Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Biosci Biotechnol Biochem ; 87(12): 1543-1550, 2023 Nov 21.
Article En | MEDLINE | ID: mdl-37715302

One of the chitinases (ChiG) derived from the chitinolytic bacterium Chitiniphilus shinanonensis SAY3T exhibited chitobiase activity cleaving dimers of N-acetyl-D-glucosamine (GlcNAc) into monomers, which is not detected in typical endo-type chitinases. Analysis of the reaction products for GlcNAc hexamers revealed that all the five internal glycosidic bonds were cleaved at the initial stage. The overall reaction catalyzed by chitobiases toward GlcNAc dimers was similar to that catalyzed by N-acetyl-D-glucosaminidases (NAGs). SAY3 possesses two NAGs (ChiI and ChiT) that are thought to be important in chitin catabolism. Unexpectedly, a triple gene-disrupted mutant (ΔchiIΔchiTΔchiG) was still able to grow on synthetic medium containing GlcNAc dimers or powdered chitin, similar to the wild-type SAY3, although it exhibited only 3% of total cellular NAG activity compared to the wild-type. This indicates the presence of unidentified enzyme(s) capable of supporting normal bacterial growth on the chitin medium by NAG activity compensation.


Betaproteobacteria , Chitinases , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Chitinases/metabolism , Betaproteobacteria/metabolism , Chitin/metabolism
2.
Int J Mol Sci ; 24(9)2023 May 06.
Article En | MEDLINE | ID: mdl-37176075

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.


Fentanyl , Receptors, Opioid , Receptors, Opioid/metabolism , Fentanyl/pharmacology , Remifentanil/pharmacology , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , beta-Arrestins/metabolism , Morphine
3.
Molecules ; 27(20)2022 Oct 19.
Article En | MEDLINE | ID: mdl-36296658

Opioid receptors (ORs) are classified into three types (µ, δ, and κ), and opioid analgesics are mainly mediated by µOR activation; however, their use is sometimes restricted by unfavorable effects. The selective κOR agonist nalfurafine was initially developed as an analgesic, but its indication was changed because of the narrow safety margin. The activation of ORs mainly induces two intracellular signaling pathways: a G-protein-mediated pathway and a ß-arrestin-mediated pathway. Recently, the expectations for κOR analgesics that selectively activate these pathways have increased; however, the structural properties required for the selectivity of nalfurafine are still unknown. Therefore, we evaluated the partial structures of nalfurafine that are necessary for the selectivity of these two pathways. We assayed the properties of nalfurafine and six nalfurafine analogs (SYKs) using cells stably expressing κORs. The SYKs activated κORs in a concentration-dependent manner with higher EC50 values than nalfurafine. Upon bias factor assessment, only SYK-309 (possessing the 3S-hydroxy group) showed higher selectivity of G-protein-mediated signaling activities than nalfurafine, suggesting the direction of the 3S-hydroxy group may affect the ß-arrestin-mediated pathway. In conclusion, nalfurafine analogs having a 3S-hydroxy group, such as SYK-309, could be considered G-protein-biased κOR agonists.


Analgesics, Opioid , Receptors, Opioid, kappa , Analgesics , Analgesics, Opioid/pharmacology , beta-Arrestins/metabolism , GTP-Binding Proteins/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/metabolism
4.
Anesth Analg ; 134(5): 1082-1093, 2022 05 01.
Article En | MEDLINE | ID: mdl-35427270

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.


Fentanyl , Morphine , Analgesics, Opioid , Animals , GTP-Binding Proteins/metabolism , Humans , Mice , Receptors, Opioid/metabolism , Receptors, Opioid, mu/agonists , beta-Arrestins/metabolism
5.
Biomolecules ; 12(3)2022 03 10.
Article En | MEDLINE | ID: mdl-35327617

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.


Ketamine , Morphine , Analgesics, Opioid/pharmacology , Drug Tolerance , Fentanyl/pharmacology , Humans , Ketamine/pharmacology , Morphine/pharmacology , Receptors, Opioid/metabolism , beta-Arrestins/metabolism
6.
Molecules ; 26(19)2021 Oct 08.
Article En | MEDLINE | ID: mdl-34641621

Activated opioid receptors transmit internal signals through two major pathways: the G-protein-mediated pathway, which exerts analgesia, and the ß-arrestin-mediated pathway, which leads to unfavorable side effects. Hence, G-protein-biased opioid agonists are preferable as opioid analgesics. Rubiscolins, the spinach-derived naturally occurring opioid peptides, are selective δ opioid receptor agonists, and their p.o. administration exhibits antinociceptive effects. Although the potency and effect of rubiscolins as G-protein-biased molecules are partially confirmed, their in vitro profiles remain unclear. We, therefore, evaluated the properties of rubiscolins, in detail, through several analyses, including the CellKeyTM assay, cADDis® cAMP assay, and PathHunter® ß-arrestin recruitment assay, using cells stably expressing µ, δ, κ, or µ/δ heteromer opioid receptors. In the CellKeyTM assay, rubiscolins showed selective agonistic effects for δ opioid receptor and little agonistic or antagonistic effects for µ and κ opioid receptors. Furthermore, rubiscolins were found to be G-protein-biased δ opioid receptor agonists based on the results obtained in cADDis® cAMP and PathHunter® ß-arrestin recruitment assays. Finally, we found, for the first time, that they are also partially agonistic for the µ/δ dimers. In conclusion, rubiscolins could serve as attractive seeds, as δ opioid receptor-specific agonists, for the development of novel opioid analgesics with reduced side effects.


Opioid Peptides/pharmacology , Receptors, Opioid, delta/agonists , Signal Transduction/drug effects , Spinacia oleracea/chemistry , GTP-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Molecular Structure , Opioid Peptides/chemistry , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Receptors, Opioid, mu/metabolism , Ribulose-Bisphosphate Carboxylase/chemistry , Ribulose-Bisphosphate Carboxylase/pharmacology , beta-Arrestins/metabolism
7.
Cells ; 10(10)2021 10 04.
Article En | MEDLINE | ID: mdl-34685631

Oxytocin (OT) influences various physiological functions such as uterine contractions, maternal/social behavior, and analgesia. Opioid signaling pathways are involved in one of the analgesic mechanisms of OT. We previously showed that OT acts as a positive allosteric modulator (PAM) and enhances µ-opioid receptor (MOR) activity. In this study, which focused on other opioid receptor (OR) subtypes, we investigated whether OT influences opioid signaling pathways as a PAM for δ-OR (DOR) or κ-OR (KOR) using human embryonic kidney-293 cells expressing human DOR or KOR, respectively. The CellKeyTM results showed that OT enhanced impedance induced by endogenous/exogenous KOR agonists on KOR-expressing cells. OT did not affect DOR activity induced by endogenous/exogenous DOR agonists. OT potentiated the KOR agonist-induced Gi/o protein-mediated decrease in intracellular cAMP, but did not affect the increase in KOR internalization caused by the KOR agonists dynorphin A and (-)-U-50488 hydrochloride (U50488). OT did not bind to KOR orthosteric binding sites and did not affect the binding affinities of dynorphin A and U50488 for KOR. These results suggest that OT is a PAM of KOR and MOR and enhances G protein signaling without affecting ß-arrestin signaling. Thus, OT has potential as a specific signaling-biased PAM of KOR.


GTP-Binding Proteins/metabolism , Oxytocin/pharmacology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Signal Transduction , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Allosteric Regulation/drug effects , Animals , Binding Sites , CHO Cells , Cricetulus , Cyclic AMP/metabolism , Diprenorphine/pharmacology , Dynorphins/pharmacology , Electric Impedance , Endocytosis/drug effects , HEK293 Cells , Humans , Inhibitory Concentration 50 , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Signal Transduction/drug effects
8.
Biomed Pharmacother ; 141: 111800, 2021 Sep.
Article En | MEDLINE | ID: mdl-34175819

BACKGROUND: The misuse of opioids has led to an epidemic in recent times. The endothelin A receptor (ETAR) has recently attracted attention as a novel therapeutic target to enhance opioid analgesia. We hypothesized that endothelin A receptors may affect pain mechanisms by heterodimerization with µ opioid receptors. We examined the mechanisms of ETAR-mediated pain and the potential therapeutic effects of an ETAR antagonist, Compound-E, as an agent for analgesia. METHODS: Real-time in vitro effect of Compound-E on morphine response was assessed in HEK293 cells expressing both endothelin A and µ opioid receptors through CellKey™ and cADDis cAMP assays. Endothelin A/µ opioid receptor dimerization was assessed by immunoprecipitation and live cell imaging. The in vivo effect of Compound-E was evaluated using a morphine analgesia mouse model that observed escape response behavior, body temperature, and locomotor activity. RESULTS: In CellKey™ and cAMP assays, pretreatment of cells with endothelin-1 attenuated morphine-induced responses. These responses were improved by Compound-E, but not by BQ-123 nor by bosentan, an ETAR and endothelin B receptor antagonist. Dimerization of ETARs and µ opioid receptors was confirmed by Western blot and total internal reflection fluorescence microscopy in live cells. In vivo, Compound-E potentiated and prolonged the analgesic effects of morphine, enhanced hypothermia, and increased locomotor activity compared to morphine alone. CONCLUSION: The results suggest that attenuation by endothelin-1 of morphine analgesia may be caused by dimerization of Endothelin A/µ opioid receptors. The novel ETAR antagonist Compound-E could be an effective adjunct to reduce opioid use.


Analgesics, Opioid/administration & dosage , Endothelin A Receptor Antagonists/administration & dosage , Morphine/administration & dosage , Protein Multimerization/physiology , Receptor, Endothelin A/metabolism , Receptors, Opioid, mu/metabolism , Animals , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Pain Measurement/drug effects , Pain Measurement/methods , Peptides, Cyclic/administration & dosage , Protein Multimerization/drug effects
9.
Curr Top Med Chem ; 20(31): 2822-2829, 2020.
Article En | MEDLINE | ID: mdl-33115393

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.


Analgesics, Opioid/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Spiro Compounds/pharmacology , Thiophenes/pharmacology , Analgesics, Opioid/adverse effects , Analgesics, Opioid/chemistry , GTP-Binding Proteins/metabolism , Humans , Signal Transduction/drug effects , Spiro Compounds/adverse effects , Spiro Compounds/chemistry , Thiophenes/adverse effects , Thiophenes/chemistry
...