Article Data

  • Views 376
  • Dowloads 70

Original Research

Open Access

Orofacial Antinociceptive Effect of Nifedipine in Rodents Is Mediated by TRPM3, TRPA1, and NMDA Processes

  • Breytiner Amaro de Oliveira1
  • Sacha Aubrey Alves Rodrigues Santos1
  • Erik Willyame Menezes Pereira2
  • Andressa Barros Nogueira1
  • Antônio Eufrásio Vieira Neto1,3
  • José de Maria de Albuquerque de Melo Júnior1
  • Marina de Barros Mamede Vidal Damasceno1
  • Lucindo José Quintans-Júnior2
  • Barry John Sessle4,5,6
  • Francisco Ernani Alves Magalhães1,7
  • Adriana Rolim Campos1,*,

1Experimental Biology Center, University of Fortaleza, Fortaleza, Brazil

2Department of Physiology, Federal University of Sergipe, Aracaju, Brazil

3Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceara, Brazil

4Faculty of Dentistry, University of Toronto, Toronto, Canada

5Faculty of Medicine, Department of Physiology, University of Toronto, Toronto, Canada

6Centre for the Study of Pain, University of Toronto, Toronto, Canada

7State University of Ceará, Tauá, Brazil

DOI: 10.11607/ofph.2491 Vol.34,Issue 2,June 2020 pp.174-186

Submitted: 02 April 2019 Accepted: 22 October 2019

Published: 30 June 2020

*Corresponding Author(s): Adriana Rolim Campos E-mail: adrirolim@unifor.br

Abstract

Aims: To test for the possible antinociceptive effect of nifedipine in rodent models of acute and chronic neuropathic orofacial pain and the possible involvement of TRP- and NMDA-related processes in this effect. Methods: Acute nociceptive behavior was induced by administering formalin, cinnamaldehyde, glutamate, capsaicin, or acidified saline to the upper lip or hypertonic saline to the cornea of Swiss mice. Acute nociceptive behavior was also induced by formalin injected into the TMJ or mustard oil injected into the masseter muscle of Wistar rats. The chronic pain model involved infraorbital nerve transection (IONX) in Wistar rats to induce mechanical hypersensitivity, which was assessed with von Frey hair stimulation of the upper lip. The effects of pretreatment with nifedipine or vehicle (control) were tested on the nociceptive behaviors. Docking experiments were also performed. Statistical analysis included one-way ANOVA followed by Tukey post hoc test and two-way ANOVA followed by Bonferroni post hoc test (statistical significance P < .05). Results: Nifedipine produced significant antinociceptive effects in all of the acute nociceptive behaviors except that induced by capsaicin. The antinociceptive effects were attenuated by NMDA, TRPA1, or TRPM3 receptor antagonists. The IONX animals developed facial mechanical hypersensitivity, which was significantly reduced by nifedipine. The docking experiments suggested that nifedipine may interact with TRPM3 and NMDA receptors. Conclusion: The present study has provided novel findings in a variety of acute and chronic orofacial pain models showing that nifedipine, a selective inhibitor of L-type Ca2+ channels, can suppress orofacial nociceptive behavior through NMDA, TRPA1, and TRPM3 receptor systems.

Keywords

acute; neuropathic; orofacial pain; rodents

Cite and Share

Breytiner Amaro de Oliveira,Sacha Aubrey Alves Rodrigues Santos,Erik Willyame Menezes Pereira,Andressa Barros Nogueira,Antônio Eufrásio Vieira Neto,José de Maria de Albuquerque de Melo Júnior,Marina de Barros Mamede Vidal Damasceno,Lucindo José Quintans-Júnior,Barry John Sessle,Francisco Ernani Alves Magalhães,Adriana Rolim Campos. Orofacial Antinociceptive Effect of Nifedipine in Rodents Is Mediated by TRPM3, TRPA1, and NMDA Processes. Journal of Oral & Facial Pain and Headache. 2020. 34(2);174-186.

References

1. Sessle BJ. Are cannabinoids effective for orofacial pain states? J Oral Facial Pain Headache 2015;29:5–6.

2. Bannister K, Kucharczyk M, Dickenson AH. Hopes for the future of pain control. Pain Ther 2017;6:117–128.

3. Goto T, Oh SB, Takeda M, et al. Recent advances in basic research on the trigeminal ganglion. J Physiol Sci 2016;66: 381–386.

4. Li KW, Yu YP, Zhou C, et al. Calcium channel α2δ1 proteins mediate trigeminal neuropathic pain states associated with aberrant excitatory synaptogenesis. J Biol Chem 2014;289: 7025–7037.

5. Martín MI, del Val VL, Colado MI, Goicoechea C, Alfaro MJ. Behavioral and analgesic effects induced by administration of nifedipine and nimodipine. Pharmacol Biochem Behav 1996;55:93–98.

6. Zbuzek VK, Cohen B, Wu W. Antinociceptive effect of nifedipine and verapamil tested on rats chronically exposed to nicotine and after its withdrawal. Life Sci 1997;60:1651–1658.

7. Hunt TE, Wu WH, Zbuzek VK. The effects of serotonin biosynthesis inhibition on nicotine and nifedipine-induced analgesia in rats. Anesth Analg 1998;87:1109–1112.

8. Chang E, Chen X, Kim M, Gong N, Bhatia S, Luo ZD. Differential effects of voltage-gated calcium channel blockers on calcium channel alpha-2-delta-1 subunit protein-mediated nociception. Eur J Pain 2015;19:639–648.

9. Esmaeili-Mahani S, Vahedi S, Motamedi F, Pourshanazari A, Khaksari M, Ahmadiani A. Involvement of hypothalamic pituitary adrenal axis on the analgesic cross-tolerance between morphine and nifedipine. Pharmacol Biochem Behav 2007; 86:806–812.

10. Wong CH, Wu WH, Yarmush J, Zbuzek VK. An antinociceptive effect of the intraperitoneal injection of nifedipine in rats, measured by tail-flick test. Life Sci 1993;53:PL249–PL253.

11. Bustamante D, Miranda HF, Pelissier T, Paeile C. Analgesic action of clonixin, nifedipine and morphine using the formalin test. Gen Pharmacol 1989;20:319–322.

12. Kawashiri T, Egashira N, Kurobe K, et al. L type Ca²+ chan-nel blockers prevent oxaliplatin-induced cold hyperalgesia and TRPM8 overexpression in rats. Mol Pain 2012;8:7.

13. Kania BF, Kowalczyk M, Brytan M, Tomaszewska D, Przekop F. The inhibition of experimentally induced visceral hyperalge-sia by nifedipine—A a voltage-gated Ca(2+) channels blocker (VGCCs) in sheep. Res Vet Sci 2009;86:285–292.

14. Magalhães FEA, Batista FLA, Serpa OF, et al. Orofacial antinociceptive effect of Mimosa tenuiflora (Willd.) Poiret. Biomed Pharmacother 2018;97:1575–1585.

15. Farazifard R, Safapour F, Sheibani V, Javan M. Eye-wiping test: A sensitive animal model for acute trigeminal pain studies. Brain Res Brain Res Protoc 2005;16:44–49.

16. Roveroni RC, Parada CA, Cecília M, Veiga FA, Tambeli CH. Development of a behavioral model of TMJ pain in rats: The TMJformalin test. Pain 2001;94:185–191.

17. Ro JY, Capra N, Masri R. Development of a behavioral assessment of craniofacial muscle pain in lightly anesthetized rats. Pain 2003;104:179–185.

18. Kumar N, Cherkas PS, Varathan V, et al. Systemic pregabalin attenuates facial hypersensitivity and noxious stimulus-evoked release of glutamate in medullary dorsal horn in a rodent model of trigeminal neuropathic pain. Neurochem Int 2013; 62:831–835.

19. Cairns BE, Sessle BJ, Hu JW. Evidence that excitatory amino acid receptors within the temporomandibular joint region are involved in the reflex activation of the jaw muscles. J Neurosci 1998;18:8056–8064.

20. Araújo JRC, Júnior JMAM, Damasceno MBMV, et al. Neuropharmacological characterization of frutalin in mice: Evidence ofan antidepressant-like effect mediated by the NMDA receptor/NO/cGMP pathway. Int J Biol Macromol 2018;112: 548–554.

21. Klose C, Straub I, Riehle M, et al. Fenamates as TRP channel blockers: Mefenamic acid selectively blocks TRPM3. Br J Pharmacol 2011;162:1757–1769.

22. Quintans Júnior LJ, Santana MT, Melo MS, et al. Antinociceptive and anti-inflammatory effects of Costus spicatus in experimental animals. Pharm Biol 2010;48:1097–1102.

23. Malafoglia V, Traversetti L, Del Grosso F, et al. Transient Receptor Potential Melastatin-3 (TRPM3) mediates nociceptive-like responses in hydra vulgaris. PLoS One 2016;11:e0151386.

24. Zamudio-Bulcock PA, Everett J, Harteneck C, Valenzuela CF. Activation of steroid-sensitive TRPM3 channels potentiates glutamatergic transmission at cerebellar Purkinje neurons from developing rats. J Neurochem 2011;119:474–485.

25. Nascimento SS, Camargo EA, DeSantana JM, et al. Linalool and linalool complexed in β-cyclodextrin produce anti-hyperalgesic activity and increase Fos protein expression in animal model for fibromyalgia. Naunyn Schmiedebergs Arch Pharmacol 2014;387:935–942.

26. Meng XY, Zhang HX, Mezei M, Cui M. Molecular docking: A powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des 2011;7:146–157.

27. Vilar S, Sobarzo-Sanchez E, Santana L, Uriarte E. Molecular docking and drug discovery in β-adrenergic receptors. Curr Med Chem 2017;24:4340–4359.

28. Okkerse P, van Amerongen G, de Kam ML, et al. The use of a battery of pain models to detect analgesic properties of compounds: A two-part, four-way, randomised, placebo-controlled, crossover study. Clin Ther 2015;37:e14.

29. Berge OG. Predictive validity of behavioural animal models for chronic pain. Br J Pharmacol 2011;164:1195–1206.

30. Le Bars D, Gozariu D, Cadden SW. Animal models of nociception. Pharmacol Rev 2001;53:597–652.

31. Raboisson P, Dallel R. The orofacial formalin test. Neurosci Biobehav Rev 2004;28:219–226.

32. McNamara CR, Mandel-Brehm J, Bautista DM, et al. TRPA1 mediates formalin-induced pain. Proc Natl Acad Sci U S A 2007;104:13525–13530.

33. Fajardo O, Meseguer, V, Belmonte, C, Viana F. TRPA1 channels: Novel targets of 1,4-dihydropyridines. Channels (Austin) 2008;2:429–438.

34. Shields SD, Cavanaugh DJ, Lee H, Anderson DJ, Basbaum AI. Pain behavior in the formalin test persists after ablation of the great majority of C-fiber nociceptors. Pain 2010;151:422–429.

35. Fischer MJ, Soller KJ, Sauer SK, Kalucka J, Veglia G, Reeh PW. Formalin evokes calcium transients from the endoplasmatic reticulum. PLoS One 2015;10:e0123762.

36. Pelissier T, Pajot J, Dallel R. The orofacial capsaicin test in rats: Effects of different capsaicin concentrations and morphine. Pain 2002;96:81–87.

37. Castro-Junior CJ, Milano J, Souza AH, et al. Phα1β toxin prevents capsaicin-induced nociceptive behavior and mechanical hypersensitivity without acting on TRPV1 channels. Neuropharmacology 2013;71:237–246.

38. Hargreaves KM. Orofacial pain. Pain 2011;152(3 suppl): s25–s32.

39. Carstens E, Kuenzler N, Handwerker HO. Activation of neurons in rat trigeminal subnucleus caudalis by different irritant chemicals applied to oral or ocular mucosa. J Neurophysiol 1998;80:465–492.

40. Ciura S, Bourque CW. Transient receptor potential vanilloid 1 is required for intrinsic osmoreception in organum vasculosum lamina terminalis neurons and for normal thirst responses to systemic hyperosmolality. J Neurosci 2006;26:9069–9075.

41. Carnevale V, Rohacs T. TRPV1: A target for rational drug design. Pharmaceuticals (Basel) 2016;9. pii: E52.

42. Gu Q, Lee LY. Acid-sensing ion channels and pain. Pharmaceuticals (Basel) 2010;3:1411–1425.

43. Gan X, Wu J, Ren C, Qiu CY, Li YK, Hu WP. Potentiation of acid-sensing ion channel activity by peripheral group I metabotropic glutamate receptor signaling. Pharmacol Res 2016;107:19–26.

44. Tikhonova TB, Nagaeva EI, Barygin OI, Potapieva NN, Bolshakov KV, Tikhonov DB. Monoamine NMDA receptor channel blockers inhibit and potentiate native and recombinant proton-gated ion channels. Neuropharmacol 2015;89:1–10.

45. Honda K, Noma N, Shinoda M, et al. Involvement of peripheral ionotropic glutamate receptors in orofacial thermal hyperalgesia in rats. Mol Pain 2011;7:75.

46. Emara LH, Badr RM, Elbary AA. Improving the dissolution and bioavailability of nifedipine using solid dispersions and solubilizers. Drug Dev Ind Pharm 2002;28:795–807.

47. Janicki PK, Siembab D, Paulo EA, Krzas´cik P. Single-dose kinetics of nifedipine in rat plasma and brain. Pharmacology 1988;36:183–187.

48. Fossat P, Sibon I, Le Masson G, Landry M, Nagy F. L-type calcium channels and NMDA receptors: A determinant duo for shortterm nociceptive plasticity. Eur J Neurosci 2007;25:127–135.

49. Thiel G, Rubil S, Lesch A, Guethlein LA, Rössler OG. Transient receptor potential TRPM3 channels: Pharmacology, signaling,and biological functions. Pharmacol Res 2017;124:92–99.

50. Wu SW, Fenwick AJ, Peters JH. Channeling satiation: A primer on the role of TRP channels in the control of glutamate release from vagal afferent neurons. Physiol Behav 2014;136:179–184.

51. Wagner TF, Loch S, Lambert S, et al. Transient receptor potential M3 channels are ionotropic steroid receptors in pancreatic beta cells. Nat Cell Biol 2008;10:1421–1430.

52. Iwata K, Imamura Y, Honda K, Shinoda M. Physiological mechanisms of neuropathic pain: The orofacial region. Int Rev Neurobiol 2011;97:227–250.

53. Liu Y, Harding M, Dore J, Chen X. Cav1.2, but not Cav1.3, L-type calcium channel subtype mediates nicotine-induced conditioned place preference in mice. Prog Neuropsychopharmacol Biol Psychiatry 2017;75:176–182.

54. Catterall WA. Structure and regulation of voltage-gated Ca2+ channels. Annu Rev Cell Dev Biol 2000;16:521–555.

55. Sisignano M, Parnham MJ, Geisslinger G. Drug repurposing for the development of novel analgesics. Trends Pharmacol Sci 2016;37:172–183.

Abstracted / indexed in

Science Citation Index (SCI)

Science Citation Index Expanded (SCIE)

BIOSIS Previews

Scopus

Cumulative Index to Nursing and Allied Health Literature (CINAHL)

Submission Turnaround Time

Conferences

Top