Korean J Pain.  2023 Jul;36(3):335-346. 10.3344/kjp.23039.

Perampanel ameliorates nitroglycerin-induced migraine through inhibition of the cAMP/ PKA/CREB signaling pathway in the trigeminal ganglion in rats

Affiliations
  • 1Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
  • 2Department of Neurology, The Third Hospital of Jinan Shandong, Jinan, China
  • 3Department of Neurology, Shengli Oilfield Central Hospital, Shandong, China
  • 4Department of Neurology, Binzhou Medical University Hospital, Shandong, China

Abstract

Background
Perampanel, a highly selective glutamate AMPA receptor antagonist, is widely used to treat epilepsy. Since the existence of common pathophysiological features between epilepsy and migraine, the aim of this study was to investigate whether perampanel could exert an antimigraine effect.
Methods
Nitroglycerin (NTG) was used to induce a migraine model in rats, and the model animals were pretreatment with 50 μg/kg and 100 μg/kg perampanel. The expression of pituitary adenylate-cyclase-activating polypeptide (PACAP) was quantified by western blot and quantitative real-time PCR in the trigeminal ganglion, and rat-specific enzyme-linked immunosorbent assay in serum. Western blot was also conducted to explore the effects of perampanel treatment on the phospholipase C (PLC)/protein kinase C (PKC) and protein kinase A (PKA)/ cAMP-responsive-element-binding protein (CREB) signaling pathways. Moreover, the cAMP/PKA/CREB-dependent mechanism was evaluated via in vitro stimulation of hippocampal neurons. The cells were treated with perampanel, antagonists and agonists for 24 hours and cell lysates were prepared for western blot analysis.
Results
Perampanel treatment notably increased the mechanical withdrawal threshold and decreased head grooming and light-aversive behaviors in NTG-treated rats. It also decreased PACAP expression and affected cAMP/ PKA/CREB signaling pathway. However, PLC/PKC signaling pathway may not be involved in this treatment. In in vitro studies, perampanel notably decreased PACAP expression by inhibiting cAMP/PKA/CREB signaling pathway.
Conclusions
This study shows that perampanel inhibits the migraine-like pain response and that this beneficial effect might be attributable to regulation of the cAMP/PKA/CREB signaling pathway.

Keyword

Chronic Pain; Cyclic AMP-Dependent Protein Kinases; Glutamic Acid; Hippocampus; Hyperalgesia; Migraine Disorders; Nitroglycerin; Pituitary Adenylate Cyclase-Activating Polypeptide; Rats; Receptors, AMPA

Figure

  • Fig. 1 Experimental design of the study and the anatomical location of trigeminal ganglions. NTG: nitroglycerin, i.p.: intraperitoneal.

  • Fig. 2 Effects of perampanel treatment on photophobia. (A) Time spent in the light compartment, Kruskal–Wallis statistic = 24.30, ***P < 0.001; (B) Latency to re-enter the light compartment, Kruskal–Wallis statistic = 16.33, ***P = 0.001; (C) Transition, Kruskal–Wallis statistic = 20.55, ***P < 0.001. Control, n = 8; NTG, n = 8; PER 50 μg/kg + NTG, n = 8; PER 100 μg/kg + NTG, n = 8. Data are presented as means ± SEM. NTG: nitroglycerin, PER: perampanel. $$P < 0.01 vs. NTG group; **P < 0.01, ***P < 0.001 vs. control group; #P < 0.05 vs. NTG group in corresponding days.

  • Fig. 3 Effects of perampanel treatment on NTG-induced migraine-like pain and nociceptive behaviors. (A) Head grooming, subgroups, F (3, 28) = 25.95, P < 0.001; time, F (4, 112) = 0.295, P = 0.880; subgroups × time, F (12, 112) = 0.629, P = 0.814. Control, n = 8; NTG, n = 8; PER 50 μg/kg + NTG, n = 8; PER 100 μg/kg + NTG, n = 8. In periorbital withdrawal thresholds test, NTG were decreased periorbital withdrawal thresholds, while perampanel treatment, at both doses of 50 and 100 μg/kg significantly increased the thresholds. (B) Periorbital withdrawal thresholds, subgroups, F (3, 28) = 23.28, P < 0.001; time, F (4, 112) = 4.397, P = 0.002; subgroups × time, F (12, 112) = 0.9606, P = 0.491. Data are presented as means ± SEM. NTG: nitroglycerin, PER: perampanel. $P < 0.05, $$P < 0.01, $$$P < 0.001 vs. NTG group; *P < 0.05, **P < 0.01, ***P < 0.001 vs. control group; #P < 0.05, ##P < 0.01, ###P < 0.001 vs. NTG group in corresponding days.

  • Fig. 4 Effects of perampanel treatment on eating disorders. (A) Body weight, subgroups, F (3, 28) = 4.720, P = 0.009; time, F (4, 112) = 67.16, P < 0.001; subgroups × time, F (12, 112) = 0.652, P = 0.793; (B) Food intake, subgroups, F (3, 28) = 4.311, P = 0.013; time, F (4, 112) = 5.916, P < 0.001; subgroups × time, F (12, 112) = 1.637, P = 0.091. Control, n = 8; NTG, n = 8; PER 50 μg/kg + NTG, n = 8; PER 100 μg/kg + NTG, n = 8. Data are presented as means ± SEM. NTG: nitroglycerin, PER: perampanel.

  • Fig. 5 Effects of perampanel treatment on PACAP level in NTG-induced migraine. (A) Western blot showing the protein levels of PACAP; (B) Western bolt analysis of PACAP expression in TG. Kruskal–Wallis statistic = 12.15, **P = 0.007; (C) qRT-PCR analysis of PACAP gene expression in TG. Kruskal–Wallis statistic = 14.16, **P = 0.003, P < 0.001; (D) Elisa analysis of PACAP level in serum. Kruskal–Wallis statistic = 9.828, *P = 0.020. Control, n = 4; NTG, n = 6; PER 50 μg/kg + NTG, n = 6; PER 100 μg/kg + NTG, n = 6. Data are presented as mean ± SEM. PACAP: pituitary adenylate-cyclase-activating polypeptide, NTG: nitroglycerin, TG: trigeminal ganglion, PER: perampanel. Significant differences: *P < 0.05, ***P < 0.001.

  • Fig. 6 Effects of perampanel on PKA/CREB and PLC/PKC signaling pathways in NTG-induced migraine. (A) Western blot showing the protein levels of PKA, CREB and p-CREB; (B) Western blot analysis of the expression of PKA in TG. Kruskal–Wallis statistic = 17.20, ***P < 0.001; (C) Western blot analysis of the expression of P-CREB in TG. Kruskal–Wallis statistic = 11.52, **P = 0.009; (D) Western blot analysis of the expression of CREB in TG. Kruskal–Wallis statistic = 9.125, *P = 0.028; (E) Western blot showing the protein levels of PLC and PKC. (F) Western blot analysis of the expression of PLC in TG. Kruskal–Wallis statistic = 3.631, P = 0.304; (G) Western blot analysis of the expression of PKC in TG. Kruskal–Wallis statistic = 7.030, P = 0.071. Control, n = 4; NTG, n = 6; PER 50 μg/kg + NTG, n = 6; PER 100 μg/kg + NTG, n = 6. Data are presented as mean ± SEM. PKA: protein kinase A, CREB: cAMP-responsive-element-binding protein, PLC: phospholipase C, PKC: protein kinase C, NTG: nitroglycerin, TG: trigeminal ganglion, PER: perampanel. Significant differences: *P < 0.05, **P < 0.01.

  • Fig. 7 Effects of perampanel on cAMP/PKA/CREB signaling pathways in vitro. (A) Western blot showing the protein levels of PACAP in HT-22 cell incubated with forskolin 10 µM for 24 hours; (B) Western blot analysis of the expression of PACAP. Kruskal–Wallis statistic = 7.538, *P = 0.011. Control, n = 4; Forskolin 10 µM, n = 4; PER 100 μM, n = 4; (C) Western blot showing the protein levels of PACAP in HT-22 cell incubated with 8-bromo-cAMP sodium salt 0.1 mM for 24 hours; (D) Western blot analysis of the expression of PACAP. Kruskal–Wallis statistic = 16.33, P = 0.001. Control, n = 4; 8-bromo-cAMP sodium salt 0.1 mM, n = 4; PER 100 μM, n = 4; (E) Western blot showing the protein levels of PACAP in HT-22 cell incubated with SQ22536 100 μM for 24 hours; (F) Western blot analysis of the expression of PACAP. Kruskal–Wallis statistic = 9.269, ***P < 0.001. Control, n = 4; SQ22536 100 μM, n = 4; PER 100 μM, n = 4. (G) Western blot showing the protein levels of PACAP in HT-22 cell incubated with H-89 10 µM for 24 hours; (H) Western blot analysis of the expression of PACAP. Kruskal–Wallis statistic = 9.846, ***P < 0.001. Control, n = 4; H-89 10 µM, n = 4; PER 100 μM, n = 4. (I) Western blot showing the protein levels of PACAP in HT-22 cell incubated with KG501 25 µM for 24 hours; (J) Western blot analysis of the expression of PACAP. Kruskal–Wallis statistic = 8.769, **P = 0.001. Control, n = 4; KG501 25 µM, n = 4; PER 100 μM, n = 4. Data are presented as mean ± SEM. PKA: protein kinase A, CREB: cAMP-responsive-element-binding protein, PER: perampanel. Significant differences: *P < 0.05, **P < 0.01.


Reference

1. Global Burden of Disease Study 2013 Collaborators. 2015; Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 386:743–800. DOI: 10.1016/S0140-6736(15)60692-4. PMID: 26063472. PMCID: PMC4561509.
2. GBD 2015 Neurological Disorders Collaborator Group. 2017; Global, regional, and national burden of neurological disorders during 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet Neurol. 16:877–97. DOI: 10.1016/S1474-4422(17)30299-5. PMID: 28931491. PMCID: PMC5641502.
3. 2018; Headache Classification Committee of the International Headache Society (IHS) The International Classification of Headache Disorders, 3rd edition. Cephalalgia. 38:1–211. DOI: 10.1177/0333102417738202. PMID: 29368949.
4. Pradhan AA, Smith ML, McGuire B, Tarash I, Evans CJ, Charles A. 2014; Characterization of a novel model of chronic migraine. Pain. 155:269–74. DOI: 10.1016/j.pain.2013.10.004. PMID: 24121068. PMCID: PMC3920577.
Article
5. Tuka B, Szabó N, Tóth E, Kincses ZT, Párdutz Á, Szok D, et al. 2016; Release of PACAP-38 in episodic cluster headache patients - an exploratory study. J Headache Pain. 17:69. DOI: 10.1186/s10194-016-0660-7. PMID: 27475101. PMCID: PMC4967416.
Article
6. Amin FM, Hougaard A, Schytz HW, Asghar MS, Lundholm E, Parvaiz AI, et al. 2014; Investigation of the pathophysiological mechanisms of migraine attacks induced by pituitary adenylate cyclase-activating polypeptide-38. Brain. 137:779–94. DOI: 10.1093/brain/awt369. PMID: 24501094.
Article
7. Markovics A, Kormos V, Gaszner B, Lashgarara A, Szoke E, Sandor K, et al. 2012; Pituitary adenylate cyclase-activating polypeptide plays a key role in nitroglycerol-induced trigeminovascular activation in mice. Neurobiol Dis. 45:633–44. DOI: 10.1016/j.nbd.2011.10.010. PMID: 22033344.
Article
8. Hanada T, Hashizume Y, Tokuhara N, Takenaka O, Kohmura N, Ogasawara A, et al. 2011; Perampanel: a novel, orally active, noncompetitive AMPA-receptor antagonist that reduces seizure activity in rodent models of epilepsy. Epilepsia. 52:1331–40. DOI: 10.1111/j.1528-1167.2011.03109.x. PMID: 21635236.
Article
9. Roche KW, O'Brien RJ, Mammen AL, Bernhardt J, Huganir RL. 1996; Characterization of multiple phosphorylation sites on the AMPA receptor GluR1 subunit. Neuron. 16:1179–88. DOI: 10.1016/S0896-6273(00)80144-0. PMID: 8663994.
Article
10. Nye BL, Thadani VM. 2015; Migraine and epilepsy: review of the literature. Headache. 55:359–80. DOI: 10.1111/head.12536. PMID: 25754865.
Article
11. Chan K, MaassenVanDenBrink A. 2014; Glutamate receptor antagonists in the management of migraine. Drugs. 74:1165–76. DOI: 10.1007/s40265-014-0262-0. PMID: 25030431.
Article
12. Park JS, Yaster M, Guan X, Xu JT, Shih MH, Guan Y, et al. 2008; Role of spinal cord alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors in complete Freund's adjuvant-induced inflammatory pain. Mol Pain. 4:67. DOI: 10.1186/1744-8069-4-67. PMID: 19116032. PMCID: PMC2628655. PMID: 549e0037c15b4a679e8f4cc0f1081361.
Article
13. Tringali G, Currò D, Navarra P. 2018; Perampanel inhibits calcitonin gene-related peptide release from rat brainstem in vitro. J Headache Pain. 19:107. DOI: 10.1186/s10194-018-0940-5. PMID: 30419806. PMCID: PMC6755590. PMID: f5224fb2b3904254b4393801e9c75cc7.
Article
14. Villalón CM, Olesen J. 2009; The role of CGRP in the pathophysiology of migraine and efficacy of CGRP receptor antagonists as acute antimigraine drugs. Pharmacol Ther. 124:309–23. DOI: 10.1016/j.pharmthera.2009.09.003. PMID: 19796656.
Article
15. Hara K, Haranishi Y, Terada T. 2020; Intrathecally administered perampanel alleviates neuropathic and inflammatory pain in rats. Eur J Pharmacol. 872:172949. DOI: 10.1016/j.ejphar.2020.172949. PMID: 31991141.
Article
16. Askari-Zahabi K, Abbasnejad M, Kooshki R, Esmaeili-Mahani S. 2021; Orexin one receptors within the basolateral amygdala are involved in the modulation of cognitive deficits associated with a migraine-like state in rats. Neurol Res. 43:1087–97. DOI: 10.1080/01616412.2021.1949687. PMID: 34233602.
Article
17. Mahmoudi J, Mohaddes G, Erfani M, Sadigh-Eteghad S, Karimi P, Rajabi M, et al. 2018; Cerebrolysin attenuates hyperalgesia, photophobia, and neuroinflammation in a nitroglycerin-induced migraine model in rats. Brain Res Bull. 140:197–204. DOI: 10.1016/j.brainresbull.2018.05.008. PMID: 29752991.
Article
18. Tang Y, Liu S, Shu H, Xing Y, Tao F. 2018; AMPA receptor GluA1 Ser831 phosphorylation is critical for nitroglycerin-induced migraine-like pain. Neuropharmacology. 133:462–9. DOI: 10.1016/j.neuropharm.2018.02.026. PMID: 29486167. PMCID: PMC5858972.
Article
19. Mustelin L, Raevuori A, Kaprio J, Keski-Rahkonen A. 2014; Association between eating disorders and migraine may be explained by major depression. Int J Eat Disord. 47:884–7. DOI: 10.1002/eat.22311. PMID: 24888633.
Article
20. Wang K, Zhai Q, Wang S, Li Q, Liu J, Meng F, et al. 2021; Cryptotanshinone ameliorates CUS-induced depressive-like behaviors in mice. Transl Neurosci. 12:469–81. DOI: 10.1515/tnsci-2020-0198. PMID: 34900345. PMCID: PMC8633587. PMID: c89ef6bd6b7d4da991f44e4049713ab2.
Article
21. Liu L, Zheng J, Huang XF, Zhu X, Ding SM, Ke HM, et al. 2018; The neuroprotective and antidepressant-like effects of Hcyb1, a novel selective PDE2 inhibitor. CNS Neurosci Ther. 24:652–60. DOI: 10.1111/cns.12863. PMID: 29704309. PMCID: PMC6489804.
Article
22. Carruthers AM, Sellers LA, Jenkins DW, Jarvie EM, Feniuk W, Humphrey PP. 2001; Adenosine A(1) receptor-mediated inhibition of protein kinase A-induced calcitonin gene-related peptide release from rat trigeminal neurons. Mol Pharmacol. 59:1533–41. DOI: 10.1124/mol.59.6.1533. PMID: 11353815.
Article
23. Baratloo A, Mirbaha S, Delavar Kasmaei H, Payandemehr P, Elmaraezy A, Negida A. 2017; Intravenous caffeine citrate vs. magnesium sulfate for reducing pain in patients with acute migraine headache; a prospective quasi-experimental study. Korean J Pain. 30:176–82. DOI: 10.3344/kjp.2017.30.3.176. PMID: 28757917. PMCID: PMC5532524.
Article
24. Casili G, Lanza M, Filippone A, Campolo M, Paterniti I, Cuzzocrea S, et al. 2020; Dimethyl fumarate alleviates the nitroglycerin (NTG)-induced migraine in mice. J Neuroinflammation. 17:59. DOI: 10.1186/s12974-020-01736-1. PMID: 32066464. PMCID: PMC7469611. PMID: 489394dbd10c4f849f6c6eaa63a1fa31.
Article
25. Deen M, Correnti E, Kamm K, Kelderman T, Papetti L, Rubio-Beltrán E, et al. European Headache Federation School of Advanced Studies (EHF-SAS). 2017; Blocking CGRP in migraine patients - a review of pros and cons. J Headache Pain. 18:96. DOI: 10.1186/s10194-017-0807-1. PMID: 28948500. PMCID: PMC5612904. PMID: d2ba45670bee4a83b7dd02bf83f5592e.
Article
26. Goadsby PJ, Lipton RB, Ferrari MD. 2002; Migraine--current understanding and treatment. N Engl J Med. 346:257–70. DOI: 10.1056/NEJMra010917. PMID: 11807151.
27. Han X, Ran Y, Su M, Liu Y, Tang W, Dong Z, et al. 2017; Chronic changes in pituitary adenylate cyclase-activating polypeptide and related receptors in response to repeated chemical dural stimulation in rats. Mol Pain. 13:1744806917720361. DOI: 10.1177/1744806917720361. PMID: 28776455. PMCID: PMC5546650.
Article
28. Brewerton TD, George MS. 1993; Is migraine related to the eating disorders? Int J Eat Disord. 14:75–9. DOI: 10.1002/1098-108X(199307)14:1<75::AID-EAT2260140110>3.0.CO;2-D. PMID: 8339102.
Article
29. D'Andrea G, Ostuzzi R, Bolner A, Colavito D, Leon A. 2012; Is migraine a risk factor for the occurrence of eating disorders? Prevalence and biochemical evidences. Neurol Sci. 33 Suppl 1:S71–6. DOI: 10.1007/s10072-012-1045-6. PMID: 22644175.
30. Catterall WA. 2015; Regulation of cardiac calcium channels in the fight-or-flight response. Curr Mol Pharmacol. 8:12–21. DOI: 10.2174/1874467208666150507103417. PMID: 25966697. PMCID: PMC4664455.
Article
31. Ahuja M, Jha A, Maléth J, Park S, Muallem S. 2014; cAMP and Ca²+ signaling in secretory epithelia: crosstalk and synergism. Cell Calcium. 55:385–93. DOI: 10.1016/j.ceca.2014.01.006. PMID: 24613710. PMCID: PMC4058382.
Article
32. Chen T, Koga K, Descalzi G, Qiu S, Wang J, Zhang LS, et al. 2014; Postsynaptic potentiation of corticospinal projecting neurons in the anterior cingulate cortex after nerve injury. Mol Pain. 10:33. DOI: 10.1186/1744-8069-10-33. PMID: 24890933. PMCID: PMC4060852.
Article
33. Yue X, Tumati S, Navratilova E, Strop D, St John PA, Vanderah TW, et al. 2008; Sustained morphine treatment augments basal CGRP release from cultured primary sensory neurons in a Raf-1 dependent manner. Eur J Pharmacol. 584:272–7. DOI: 10.1016/j.ejphar.2008.02.013. PMID: 18328477. PMCID: PMC2375088.
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