Selective δ-opioid receptor antagonism by ICI 174,864 in the central nervous system

Peptides ◽  
1984 ◽  
Vol 5 (5) ◽  
pp. 1015-1016 ◽  
Author(s):  
A. Dray ◽  
L. Nunan
2019 ◽  
Vol 131 (3) ◽  
pp. 649-663 ◽  
Author(s):  
Daniel J. Bruce ◽  
Cristina D. Peterson ◽  
Kelley F. Kitto ◽  
Eyup Akgün ◽  
Sophia Lazzaroni ◽  
...  

Abstract Editor’s Perspective What We Already Know about This Topic What This Article Tells Us That Is New Background The long-term use of opioids for analgesia carries significant risk for tolerance, addiction, and diversion. These adverse effects are largely mediated by μ-opioid receptors in the central nervous system. Based on the authors’ previous observation that morphine and δ-opioid receptor agonists synergize in spinal cord in a protein kinase Cε–dependent manner, they predicted that this μ-opioid receptor–δ-opioid receptor synergy would take place in the central terminals of nociceptive afferent fibers and generalize to their peripheral terminals. Therefore, the authors hypothesized that loperamide, a highly efficacious μ-opioid receptor agonist that is excluded from the central nervous system, and oxymorphindole, a δ-opioid receptor agonist that was shown to synergize with morphine spinally, would synergistically reverse complete Freund’s adjuvant–induced hyperalgesia. Methods Using the Hargreaves assay for thermal nociception, the von Frey assay for mechanical nociception and the complete Freund’s adjuvant–induced model of inflammatory pain, we tested the antinociceptive and antihyperalgesic effect of loperamide, oxymorphindole, or the loperamide–oxymorphindole combination. Animals (Institute for Cancer Research [ICR] CD1 strain mice; n = 511) received drug by systemic injection, intraplantar injection to the injured paw, or a transdermal solution on the injured paw. Dose–response curves for each route of administration and each nociceptive test were generated, and analgesic synergy was assessed by isobolographic analysis. Results In naïve animals, the loperamide–oxymorphindole combination ED50 value was 10 times lower than the theoretical additive ED50 value whether given systemically or locally. In inflamed animals, the combination was 150 times more potent systemically, and 84 times more potent locally. All combinations showed statistically significant synergy when compared to the theoretical additive values, as verified by isobolographic analysis. The antihyperalgesia was ablated by a peripherally-restricted opioid antagonist. Conclusions From these data we conclude that the loperamide–oxymorphindole combination synergistically reverses complete Freund’s adjuvant–induced inflammatory hyperalgesia. The authors also conclude that this interaction is mediated by opioid receptors located in the peripheral nervous system.


2013 ◽  
Vol 2013 ◽  
pp. 1-17 ◽  
Author(s):  
Dasiel O. Borroto-Escuela ◽  
Wilber Romero-Fernandez ◽  
Alicia Rivera ◽  
Kathleen Van Craenenbroeck ◽  
Alexander O. Tarakanov ◽  
...  

The modulatory role of allosteric receptor-receptor interactions in the pain pathways of the Central Nervous System and the peripheral nociceptors has become of increasing interest. As integrators of nociceptive and antinociceptive wiring and volume transmission signals, with a major role for the opioid receptor heteromers, they likely have an important role in the pain circuits and may be involved in acupuncture. The delta opioid receptor (DOR) exerts an antagonistic allosteric influence on the mu opioid receptor (MOR) function in a MOR-DOR heteromer. This heteromer contributes to morphine-induced tolerance and dependence, since it becomes abundant and develops a reduced G-protein-coupling with reduced signaling mainly operating viaβ-arrestin2 upon chronic morphine treatment. A DOR antagonist causes a return of the Gi/o binding and coupling to the heteromer and the biological actions of morphine. The gender- and ovarian steroid-dependent recruitment of spinal cord MOR/kappa opioid receptor (KOR) heterodimers enhances antinociceptive functions and if impaired could contribute to chronic pain states in women. MOR1D heterodimerizes with gastrin-releasing peptide receptor (GRPR) in the spinal cord, mediating morphine induced itch. Other mechanism for the antinociceptive actions of acupuncture along meridians may be that it enhances the cross-desensitization of the TRPA1 (chemical nociceptor)-TRPV1 (capsaicin receptor) heteromeric channel complexes within the nociceptor terminals located along these meridians. Selective ionotropic cannabinoids may also produce cross-desensitization of the TRPA1-TRPV1 heteromeric nociceptor channels by being negative allosteric modulators of these channels leading to antinociception and antihyperalgesia.


Author(s):  
A. Porteros ◽  
M. Garc�a-Isidoro ◽  
A. Barrallo ◽  
R. Gonz�lez-Sarmiento ◽  
R.E. Rodr�guez

Life Sciences ◽  
1983 ◽  
Vol 33 ◽  
pp. 167-173 ◽  
Author(s):  
G.W. Pasternak ◽  
A.R. Gintzler ◽  
R.A. Houghten ◽  
G.S.F. Ling ◽  
R.R. Goodman ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document