Anesth Pain Med.  2018 Jan;13(1):82-92. 10.17085/apm.2018.13.1.82.

Differential expression of spinal γ-aminobutyric acid and opioid receptors modulates the analgesic effects of intrathecal curcumin on postoperative/inflammatory pain in rats

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

Abstract

BACKGROUND
Curcumin is traditionally used as an herbal medicine. We explored the efficacy of intrathecal curcumin in relieving both postoperative and inflammatory pain and elucidated the mechanisms of action of curcumin interacting with γ-aminobutyric acid (GABA) and opioid receptors at the spinal level.
METHODS
Experimental pain was induced in male Sprague-Dawley rats via paw incision or injection of intraplantar carrageenan. After examination of the effects of intrathecal curcumin on the pain, GABA and opioid receptor antagonists were intrathecally administered to explore the involvement of GABA or opioid receptors on the effect of curcumin. Additionally, the expression levels of the GABA and opioid receptors were assessed.
RESULTS
Intrathecal curcumin reduced the withdrawal threshold of both incisional surgery- and carrageenan injection-induced nociception. Intrathecal GABA and opioid receptor antagonists reversed the curcumin-mediated antinociception. Incisional surgery decreased the levels of the GABA receptors mRNA, but little changed the levels of the opioid receptors mRNA. Carrageenan injection increased the levels of the opioid receptors mRNA, but not the GABA receptors mRNA levels. Intrathecal curcumin increased or decreased the levels of GABA receptors mRNA and opioid receptors mRNA in the spinal cords of incised or carrageenan-injected rats, respectively.
CONCLUSIONS
Intrathecal curcumin was effective to postoperative and inflammatory pain and such antinociception of curcumin was antagonized by both GABA and opioid receptor antagonists. Also, intrathecal curcumin altered the levels of GABA and opioid receptors. Thus, spinal GABA and opioid receptors may, respectively, be directly or indirectly involved when curcumin alleviates postoperative and inflammatory pain.

Keyword

Curcumin; γ-aminobutyric acid and opioid receptor; Postoperative and inflammatory pain; Spinal cord

MeSH Terms

Animals
Carrageenan
Curcumin*
gamma-Aminobutyric Acid
Herbal Medicine
Humans
Male
Narcotic Antagonists
Nociception
Rats*
Rats, Sprague-Dawley
Receptors, GABA
Receptors, Opioid*
RNA, Messenger
Spinal Cord
Carrageenan
Curcumin
Narcotic Antagonists
RNA, Messenger
Receptors, GABA
Receptors, Opioid
gamma-Aminobutyric Acid

Figure

  • Fig. 1 Time course of hindpaw withdrawal responses to von Frey filament pressure after (A) incisional surgery or (B) carrageenan injection. Data are presented as withdrawal thresholds. Each line represents the mean ± SEM of data from five or six rats. BL: baseline withdrawal threshold measured prior to paw incision or carrageenan injection. Significant differences between data from the ipsilateral and contralateral sites are indicated, *P < 0.05, †P < 0.01, ‡P < 0.001.

  • Fig. 2 Expression levels of mRNAs encoding γ-aminobutyric acid (GABA) and opioid receptors in the left dorsal horn of the spinal cord in naïve, incised, and carrageenan-injected rats. Data are presented as relative values of GABA and opioid receptors to that of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Each bar represents the mean ± SEM of data from four rats. Reverse transcriptase polymerase chain reaction detected mRNA bands corresponding to the GABA and opioid receptors in naïve animals. (A) Quantitative analysis indicated that incisional surgery reduced the levels of expression of mRNAs encoding GABA receptors, but not of those encoding opioid receptors. (B) Carrageenan injection increased expression of opioid receptor mRNAs, but not those of GABA receptor mRNAs. *P < 0.05, †P < 0.01, ‡P < 0.001 compared to naïve rats.

  • Fig. 3 (A) The time effect and (B) dose-response of intrathecal curcumin on the hindpaw withdrawal response to von Frey filament pressure applied after incisional surgery. Data are presented as withdrawal thresholds or percentages of maximal possible effects (%MPEs). Each line represents the mean ± SEM of data from five-to-seven rats. Control data were obtained 2 h after incision. Curcumin was administered immediately after measurement of control post-incisional thresholds. BL: baseline withdrawal threshold measured before paw incision. Curcumin produced dose-dependent increases in withdrawal thresholds of injured paws after incision. *P < 0.05, †P < 0.01 compared to vehicle (dimethylsulfoxide, DMSO) alone.

  • Fig. 4 (A) The time effect of and (B) dose-response to intrathecal curcumin, assessed by measuring hindpaw withdrawal responses to von Frey filament pressure applied after carrageenan injection. Data are presented as withdrawal thresholds or percentages of maximal possible effects (%MPEs). Each line represents the mean ± SEM of data from five-to-seven rats. Control data were obtained 4 h after carrageenan injection. Curcumin was administered immediately after measurement of control post-injectional thresholds. BL: baseline withdrawal threshold measured before carrageenan injection. Curcumin caused a dose-dependent increase in the withdrawal thresholds of injected paws after carrageenan injection. *P < 0.05 compared to vehicle (dimethylsulfoxide, DMSO) alone.

  • Fig. 5 (A) The effects of intrathecal bicuculline (a γ-aminobutyric acid [GABA]-A receptor antagonist, 0.3 μg), and saclofen (a GABA-B receptor antagonist, 100 μg), on the antinociceptive effects of intrathecal curcumin (100 μg) in rats experiencing postoperative pain; and (B) the effects of intrathecal curcumin (100 μg) on the expression levels of mRNAs encoding GABA receptors in the left dorsal horn of the spinal cord in incised rats. Data are presented as percentages of maximal possible effects (%MPEs) or the relative values of GABA receptor levels to that of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Each bar represents the mean ± SEM of data from four-to-six rats. DMSO: dimethylsulfoxide, CUR: curcumin, BI: Bicuculline, SA: saclofen. (A) Both bicuculline and saclofen reversed the effects of curcumin, †P < 0.01, ‡P < 0.001 compared with curcumin. (B) Intrathecal curcumin increased the expression levels of mRNAs encoding GABA receptors, †P < 0.01, ‡P < 0.001 compared with the incised group.

  • Fig. 6 (A) The effects of intrathecal CTOP (a μ opioid receptor antagonist, 15 μg), naltrindole (a δ opioid receptor antagonist, 10 g), and GNTI (a κ opioid receptor antagonist, 50 μg) on the antinociceptive effects of intrathecal curcumin (100 μg) on postoperative pain, and (B) the effects of intrathecal curcumin (100 μg) on the expression levels of mRNAs encoding opioid receptors in the left dorsal horn of the spinal cord in incised rats. Data are presented as percentages of maximal possible effects (%MPEs) or the relative expression levels of opioid receptors compared to that of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Each bar represents the mean ± SEM of data from four-to-six rats. DMSO: dimethylsulfoxide, CUR: curcumin, NALT: naltrindole. (A) CTOP, naltrindole and GNTI reversed the effects of curcumin, ‡P < 0.001 compared with curcumin. (B) Intrathecal curcumin only marginally affected the expression levels of mRNAs encoding opioid receptors in the incised group.

  • Fig. 7 (A) The effects of intrathecal CTOP (a μ opioid receptor antagonist, 15 μg), naltrindole (a δ opioid receptor antagonist, 10 μg) and GNTI (a κ opioid receptor antagonist, 50 μg) on the antinociceptive effects of intrathecal curcumin (300 μg) on inflammatory pain, and (B) the effects of intrathecal curcumin (300 μg) on the expression levels of mRNAs encoding opioid receptors in the left dorsal horn of the spinal cord in carrageenan-injected rats. Data are presented as percentages of maximal possible effects (%MPEs) or the relative expression levels of opioid receptors compared to that of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Each bar represents the mean ± SEM of data from four-to-six rats. DMSO: dimethylsulfoxide, CUR: curcumin, NALT: naltrindole. (A) All of CTOP, naltrindole, and GNTI reversed the effects of curcumin. ‡P < 0.001 compared with curcumin. (B) Intrathecal curcumin decreased the expression levels of mRNAs encoding all three opioid receptors, *P < 0.05, †P < 0.01, compared to the inflammatory group.

  • Fig. 8 (A) The effects of intrathecal bicuculline (a γ-aminobutyric acid [GABA]-A receptor antagonist, 0.3 μg) and saclofen (a GABA-B receptor antagonist, 100 μg) on the antinociceptive effects of intrathecal curcumin (300 μg) on inflammatory pain, and (B) the effects of intrathecal curcumin (300 μg) on the expression levels of mRNAs encoding GABA receptors in the left dorsal horn of the spinal cord in carrageenan-injected rats. Data are presented as percentages of maximal possible effects (%MPEs) or the relative expression levels of GABA receptors to that of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Each bar represents the mean ± SEM of data from four-to-six rats. DMSO: dimethylsulfoxide, CUR: curcumin, BI: Bicuculline, SA: saclofen. (A) Both bicuculline and saclofen reversed the effects of curcumin. †P < 0.01 compared with curcumin. (B) Intrathecal curcumin only marginally affected the expression levels of mRNAs encoding GABA receptors in the inflammatory group.


Reference

1. Gold MS, Gebhart GF. Nociceptor sensitization in pain pathogenesis. Nat Med. 2010; 16:1248–57. DOI: 10.1038/nm.2235. PMID: 20948530. PMCID: PMC5022111.
2. Woolf CJ. Central sensitization: implications for the diagnosis and treatment of pain. Pain. 2011; 152(3 Suppl):S2–15. DOI: 10.1016/j.pain.2010.09.030. PMID: 20961685. PMCID: PMC3268359.
3. Fishman SM, Ballantyne JE, Rathmell JP, Bonica JJ. Bonica's management of pain. 4th ed. Baltimore, MD: Lippincott Williams & Wilkins;2010.
4. Chainani-Wu N. Safety and anti-inflammatory activity of curcumin: a component of tumeric (Curcuma longa). J Altern Complement Med. 2003; 9:161–8. DOI: 10.1089/107555303321223035. PMID: 12676044.
5. Chen JJ, Dai L, Zhao LX, Zhu X, Cao S, Gao YJ. Intrathecal curcumin attenuates pain hypersensitivity and decreases spinal neuroinflammation in rat model of monoarthritis. Sci Rep. 2015; 5:10278. DOI: 10.1038/srep10278. PMID: 25988362. PMCID: PMC4437313.
6. Lee JH, Kim YD, Jung HC, Cheong YK. The effect of intrathecal curcumin on mechanical allodynia in rats after L5 spinal nerve ligation. Korean J Anesthesiol. 2014; 67(Suppl):S122–3. DOI: 10.4097/kjae.2014.67.S.S122. PMID: 25598884. PMCID: PMC4295958.
7. Yaksh TL, Rudy TA. Chronic catheterization of the spinal subarachnoid space. Physiol Behav. 1976; 17:1031–6. DOI: 10.1016/0031-9384(76)90029-9.
8. Lin H, Lee HG, Yoon MH. Antiallodynic effects of intrathecal tianeptine in a neuropathic pain rat. Anesth Pain Med. 2014; 9:93–7.
9. Brennan TJ, Vandermeulen EP, Gebhart GF. Characterization of a rat model of incisional pain. Pain. 1996; 64:493–501. DOI: 10.1016/0304-3959(95)01441-1.
10. Yang J, Bae HB, Ki HG, Oh JM, Kim WM, Lee HG, et al. Different role of spinal 5-HT(hydroxytryptamine)7 receptors and descending serotonergic modulation in inflammatory pain induced in formalin and carrageenan rat models. Br J Anaesth. 2014; 113:138–47. DOI: 10.1093/bja/aet336. PMID: 24129596.
11. Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 1994; 53:55–63. DOI: 10.1016/0165-0270(94)90144-9.
12. Castro AR, Pinto M, Lima D, Tavares I. Secondary hyperalgesia in the monoarthritic rat is mediated by GABAB and NK1 receptors of spinal dorsal horn neurons: a behavior and c-fos study. Neuroscience. 2006; 141:2087–95. DOI: 10.1016/j.neuroscience.2006.05.048. PMID: 16809001.
13. Yoon MH, Kim KS, Lee HG, Kim CM, Kim WM, Choi JI, et al. Synergistic interaction between intrathecal ginsenosides and morphine on formalin-induced nociception in rats. J Pain. 2011; 12:774–81. DOI: 10.1016/j.jpain.2010.12.012. PMID: 21459679.
14. Goodchild CS, Nadeson R, Cohen E. Supraspinal and spinal cord opioid receptors are responsible for antinociception following intrathecal morphine injections. Eur J Anaesthesiol. 2004; 21:179–85. DOI: 10.1097/00003643-200403000-00003. PMID: 15055889.
15. Malcangio M, Bowery NG. GABA and its receptors in the spinal cord. Trends Pharmacol Sci. 1996; 17:457–62. DOI: 10.1016/S0165-6147(96)01013-9.
16. Malan TP, Mata HP, Porreca F. Spinal GABA(A) and GABA(B) receptor pharmacology in a rat model of neuropathic pain. Anesthesiology. 2002; 96:1161–7. DOI: 10.1097/00000542-200205000-00020. PMID: 11981157.
17. Reichl S, Augustin M, Zahn PK, Pogatzki-Zahn EM. Peripheral and spinal GABAergic regulation of incisional pain in rats. Pain. 2012; 153:129–41. DOI: 10.1016/j.pain.2011.09.028. PMID: 22054599.
18. Lee HG, Park SK, Yoon MH. Potentiation of morphine antiallodynic efficacy by ACPT-III, a group III metabotropic glutamate receptor agonist, in rat spinal nerve ligation-induced neuropathic pain. Pharmacol Biochem Behav. 2010; 96:108–13. DOI: 10.1016/j.pbb.2010.04.014. PMID: 20434481.
19. Cabañero D, Célérier E, García-Nogales P, Mata M, Roques BP, Maldonado R, et al. The pro-nociceptive effects of remifentanil or surgical injury in mice are associated with a decrease in delta-opioid receptor mRNA levels: Prevention of the nociceptive response by on-site delivery of enkephalins. Pain. 2009; 141:88–96. DOI: 10.1016/j.pain.2008.10.011. PMID: 19058913.
20. Castro-Lopes JM, Malcangio M, Pan BH, Bowery NG. Complex changes of GABAA and GABAB receptor binding in the spinal cord dorsal horn following peripheral inflammation or neurectomy. Brain Res. 1995; 679:289–97. DOI: 10.1016/0006-8993(95)00262-O.
21. Obara I, Parkitna JR, Korostynski M, Makuch W, Kaminska D, Przewlocka B, et al. Local peripheral opioid effects and expression of opioid genes in the spinal cord and dorsal root ganglia in neuropathic and inflammatory pain. Pain. 2009; 141:283–91. DOI: 10.1016/j.pain.2008.12.006. PMID: 19147290.
22. Cahill CM, Morinville A, Hoffert C, O’Donnell D, Beaudet A. Up-regulation and trafficking of delta opioid receptor in a model of chronic inflammation: implications for pain control. Pain. 2003; 101:199–208. DOI: 10.1016/S0304-3959(02)00333-0.
23. Maekawa K, Minami M, Masuda T, Satoh M. Expression of mu- and kappa-, but not delta-, opioid receptor mRNAs is enhanced in the spinal dorsal horn of the arthritic rats. Pain. 1996; 64:365–71. DOI: 10.1016/0304-3959(95)00132-8.
24. Han YK, Lee SH, Jeong HJ, Kim MS, Yoon MH, Kim WM. Analgesic effects of intrathecal curcumin in the rat formalin test. Korean J Pain. 2012; 25:1–6. DOI: 10.3344/kjp.2012.25.1.1. PMID: 22259709. PMCID: PMC3259131.
25. De Paz-Campos MA, Ortiz MI, Chávez Piña AE, Zazueta-Beltrán L, Castañeda-Hernández G. Synergistic effect of the interaction between curcumin and diclofenac on the formalin test in rats. Phytomedicine. 2014; 21:1543–8. DOI: 10.1016/j.phymed.2014.06.015. PMID: 25442263.
26. Nurullahoglu KE, Okudan N, Belviranli M, Oz M. The comparison of preemptive analgesic effects of curcumin and diclofenac. Bratisl Lek Listy. 2014; 115:757–60. DOI: 10.4149/BLL_2014_146.
27. Zhao X, Wang C, Zhang JF, Liu L, Liu AM, Ma Q, et al. Chronic curcumin treatment normalizes depression-like behaviors in mice with mononeuropathy: involvement of supraspinal serotonergic system and GABAA receptor. Psychopharmacology (Berl). 2014; 231:2171–87. DOI: 10.1007/s00213-013-3368-2. PMID: 24297305.
28. Zhi L, Dong L, Kong D, Sun B, Sun Q, Grundy D, et al. Curcumin acts via transient receptor potential vanilloid-1 receptors to inhibit gut nociception and reverses visceral hyperalgesia. Neurogastroenterol Motil. 2013; 25:e429–40. DOI: 10.1111/nmo.12145. PMID: 23638900.
29. Greeshma N, Prasanth KG, Balaji B. Tetrahydrocurcumin exerts protective effect on vincristine induced neuropathy: Behavioral, biochemical, neurophysiological and histological evidence. Chem Biol Interact. 2015; 238:118–28. DOI: 10.1016/j.cbi.2015.06.025. PMID: 26102012.
30. Lee JY, Shin TJ, Choi JM, Seo KS, Kim HJ, Yoon TG, et al. Antinociceptive curcuminoid, KMS4034, effects on inflammatory and neuropathic pain likely via modulating TRPV1 in mice. Br J Anaesth. 2013; 111:667–72. DOI: 10.1093/bja/aet176. PMID: 23719767.
Full Text Links
  • APM
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