Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(9)2023 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-37176075

RESUMEN

Remifentanil (REM) and fentanyl (FEN) are commonly used analgesics that act by activating a µ-opioid receptor (MOR). Although optimal concentrations of REM can be easily maintained during surgery, it is sometimes switched to FEN for optimal pain regulation. However, standards for this switching protocol remain unclear. Opioid anesthetic efficacy is decided in part by MOR desensitization; thus, in this study, we investigated the desensitization profiles of REM and FEN to MOR. The efficacy and potency during the 1st administration of REM or FEN in activating the MOR were almost equal. Similarly, in ß arrestin recruitment, which determines desensitization processes, they showed no significant differences. In contrast, the 2nd administration of FEN resulted in a stronger MOR desensitization potency than that of REM, whereas REM showed a higher internalization potency than FEN. These results suggest that different ß arrestin-mediated signaling caused by FEN or REM led to their distinct desensitization and internalization processes. Our three-dimensional analysis, with in silico binding of REM and FEN to MOR models, highlighted that REM and FEN bound to similar but distinct sites of MOR and led to distinct ß arrestin-mediated profiles, suggesting that distinct binding profiles to MOR may alter ß arrestin activity, which accounts for MOR desensitization and internalization.


Asunto(s)
Fentanilo , Receptores Opioides , Receptores Opioides/metabolismo , Fentanilo/farmacología , Remifentanilo/farmacología , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacología , beta-Arrestinas/metabolismo , Morfina
2.
Anesth Analg ; 134(5): 1082-1093, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35427270

RESUMEN

BACKGROUND: Transdermal fentanyl is widely used in the treatment of severe pain because of convenience, safety, and stable blood concentrations. Nevertheless, patients often develop tolerance to fentanyl, necessitating the use of other opioids; transdermal buprenorphine patch is widely used as an analgesic agent, though available formulation does not provide comparable analgesic effect as transdermal fentanyl patch. Opioids bind to the opioid receptor (OR) to activate both G protein-mediated and ß-arrestin-mediated pathways. We synthesized morphine-related compounds with high transdermal absorbability (N1 and N2) and evaluated their OR activities pharmacologically in comparison with fentanyl and morphine. METHODS: In cells stably expressing µ-opioid receptor (MOR), δ-opioid receptor (DOR), and κ-opioid receptor (KOR), G protein-mediated pathways were assessed using the CellKey and an intracellular cyclic adenosine monophosphate (cAMP) assay, while ß-arrestin-mediated pathways were analyzed with ß-arrestin recruitment and receptor internalization assays. Furthermore, analgesic effects were evaluated using a tail-flick test in mice, and the analgesic effect on fentanyl-tolerant mice was evaluated. RESULTS: In the CellKey and cAMP assays, both N1 and N2 showed the highest affinity for MOR and acted as full agonists as well as partial agonists for DOR and KOR. In the ß-arrestin and internalization assays, only fentanyl acted as a full agonist; N1 and N2 acted as partial agonists of MOR. In the mouse tail-flick test, N1 and N2 showed analgesic effects equivalent to those of fentanyl and morphine. In fentanyl-tolerant mice, fentanyl showed a diminished analgesic effect, whereas N1 and N2 as well as morphine retained their analgesic effects. CONCLUSIONS: While N1 and N2 have higher transdermal absorbability than fentanyl, they also have analgesic effects comparable to those of morphine, suggesting that they may be attractive compounds for the development of novel opioid patches for transitioning from fentanyl patches.


Asunto(s)
Fentanilo , Morfina , Analgésicos Opioides , Animales , Proteínas de Unión al GTP/metabolismo , Humanos , Ratones , Receptores Opioides/metabolismo , Receptores Opioides mu/agonistas , beta-Arrestinas/metabolismo
3.
Masui ; 65(8): 850-852, 2016 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-30351602

RESUMEN

We experienced anesthetic management of open reduction for a femoral neck fracture in a patient com- plicated with fat embolism syndrome. An 83-year-old woman with a femoral neck fracture was admitted to our hospital after suffering an injury. She developed hypoxemia on admission. Chest X-ray showed a decrease in permeability of the right lung and chest CT scan showed ground glass opacities of the right lung. A blood test showed anemia, thrombocytopenia, and elevation of C-reactive protein. She was diagnosed with fat embolism syndrome using the classification of Tsuruta. Oxygen was administered. C-reactive protein decreased gradually after hospitalization. Echocardiog- raphy showed normal left ventricular function without pulmonary hypertension. She was scheduled for open reduction for the femoral neck fractu-e with artificial grit insertion under general anesthesia 6 days after hospitalization. Her operation was performed with- out exacerbation of the fat embolism syndrome. She was extubated in the operating room, and was dis- charged from the recovery room without any conpli- cations. After surgery, chest X-ray showed further improvement and she was transferred to a rehabilita- tion hospital on the 27th hospital day.


Asunto(s)
Embolia Grasa/complicaciones , Fracturas del Cuello Femoral/cirugía , Anciano de 80 o más Años , Anestesia General , Femenino , Fracturas del Cuello Femoral/complicaciones , Humanos , Tomografía Computarizada por Rayos X
4.
Biomolecules ; 12(3)2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35327617

RESUMEN

The issue of tolerance to continuous or repeated administration of opioids should be addressed. The ability of ketamine to improve opioid tolerance has been reported in clinical studies, and its mechanism of tolerance may involve improved desensitization of µ-opioid receptors (MORs). We measured changes in MOR activity and intracellular signaling induced by repeated fentanyl and morphine administration and investigated the effects of ketamine on these changes with human embryonic kidney 293 cells expressing MOR using the CellKey™, cADDis cyclic adenosine monophosphate, and PathHunter® ß-arrestin recruitment assays. Repeated administration of fentanyl or morphine suppressed the second MOR responses. Administration of ketamine before a second application of opioids within clinical concentrations improved acute desensitization and enhanced ß-arrestin recruitment elicited by fentanyl but not by morphine. The effects of ketamine on fentanyl were suppressed by co-treatment with an inhibitor of G-protein-coupled receptor kinase (GRK). Ketamine may potentially reduce fentanyl tolerance but not that of morphine through modulation of GRK-mediated pathways, possibly changing the conformational changes of ß-arrestin to MOR.


Asunto(s)
Ketamina , Morfina , Analgésicos Opioides/farmacología , Tolerancia a Medicamentos , Fentanilo/farmacología , Humanos , Ketamina/farmacología , Morfina/farmacología , Receptores Opioides/metabolismo , beta-Arrestinas/metabolismo
5.
Curr Top Med Chem ; 20(31): 2822-2829, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33115393

RESUMEN

Opioid agonists elicit their analgesic action mainly via µ opioid receptors; however, their use is limited because of adverse events including constipation and respiratory depression. It has been shown that analgesic action is transduced by the G protein-mediated pathway whereas adverse events are by the ß-arrestin-mediated pathway through µ opioid receptor signaling. The first new-generation opioid TRV130, which preferentially activates G protein- but not ß-arrestin-mediated signal, was constructed and developed to reduce adverse events. TRV130 and other G protein-biased compounds tend to elicit desirable analgesic action with less adverse effects. In clinical trials, the intravenous TRV130 (oliceridine) was evaluated in Phase I, II and III clinical studies. Here we review the discovery and synthesis of TRV130, its main action as a novel analgesic having less adverse events, its up-to-date status in clinical trials, and additional concerns about TRV130 as demonstrated in the literature.


Asunto(s)
Analgésicos Opioides/farmacología , Proteínas de Unión al GTP/antagonistas & inhibidores , Compuestos de Espiro/farmacología , Tiofenos/farmacología , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/química , Proteínas de Unión al GTP/metabolismo , Humanos , Transducción de Señal/efectos de los fármacos , Compuestos de Espiro/efectos adversos , Compuestos de Espiro/química , Tiofenos/efectos adversos , Tiofenos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA