• Title/Summary/Keyword: opioid receptor

Search Result 151, Processing Time 0.029 seconds

Can oliceridine (TRV130), an ideal novel µ receptor G protein pathway selective (µ-GPS) modulator, provide analgesia without opioid-related adverse reactions?

  • Ok, Hwoe Gyeong;Kim, Su Young;Lee, Su Jung;Kim, Tae Kyun;Huh, Billy K;Kim, Kyung Hoon
    • The Korean Journal of Pain
    • /
    • v.31 no.2
    • /
    • pp.73-79
    • /
    • 2018
  • All drugs have both favorable therapeutic and untoward adverse effects. Conventional opioid analgesics possess both analgesia and adverse reactions, such as nausea, vomiting, and respiratory depression. The opioid ligand binds to ${\mu}$ opioid receptor and non-selectively activates two intracellular signaling pathways: the G protein pathway induce analgesia, while the ${\beta}$-arrestin pathway is responsible for the opioid-related adverse reactions. An ideal opioid should activate the G protein pathway while deactivating the ${\beta}$-arrestin pathway. Oliceridine (TRV130) has a novel characteristic mechanism on the action of the ${\mu}$ receptor G protein pathway selective (${\mu}$-GPS) modulation. Even though adverse reactions (ADRs) are significantly attenuated, while the analgesic effect is augmented, the some residual ADRs persist. Consequently, a G protein biased ${\mu}$ opioid ligand, oliceridine, improves the therapeutic index owing to increased analgesia with decreased adverse events. This review article provides a brief history, mechanism of action, pharmacokinetics, pharmacodynamics, and ADRs of oliceridine.

Roles of Opioid Receptor Subtype in the Spinal Antinociception of Selective Cyclooxygenase 2 Inhibitor

  • Choi, Cheol-Hun;Kim, Woong-Mo;Lee, Hyung-Gon;Jeong, Cheol-Won;Kim, Chang-Mo;Lee, Seong-Heon;Yoon, Myung-Ha
    • The Korean Journal of Pain
    • /
    • v.23 no.4
    • /
    • pp.236-241
    • /
    • 2010
  • Background: Selective inhibitors of cycloosygenase (COX)-2 are commonly used analgesics in various pain conditions. Although their actions are largely thought to be mediated by the blockade of prostaglandin (PG) biosynthesis, evidences suggesting endogenous opioid peptide link in spinal antinociception of COX inhibitor have been reported. We investigated the roles of opioid receptor subtypes in the spinal antionociception of selective COX-2 inhibitor. Methods: To examine the antionociception of a selective COX-2 inhibitor, DUP-697 was delivered through an intrathecal catheter, 10 minutes before the formalin test in male Sprague-Dawley rats. Then, the effect of intrathecal pretreatment with CTOP, naltrindole and GNTI, which are ${\mu}$, $\delta$, and k opioid receptor antagonist, respectively, on the analgesia induced by DUP-697 was assessed. Results: Intrathecal DUP-697 reduced the flinching response evoked by formalin injection during phase 1 and 2 Naltrindole and GNTI attenuated the antinociceptive effect of intrathecal DUP-697 during both phases of the formalin test, CTOP reversed the antinociception of DUP-697 during phase 2, but not during phase 1, Conclusions: Intrathecal DUP-697, a selective COX-2 inhibitor, effectively relieved inflammatory pain in rats. The $\delta$ and $\kappa$ opioid receptors are involved in the activity of COX-2 inhibitor on the facilitated state as well as acute pain at the spinal level, whereas the ${\mu}$ opioid receptor is related only to facilitated pain.

Electrophysiological Responses of ${\delta}-Opioid$ Receptor Expressed on HEK293 Cells

  • Kim, Jin-Hyuk;Koh, Young-Ik;Chin, He-Min;Lee, Yong-Sung;Cho, Yeul-Hee;Kim, Kee-Soon
    • The Korean Journal of Physiology
    • /
    • v.29 no.2
    • /
    • pp.301-307
    • /
    • 1995
  • To explore electrophysiological properties of the ${\delta}-Opioid$ receptors artificially expressed in the mammalian cell, effect of an opioid agonist DPDPE $(1\;{\mu}M)$ on the voltage-sensitive outward currents was examined in the HEK293 (human embryonic kidney) cells transfected with ${\delta}-Opioid$ receptor cDNA cloned from NG-108-15 $(neuroblastoma\;{\times}\;glioma\;hybrid)$ cDNA library. Also studied were effects of 8-bromo-cyclic AMP and naloxone on DPDPE-induced changes in the voltage sensitive outward current. The voltage sensitive outward currents were recorded using perforated patch technique at room temperature. In the non-transformed HEK293 cells, DPDPE did not alter voltage sensitive outward current, indicating that no native ${\delta}-Opioid$ receptor had been developed. However, $(1\;{\mu}M)$ DPDPE remarkably increased the voltage sensitive outward current in the transformed HEK293 cells. The increment in voltage sensitive outward current peaked in $7{\sim}10\;minutes$ after DPDPE application, and the maximum DPDPE-activated outward current $(313.1{\pm}12.3\;pA)$ was recorded when the membrane potential was depolarized to +70mv. Following pretreatment of the transformed HEK293 cells with 1 mM 8-bromo-cyclic AMP, DPDPE failed to increase the voltage sensitive outward currents. On the other hand, naloxone completely abolished DPDPE-activated voltage sensitive outward current in the transformed HEK293 cells. The results of present study suggest that in the transformed HEK293 cells an activation of the ${\delta}-Opioid$ receptors by an opioid agonist DPDPE increases the voltage-sensitive potassium current as a result of decrement in cyclic AMP level.

  • PDF

Opioid Receptor Selectivity and General Pharmacology of DK1001, New Alkaloid Analgesic (알칼로이드 진통제 DK 1001의 opioid 수용체 선택성 및 일반약리)

  • Kim, Jin-Sook;Kim, Dae-Kyung;Kwon, Tae-Hyub;Yong, Chul-Soon;Ha, Jeoung-Hee;Huh, Keon;Kim, Jung-Ae
    • Biomolecules & Therapeutics
    • /
    • v.7 no.3
    • /
    • pp.278-284
    • /
    • 1999
  • DK1001 is a thebain derivative, which is newly synthesized as an alkaloid analgesic. This study was designed to study effects of DK1001 on the ligands binding to the opioid receptor subtypes, and general pharmacology of DK1001. DK1001 inhibited the binding of [$^3H$]DAMGO, a selective mu-subtype agonist, to the opioid receptor of rat forebrain in a concentration-dependent manner. $EC_{50}$ of DK1001 was significantly lower than that of morphine. DK1001 inhibited the binding of 〔$^3$H〕DPDPE, a selective delta-subtype agonist concentration-dependently. DK1001(0.5 mg/kg) had no effects on behavior, body temperature, blood pressure. respiratory rate, and intestinal charcoal propulsion of mice. In addition, DK1001 did not affect on the contractilities of isolated muscle strips of aorta, ileum, and trachea of rats. These results suggest that DK1001 might be a potent analgesic without serious side effects.

  • PDF

Medial prefrontal cortex nitric oxide modulates neuropathic pain behavior through mu opioid receptors in rats

  • Raisian, Dorsa;Erfanparast, Amir;Tamaddonfard, Esmaeal;Soltanalinejad-Taghiabad, Farhad
    • The Korean Journal of Pain
    • /
    • v.35 no.4
    • /
    • pp.413-422
    • /
    • 2022
  • Background: The neocortex, including the medial prefrontal cortex (mPFC), contains many neurons expressing nitric oxide synthase (NOS). In addition, increasing evidence shows that the nitric oxide (NO) and opioid systems interact in the brain. However, there have been no studies on the interaction of the opioid and NO systems in the mPFC. The objective of this study was to investigate the effects of administrating L-arginine (L-Arg, a precursor of NO) and N(gamma)-nitro-L-arginine methyl ester (L-NAME, an inhibitor of NOS) into the mPFC for neuropathic pain in rats. Also, we used selective opioid receptor antagonists to clarify the possible participation of the opioid mechanism. Methods: Complete transection of the peroneal and tibial branches of the sciatic nerve was applied to induce neuropathic pain, and seven days later, the mPFC was cannulated bilaterally. The paw withdrawal threshold fifty percent (50% PWT) was recorded on the 14th day. Results: Microinjection of L-Arg (2.87, 11.5 and 45.92 nmol per 0.25 µL) increased 50% PWT. L-NAME (17.15 nmol per 0.25 µL) and naloxonazine (an antagonist of mu opioid receptors, 1.54 nmol per 0.25 µL) inhibited anti-allodynia induced by L-Arg (45.92 nmol per 0.25 µL). Naltrindole (a delta opioid receptor antagonist, 2.45 nmol per 0.25 µL) and nor-binaltorphimine (a kappa opioid receptor antagonist, 1.36 nmol per 0.25 µL) were unable to prevent L-Arg (45.92 nmol per 0.25 µL)-induced antiallodynia. Conclusions: Our results indicate that the NO system in the mPFC regulates neuropathic pain. Mu opioid receptors of this area might participate in pain relief caused by L-Arg.

Role of nociceptin/orphanin FQ and nociceptin opioid peptide receptor in depression and antidepressant effects of nociceptin opioid peptide receptor antagonists

  • Park, Jong Yung;Chae, Suji;Kim, Chang Seop;Kim, Yoon Jae;Yi, Hyun Joo;Han, Eunjoo;Joo, Youngshin;Hong, Surim;Yun, Jae Won;Kim, Hyojung;Shin, Kyung Ho
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.23 no.6
    • /
    • pp.427-448
    • /
    • 2019
  • Nociceptin/orphanin FQ (N/OFQ) and its receptor, nociceptin opioid peptide (NOP) receptor, are localized in brain areas implicated in depression including the amygdala, bed nucleus of the stria terminalis, habenula, and monoaminergic nuclei in the brain stem. N/OFQ inhibits neuronal excitability of monoaminergic neurons and monoamine release from their terminals by activation of G protein-coupled inwardly rectifying $K^+$ channels and inhibition of voltage sensitive calcium channels, respectively. Therefore, NOP receptor antagonists have been proposed as a potential antidepressant. Indeed, mounting evidence shows that NOP receptor antagonists have antidepressant-like effects in various preclinical animal models of depression, and recent clinical studies again confirmed the idea that blockade of NOP receptor signaling could provide a novel strategy for the treatment of depression. In this review, we describe the pharmacological effects of N/OFQ in relation to depression and explore the possible mechanism of NOP receptor antagonists as potential antidepressants.

Hair Growth Promotion by δ-Opioid Receptor Activation

  • Zheng, Mei;Choi, Nahyun;Balboni, Gianfranco;Xia, Ying;Sung, Jong-Hyuk
    • Biomolecules & Therapeutics
    • /
    • v.29 no.6
    • /
    • pp.643-649
    • /
    • 2021
  • Literature has revealed that the delta opioid receptor (DOR) exhibited diverse pharmacological effects on neuron and skin. In the present study, we have investigated whether the activation of DOR has hair-growth promotion effects. Compared with other opioid receptor, DOR was highly expressed in epidermal component of hair follicle in human and rodents. The expression of DOR was high in the anagen phase, but it was low in the catagen and telogen phases during mouse hair cycle. Topical application of UFP-512, a specific DOR agonist, significantly accelerated the induction of the anagen in C3H mice. Topical application of UFP-512 also increased the hair length in hair organ cultures and promoted the proliferation and the migration of outer root sheath (ORS) cells. Similarly, pharmacological inhibition of DOR by naltrindole significantly inhibited the anagen transition process and decreased hair length in hair organ cultures. Thus, we further examined whether Wnt/β-catenin pathway was related to the effects of DOR on hair growth. We found that Wnt/β-catenin pathway was activated by UFP-512 and siRNA for β-catenin attenuated the UFP-512 induced proliferation and migration of ORS cells. Collectively, result established that DOR was involved in hair cycle regulation, and that DOR agonists such as UFP-512 should be developed for novel hair-loss treatment.

Effects of Neuroleptics on the Opioid Receptor Binding in the Mouse Striatum (마우스 선조체에서 Opioid 수용체 결합에 대한 Neuroleptics의 영향)

  • Kim, Soo-Kyung;Lee, Seong-Ryong;Park, Chang-Gyo
    • The Korean Journal of Pharmacology
    • /
    • v.30 no.3
    • /
    • pp.291-297
    • /
    • 1994
  • Our purpose was to gain insight into a possible modulatory role for ${\mu},\;{\delta},\;and\;{\kappa}$ opioid receptors by neuroleptics (chlorpromazine, thioridazine, haloperidol, sulpiride, and pimozide) in chronic morphine 5 mg/kg and 20 mg/kg treated mouse striatum. We attempted quantitative receptor assays using highly specific radioligands, $[^3H]\;DAGO\;([D-Ala^2,\;N-Mephe^4,\;Glycol^5]\;enkephalin)$, $[^3H]DPDPE\;([D-Pen^2,\;D-Pen^5]\;enkephalin)$ and $[^3H]\;DPN(diprenorphine)$ to measure the binding affinity in the experimental groups. The decrease of $[^3H]DAGO$ binding was potentiated by sulpiride and pimozide in the chronic morphine treatment (5 mg/kg and 20 mg/kg). The decrease of $[^3H]DPDPE$ binding was inhibited by chlorpromazine, thioridazine, haloperidol, sulpiride, and pimozide in chronic morphine treatment (5 mg/kg and 20 mg/kg). The decrease of $[^3H]\;DPN$ binding was significantly inhibited by chlorpromazine, thioridazine, sulpiride, and pimozide in chronic morphine 20 mg/kg treatment. $[^3H]\;DPN$ binding on the neuroleptics was antagonized by naloxone pretreatment in chronic morphine 20 mg/kg treatment. These findings suggest that neuroleptics influence opposing tonically active on the ${\delta},\;and\;{\kappa}$ opioid receptor compared with ${\mu}$ opioid receptor in the chronic morphine treated mouse striatum.

  • PDF

LIGAND BINDING CHARACTERISTICS OF $K_2$- OPIOID RECEPTOR AND ITS ROLE IN REGULATION OF 〔$^3$H〕HISTAMINE RELEASE IN FRONTAL CORTEX OF THE RAT

  • Kim, Kee-Won-;Park, Kyu--Cho
    • Proceedings of the Korean Society of Applied Pharmacology
    • /
    • 1994.04a
    • /
    • pp.305-305
    • /
    • 1994
  • It has been shown that there are several subtypes of $\kappa$ opioid receptor, We have evaluated the properties of non-${\mu}$, non-$\delta$ binding of 〔$^3$H〕DIP, a nonselective opioid antagonist, in rat cortex membranes. Binding to ${\mu}$ and $\delta$ sites was inhibited by the use of an excess of competing selective agonists (DAMGO, DPDPE) for these sites. (-)Ethylketocyclazocine(EKC) inhibited 〔$^3$H〕DIP binding with Ki. of 70 nM. However, arylacetamides (U69593 and U50488H) gave little inhibition. Also, we have examined the opioid modulation of K$\^$+/(30 mM)-induced histamine release in rat frontal cortex slices labeled with 1-〔$^3$H〕histidine. The 〔$^3$H〕histamine release from cortex slices was inhibited by EKC, a $\kappa$$_1$-and $\kappa$$_2$-agonist, in a concentration-dependent manner(10 to 10,000 nM). The IC$\sub$50/ of EKC was 107 ${\pm}$ 6 nM. However, the $\delta$ receptor selective agonists, DPDPE and deltorphine II, ${\mu}$ receptor agonists, DAMGO and TAPS, $\kappa$$_1$-agonists, U69593 and U50488H, and $\varepsilon$-agonist, ${\beta}$-endorphin, did not inhibit histamine release even in micromoiar dose, indicating that ${\mu}$, $\delta$ or $\kappa$$_1$ receptors are not involved. The concentration-response curve of EKC was shifted to right in the presence of naloxone (300 nM), a ${\mu}$ preferential antagonist, norbinaltorphimine(300 nM), a $\kappa$$_1$ preferential antagonist and bremazocine(1 nM), a $\kappa$$_1$-agonist and $\kappa$$_2$-antagonist. These results suggest that $\kappa$$_2$ opioid receptor regulates histamine release in the frontal cortex of the rat.

  • PDF

Role of $\mu$-Opioid Receptors on Neurobehaviors (뇌신경행동에 미치는 $\mu$-opioid 수용체의 역할)

  • Jang, Choon-Gon
    • Proceedings of the Korean Society of Applied Pharmacology
    • /
    • 2003.04a
    • /
    • pp.21-37
    • /
    • 2003
  • 1. Stimulation of dopaminergic system by morphine was abolished in ${\mu}$-opioid receptor knockout mice. 2. Dopaminergic stimulation by opioid agonists, morphine, DPDPE, and U50488, acts independently. 3. Loss of ${\mu}$-opioid receptors is more sensitive to the response of NMDA-induced convulsion and increase in the expression of mRNA for NMDA receptors.

  • PDF