergic receptor, expressing properties of neurons, which is a good cell model in vitro for investigating the neural molecular mechanism of opioid addition and regulating the opioid receptor function by the system of agmatine and I 1 Rin the future.

12.
Rev. dor ; 13(1): 75-79, jan.-mar. 2012.
Article in Portuguese | LILACS | ID: lil-624936

ABSTRACT

JUSTIFICATIVA E OBJETIVOS: Devido à finalidade do ambulatório e os fármacos utilizados rotineiramente, o objetivo deste estudo foi rever e atualizar os conhecimentos sobre os receptores opioides e como complemento de estudo após palestra apresentada aos integrantes da equipe. CONTEÚDO: Foram revisados desde os aspectos históricos até os conhecimentos mais recentes sobre receptores opioides, descritos seus subtipos e mecanismos de ação. Para tal, foram consultadas referências indexadas pelo Pubmed. CONCLUSÃO: Com os dados presentes na literatura atual, concluiu-se que ainda existe muito a ser pesquisado sobre o tópico, visando medicações mais seguras e novas técnicas biomoleculares ainda são necessárias.


BACKGROUND AND OBJECTIVES: Due to the objective of the outpatient setting and to routinely used drugs, this study aimed at reviewing and updating the knowledge about opioid receptors and worked as a study complement after a lecture presented to team members. CONTENTS: We have reviewed from historical aspects to most recent developments about opioid receptors, in addition to describing subtypes and action mechanisms. For such, Pubmed-indexed references were queried. CONCLUSION: After reviewing current literature data, we have concluded that there is still a lot to be researched about the topic, aiming at safer drugs, and new biomolecular techniques are still needed.


Subject(s)
Analgesics, Opioid , History of Medicine , Morphine , Narcotic Antagonists , Opium , Papaver , Receptors, Opioid
13.
São Paulo; s.n; 2011. 181 p.
Thesis in Portuguese | LILACS, SES-SP, SESSP-IBPROD, SES-SP, SESSP-IBACERVO | ID: biblio-1080916

ABSTRACT

Este trabalho teve como objetivo avaliar os mecanismos envolvidos no aumento da eficácia analgesia da crotalfina (CRF), um peptídeo com atividade tipo opióide, na vigência de inflamação (prostaglandina E2/PGE2) ou lesão tecidual (constrição crônica do nervo isquiático/CCI). A PGE2 (intraplantar), em ratos, aumentou a expressão gênica e protéica de receptores opióides do tipo m e k, no gânglio da raiz dorsal (DRG) e nervo da pata (NP), quando comparado a animais naive. A CCI aumentou a expressão de receptores m e d, no DRG e diminuiu os níveis de receptores k. Embora a PGE2 e CCI, per se, não ativem receptores opióides o CRF e agonistas opióides acarretam maior ativação destes receptores na vigência de sensibilização por PGE2 e CCI. Ainda, o CRF ativa a via das MAPKs (ERK1/2 e JNK), apenas na presença de PGE2, efeito mediado por receptores do tipo k e PKCz. Estes dados mostram que a expressão e ativação de receptores opióides são diferentemente regulados pela injúria aguda ou crônica.


This study aimed to evaluate the mechanisms involved in the increased analgesic efficacy of crotalphine (CRP), an opioid-like peptide, under inflammation prostaglandin E2 (PGE2) and tissue injury (chronic constriction injury/CCI). PGE2 (intraplantar) in rats, increases the genic and proteic expression of m- and k-opioid receptors in the dorsal root ganglia (DRG) and nerve paw (NP), when compared to naïve rats. CCI up-regulates the expression of m and d -opioid receptors in DRG and NP. In contrast, k-opioid receptors were down-regulated by CCI. Although PGE2 and CCI, per se, do not activate opioid receptors, CRP and opioid agonists lead to a higher activation of these receptors in NP slices under PGE2 or CCI sensitization or in DRG cells incubated with PGE2. Moreover, CRP activates ERK1/2 and JNK MAPKs pathways, k-receptor and PKCz-mediated, only when the cells were pre-incubated with PGE2. These data indicate that the expression and activation of opioid receptors are distinctly regulated by the presence of acute or chronic injury.


Subject(s)
Animals , Rats , Analgesia , Analgesics, Opioid/therapeutic use , Receptors, Opioid/therapeutic use , Inflammation
14.
Korean Journal of Anesthesiology ; : 69-74, 2011.
Article in English | WPRIM | ID: wpr-171786

ABSTRACT

BACKGROUND: It is generally accepted that morphine affords cardioprotection against ischemia/reperfusion injury. Inhibition of the mitochondrial permeability transition pore (MPTP) is considered an end target for cardioprotection. The aim of this study was to investigate the involvement of opioid receptors (OR) and MPTP in morphine-induced postconditioning (M-Post). METHODS: Isolated rat hearts were subjected to 30 min of regional ischemia and 2 h of reperfusion. Hearts were treated with 1 microM morphine, with or without the OR antagonists or a MPTP opener at early reperfusion. Infarct size was measured with 2,3,5-triphenyltetrazolium chloride staining. RESULTS: There were no significant differences in cardiodynamic variables except a decrease in heart rate in the M-Post group (P 0.05). The nonspecific OR antagonist naloxone (25.7 +/- 1.9%, P < 0.01), the delta-OR antagonist naltrindole (27.8 +/- 4.3%, P < 0.05) and delta1-OR antagonist 7-benzylidenenaltrexone (24.7 +/- 3.7%, P < 0.01) totally abrogated the anti-infarct effect of M-Post. In addition, the anti-infarct effect by M-Post was also totally blocked by the MPTP opener atractyloside (26.3 +/- 5.2%, P < 0.05). CONCLUSIONS: M-Post effectively reduces myocardial infarction. The anti-infarct effect by M-Post is mediated via activation of delta-OR, especially delta1-OR, and inhibition of the MPTP opening.


Subject(s)
Animals , Rats , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Atractyloside , Benzylidene Compounds , Heart , Heart Rate , Ischemia , Ischemic Postconditioning , Mitochondrial Membrane Transport Proteins , Morphine , Myocardial Infarction , Naloxone , Naltrexone , Permeability , Receptors, Opioid , Reperfusion , Reperfusion Injury , Tetrazolium Salts
15.
Korean Journal of Anesthesiology ; : 351-356, 2011.
Article in English | WPRIM | ID: wpr-224612

ABSTRACT

BACKGROUND: We investigated whether p42/p44 extracellular signal-regulated kinases (ERK1/2) and/or phosphatidylinositol-3-OH kinase (PI3K)-Akt play a crucial role in cardioprotection by kappa-opioid receptor (KOP) activation. METHODS: Langendorff perfused rat hearts were subjected to 30 min of regional ischemia and 2 h of reperfusion. Antagonists of ERK1/2 and PI3K were perfused in hearts treated with the KOP agonist U50488H (U50). Infarct size was measured after 2 h of reperfusion. The phosphorylation states of ERK1/2 and Akt by Western immunoblots were determined. Drugs were perfused for a period of 5 min before and 30 min after reperfusion. RESULTS: Inhibition of ERK1/2 (26.8 +/- 2.9%, P 0.05 vs. U50) completely abrogated the anti-infarct effect of U50488H. Western blot analysis revealed a significant increase in ERK1/2 but not Akt phsophorylation in U50488H-treated hearts as compared to control hearts when measured immediately after reperfusion. CONCLUSIONS: KOP activation effectively reduces myocardial infarction. The anti-infarct effect of U50488H is mediated by the ERK1/2, but not the PI3K-Akt pathway.


Subject(s)
Animals , Rats , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer , Blotting, Western , Coronary Occlusion , Extracellular Signal-Regulated MAP Kinases , Heart , Ischemia , Myocardial Infarction , Phosphatidylinositol 3-Kinases , Phosphorylation , Receptors, Opioid , Reperfusion
16.
Braz. j. med. biol. res ; 43(9): 906-909, Sept. 2010. ilus
Article in English | LILACS | ID: lil-556855

ABSTRACT

Exercise is a low-cost intervention that promotes health and contributes to the maintenance of the quality of life. The present study was designed to investigate the influence of different resistance exercise protocols on the nociceptive threshold of rats. Female Wistar rats were used to perform exercises in a weight-lifting exercise model. The following groups were examined (N = 6 per group): untrained rats (control group); an acute protocol group consisting of rats submitted to 15 sets of 15 repetitions of resistance exercise (acute group); rats exercised with 3 sets of 10 repetitions, three times per week for 12 weeks (trained group), and a group consisting of trained rats that were further submitted to the acute protocol (trained-acute group). The nociceptive threshold was measured by the paw-withdrawal test, in which the withdrawal threshold (escape reaction) was measured by an apparatus applying force to the plantar surface of the animal paw. The opioid antagonist naloxone (2 mg/kg) was administered subcutaneously 10 min before the exercise protocols. The trained group demonstrated antinociception only up to day 45 of the 12-week training period. A significant increase (37 percent, P < 0.05) in the nociceptive threshold was produced immediately after exercise, decreasing to 15 percent after 15 min, when the acute exercise protocol was used. Naloxone reversed this effect. These data show that the acute resistance exercise protocol was effective in producing antinociception for 15 min. This antinociceptive effect is mediated by the activation of opioid receptors.


Subject(s)
Animals , Female , Rats , Analgesia , Physical Conditioning, Animal , Pain Threshold/drug effects , Resistance Training , Receptors, Opioid/physiology , Pain Measurement , Pain Threshold/physiology , Rats, Wistar
17.
International Journal of Oral Biology ; : 69-74, 2010.
Article in Korean | WPRIM | ID: wpr-75409

ABSTRACT

The mu opioid receptor (MOR) has been regarded as the main site of interaction with analgesics in major clinical use, particularly morphine. The repressor element-1 silencing transcription factor (REST) functions as a transcriptional repressor of neuronal genes in non-neuronal cells. However, it is expressed in certain mature neurons, suggesting that it may have complex and novel roles. In addition, the interactions between MOR and REST and their functions remain unclear. In this study, we examined the effects of morphine on the expression of REST mRNA and protein in human neuroblastoma NMB cells to investigate the roles of REST induced by MOR activation in neuronal cells. To determine the effects of morphine on REST expression, we performed RT-PCR, real-time quantitative RT-PCR, western blot analysis and radioligand binding assays in NMB cells. By RT-PCR and real-time quantitative RT-PCR, the expression of REST was found to be unchanged by either the MOR agonist morphine or the MOR specific antagonist CTOP. By western blot, morphine was shown to significantly inhibit the expression of REST, but this suppression was completely blocked by treatment with CTOP. In the radioligand binding assay, the overexpression of REST led to an increased opioid ligand binding activity of endogenous MOR in the NMB cells. These results together suggest that morphine inhibits the expression of REST in human neuroblastoma cells through a post-transcriptional regulatory mechanism mediated through MOR.


Subject(s)
Humans , Analgesics , Blotting, Western , Morphine , Neuroblastoma , Neurons , Receptors, Opioid, mu , RNA, Messenger , Somatostatin , Transcription Factors
18.
Korean Journal of Anesthesiology ; : 162-168, 2010.
Article in English | WPRIM | ID: wpr-138719

ABSTRACT

BACKGROUND: Because the kappa-opioid receptor (OR) agonist U50488H stimulates opioidergic regulation and inhibits L-type Ca2+ channels, this study was aimed at assessing the roles of OR and L-type Ca2+ channels on U50488H-induced cardioprotection. METHODS: Langendorff-perfused rat hearts were subjected to 30 min of regional ischemia and 2 h of reperfusion. Isolated hearts were treated with U50488H with or without the kappa-OR antagonist nor-binaltorphimine (nor-BNI) or the Ca2+ channels activator BAY K 8644. Infarct size was measured with 2,3,5-triphenyltetrazolium chloride staining. RESULTS: U50488H treatment at reperfusion: (1) significantly reduced infarct size (11.3 +/- 1.3%) compared to control hearts (27.7 +/- 1.1%, P 0.05 vs. U50488H); (2) significantly increased left ventricular developed pressure (65.3 +/- 4.8%) after 2 h of reperfusion compared to control hearts (44.8 +/- 3.6%, P 0.05 vs. control) but not by BAY K 8644 (64.3 +/- 5.6%, P < 0.01 vs. control); and (3) significantly decreased heart rate (P < 0.01 vs. control), an effect that was completely abrogated by both nor-BNI and BAY K 8644. CONCLUSIONS: U50488H significantly limits myocardial infarction and stunning in isolated rat hearts after ischemia-reperfusion induction. The infarct size limitation and contractility improvement observed with U50488H treatment during reperfusion are entirely mediated by OR stimulation and not by Ca2+ channel modulation.


Subject(s)
Animals , Rats , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester , Calcium , Coronary Occlusion , Heart , Heart Rate , Ischemia , Myocardial Infarction , Naltrexone , Receptors, Opioid , Reperfusion , Tetrazolium Salts
19.
Korean Journal of Anesthesiology ; : 162-168, 2010.
Article in English | WPRIM | ID: wpr-138718

ABSTRACT

BACKGROUND: Because the kappa-opioid receptor (OR) agonist U50488H stimulates opioidergic regulation and inhibits L-type Ca2+ channels, this study was aimed at assessing the roles of OR and L-type Ca2+ channels on U50488H-induced cardioprotection. METHODS: Langendorff-perfused rat hearts were subjected to 30 min of regional ischemia and 2 h of reperfusion. Isolated hearts were treated with U50488H with or without the kappa-OR antagonist nor-binaltorphimine (nor-BNI) or the Ca2+ channels activator BAY K 8644. Infarct size was measured with 2,3,5-triphenyltetrazolium chloride staining. RESULTS: U50488H treatment at reperfusion: (1) significantly reduced infarct size (11.3 +/- 1.3%) compared to control hearts (27.7 +/- 1.1%, P 0.05 vs. U50488H); (2) significantly increased left ventricular developed pressure (65.3 +/- 4.8%) after 2 h of reperfusion compared to control hearts (44.8 +/- 3.6%, P 0.05 vs. control) but not by BAY K 8644 (64.3 +/- 5.6%, P < 0.01 vs. control); and (3) significantly decreased heart rate (P < 0.01 vs. control), an effect that was completely abrogated by both nor-BNI and BAY K 8644. CONCLUSIONS: U50488H significantly limits myocardial infarction and stunning in isolated rat hearts after ischemia-reperfusion induction. The infarct size limitation and contractility improvement observed with U50488H treatment during reperfusion are entirely mediated by OR stimulation and not by Ca2+ channel modulation.


Subject(s)
Animals , Rats , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester , Calcium , Coronary Occlusion , Heart , Heart Rate , Ischemia , Myocardial Infarction , Naltrexone , Receptors, Opioid , Reperfusion , Tetrazolium Salts
20.
The Korean Journal of Physiology and Pharmacology ; : 223-230, 2005.
Article in English | WPRIM | ID: wpr-728726

ABSTRACT

The purpose of the present study was to examine the effect of naltrexone, an opioid antagonist, on secretion of catecholamines (CA) evoked by cholinergic nicotinic stimulation and membrane-depolarization from the isolated perfused rat adrenal gland and to establish the mechanism of its action. Naltrexone (3x10 (-6) M) perfused into an adrenal vein for 60 min produced time-dependent inhibition in CA secretory responses evoked by ACh (5.32x10 (-3) M), high K+ (5.6x10 (-2) M), DMPP (10 (-4) M) and McN-A-343 (10 (-4) M). Naltrexone itself did also fail to affect basal CA output. In adrenal glands loaded with naltrexone (3x10 (-6) M), the CA secretory responses evoked by Bay-K-8644, an activator of L-type Ca2+ channels and cyclopiazonic acid, an inhibitor of cytoplasmic Ca2+-ATPase, were also inhibited. However, in the presence of met-enkephalin (5x10 (-6) M), a well-known opioid agonist, the CA secretory responses evoked by ACh, high K+, DMPP, McN-A-343, Bay-K-8644 and cyclopiazonic acid were also significantly inhibited. Collectively, these experimental results demonstrate that naltrexone inhibits greatly CA secretion evoked by stimulation of cholinergic (both nicotinic and muscarinic) receptors as well as that by membrane depolarization. It seems that this inhibitory effect of naltrexone does not involve opioid receptors, but might be mediated by blocking both the calcium influx into the rat adrenal medullary chromaffin cells and the uptake of Ca2+ into the cytoplasmic calcium store, which are at least partly relevant to the direct interaction with the nicotinic receptor itself.


Subject(s)
Animals , Rats , (4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester , Adrenal Glands , Adrenal Medulla , Calcium , Catecholamines , Chromaffin Cells , Cytoplasm , Dimethylphenylpiperazinium Iodide , Enkephalin, Methionine , Membranes , Naltrexone , Receptors, Nicotinic , Receptors, Opioid , Veins
SELECTION OF CITATIONS
SEARCH DETAIL