• Title/Summary/Keyword: mu opioid receptors

Search Result 49, Processing Time 0.021 seconds

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

Effects of Whole Body Irradiation on Morphine, DAMGO, DPDPE, U50,488H and $\beta$-endorphin-Induced Antinociception

  • Park, Tae-Won;Kim, Jin-Kyu;Jeong, Jae-Soo;Kim, Tae-Wan;Cho, Young-Kyung;Kim, Kyung-Nyun;Chung, Ki-Myung
    • International Journal of Oral Biology
    • /
    • v.37 no.1
    • /
    • pp.1-7
    • /
    • 2012
  • Opioid receptors have been pharmacologically classified as ${\mu}$, ${\delta}$, ${\kappa}$ and ${\varepsilon}$. We have recently reported that the antinociceptive effect of morphine (a ${\mu}$-opioid receptor agonist), but not that of ${\beta}$-endorphin (a novel ${\mu}/{\varepsilon}$-opioid receptor agonist), is attenuated by whole body irradiation (WBI). It is unclear at present whether WBI has differential effects on the antinociceptive effects of ${\mu}-$, ${\delta}-$, ${\kappa}-$ and ${\varepsilon}$-opioid receptor agonists. In our current experiments, male ICR mice were exposed to WBI (5Gy) from a $^{60}Co$ gamma-source and the antinociceptive effects of opioid receptor agonists were assessed two hours later using the hot water ($52^{\circ}C$) tail-immersion test. Morphine and $D-Ala^2$, $N-Me-Phe^4$, Gly-olenkephalin (DAMGO), [$D-Pen^2-D-Pen^5$] enkephalin (DPDPE), trans-3,4-Dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl]-benzeneacetamide (U50,488H), and ${\beta}$-endorphin were tested as agonists for ${\mu}$, ${\delta}$, ${\kappa}$, and ${\varepsilon}$-opioid receptors, respectively. WBI significantly attenuated the antinociceptive effects of morphine and DAMGO, but increased those of ${\beta}$-endorphin. The antinociceptive effects of DPDPE and U50,488H were not affected by WBI. In addition, to more preciously understand the differential effects of WBI on ${\mu}-$ and ${\varepsilon}$-opioid receptor agonists, we assessed pretreatment effects of ${\beta}$-funaltrexamine (${\beta}$-FNA, a ${\mu}$-opioid receptor antagonist) or ${\beta}$-$endorphin_{1-27}$ (${\beta}$-$EP_{1-27}$, an ${\varepsilon}$-opioid receptor antagonist), and found that pretreatment with ${\beta}$-FNA significantly attenuated the antinociceptive effects of morphine and ${\beta}$-endorphin by WBI. ${\beta}$-$EP_{1-27}$ significantly reversed the attenuation of morphine by WBI and significantly attenuated the increased effects of ${\beta}$-endorphin by WBI. The results demonstrate differential sensitivities of opioid receptors to WBI, especially for ${\mu}-$ and ${\varepsilon}$-opioid receptors.

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.

Study on Peripheral Mechanism and Opioid Receptors Implicated in Electroacupunture-induced Inbibition of Chronic Pain (만성통증을 억제하는 전침효과의 말초성 기전과 아편양물질수용기에 관한 연구)

  • 신홍기;이서은;박동석
    • The Journal of Korean Medicine
    • /
    • v.24 no.3
    • /
    • pp.108-117
    • /
    • 2003
  • Objective : The central opioid mechanism of acupuncture analgesia has been fairly well documented in acute behavioral experiments, but little electrophysiological study has been performed on the peripheral mechanism and subtypes of opioid receptors responsible for acupuncture-induced antinociception in chronic animal models. In the present electrophysiological experiment, we studied the peripheral mechanism and opioid receptor subtypes which Were implicated in electroacupuncture-induced antinociception in the rat with chronic inflammatory and neurogenic pain. Methods : In the rat with complete Freund's adjuvant-induced inflammation and spinal nerve injury, dorsal horn cell responses to afferent C fiber stimulation were recorded before and after electroacupuncture (EA) stimulation applied to the contralateral Zusanli point for 30 minutes. Also studied Were the effects of specific opioid receptor antagonists and naloxone methiodide, which can not cross the blood-brain barrier, on EA-induced inhibitory action. Results : EA-induced inhibitory action was significantly attenuated by naloxone methiodide, suggesting that EA-induced inhibition was mediated through peripheral mechanism. Pretreatment, but not posttreatment of naltrexone and spinal application significantly blocked EA-induced inhibitory actions. In inflammatory and neurogenic pain models, ${\mu}-$ and ${\delta}-opioid$ receptor antagonists (${\beta}-funaltrexamine$ & naltrindole) significantly reduced EA-induced inhibitory action, but ${\kappa}-opioid$ receptor antagonist had weak inhibitory effect on EA-induced antinociception. Conclusion : These results suggest that 2Hz EA-stimulation induced antinoeiceptive action is mediated through peripheral as well as central mechanism, and mainly through ${\mu}-$ and ${\delta}-opioid$ receptors.

  • PDF

Effects of Morphine and Panax ginseng on the Opioid Receptor-G protein Interactions

  • Kim, Young-Ran;Kim, Ae-young;Kim, Kyeong-Man
    • Biomolecules & Therapeutics
    • /
    • v.7 no.1
    • /
    • pp.1-6
    • /
    • 1999
  • Effects of Panax ginseng on the morphine toxicity were studied in relation to its effects on the opioid receptor-G protein interactions. Morphine treatments (3 days) reduced the body weight increment rate and the weight of the thymus and spleen. These changes were usually recovered by the concomitant administration of ginseng total saponin (GTS) but occasionally further deteriorated. This discrepancy was studied in relation to the opioid receptor coupling to G protein, that is, the effects of morphine and GTS on the opioid receptors were studied using the antagonist-agonist competitive binding studies. When GTS recovered the morphine toxicity, morphine shifted the striatal $\delta$ receptors to slightly higher affinity state, and this was partly recovered by the GTS treatment. However, morphine did not have any effect on the affinity state of $\delta$ receptor from NG108-15 cells, suggesting that additional factors were needed for the modulation of the affinity states of $\delta$ receptor. Effects of morphine and GTS on $\mu$ receptor were complicate and variable, and we could not reach a clear conclusion. The morphine toxicity might accompany complicate biological involvements, and the modulation of the affinity states of the opioid receptors might explain a part of the effects of GTS on the morphine toxicity.

  • PDF

Differential Actions of Intracerebroventricular Opioid Receptor Agonists on the Activity of Dorsal Horn Neurons in the Cat Spinal Cord (Opioid 수용체 효능제의 뇌실 내 주입이 고양이 척수후각세포의 활성에 미치는 영향)

  • 문태상;오우택
    • YAKHAK HOEJI
    • /
    • v.43 no.4
    • /
    • pp.411-418
    • /
    • 1999
  • Intracerebroventricular (ICV) infusion of morphine (MOR) produces strong analgesia in man and animals. The analgesic effect is thought to be mediated by the centrifugal inhibitory control. But neural mechanisms of the analgesic effect of ICV morphine are not well understood. In the present study, we found that ICV MOR had dual actions on the activity of dorsal horn heurons: it produced both inhibition and excitation of dorsal horn neurons. Since MOR exerts its action via three different types of opioid receptors, we further sought to investigate if there are differential effects of opioid receptor agonists on dorsal horn neurons when administered intracerebroventricularly. Effects of ICV MOR were tested in 28 dorsal horn neurons of the spinal cord in the cat. ICV MOR inhibited, excited and did not affect the heat responses of dorsal horn neurons. ICV DAMGO and DADLE, $\mu$- and $\delta$-opioid agonist, respectively, exhibited the excitation of dorsal horn neurons. In contract, U-50488, a k-opioid agonist, exhibited both the inhibition and excitation of dorsal horn neurons. These results suggest that opioid receptors have different actions on activity of dorsal horn neuron and that the inhibitory action of k-opioid agonist may subserve the analgesia often produced by ICV MOR.

  • PDF

Do Opioid Receptors Play a Role in Blood Pressure Regulation?

  • Rhee, H.M.;Holaday, J.W.;Long, J.B.;Gaumann, M.D.;Yaksh, T.L.;Tyce, G.M.;Dixon, W.R.;Chang, A.P.;Mastrianni, J.A.;Mosqueda-Garcia, R.;Kunos, G.
    • The Korean Journal of Pharmacology
    • /
    • v.24 no.2
    • /
    • pp.153-164
    • /
    • 1988
  • The potential role of endogenous opioid peptides (EOPS) in cardiovascular regulation has only recently been entertained. EOPS have been localized in brain, spinal cord, autonomic ganglia, particularly the adrenal gland, and many other peripheral tissues. There are at least five major types of opioid receptors; namely ${\mu},\;{\delta},\;k,\;{\sigma},\;and\;{\varepsilon}$ and Experimental evidence indicates that cardiovascular actions of the peptide are mediated primarily by ${\mu},\;{\delta}$ and k receptors, and that these receptor types may be allosterically coupled. In anesthetized rabbits met-enkephalin decreased blood pressure and heart rate, which closely paralleled a reduction in sympathetic discharge. Naloxone, but not naloxone methobromide, antagonized these effects, which suggests a central site of action of met-enkephalin. A number of autonomic agents, particularly adrenergic ${\alpha}$-and, ${\beta}-agonists$ and antagonists modify the cardiovascular actions of met-enkephalin. Experiments in reserpine-treated and adrenalectomized rats provide no evidence of sympathetic nervous system involvement in the pressor responses to intravenous injection of opioid peptides, but rather suggest a direct peripheral action. Finally, activation of a beta-endorphinergic pathway projecting from the arcuate nucleus to the nucleus tractos solitarii in rats can cause naloxone reversible hypotension and bradycardia. There is evidence to implicate this pathway in antihypertensive drug action and in the modulation of baroreflex activity.

  • PDF

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

Effects of Ginseng Total Saponin on [3H]DAGO Bindings of Opioid μ-Receptors

  • Oh, Ki-Wan;Lim, Hwa-Kyung;Park, Cheol-Beum;Shin, Im-Chul;Hong, Jin-Tae
    • Journal of Ginseng Research
    • /
    • v.26 no.4
    • /
    • pp.187-190
    • /
    • 2002
  • Crude synaptic membrane fractions from the frontal cortex, striatum, brain stem and whole brain of rat were prepared to assay the effects of ginseng total saponin (GTS) on [$^3$H]DAGO bindings of the opioid $\mu$-receptors. Scatchard plots analysis binding data demonstrated that GTS (0.1 mg/ml) decreased the affinity of specific [$^3$H]DAGO bindings without changes in B$\_$max/ in the frontal cortex and striatum. On the other hand, GTS did not affect the [$^3$H]DAGO bindings iii the brain stem and whole brain. These results suggest that the regulation of [$^3$H]DAGO bindings by GTS may play roles in the change of the pharmacological responses of $\mu$-opioids.

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.