Korean J Pain.  2016 Jul;29(3):164-171. 10.3344/kjp.2016.29.3.164.

The Role of Spinal Dopaminergic Transmission in the Analgesic Effect of Nefopam on Rat Inflammatory Pain

Affiliations
  • 1Department of Anesthesiology and Pain Medicine, Chonnam National University Medical School, Gwangju, Korea. kimwm@jnu.ac.kr
  • 2Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea.

Abstract

BACKGROUND
Nefopam has been known as an inhibitor of the reuptake of monoamines, and the noradrenergic and/or serotonergic system has been focused on as a mechanism of its analgesic action. Here we investigated the role of the spinal dopaminergic neurotransmission in the antinociceptive effect of nefopam administered intravenously or intrathecally.
METHODS
The effects of intravenously and intrathecally administered nefopam were examined using the rat formalin test. Then we performed a microdialysis study to confirm the change of extracellular dopamine concentration in the spinal dorsal horn by nefopam. To determine whether the changes of dopamine level are associated with the nefopam analgesia, its mechanism was investigated pharmacologically via pretreatment with sulpiride, a dopaminergic D2 receptor antagonist.
RESULTS
When nefopam was administered intravenously the flinching responses in phase I of the formalin test were decreased, but not those in phase II of the formalin test were decreased. Intrathecally injected nefopam reduced the flinching responses in both phases of the formalin test in a dose dependent manner. Microdialysis study revealed a significant increase of the level of dopamine in the spinal cord by intrathecally administered nefopam (about 3.8 fold the baseline value) but not by that administered intravenously. The analgesic effects of intrathecally injected nefopam were not affected by pretreatment with sulpiride, and neither were those of the intravenous nefopam.
CONCLUSIONS
Both the intravenously and intrathecally administered nefopam effectively relieved inflammatory pain in rats. Nefopam may act as an inhibitor of dopamine reuptake when delivered into the spinal cord. However, the analgesic mechanism of nefopam may not involve the dopaminergic transmission at the spinal level.

Keyword

Analgesia; Dopamine; Microdialysis; Nefopam; Spinal cord; Sulpiride

MeSH Terms

Analgesia
Animals
Dopamine
Microdialysis
Nefopam*
Pain Measurement
Rats*
Spinal Cord
Spinal Cord Dorsal Horn
Sulpiride
Synaptic Transmission
Dopamine
Nefopam
Sulpiride

Figure

  • Fig. 1 Time-response (A) and dose-response data (B) of intravenously administered nefopam on flinching behavior during the formalin test. Each drug was administered 10 min before the formalin test. Data are presented as the number of flinching or the percentage of control. Intravenously administered nefopam reduced the flinching responses in phase I at a dose of 3 mg/kg. Each line or bar represents mean ± SEM of 5 rats. *P < 0.05 compared to control.

  • Fig. 2 Time-response (A) and dose-response data (B) of intrathecally administered nefopam on flinching behavior during the formalin test. Each drug was administered 10 min before the formalin test. Data are presented as the number of flinching or the percentage of control. Intrathecally administered nefopam reduced the flinching responses in both phases of formalin test in a dose dependent manner. Each line or bar represents mean ± SEM of 5 rats. *P < 0.05 †P < 0.001 compared to control.

  • Fig. 3 Microdialysis study measuring extracellular dopamine level at the spinal dorsal horn after intravenous (3 mg/kg) or intrathecal (30 µg) delivery of nefopam. Data are presented over time as a percent change from the baseline (n = 5 in each group). *P < 0.05 compared to baseline (BL) value.

  • Fig. 4 The effects of intravenous pretreatment with intravenous sulpiride (3 mg/kg) on the analgesic effect of intravenous (3 mg/kg) nefopam (A), or those with intrathecal sulpiride (100 µg) on intrathecal (30 µg) nefopam (B). The antinociception produced by nefopam was not attenuated by sulpiride. Each line or bar represents mean ± SEM of 5 rats.


Reference

1. Kim KH, Abdi S. Rediscovery of nefopam for the treatment of neuropathic pain. Korean J Pain. 2014; 27:103–111. PMID: 24748937.
Article
2. Esposito E, Romandini S, Merlo-Pich E, Mennini T, Samanin R. Evidence of the involvement of dopamine in the analgesic effect of nefopam. Eur J Pharmacol. 1986; 128:157–164. PMID: 3098570.
Article
3. Fuller RW, Snoddy HD. Evaluation of nefopam as a monoamine uptake inhibitor in vivo in mice. Neuropharmacology. 1993; 32:995–999. PMID: 7507578.
Article
4. Hunskaar S, Fasmer OB, Broch OJ, Hole K. Involvement of central serotonergic pathways in nefopam-induced antinociception. Eur J Pharmacol. 1987; 138:77–82. PMID: 2442003.
Article
5. Rosland JH, Hole K. The effect of nefopam and its enantiomers on the uptake of 5-hydroxytryptamine, noradrenaline and dopamine in crude rat brain synaptosomal preparations. J Pharm Pharmacol. 1990; 42:437–438. PMID: 1979627.
Article
6. Vonvoigtlander PF, Lewis RA, Neff GL, Triezenberg HJ. Involvement of biogenic amines with the mechanisms of novel analgesics. Prog Neuropsychopharmacol Biol Psychiatry. 1983; 7:651–656. PMID: 6141608.
Article
7. Gray AM, Nevinson MJ, Sewell RD. The involvement of opioidergic and noradrenergic mechanisms in nefopam antinociception. Eur J Pharmacol. 1999; 365:149–157. PMID: 9988097.
Article
8. Girard P, Coppé MC, Verniers D, Pansart Y, Gillardin JM. Role of catecholamines and serotonin receptor subtypes in nefopam-induced antinociception. Pharmacol Res. 2006; 54:195–202. PMID: 16750379.
Article
9. Jeong SH, Heo BH, Park SH, Kim WM, Lee HG, Yoon MH, et al. Spinal noradrenergic modulation and the role of the alpha-2 receptor in the antinociceptive effect of intrathecal nefopam in the formalin test. Korean J Pain. 2014; 27:23–29. PMID: 24478897.
Article
10. Yaksh TL, Rudy TA. Chronic catheterization of the spinal subarachnoid space. Physiol Behav. 1976; 17:1031–1036. PMID: 14677603.
Article
11. Cho SY, Park AR, Yoon MH, Lee HG, Kim WM, Choi JI. Antinociceptive effect of intrathecal nefopam and interaction with morphine in formalin-induced pain of rats. Korean J Pain. 2013; 26:14–20. PMID: 23342202.
Article
12. Coderre TJ, Melzack R. The contribution of excitatory amino acids to central sensitization and persistent nociception after formalin-induced tissue injury. J Neurosci. 1992; 12:3665–3670. PMID: 1326610.
Article
13. Lee HG, Choi JI, Yoon MH, Obata H, Saito S, Kim WM. The antiallodynic effect of intrathecal tianeptine is exerted by increased serotonin and norepinephrine in the spinal dorsal horn. Neurosci Lett. 2014; 583:103–107. PMID: 25233863.
Article
14. Smith DF, Glaser R, Gee A, Gjedde A. [11C]Nefopam as a potential PET tracer of serotonin reuptake sites. In : Myers R, Cunningham V, Bailey D, Jones T, editors. Quantification of brain function using PET. San Diego (CA): Academic Press;1996. p. 38–41.
15. Millan MJ. Descending control of pain. Prog Neurobiol. 2002; 66:355–474. PMID: 12034378.
Article
16. Potvin S, Grignon S, Marchand S. Human evidence of a supra-spinal modulating role of dopamine on pain perception. Synapse. 2009; 63:390–402. PMID: 19173266.
Article
17. Franklin KB. Analgesia and abuse potential: an accidental association or a common substrate? Pharmacol Biochem Behav. 1998; 59:993–1002. PMID: 9586860.
Article
18. Altier N, Stewart J. The role of dopamine in the nucleus accumbens in analgesia. Life Sci. 1999; 65:2269–2287. PMID: 10597883.
Article
19. Cobacho N, de la Calle JL, Paíno CL. Dopaminergic modulation of neuropathic pain: analgesia in rats by a D2-type receptor agonist. Brain Res Bull. 2014; 106:62–71. PMID: 24959942.
Article
20. Fairbanks CA. Spinal delivery of analgesics in experimental models of pain and analgesia. Adv Drug Deliv Rev. 2003; 55:1007–1041. PMID: 12935942.
Article
21. Xu JJ, Walla BC, Diaz MF, Fuller GN, Gutstein HB. Intermittent lumbar puncture in rats: a novel method for the experimental study of opioid tolerance. Anesth Analg. 2006; 103:714–720. PMID: 16931686.
Article
22. Ohkubo Y, Nomura K, Yamaguchi I. Involvement of dopamine in the mechanism of action of FR64822, a novel non-opioid antinociceptive compound. Eur J Pharmacol. 1991; 204:121–125. PMID: 1839620.
Article
23. Novelli A, Díaz-Trelles R, Groppetti A, Fernández-Sánchez MT. Nefopam inhibits calcium influx, cGMP formation, and NMDA receptor-dependent neurotoxicity following activation of voltage sensitive calcium channels. Amino Acids. 2005; 28:183–191. PMID: 15714253.
Article
24. Verleye M, André N, Heulard I, Gillardin JM. Nefopam blocks voltage-sensitive sodium channels and modulates glutamatergic transmission in rodents. Brain Res. 2004; 1013:249–255. PMID: 15193535.
Article
Full Text Links
  • KJP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr