DOI QR코드

DOI QR Code

Neuroprotection by Valproic Acid in Mouse Models of Permanent and Transient Focal Cerebral Ischemia

  • Qian, Yong Ri (Department of Pharmacology, School of Basic Medicine, Yanbian University) ;
  • Lee, Mu-Jin (Department of Pharmacology, Chonnam National University Medical School) ;
  • Hwang, Shi-Nae (Department of Pharmacology, Chonnam National University Medical School) ;
  • Kook, Ji-Hyun (Department of Pharmacology, Chonnam National University Medical School) ;
  • Kim, Jong-Keun (Department of Pharmacology, Chonnam National University Medical School) ;
  • Bae, Choon-Sang (Department of Anatomy, Chonnam National University Medical School)
  • Received : 2010.11.18
  • Accepted : 2010.12.10
  • Published : 2010.12.31

Abstract

Valproic acid (VPA) is a well-known anti-epileptic and mood stabilizing drug. A growing number of reports demonstrate that VPA is neuroprotective against various insults. Despite intensive efforts to develop new therapeutics for stroke over the past two decades, all treatments have thus far failed to show clinical effect because of treatment-limiting side effects of the drugs. Therefore, a safety-validated drug like VPA would be an attractive candidate if it has neuroprotective effects against ischemic insults. The present study was undertaken to examine whether pre- and post-insult treatments with VPA protect against brain infarct and neurological deficits in mouse transient (tMCAO) and permanent middle cerebral artery occlusion (pMCAO) models. In the tMCAO (2 hr MCAO and 22 hr reperfusion) model, intraperitoneal injection of VPA (300 mg/kg, Lp.) 30 min prior to MCAO significantly reduced the infarct size and the neurological deficit. VPA treatment immediately after reperfusion significantly reduced the infarct size. The administration of VPA at 4 hr after reperfusion failed to reduce the infarct size and the neurological deficit. In the pM CAO model, treatment with VPA (300 mg/kg, i.p.) 30 min prior to MCAO significantly attenuated the infarct size, but did not affect the neurological deficit. Western blot analysis of acetylated H3 and H4 protein levels in extracts from the ischemic cortical area showed that treatment with VPA increased the expression of acetylated H3 and H4 at 2 hrs after MCAO. These results demonstrated that treatment with VPA prior to ischemia attenuated ischemic brain damage in both mice tMCAO and pMCAO models and treatment with VPA immediately after reperfusion reduced the infarct area in the tMCAO model. VPA could therefore be evaluated for clinical use in stroke patients.

Keywords

References

  1. Rogawski MA, Loscher W. The neurobiology of antiepileptic drugs. Nat Rev Neurosci. 2004;5:553-564. https://doi.org/10.1038/nrn1430
  2. Hashimoto R, Hough C, Nakazawa T, Yamamoto T, Chuang DM. Lithium protection against glutamate excitotoxicity in rat cerebral cortical neurons: involvement of NMDA receptor inhibition possibly by decreasing NR2B tyrosine phosphorylation. J Neurochem. 2002;80:589-597. https://doi.org/10.1046/j.0022-3042.2001.00728.x
  3. Kanai H, Sawa A, Chen R-W, Leeds P, Chuang DM. Valproic acid inhibits histone deacetylase activity and suppresses excitotoxicity-induced GAPDH nuclear accumulation and apoptotic death in neuron. Pharmacogenomics J. 2004;4:336-344. https://doi.org/10.1038/sj.tpj.6500269
  4. Rekling JC. Neuroprotective effects of anticonvulsants in rat hippocampal slice cultures exposed to oxygen/glucose deprivation. Neurosci Lett. 2003;335:167-170. https://doi.org/10.1016/S0304-3940(02)01193-X
  5. Jeong MR. Hashimoto R. Senatorov VV, Fujimaki K, Ren M, Lee MS, Chuang DM. Valproic acid, a mood stabilizer and anticonvulsant, protects rat cerebral cortical neurons from spontaneous cell death: a role of histone deacetylase inhibition. FEES Lett. 2003;542:74-78. https://doi.org/10.1016/S0014-5793(03)00350-8
  6. Kim HJ, Rowe M, Ren M, Hong JS, Chen PS, Chuang DM. Histone deacetylase inhibitors exhibit anti-inflammatory and neuroprotective effects in a rat permanent ischemic model of stroke: multiple mechanisms of action. J Pharmacol Exp Ther. 2007;321:892-901. https://doi.org/10.1124/jpet.107.120188
  7. Ren M, Leng Y. Jeong M, Leeds PR, Chuang DW. Valproic acid reduces brain damage induced by transient focal cerebral ischemia in rats: potential roles of histone deacetylase inhibition and heat shock protein induction. J Neurochem. 2004;89:1358-1367. https://doi.org/10.1111/j.1471-4159.2004.02406.x
  8. Sinn DI, Kim SJ, Chu K, Jung KH, Lee ST, Song EC, Kim JM, Park DK, Kun Lee S, Kim M, Roh JK. Valproic acid-mediated neuroprotection in intracerebral hemorrhage via histone deacetylase inhibition and transcriptional activation. Neurobiol Dis. 2007;26:464-472. https://doi.org/10.1016/j.nbd.2007.02.006
  9. Zaleska MM, Mercado MLT, Chavez J, Feuerstein Gz, Pangalos MN, Wood A. The development of stroke therapeutics: Promising mechanisms and translational challenges. Neuropharmacology. 2009;56:329-341. https://doi.org/10.1016/j.neuropharm.2008.10.006
  10. Dirrtagl U, Macleod MR. Stroke research at a road block: the streets from adversity should be paved with meta-analysis and good laboratory practice. Br J Pharmacol. 2009;157:1154-1156. https://doi.org/10.1111/j.1476-5381.2009.00211.x
  11. Qian YR. Kook JH, Hwang S, Kim DK, Kim JK. Effects of (-)-epigallocatechin-3-gallate on brain infarction and the activity change of matrix metalloproteinase-9 induced by middle cerebral artery occlusion in mice. Korean J Physiol Phamacol. 2007;11:85-88.
  12. Gidday JM, Gasche YG, Copin JC, Shah AN, Perez RS, Shapiro SD, Chan PH, Park TS. Leukocyte-derived matrix metalloproteinase-9 mediates blood-brain barrier breakdown and is proinflammatory after transient focal cerebral ischemia. Am J Physiol Heart Cire Physiol. 2005;289:H558-568. https://doi.org/10.1152/ajpheart.01275.2004
  13. Shin TK, Kang MS, Lee HY, Seo MS, Kim SG, Kim CD, Lee WS. Fluoxetine and sertraline attenuate postischemic brain injury in mice. Korean J Physiol Phamacol. 2009;13:257-263. https://doi.org/10.4196/kjpp.2009.13.3.257
  14. Mao Y, Yang G, Zhou L. Temporary and permanent focal cerebral ischemia in the mouse: assessment of cerebral blood flow, brain damage and blood-brain barrier permeability. Chin Med J (Engl). 2000;113:361-366.
  15. Lo EH, Dalkara T, Moskowitz MA. Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci. 2003;4:399-415.
  16. Klitgaard H. Levetiracetam: the preclinical profile of a new class of antiepileptic drugs? Epilepsia. 2001;42 Suppl 4:13-18. https://doi.org/10.1046/j.1528-1157.2001.0420s6013.x
  17. Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem. 2001;276:36734-36741. https://doi.org/10.1074/jbc.M101287200
  18. Hsieh J, Gage FH. Chromatin remodeling in neural development and plasticity. Curr Opin Cell Biol. 2005;17:664-671. https://doi.org/10.1016/j.ceb.2005.09.002
  19. STAIR. Recommendations for standards regarding preclinical neuroprotective and restorative drug development. Stroke. 1999;30:2752-2758. https://doi.org/10.1161/01.STR.30.12.2752

Cited by

  1. Beneficial effects of mood stabilizers lithium, valproate and lamotrigine in experimental stroke models vol.32, pp.12, 2010, https://doi.org/10.1038/aps.2011.140
  2. Effect of Sodium Valproate Administration on Brain Neprilysin Expression and Memory in Rats vol.46, pp.3, 2010, https://doi.org/10.1007/s12031-011-9644-x
  3. Clusterin in neurological disorders: Molecular perspectives and clinical relevance vol.88, pp.5, 2010, https://doi.org/10.1016/j.brainresbull.2012.05.006
  4. Therapeutic Potential of Mood Stabilizers Lithium and Valproic Acid: Beyond Bipolar Disorder vol.65, pp.1, 2013, https://doi.org/10.1124/pr.111.005512
  5. Valproic Acid Protects Motor Neuron Death by Inhibiting Oxidative Stress and Endoplasmic Reticulum Stress-Mediated Cytochrome C Release after Spinal Cord Injury vol.31, pp.6, 2014, https://doi.org/10.1089/neu.2013.3146
  6. Purpurogallin, a Natural Phenol, Attenuates High-Mobility Group Box 1 in Subarachnoid Hemorrhage Induced Vasospasm in a Rat Model vol.2014, pp.None, 2014, https://doi.org/10.1155/2014/254270
  7. Valproic Acid: A New Candidate of Therapeutic Application for the Acute Central Nervous System Injuries vol.39, pp.9, 2010, https://doi.org/10.1007/s11064-014-1241-2
  8. Valproic Acid Neuroprotection in the 6-OHDA Model of Parkinson's Disease Is Possibly Related to Its Anti-Inflammatory and HDAC Inhibitory Properties vol.2015, pp.None, 2010, https://doi.org/10.1155/2015/313702
  9. Method parameters’ impact on mortality and variability in mouse stroke experiments: a meta-analysis vol.6, pp.None, 2010, https://doi.org/10.1038/srep21086
  10. Neuroprotective Effects of Valproic Acid on Blood-Brain Barrier Disruption and Apoptosis-Related Early Brain Injury in Rats Subjected to Subarachnoid Hemorrhage Are Modulated by Heat Shock Protein 70/ vol.79, pp.2, 2010, https://doi.org/10.1227/neu.0000000000001264
  11. Valproic Acid Combined With Postcardiac Arrest Hypothermic-Targeted Temperature Management Prevents Delayed Seizures and Improves Survival in a Rat Cardiac Arrest Model vol.45, pp.11, 2017, https://doi.org/10.1097/ccm.0000000000002690
  12. Valproate Sodium Protects Blood Brain Barrier Integrity in Intracerebral Hemorrhage Mice vol.2020, pp.None, 2010, https://doi.org/10.1155/2020/8884320
  13. Blood-Spinal Cord Barrier in Spinal Cord Injury: A Review vol.38, pp.9, 2021, https://doi.org/10.1089/neu.2020.7413
  14. The Effect of Histone Deacetylase Inhibitors Panobinostat or Entinostat on Motor Recovery in Mice After Ischemic Stroke vol.23, pp.4, 2021, https://doi.org/10.1007/s12017-021-08647-1