DOI QR코드

DOI QR Code

Pachymic Acid Enhances Pentobarbital-Induced Sleeping Behaviors via GABAA-ergic Systems in Mice

  • Shah, Vikash Kumar (College of Pharmacy, Chungbuk National University) ;
  • Choi, Jae Joon (College of Pharmacy, Chungbuk National University) ;
  • Han, Jin-Yi (Institute of Veterinary Medicine, Chungbuk National University) ;
  • Lee, Mi Kyeong (College of Pharmacy, Chungbuk National University) ;
  • Hong, Jin Tae (College of Pharmacy, Chungbuk National University) ;
  • Oh, Ki-Wan (College of Pharmacy, Chungbuk National University)
  • Received : 2014.04.21
  • Accepted : 2014.06.13
  • Published : 2014.07.31

Abstract

This study was investigated to know whether pachymic acid (PA), one of the predominant triterpenoids in Poria cocos (Hoelen) has the sedative-hypnotic effects, and underlying mechanisms are mediated via ${\gamma}$-aminobutyric acid (GABA)-ergic systems. Oral administration of PA markedly suppressed locomotion activity in mice. This compound also prolonged sleeping time, and reduced sleep latency showing synergic effects with muscimol (0.2 mg/kg) in shortening sleep onset and enhancing sleep time induced by pentobarbital, both at the hypnotic (40 mg/kg) and sub-hypnotic (28 mg/kg) doses. Additionally, PA elevated intracellular chloride levels in hypothalamic primary cultured neuronal cells of rats. Moreover, Western blotting quantitative results showed that PA increased the amount of protein level expression of $GAD_{65/67}$ over a broader range of doses. PA increased ${\alpha}$- and ${\beta}$-subunits protein levels, but decreased ${\gamma}$-subunit protein levels in $GABA_A$ receptors. The present experiment provides evidence for the hypnotic effects as PA enhanced pentobarbital-induced sleeping behaviors via $GABA_A$-ergic mechanisms in rodents. Taken together, it is proposed that PA may be useful for the treatment of sleep disturbed subjects with insomnia.

Keywords

References

  1. Abourashed, E. A., Koetter, U. and Brattstrom, A. (2004) In vitro binding experiments with a Valerian, hops and their fixed combination extract (Ze91019) to selected central nervous system receptors. Phytomedicine 11, 633-638. https://doi.org/10.1016/j.phymed.2004.03.005
  2. Barnard, E. A., Skolnick, P., Olsen, R. W., Mohler, H., Sieghart, W., Biggio, G., Braestrup, C., Bateson, A. N. and Langer, S. Z. (1998) International Union of Pharmacology. XV. Subtypes of gammaaminobutyric acidA receptors: classification on the basis of subunit structure and receptor function. Pharmacol. Rev. 50, 291-313.
  3. Chen, F. P., Jong, M. S., Chen, Y. C., Kung, Y. Y., Chen, T. J., Chen, F. J. and Hwang, S. J. (2011) Prescriptions of Chinese Herbal Medicines for Insomnia in Taiwan during 2002. Evid. Based Complement Alternat. Med. 2011, 236341.
  4. Chistina Grobin, A., Inglefi eld, J. R., Schwartz-Bloom, R. D., Devaud, L. L. and Morrow, A. L. (1999) Fluorescence imaging of GABAA receptor-mediated intracellular [Cl-] in P19-N cells reveals unique pharmacological properties. Brain Res. 827, 1-11. https://doi.org/10.1016/S0006-8993(99)01223-8
  5. Cuella, M. J., Giner, R. M., Recio, M. C., Just, M. J., Manez, S. and Rios, J. L. (1996) Two fungal lanostane derivatives as phospholipase A2 inhibitors. J. Nat. Prod. 59, 977-979. https://doi.org/10.1021/np9604339
  6. Darias, V., Abdala, S., Martin-Herrera, D., Tello, M. L. and Vega, S. (1998) CNS effects of a series of 1,2,4-triazolyl heterocarboxylic derivatives. Pharmazie 53, 477-481.
  7. de Sousa, F. C., Pereira, B. A., Lima, V. T., Lacerda, C. D., Melo, C. T., Barbosa-Filho, J. M., Vasconcelos, S. M. and Viana, G. S. (2005) Central nervous system activity of yangambin from Ocotea duckei Vattimo (Lauraceae) in mice. Phytother. Res. 19, 282-286. https://doi.org/10.1002/ptr.1499
  8. Gapter, L., Wang, Z., Glinski, J. and Ng, K. Y. (2005) Induction of apoptosis in prostate cancer cells by pachymic acid from Poria cocos. Biochem. Biophys. Res. Commun. 332, 1153-1161. https://doi.org/10.1016/j.bbrc.2005.05.044
  9. Giner, E. M., Manez, S., Recio, M. C., Giner, R. M., Cerda-Nicolas, M. and Rios, J. L. (2000) In vivo studies on the anti-infl ammatory activity of pachymic and dehydrotumulosic acids. Planta Med. 66, 221-227. https://doi.org/10.1055/s-2000-8563
  10. Imamura, M. and Prasad, C. (1998) Increased GABA-gated chloride ion infl ux in the hypothalamus of low-anxiety rats. Physiol. Behav. 64, 415-417. https://doi.org/10.1016/S0031-9384(98)00105-X
  11. Kaminaga, T., Yasukawa, K., Kanno, H., Tai, T., Nunoura, Y. and Takido, M. (1996) Inhibitory effects of lanostane-type triterpene acids, the components of Poria cocos, on tumor promotion by 12-O-tetradecanoylphorbol-13-acetate in two-stage carcinogenesis in mouse skin. Oncology 53, 382-385. https://doi.org/10.1159/000227592
  12. Lee, S. M., Lee, Y. J., Yoon, J. J., Kang, D. G. and Lee, H. S. (2012) Effect of Poria cocos on hypertonic stress-induced water channel expression and apoptosis in renal collecting duct cells. J. Ethnopharmacol. 141, 368-376. https://doi.org/10.1016/j.jep.2012.02.048
  13. Li, G., Xu, M. L., Lee, C. S., Woo, M. H., Chang, H. W. and Son, J. K. (2004) Cytotoxicity and DNA topoisomerases inhibitory activity of constituents from the sclerotium of Poria cocos. Arch. Pharm. Res. 27, 829-833. https://doi.org/10.1007/BF02980174
  14. Ma, Y., Han, H., Eun, J. S., Kim, H. C., Hong, J. T. and Oh, K. W. (2007) Sanjoinine A isolated from Zizyphi Spinosi Semen augments pentobarbital-induced sleeping behaviors through the modification of GABA-ergic systems. Biomol. Ther. 30, 1748-1753.
  15. Ma, Y., Jo, Y. J., Woo, S. S., Hong, J. T. and Oh, K. W. (2008) Honokiol potentiates pentobarbital-induced sleeping behaviors through GABA (A) receptor CI-channel activation. J. Applied Pharmacol. 16, 328-335.
  16. Mathews, G. C., Bolos-Sy, A. M., Holland, K. D., Isenberg, K. E., Covey, D. F., Ferrendelli, J. A. and Rothman, S. M. (1994) Developmental alteration in GABAA receptor structure and physiological properties in cultured cerebellar granule neurons. Neuron 13, 149-158. https://doi.org/10.1016/0896-6273(94)90465-0
  17. McKernan, R. M. and Whiting, P. J. (1996) Which GABAA-receptor subtypes really occur in the brain? Trends. Neurosci. 19, 139-143. https://doi.org/10.1016/S0166-2236(96)80023-3
  18. Mohler, H., Fritschy, J. M. and Rudolph, U. (2002) A new benzodiazepine pharmacology. J. Pharmacol. Exp. Ther. 300, 2-8. https://doi.org/10.1124/jpet.300.1.2
  19. Morton, G. J., Kaiyala, K. J., Fisher, J. D., Ogimoto, K., Schwartz, M. W. and Wisse, B. E. (2011) Identifi cation of a physiological role for leptin in the regulation of ambulatory activity and wheel running in mice. Am. J. Physiol. Endocrinol. Metab. 300, E392-401. https://doi.org/10.1152/ajpendo.00546.2010
  20. Park, J. H., Cha, H. Y., Seo, J. J., Hong, J. T., Han, K. and Oh, K. W. (2005) Anxiolytic-like effects of ginseng in the elevated plus-maze model: comparison of red ginseng and sun ginseng. Prog. Neuropsychopharmacol. Biol. Psychiatry 29, 895-900. https://doi.org/10.1016/j.pnpbp.2005.04.016
  21. Qu, W. M., Yue, X. F., Sun, Y., Fan, K., Chen, C. R., Hou, Y. P., Urade, Y. and Huang, Z. L. (2012) Honokiol promotes non-rapid eye movement sleep via the benzodiazepine site of the GABA(A) receptor in mice. Br. J. Pharmacol. 167, 587-598. https://doi.org/10.1111/j.1476-5381.2012.02010.x
  22. Rosenberg, R. P. (2006) Sleep maintenance insomnia: strengths and weaknesses of current pharmacologic therapies. Ann. Clin. Psychiatry 18, 49-56. https://doi.org/10.1080/10401230500464711
  23. Rudolph, U. and Mohler, H. (2004) Analysis of GABAA receptor function and dissection of the pharmacology of benzodiazepines and general anesthetics through mouse genetics. Annu. Rev. Pharmacol. Toxicol. 44, 475-498. https://doi.org/10.1146/annurev.pharmtox.44.101802.121429
  24. Rudolph, U. and Mohler, H. (2006) GABA-based therapeutic approaches: GABAA receptor subtype functions. Curr. Opin. Pharmacol. 6, 18-23. https://doi.org/10.1016/j.coph.2005.10.003
  25. Sieghart, W. and Sperk, G. (2002) Subunit composition, distribution and function of GABA(A) receptor subtypes. Curr. Top. Med. Chem. 2, 795-816. https://doi.org/10.2174/1568026023393507
  26. Spelman, K., Burns, J., Nichols, D., Winters, N., Ottersberg, S. and Tenborg, M. (2006) Modulation of cytokine expression by traditional medicines: a review of herbal immunomodulators. Altern. Med. Rev. 11, 128-150.
  27. Tai, T., Akita, Y., Kinoshita, K., Koyama, K., Takahashi, K. and Watanabe, K. (1995) Anti-emetic principles of Poria cocos. Planta Med. 61, 527-530. https://doi.org/10.1055/s-2006-959363
  28. Tillakaratne, N. J., Medina-Kauwe, L. and Gibson, K. M. (1995) gamma-Aminobutyric acid (GABA) metabolism in mammalian neural and nonneural tissues. Comp. Biochem. Physiol. A Physiol. 112, 247-263. https://doi.org/10.1016/0300-9629(95)00099-2
  29. Wafford, K. A. and Ebert, B. (2008) Emerging anti-insomnia drugs: tackling sleeplessness and the quality of wake time. Nat. Rev. Drug Discov. 7, 530-540. https://doi.org/10.1038/nrd2464
  30. West, M. R. and Molloy, C. R. (1996) A microplate assay measuring chloride ion channel activity. Anal. Biochem. 241, 51-58. https://doi.org/10.1006/abio.1996.0377
  31. Wolfman, C., Viola, H., Marder, M., Wasowski, C., Ardenghi, P., Izquierdo, I., Paladini, A. C. and Medina, J. H. (1996) Anxioselective properties of 6,3'-dinitrofl avone, a high-affinity benzodiazepine receptor ligand. Eur. J. Pharmacol. 318, 23-30. https://doi.org/10.1016/S0014-2999(96)00784-4
  32. Zhang, Z. J. (2004) Therapeutic effects of herbal extracts and constituents in animal models of psychiatric disorders. Life Sci. 75, 1659-1699. https://doi.org/10.1016/j.lfs.2004.04.014

Cited by

  1. Rosmarinic Acid Potentiates Pentobarbital-Induced Sleep Behaviors and Non-Rapid Eye Movement (NREM) Sleep through the Activation of GABAA-ergic Systems vol.25, pp.2, 2017, https://doi.org/10.4062/biomolther.2016.035
  2. Quinpirole Increases Melatonin-Augmented Pentobarbital Sleep via Cortical ERK, p38 MAPK, and PKC in Mice vol.24, pp.2, 2016, https://doi.org/10.4062/biomolther.2015.097
  3. Development of an Assay Method to Search for Compounds Inhibiting Stress-Enhanced Allergy vol.39, pp.5, 2016, https://doi.org/10.1248/bpb.b15-00731
  4. Artemisia capillaris Thunberg Produces Sedative-Hypnotic Effects in Mice, Which are Probably Mediated Through Potentiation of the GABAA Receptor vol.43, pp.04, 2015, https://doi.org/10.1142/S0192415X1550041X
  5. Sleep-promoting effects of the GABA/5-HTP mixture in vertebrate models vol.310, 2016, https://doi.org/10.1016/j.bbr.2016.04.049
  6. Evodiamine Reduces Caffeine-Induced Sleep Disturbances and Excitation in Mice vol.26, pp.5, 2018, https://doi.org/10.4062/biomolther.2017.146
  7. Sleep-Aids Derived from Natural Products vol.26, pp.4, 2018, https://doi.org/10.4062/biomolther.2018.099
  8. The core pattern of Chinese herbal formulae and drug–herb concurrent usage in patients with dementia vol.98, pp.4, 2019, https://doi.org/10.1097/MD.0000000000013931
  9. Poria cocos ethanol extract and its active constituent, pachymic acid, modulate sleep architectures via activation of GABAA-ergic transmission in rats vol.16, pp.3, 2014, https://doi.org/10.12729/jbr.2015.16.3.084
  10. Ephedrine Alkaloids-Free Ephedra Herb Extract, EFE, Has No Adverse Effects Such as Excitation, Insomnia, and Arrhythmias vol.41, pp.2, 2014, https://doi.org/10.1248/bpb.b17-00803
  11. Insights into Triterpene Acids in Fermented Mycelia of Edible Fungus Poria cocos by a Comparative Study vol.24, pp.7, 2014, https://doi.org/10.3390/molecules24071331
  12. Nature brings new avenues to the therapy of central nervous system diseases-An overview of possible treatments derived from natural products vol.62, pp.10, 2014, https://doi.org/10.1007/s11427-019-9587-y
  13. Research Progress on Classical Traditional Chinese Medicine Jieyu Pills in the Treatment of Depression vol.16, pp.None, 2020, https://doi.org/10.2147/ndt.s282384
  14. Pachymic acid protects against cerebral ischemia/reperfusion injury by the PI3K/Akt signaling pathway vol.35, pp.4, 2014, https://doi.org/10.1007/s11011-020-00540-3
  15. Pachymic acid protects oocyte by improving the ovarian microenvironment in polycystic ovary syndrome mice† vol.103, pp.5, 2014, https://doi.org/10.1093/biolre/ioaa141
  16. Effects and molecular mechanism of pachymic acid on ferroptosis in renal ischemia reperfusion injury vol.23, pp.1, 2020, https://doi.org/10.3892/mmr.2020.11704
  17. The Lanostane Triterpenoids in Poria cocos Play Beneficial Roles in Immunoregulatory Activity vol.11, pp.2, 2021, https://doi.org/10.3390/life11020111
  18. Kamishoyosan potentiates pentobarbital-induced sleep in socially isolated, ovariectomized mice vol.281, pp.None, 2014, https://doi.org/10.1016/j.jep.2021.114585