Dezocine exhibits antihypersensitivity activities in neuropathy through spinal μ-opioid receptor activation and norepinephrine reuptake inhibition

Yong-Xiang Wang, Xiao-Fang Mao, Teng-Fei Li, Nian Gong, Ma-Zhong Zhang, Yong-Xiang Wang, Xiao-Fang Mao, Teng-Fei Li, Nian Gong, Ma-Zhong Zhang

Abstract

Dezocine is the number one opioid painkiller prescribed and sold in China, occupying 44% of the nation's opioid analgesics market today and far ahead of the gold-standard morphine. We discovered the mechanisms underlying dezocine antihypersensitivity activity and assessed their implications to antihypersensitivity tolerance. Dezocine, given subcutaneously in spinal nerve-ligated neuropathic rats, time- and dose-dependently produced mechanical antiallodynia and thermal antihyperalgesia, significantly increased ipsilateral spinal norepinephrine and serotonin levels, and induced less antiallodynic tolerance than morphine. Its mechanical antiallodynia was partially (40% or 60%) and completely (100%) attenuated by spinal μ-opioid receptor (MOR) antagonism or norepinephrine depletion/α2-adrenoceptor antagonism and combined antagonism of MORs and α2-adenoceptors, respectively. In contrast, antagonism of spinal κ-opioid receptors (KORs) and δ-opioid receptors (DORs) or depletion of spinal serotonin did not significantly alter dezocine antiallodynia. In addition, dezocine-delayed antiallodynic tolerance was accelerated by spinal norepinephrine depletion/α2-adenoceptor antagonism. Thus dezocine produces antihypersensitivity activity through spinal MOR activation and norepinephrine reuptake inhibition (NRI), but apparently not through spinal KOR and DOR activation, serotonin reuptake inhibition or other mechanisms. Our findings reclassify dezocine as the first analgesic of the recently proposed MOR-NRI, and reveal its potential as an alternative to as well as concurrent use with morphine in treating pain.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. The chemical structures of pentazocine…
Figure 1. The chemical structures of pentazocine and dezocine.
Figure 2
Figure 2
Time-course (A,B) and cumulative dose-response curves (C–F) of subcutaneous injection of dezocine and morphine antihypersensitivity activity in neuropathy. Neuropathic rats, induced by tight ligation of L5/L6 spinal nerves, received subcutaneous injection of dezocine or morphine. Data are summarized as means ± SEM (n = 6 and 10 in each group in the time-course and dose-response study, respectively). *P < 0.05 vs. the normal saline control, by repeats-measured two-way ANOVA followed by post-hoc student-Newman-Keuls tests.
Figure 3
Figure 3
Effects of the specific opioid receptor subtype antagonists, given intrathecally, on dezocine (A–C) and morphine antinociception (D–F) in neuropathic rats, induced by tight ligation of L5/L6 spinal nerves. Data are presented as means ± SEM (n = 6–8 in each group). *P < 0.05 vs. the respective normal saline + dezocine and normal saline + morphine group, by repeats-measured two-way ANOVA followed by post-hoc student-Newman-Keuls tests.
Figure 4
Figure 4
Effects of dezocine and morphine, given subcutaneously, on spinal levels of norepinephrine (A), serotonin (B) and dopamine (C) in neuropathic rats, induced by tight ligation of L5/L6 spinal nerves. Effects of the specific norepinephrine depletor 6-hydroxydopamine (6-OHDA), serotonin depletor p-cholrophenylalanine (PCPA) (D, E) and 5-hydroxytryptamine (5-HT) receptor antagonist metergoline (F,G), given intrathecally, on dezocine and morphine mechanical antiallodynia in neuropathy. Data are presented as means ± SEM (n = 6 in each group). *P < 0.05 vs. the normal saline control or respective normal saline + dezocine and normal saline + morphine group, by one-way or repeats-measured two-way ANOVA followed by post-hoc student-Newman-Keuls tests.
Figure 5
Figure 5
Effects of the selective noradrenergic α-adrenoceptor antagonist phentolamine (A,B), β-adrenoceptor antagonist propranolol (C,D), α1-adrenoceptor antagonist prazosin, α2-adrenoceptor receptor antagonist yohimbe, and the mixture of the μ-opioid receptor antagonist CTAP and yohimbe (E), given intrathecally, on dezocine and morphine antinociception in neuropathic rats, induced by tight ligation of L5/L6 spinal nerves. Data are presented as means ± SEM (n = 6 in each group). *P < 0.05 vs. the normal saline control or respective normal saline + dezocine and normal saline + morphine group, by repeats-measured two-way ANOVA followed by post-hoc student-Newman-Keuls tests.
Figure 6
Figure 6
Mechanical antiallodynic tolerance development of dezocine, morphine and their combination, given subcutaneously, in neuropathic rats, induced by tight ligation of L5/L6 spinal nerves (A). Effects of the specific norepinephrine depletor 6-hydroxydopamine (6-OHDA), serotonin depletor p-cholrophenylalanine (PCPA) (B,C), and the selective noradrenergic α1-adrenoceptor antagonist prazosin and α2-adrenoceptor receptor antagonist yohimbe (D), given intrathecally, on dezocine and morphine antinociceptive tolerance in neuropathy. Data are presented as means ± SEM (n = 6 in each group). *P < 0.05 vs. the normal saline control or respective normal saline + 6-OHDA and normal saline + PCPA group, by repeats-measured two-way ANOVA followed by post-hoc student-Newman-Keuls tests.

References

    1. Pandit U. A., Kothary S. P. & Pandit S. K. Intravenous dezocine for postoperative pain: a double-blind, placebo-controlled comparison with morphine. Journal of clinical pharmacology 26, 275–280 (1986).
    1. Wang C. et al.. A multicenter randomized double-blind prospective study of the postoperative patient controlled intravenous analgesia effects of dezocine in elderly patients. International journal of clinical and experimental medicine 7, 530–539 (2014).
    1. Wu F. X., Pan R. R., Yu W. F. & Liu R. The Anti-Nociception Effect of Dezocine in a Rat Neuropathic Pain Model. Translational perioperative and pain medicine 1, 5–8 (2014).
    1. Zhou X., Zhang C., Wang M., Yu L. & Yan M. Dezocine for Preventing Postoperative Pain: A Meta-Analysis of Randomized Controlled Trials. PloS one 10, e0136091 (2015).
    1. Malis J. L., Rosenthale M. E. & Gluckman M. I. Animal pharmacology of Wy-16,225, a new analgesic agent. Journal of pharmacology and experimental therapeutics 194, 488–498 (1975).
    1. Svensson B. A., Alari L. & Post C. Repeated intrathecal injections of dezocine produce antinociception without evidence for neurotoxicity in the rat: a study of morphometric evaluation of spinal cord histology. Anesthesia and analgesia 75, 392–399 (1992).
    1. Luo H., Yang H., Hu Y. & Zhang D. Preventive effects of dezocine on postoperative hyperalgesia in patients after remifentanil-based anesthesia. Chinese Journal of Anesthesiology 31, 1213–1216 (2011).
    1. Liu R., Huang X. P., Yeliseev A., Xi J. & Roth B. L. Novel molecular targets of dezocine and their clinical implications. Anesthesiology 120, 714–723 (2014).
    1. Jacobs A. M. & Youngblood F. Opioid receptor affinity for agonist-antagonist analgesics. Journal of the American Podiatric Medical Association 82, 520–524 (1992).
    1. Gal T. J. & DiFazio C. A. Ventilatory and analgesic effects of dezocine in humans. Anesthesiology 61, 716–722 (1984).
    1. Liu X. S. et al.. Dezocine prevents sufentanil-induced cough during general anesthesia induction: A randomized controlled trial. Pharmacological reports : PR 67, 52–55 (2015).
    1. Romagnoli A. & Keats A. S. Ceiling respiratory depression by dezocine. Clinical pharmacology and therapeutics 35, 367–373 (1984).
    1. Fragen R. J. & Caldwell N. Comparison of dezocine (WY 16, 225) and meperidine as postoperative analgesics. Anesthesia and analgesia 57, 563–566 (1978).
    1. Chen J. C., Smith E. R., Cahill M., Cohen R. & Fishman J. B. The opioid receptor binding of dezocine, morphine, fentanyl, butorphanol and nalbuphine. Life sciences 52, 389–396 (1993).
    1. Gharagozlou P., Demirci H., David Clark J. & Lameh J. Activity of opioid ligands in cells expressing cloned mu opioid receptors. BMC Pharmacology 3, 1 (2003).
    1. Gharagozlou P., Hashemi E., DeLorey T. M., Clark J. D. & Lameh J. Pharmacological profiles of opioid ligands at kappa opioid receptors. BMC Pharmacology 6, 3 (2006).
    1. Jasinski D. R. & Preston K. L. Assessment of dezocine for morphine-like subjective effects and miosis. Clinical pharmacology and therapeutics 38, 544–548 (1985).
    1. Walker E. A., Tiano M. J., Benyas S. I., Dykstra L. A. & Picker M. J. Naltrexone and beta-funaltrexamine antagonism of the antinociceptive and response rate-decreasing effects of morphine, dezocine, and d-propoxyphene. Psychopharmacology 144, 45–53 (1999).
    1. Picker M. J. Discriminative stimulus effects of the mixed-opioid agonist/antagonist dezocine: cross-substitution by mu and delta opioid agonists. Journal of pharmacology and experimental therapeutics 283, 1009–1017 (1997).
    1. Qiao W. et al.. The relationship of analgesia effect of Dezocine and kappa opioid receptors. Journal of Nantong University (Medical Sciences) 32, 353–355 (2012).
    1. Chung J. M., Kim H. K. & Chung K. Segmental spinal nerve ligation model of neuropathic pain. Methods in molecular medicine 99, 35–45 (2004).
    1. Zhang H., Shi Y. G., Woods J. H., Watson S. J. & Ko M. C. Central kappa-opioid receptor-mediated antidepressant-like effects of nor-Binaltorphimine: behavioral and BDNF mRNA expression studies. European journal of pharmacology 570, 89–96 (2007).
    1. Gharagozlou P., Demirci H., Clark J. D. & Lameh J. Activation profiles of opioid ligands in HEK cells expressing delta opioid receptors. BMC neuroscience 3, 19 (2002).
    1. Kogel B., De Vry J., Tzschentke T. M. & Christoph T. The antinociceptive and antihyperalgesic effect of tapentadol is partially retained in OPRM1 (mu-opioid receptor) knockout mice. Neuroscience letters 491, 104–107 (2011).
    1. Kuraishi Y., Harada Y., Aratani S., Satoh M. & Takagi H. Separate involvement of the spinal noradrenergic and serotonergic systems in morphine analgesia: the differences in mechanical and thermal algesic tests. Brain research 273, 245–252 (1983).
    1. Jiang L. Y., Li S. R., Zhao F. Y., Spanswick D. & Lin M. T. Norepinephrine can act via alpha(2)-adrenoceptors to reduce the hyper-excitability of spinal dorsal horn neurons following chronic nerve injury. Journal of the Formosan Medical Association 109, 438–445 (2010).
    1. Li J. Y. et al.. Modulations of spinal serotonin activity affect the development of morphine tolerance. Anesthesia and analgesia 92, 1563–1568 (2001).
    1. Breisch S. T., Zemlan F. P. & Hoebel B. G. Hyperphagia and obesity following serotonin depletion by intraventricular p-chlorophenylalanine. Science 192, 382–385 (1976).
    1. Eide P. K. & Hole K. Interactions between serotonin and substance P in the spinal regulation of nociception. Brain research 550, 225–230 (1991).
    1. Horiuchi H., Ogata T., Morino T., Takeba J. & Yamamoto H. Serotonergic signaling inhibits hyperalgesia induced by spinal cord damage. Brain research 963, 312–320 (2003).
    1. Kimura M., Obata H. & Saito S. Peripheral nerve injury reduces analgesic effects of systemic morphine via spinal 5-hydroxytryptamine 3 receptors. Anesthesiology 121, 362–371 (2014).
    1. Berge O. G., Fasmer O. B. & Hole K. Serotonin receptor antagonists induce hyperalgesia without preventing morphine antinociception. Pharmacology, biochemistry, and behavior 19, 873–878 (1983).
    1. Hall F. S. et al.. A greater role for the norepinephrine transporter than the serotonin transporter in murine nociception. Neuroscience 175, 315–327 (2011).
    1. Nakajima K., Obata H., Iriuchijima N. & Saito S. An increase in spinal cord noradrenaline is a major contributor to the antihyperalgesic effect of antidepressants after peripheral nerve injury in the rat. Pain 153, 990–997 (2012).
    1. Suzuki T., Ueta K., Tamagaki S. & Mashimo T. Antiallodynic and antihyperalgesic effect of milnacipran in mice with spinal nerve ligation. Anesthesia and analgesia 106, 1309–1315 (2008).
    1. Wang Y. X., Bowersox S. S., Pettus M. & Gao D. Antinociceptive properties of fenfluramine, a serotonin reuptake inhibitor, in a rat model of neuropathy. Journal of pharmacology and experimental therapeutics 291, 1008–1016 (1999).
    1. Bohn L. M., Xu F., Gainetdinov R. R. & Caron M. G. Potentiated opioid analgesia in norepinephrine transporter knock-out mice. Journal of neuroscience 20, 9040–9045 (2000).
    1. Hughes S., Hickey L., Donaldson L. F., Lumb B. M. & Pickering A. E. Intrathecal reboxetine suppresses evoked and ongoing neuropathic pain behaviours by restoring spinal noradrenergic inhibitory tone. Pain 156, 328–334 (2015).
    1. O'Neill D. J. et al.. Discovery of novel selective norepinephrine inhibitors: 1-(2-morpholin-2-ylethyl)-3-aryl-1,3-dihydro-2,1,3-benzothiadiazole 2,2-dioxides (WYE-114152). Journal of medicinal chemistry 54, 6824–6831 (2011).
    1. Hartrick C. T. Noradrenergic reuptake inhibition in the treatment of pain. Expert opinion on investigational drugs 21, 1827–1834 (2012).
    1. Tzschentke T. M., Christoph T. & Kogel B. Y. The mu-opioid receptor agonist/noradrenaline reuptake inhibition (MOR-NRI) concept in analgesia: the case of tapentadol. CNS drugs 28, 319–329 (2014).
    1. Bee L. A., Bannister K., Rahman W. & Dickenson A. H. Mu-opioid and noradrenergic alpha(2)-adrenoceptor contributions to the effects of tapentadol on spinal electrophysiological measures of nociception in nerve-injured rats. Pain 152, 131–139 (2011).
    1. Tzschentke T. M. et al.. (-)-(1R,2R)-3-(3-dimethylamino-1-ethyl-2-methyl-propyl)-phenol hydrochloride (tapentadol HCl): a novel mu-opioid receptor agonist/norepinephrine reuptake inhibitor with broad-spectrum analgesic properties. Journal of pharmacology and experimental therapeutics 323, 265–276 (2007).
    1. Zarrindast M. R., Sajedian M., Rezayat M. & Ghazi-Khansari M. Effects of 5-HT receptor antagonists on morphine-induced tolerance in mice. European journal of pharmacology 273, 203–207 (1995).
    1. Nakagawa T. et al.. Yokukansan inhibits morphine tolerance and physical dependence in mice: the role of alpha(2)A-adrenoceptor. Neuroscience 227, 336–349 (2012).
    1. Ozdogan U. K., Lahdesmaki J., Hakala K. & Scheinin M. The involvement of alpha 2A-adrenoceptors in morphine analgesia, tolerance and withdrawal in mice. European journal of pharmacology 497, 161–171 (2004).
    1. Miller G. Pot and pain: Hints are emerging that cannabis could be an alternative to opioid painkillers. Science 354(6312), 566–568 (2016).
    1. Basbaum A. I., Bautista D. M., Scherrer G. & Julius D. Cellular and molecular mechanisms of pain. Cell 139, 267–284 (2009).
    1. Huang J. L., Chen X. L., Guo C. & Wang Y. X. Contributions of spinal D-amino acid oxidase to bone cancer pain. Amino acids 43, 1905–1918 (2012).
    1. Gong N. et al.. Activation of spinal glucagon-like peptide-1 receptors specifically suppresses pain hypersensitivity. Journal of neuroscience 34, 5322–5334 (2014).
    1. Li T. F., Fan H. & Wang Y. X. Epidural sustained release ropivacaine prolongs anti-allodynia and anti-hyperalgesia in developing and established neuropathic pain. PloS one 10, e0117321 (2015).
    1. Fan H. et al.. The non-peptide GLP-1 receptor agonist WB4-24 blocks inflammatory nociception by stimulating beta-endorphin release from spinal microglia. British journal of pharmacology 172, 64–79 (2015).
    1. Steinmiller C. L. & Young A. M. Pharmacological selectivity of CTAP in a warm water tail-withdrawal antinociception assay in rats. Psychopharmacology 195, 497–507 (2008).
    1. Liu R. J., Zhang R. X., Qiao J. T. & Dafny N. Interrelations of opioids with monoamines in descending inhibition of nociceptive transmission at the spinal level: an immunocytochemical study. Brain research 830, 183–190 (1999).
    1. Satoh M., Oku R. & Akaike A. Analgesia produced by microinjection of L-glutamate into the rostral ventromedial bulbar nuclei of the rat and its inhibition by intrathecal alpha-adrenergic blocking agents. Brain research 261, 361–364 (1983).
    1. Bowersox S. S., Gadbois T., Singh T., Pettus M., Wang Y. X. & Luther R. R. Selective N-type neuronal voltage-sensitive calcium channel blocker, SNX-111, produces spinal antinociception in rat models of acute, persistent and neuropathic pain. J. Pharmacol. Exp. Ther. 279, 1243–1249 (1996).
    1. Wang Y. X. & Pang C. C. Functional integrity of the central and sympathetic nervous systems is a prerequisite for pressor and tachycardic effects of diphenyleneiodonium, a novel inhibitor of nitric oxide synthase. Journal of pharmacology and experimental therapeutics 265, 263–272 (1993).

Source: PubMed

3
Subscribe