Skip to main content
Top
Published in: Neurotoxicity Research 2/2014

01-08-2014 | Original Article

Rosiglitazone Synergizes the Neuroprotective Effects of Valproic Acid Against Quinolinic Acid-Induced Neurotoxicity in Rats: Targeting PPARγ and HDAC Pathways

Authors: Jitendriya Mishra, Tanya Chaudhary, Anil Kumar

Published in: Neurotoxicity Research | Issue 2/2014

Login to get access

Abstract

Huntington’s disease (HD) is an autosomal dominant progressive neurodegenerative disorder which affects medium spiny GABAergic neurons mainly in the striatum. Oxidative damage, neuro-inflammation, apoptosis, protein aggregation, and signaling of neurotrophic factors are some of the common cellular pathways involved in HD. Quinolinic acid (QA) causes excitotoxicity by stimulating N-methyl-d-aspartate receptors via calcium overload leading to neurodegeneration. Neuroprotective potential of peroxisome proliferator activated receptor-γ (PPARγ) agonists and histone deacetylase (HDAC) inhibitors have been well documented in experimental models of neurodegenerative disorders; however, their exact mechanisms are not clear. Therefore, present study has been designed to explore possible neuroprotective mechanism of valproic acid (VPA) and its interaction with rosiglitazone against QA induced HD-like symptoms in rats. Single bilateral intrastriatal QA (200 nmol/2 μl saline) administration significantly caused motor incoordination, memory impairment, oxidative damage, mitochondrial dysfunction (complex I, II, II and IV), cellular alterations [tumor necrosis factor-alpha (TNF-α), caspase-3, brain derived neurotrophic factor, acetylcholinesterase], and striatal neurodegeneration as compared to sham group. Treatment with rosiglitazone (5, 10 mg/kg) and VPA (100, 200 mg/kg) for 21 days significantly attenuated these behavioral, biochemical, and cellular alterations as compared to control (QA 200 nmol) group. However, VPA (100 mg/kg) treatment in combination with rosiglitazone (5 mg/kg) for 21 days synergized their neuroprotective effect, which was significant as compared to their effects per se in QA-treated animals. The present study provides an evidence of possible interplay of PPARγ agonists and HDAC inhibitors as a novel therapeutic strategy in the management of HD.
Literature
go back to reference Baitharu I, Jain V, Deep SN, Hota KB, Hota SK, Prasad D, Ilavazhagan G (2013) Withania somnifera root extract ameliorates hypobaric hypoxia induced memory impairment in rats. J Ethnopharmacol 145(2):431–441PubMedCrossRef Baitharu I, Jain V, Deep SN, Hota KB, Hota SK, Prasad D, Ilavazhagan G (2013) Withania somnifera root extract ameliorates hypobaric hypoxia induced memory impairment in rats. J Ethnopharmacol 145(2):431–441PubMedCrossRef
go back to reference Baquet ZC, Gorski JA, Jones KR (2004) Early striatal dendrite deficits followed by neuron loss with advanced age in the absence of anterograde cortical brain-derived neurotrophic factor. J Neurosci 24(17):4250–4258PubMedCrossRef Baquet ZC, Gorski JA, Jones KR (2004) Early striatal dendrite deficits followed by neuron loss with advanced age in the absence of anterograde cortical brain-derived neurotrophic factor. J Neurosci 24(17):4250–4258PubMedCrossRef
go back to reference Berman SB, Hastings TG (1999) Dopamine oxidation alters mitochondrial respiration and induces permeability transition in brain mitochondria: implications for Parkinson’s disease. J Neurochem 73(3):1127–1137PubMedCrossRef Berman SB, Hastings TG (1999) Dopamine oxidation alters mitochondrial respiration and induces permeability transition in brain mitochondria: implications for Parkinson’s disease. J Neurochem 73(3):1127–1137PubMedCrossRef
go back to reference Block F, Kunkel M, Schwarz M (1993) Quinolinic acid lesion of the striatum induces impairment in spatial learning and motor performance in rats. Neurosci Lett 149(2):126–128PubMedCrossRef Block F, Kunkel M, Schwarz M (1993) Quinolinic acid lesion of the striatum induces impairment in spatial learning and motor performance in rats. Neurosci Lett 149(2):126–128PubMedCrossRef
go back to reference Carta AR, Pisanu A, Carboni E (2011) Do PPAR-gamma agonists have a future in Parkinson’s disease therapy? Parkinsons Dis 2011:689181PubMedCentralPubMed Carta AR, Pisanu A, Carboni E (2011) Do PPAR-gamma agonists have a future in Parkinson’s disease therapy? Parkinsons Dis 2011:689181PubMedCentralPubMed
go back to reference Chen PS, Wang CC, Bortner CD, Peng GS, Wu X, Pang H, Lu RB, Gean PW, Chuang DM, Hong JS (2007) Valproic acid and other histone deacetylase inhibitors induce microglial apoptosis and attenuate lipopolysaccharide-induced dopaminergic neurotoxicity. Neuroscience 149(1):203–212PubMedCentralPubMedCrossRef Chen PS, Wang CC, Bortner CD, Peng GS, Wu X, Pang H, Lu RB, Gean PW, Chuang DM, Hong JS (2007) Valproic acid and other histone deacetylase inhibitors induce microglial apoptosis and attenuate lipopolysaccharide-induced dopaminergic neurotoxicity. Neuroscience 149(1):203–212PubMedCentralPubMedCrossRef
go back to reference Chiang MC, Chern Y, Huang RN (2012) PPARgamma rescue of the mitochondrial dysfunction in Huntington’s disease. Neurobiol Dis 45(1):322–328PubMedCrossRef Chiang MC, Chern Y, Huang RN (2012) PPARgamma rescue of the mitochondrial dysfunction in Huntington’s disease. Neurobiol Dis 45(1):322–328PubMedCrossRef
go back to reference Cristiano L, Bernardo A, Ceru MP (2001) Peroxisome proliferator-activated receptors (PPARs) and peroxisomes in rat cortical and cerebellar astrocytes. J Neurocytol 30(8):671–683PubMedCrossRef Cristiano L, Bernardo A, Ceru MP (2001) Peroxisome proliferator-activated receptors (PPARs) and peroxisomes in rat cortical and cerebellar astrocytes. J Neurocytol 30(8):671–683PubMedCrossRef
go back to reference Curtis CL, Ross GJ III, Hyde EA, Szymanski RE, Hull JS, Dunbar GL (1992) Intrastriatal injections of quinolinic acid cause spatial learning deficits in rats. Soc Neurosci Abstr 18:1602 Curtis CL, Ross GJ III, Hyde EA, Szymanski RE, Hull JS, Dunbar GL (1992) Intrastriatal injections of quinolinic acid cause spatial learning deficits in rats. Soc Neurosci Abstr 18:1602
go back to reference Dash PK, Orsi SA, Zhang M, Grill RJ, Pati S, Zhao J, Moore AN (2010) Valproate administered after traumatic brain injury provides neuroprotection and improves cognitive function in rats. PLoS ONE 5(6):e11383PubMedCentralPubMedCrossRef Dash PK, Orsi SA, Zhang M, Grill RJ, Pati S, Zhao J, Moore AN (2010) Valproate administered after traumatic brain injury provides neuroprotection and improves cognitive function in rats. PLoS ONE 5(6):e11383PubMedCentralPubMedCrossRef
go back to reference Ding G, Fu M, Qin Q, Lewis W, Kim HW, Fukai T, Bacanamwo M, Chen YE, Schneider MD, Mangelsdorf DJ, Evans RM, Yang Q (2007) Cardiac peroxisome proliferator-activated receptor gamma is essential in protecting cardiomyocytes from oxidative damage. Cardiovasc Res 76(2):269–279PubMedCrossRef Ding G, Fu M, Qin Q, Lewis W, Kim HW, Fukai T, Bacanamwo M, Chen YE, Schneider MD, Mangelsdorf DJ, Evans RM, Yang Q (2007) Cardiac peroxisome proliferator-activated receptor gamma is essential in protecting cardiomyocytes from oxidative damage. Cardiovasc Res 76(2):269–279PubMedCrossRef
go back to reference Drew PD, Xu J, Storer PD, Chavis JA, Racke MK (2006) Peroxisome proliferator-activated receptor agonist regulation of glial activation: relevance to CNS inflammatory disorders. Neurochem Int 49(2):183–189PubMedCrossRef Drew PD, Xu J, Storer PD, Chavis JA, Racke MK (2006) Peroxisome proliferator-activated receptor agonist regulation of glial activation: relevance to CNS inflammatory disorders. Neurochem Int 49(2):183–189PubMedCrossRef
go back to reference Ellman GL, Courtney KD, Andres V Jr, Feather-Stone RM (1961) A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 7:88–95PubMedCrossRef Ellman GL, Courtney KD, Andres V Jr, Feather-Stone RM (1961) A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 7:88–95PubMedCrossRef
go back to reference Escribano L, Simon AM, Perez-Mediavilla A, Salazar-Colocho P, Del Rio J, Frechilla D (2009) Rosiglitazone reverses memory decline and hippocampal glucocorticoid receptor down-regulation in an Alzheimer’s disease mouse model. Biochem Biophys Res Commun 379(2):406–410PubMedCrossRef Escribano L, Simon AM, Perez-Mediavilla A, Salazar-Colocho P, Del Rio J, Frechilla D (2009) Rosiglitazone reverses memory decline and hippocampal glucocorticoid receptor down-regulation in an Alzheimer’s disease mouse model. Biochem Biophys Res Commun 379(2):406–410PubMedCrossRef
go back to reference Escribano L, Simon AM, Gimeno E, Cuadrado-Tejedor M, de Maturana RL, Garcia-Osta A, Ricobaraza A, Perez-Mediavilla A, Del Rio J, Frechilla D (2010) Rosiglitazone rescues memory impairment in Alzheimer’s transgenic mice: mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology 35(7):1593–1604PubMedCentralPubMedCrossRef Escribano L, Simon AM, Gimeno E, Cuadrado-Tejedor M, de Maturana RL, Garcia-Osta A, Ricobaraza A, Perez-Mediavilla A, Del Rio J, Frechilla D (2010) Rosiglitazone rescues memory impairment in Alzheimer’s transgenic mice: mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology 35(7):1593–1604PubMedCentralPubMedCrossRef
go back to reference Ferrante RJ, Kowall NW, Cipolloni PB, Storey E, Beal MF (1993) Excitotoxin lesions in primates as a model for Huntington’s disease: histopathologic and neurochemical characterization. Exp Neurol 119(1):46–71PubMedCrossRef Ferrante RJ, Kowall NW, Cipolloni PB, Storey E, Beal MF (1993) Excitotoxin lesions in primates as a model for Huntington’s disease: histopathologic and neurochemical characterization. Exp Neurol 119(1):46–71PubMedCrossRef
go back to reference Ferrante RJ, Kubilus JK, Lee J, Ryu H, Beesen A, Zucker B, Smith K, Kowall NW, Ratan RR, Luthi-Carter R, Hersch SM (2003) Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington’s disease mice. J Neurosci Methods 23(28):9418–9427 Ferrante RJ, Kubilus JK, Lee J, Ryu H, Beesen A, Zucker B, Smith K, Kowall NW, Ratan RR, Luthi-Carter R, Hersch SM (2003) Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington’s disease mice. J Neurosci Methods 23(28):9418–9427
go back to reference Ferrer I, Goutan E, Marin C, Rey MJ, Ribalta T (2000) Brain-derived neurotrophic factor in Huntington disease. Brain Res 866(1–2):257–261PubMedCrossRef Ferrer I, Goutan E, Marin C, Rey MJ, Ribalta T (2000) Brain-derived neurotrophic factor in Huntington disease. Brain Res 866(1–2):257–261PubMedCrossRef
go back to reference Furtado JC, Mazurek MF (1996) Behavioral characterization of quinolinate-induced lesions of the medial striatum: relevance for Huntington’s disease. Exp Neurol 138(1):158–168PubMedCrossRef Furtado JC, Mazurek MF (1996) Behavioral characterization of quinolinate-induced lesions of the medial striatum: relevance for Huntington’s disease. Exp Neurol 138(1):158–168PubMedCrossRef
go back to reference Ganzella M, Jardim FM, Boeck CR, Vendite D (2006) Time course of oxidative events in the hippocampus following intracerebroventricular infusion of quinolinic acid in mice. Neurosci Res 55(4):397–402PubMedCrossRef Ganzella M, Jardim FM, Boeck CR, Vendite D (2006) Time course of oxidative events in the hippocampus following intracerebroventricular infusion of quinolinic acid in mice. Neurosci Res 55(4):397–402PubMedCrossRef
go back to reference Gardian G, Vecsei L (2004) Huntington’s disease: pathomechanism and therapeutic perspectives. J Neural Transm 111(10–11):1485–1494PubMedCrossRef Gardian G, Vecsei L (2004) Huntington’s disease: pathomechanism and therapeutic perspectives. J Neural Transm 111(10–11):1485–1494PubMedCrossRef
go back to reference Gaur V, Kumar A (2011) Neuroprotective potentials of candesartan, atorvastatin and their combination against stroke induced motor dysfunction. Inflammopharmacology 19(4):205–214PubMedCrossRef Gaur V, Kumar A (2011) Neuroprotective potentials of candesartan, atorvastatin and their combination against stroke induced motor dysfunction. Inflammopharmacology 19(4):205–214PubMedCrossRef
go back to reference Gauthier LR, Charrin BC, Borrell-Pages M, Dompierre JP, Rangone H, Cordelieres FP, De Mey J, MacDonald ME, Lessmann V, Humbert S, Saudou F (2004) Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 118(1):127–138PubMedCrossRef Gauthier LR, Charrin BC, Borrell-Pages M, Dompierre JP, Rangone H, Cordelieres FP, De Mey J, MacDonald ME, Lessmann V, Humbert S, Saudou F (2004) Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 118(1):127–138PubMedCrossRef
go back to reference Gornall AG, Bardawill CJ, David MM (1949) Determination of serum proteins by means of the biuret reaction. J Biol Chem 177(2):751–766PubMed Gornall AG, Bardawill CJ, David MM (1949) Determination of serum proteins by means of the biuret reaction. J Biol Chem 177(2):751–766PubMed
go back to reference Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR (1982) Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem 126(1):131–138PubMedCrossRef Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR (1982) Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem 126(1):131–138PubMedCrossRef
go back to reference Hashimoto R, Hough C, Nakazawa T, Yamamoto T, Chuang DM (2002) Lithium protection against glutamate excitotoxicity in rat cerebral cortical neurons: involvement of NMDA receptor inhibition possibly by decreasing NR2B tyrosine phosphorylation. J Neurochem 80(4):589–597PubMedCrossRef Hashimoto R, Hough C, Nakazawa T, Yamamoto T, Chuang DM (2002) Lithium protection against glutamate excitotoxicity in rat cerebral cortical neurons: involvement of NMDA receptor inhibition possibly by decreasing NR2B tyrosine phosphorylation. J Neurochem 80(4):589–597PubMedCrossRef
go back to reference Heneka MT, Landreth GE, Hull M (2007) Drug insight: effects mediated by peroxisome proliferator-activated receptor-gamma in CNS disorders. Nat Clin Pract Neurol 3(9):496–504PubMedCrossRef Heneka MT, Landreth GE, Hull M (2007) Drug insight: effects mediated by peroxisome proliferator-activated receptor-gamma in CNS disorders. Nat Clin Pract Neurol 3(9):496–504PubMedCrossRef
go back to reference Hwang J, Kleinhenz DJ, Lassegue B, Griendling KK, Dikalov S, Hart CM (2005) Peroxisome proliferator-activated receptor-gamma ligands regulate endothelial membrane superoxide production. Am J Physiol Cell Physiol 288(4):C899–C905PubMedCrossRef Hwang J, Kleinhenz DJ, Lassegue B, Griendling KK, Dikalov S, Hart CM (2005) Peroxisome proliferator-activated receptor-gamma ligands regulate endothelial membrane superoxide production. Am J Physiol Cell Physiol 288(4):C899–C905PubMedCrossRef
go back to reference Jeong MR, Hashimoto R, Senatorov VV, Fujimaki K, Ren M, Lee MS, Chuang DM (2003) Valproic acid, a mood stabilizer and anticonvulsant, protects rat cerebral cortical neurons from spontaneous cell death: a role of histone deacetylase inhibition. FEBS Lett 542(1–3):74–78PubMedCrossRef Jeong MR, Hashimoto R, Senatorov VV, Fujimaki K, Ren M, Lee MS, Chuang DM (2003) Valproic acid, a mood stabilizer and anticonvulsant, protects rat cerebral cortical neurons from spontaneous cell death: a role of histone deacetylase inhibition. FEBS Lett 542(1–3):74–78PubMedCrossRef
go back to reference Jiang M, Peng Q, Liu X, Jin J, Hou Z, Zhang J, Mori S, Ross CA, Ye K, Duan W (2013) Small-molecule TrkB receptor agonists improve motor function and extend survival in a mouse model of Huntington’s disease. Hum Mol Genet 22(12):2462–2470PubMedCentralPubMedCrossRef Jiang M, Peng Q, Liu X, Jin J, Hou Z, Zhang J, Mori S, Ross CA, Ye K, Duan W (2013) Small-molecule TrkB receptor agonists improve motor function and extend survival in a mouse model of Huntington’s disease. Hum Mol Genet 22(12):2462–2470PubMedCentralPubMedCrossRef
go back to reference Jin J, Albertz J, Guo Z, Peng Q, Rudow G, Troncoso JC, Ross CA, Duan W (2013) Neuroprotective effects of PPAR-γ agonist rosiglitazone in N171-82Q mouse model of Huntington’s disease. J Neurochem 125(3):410–419PubMedCentralPubMedCrossRef Jin J, Albertz J, Guo Z, Peng Q, Rudow G, Troncoso JC, Ross CA, Duan W (2013) Neuroprotective effects of PPAR-γ agonist rosiglitazone in N171-82Q mouse model of Huntington’s disease. J Neurochem 125(3):410–419PubMedCentralPubMedCrossRef
go back to reference Kalonia H, Kumar P, Kumar A, Nehru B (2009) Effect of caffeic acid and rofecoxib and their combination against intrastriatal quinolinic acid induced oxidative damage, mitochondrial and histological alterations in rats. Inflammopharmacology 17(4):211–219PubMedCrossRef Kalonia H, Kumar P, Kumar A, Nehru B (2009) Effect of caffeic acid and rofecoxib and their combination against intrastriatal quinolinic acid induced oxidative damage, mitochondrial and histological alterations in rats. Inflammopharmacology 17(4):211–219PubMedCrossRef
go back to reference Kalonia H, Kumar P, Kumar A (2010a) Pioglitazone ameliorates behavioral, biochemical and cellular alterations in quinolinic acid induced neurotoxicity: possible role of peroxisome proliferator activated receptor-γ (PPARγ) in Huntington’s disease. Pharmacol Biochem Behav 96(2):115–124PubMedCrossRef Kalonia H, Kumar P, Kumar A (2010a) Pioglitazone ameliorates behavioral, biochemical and cellular alterations in quinolinic acid induced neurotoxicity: possible role of peroxisome proliferator activated receptor-γ (PPARγ) in Huntington’s disease. Pharmacol Biochem Behav 96(2):115–124PubMedCrossRef
go back to reference Kalonia H, Kumar P, Kumar A, Nehru B (2010b) Protective effect of montelukast against quinolinic acid/malonic acid induced neurotoxicity: possible behavioral, biochemical, mitochondrial and tumor necrosis factor-α level alterations in rats. Neuroscience 171(1):284–299PubMedCrossRef Kalonia H, Kumar P, Kumar A, Nehru B (2010b) Protective effect of montelukast against quinolinic acid/malonic acid induced neurotoxicity: possible behavioral, biochemical, mitochondrial and tumor necrosis factor-α level alterations in rats. Neuroscience 171(1):284–299PubMedCrossRef
go back to reference Kalonia H, Kumar P, Kumar A (2011) Attenuation of proinflammatory cytokines and apoptotic process by verapamil and diltiazem against quinolinic acid induced Huntington like alterations in rats. Brain Res 1372:115–126PubMedCrossRef Kalonia H, Kumar P, Kumar A (2011) Attenuation of proinflammatory cytokines and apoptotic process by verapamil and diltiazem against quinolinic acid induced Huntington like alterations in rats. Brain Res 1372:115–126PubMedCrossRef
go back to reference Kalonia H, Mishra J, Kumar A (2012) Targeting neuro-inflammatory cytokines and oxidative stress by minocycline attenuates quinolinic-acid-induced Huntington’s disease-like symptoms in rats. Neurotox Res 22(4):310–320PubMedCrossRef Kalonia H, Mishra J, Kumar A (2012) Targeting neuro-inflammatory cytokines and oxidative stress by minocycline attenuates quinolinic-acid-induced Huntington’s disease-like symptoms in rats. Neurotox Res 22(4):310–320PubMedCrossRef
go back to reference Kiaei M, Kipiani K, Chen J, Calingasan NY, Beal MF (2005) Peroxisome proliferator–activated receptor-gamma agonist extends survival in transgenic mouse model of amyotrophic lateral sclerosis. Exp Neurol 191(2):331–336PubMedCrossRef Kiaei M, Kipiani K, Chen J, Calingasan NY, Beal MF (2005) Peroxisome proliferator–activated receptor-gamma agonist extends survival in transgenic mouse model of amyotrophic lateral sclerosis. Exp Neurol 191(2):331–336PubMedCrossRef
go back to reference Kim J-H, Kim S, Yoon I-S, Lee J-H, Jang B-J, Jeong SM, Lee J-H, Lee B-H, Han J-S, Oh S, Kim H-C, Park TK, Rhim H, Nah S-Y (2005) Protective effects of ginseng saponins on 3-nitropropionic acid-induced striatal degeneration in rats. Neuropharmacology 48(5):743–756PubMedCrossRef Kim J-H, Kim S, Yoon I-S, Lee J-H, Jang B-J, Jeong SM, Lee J-H, Lee B-H, Han J-S, Oh S, Kim H-C, Park TK, Rhim H, Nah S-Y (2005) Protective effects of ginseng saponins on 3-nitropropionic acid-induced striatal degeneration in rats. Neuropharmacology 48(5):743–756PubMedCrossRef
go back to reference Kim HJ, Rowe M, Ren M, Hong JS, Chen PS, Chuang DM (2007) 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 321(3):892–901PubMedCrossRef Kim HJ, Rowe M, Ren M, Hong JS, Chen PS, Chuang DM (2007) 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 321(3):892–901PubMedCrossRef
go back to reference King TE (1967) Preparation of succinate dehydrogenase and reconstitution of succinate oxidase. In: Estabrook RW, Pullman ME (eds) Methods in enzymology, vol 10. Academic Press, New York, pp 322–331 King TE (1967) Preparation of succinate dehydrogenase and reconstitution of succinate oxidase. In: Estabrook RW, Pullman ME (eds) Methods in enzymology, vol 10. Academic Press, New York, pp 322–331
go back to reference King TE, Howard RL (1967) Preparations and properties of soluble NADH dehydrogenases from cardiac muscle. In: Estabrook RW, Pullman ME (eds) Methods in enzymololgy, vol 10. Academic Press, New York, pp 275–294 King TE, Howard RL (1967) Preparations and properties of soluble NADH dehydrogenases from cardiac muscle. In: Estabrook RW, Pullman ME (eds) Methods in enzymololgy, vol 10. Academic Press, New York, pp 275–294
go back to reference Kono Y (1978) Generation of superoxide radical during autoxidation of hydroxylamine and an assay for superoxide dismutase. Archiv Biochem Biophy 186(1):189–195CrossRef Kono Y (1978) Generation of superoxide radical during autoxidation of hydroxylamine and an assay for superoxide dismutase. Archiv Biochem Biophy 186(1):189–195CrossRef
go back to reference Kukreja RC, Kontos HA, Hess ML, Ellis EF (1986) PGH synthase and lipoxygenase generate superoxide in the presence of NADH or NADPH. Circ Res 59(6):612–619PubMedCrossRef Kukreja RC, Kontos HA, Hess ML, Ellis EF (1986) PGH synthase and lipoxygenase generate superoxide in the presence of NADH or NADPH. Circ Res 59(6):612–619PubMedCrossRef
go back to reference Kumar A, Prakash A, Pahwa D, Mishra J (2012a) Montelukast potentiates the protective effect of rofecoxib against kainic acid-induced cognitive dysfunction in rats. Pharmacol Biochem Behav 103(1):43–52PubMedCrossRef Kumar A, Prakash A, Pahwa D, Mishra J (2012a) Montelukast potentiates the protective effect of rofecoxib against kainic acid-induced cognitive dysfunction in rats. Pharmacol Biochem Behav 103(1):43–52PubMedCrossRef
go back to reference Kumar A, Sharma N, Gupta A, Kalonia H, Mishra J (2012b) Neuroprotective potential of atorvastatin and simvastatin (HMG-CoA reductase inhibitors) against 6-hydroxydopamine (6-OHDA) induced Parkinson-like symptoms. Brain Res 1471:13–22PubMedCrossRef Kumar A, Sharma N, Gupta A, Kalonia H, Mishra J (2012b) Neuroprotective potential of atorvastatin and simvastatin (HMG-CoA reductase inhibitors) against 6-hydroxydopamine (6-OHDA) induced Parkinson-like symptoms. Brain Res 1471:13–22PubMedCrossRef
go back to reference Kumar A, Sharma N, Mishra J, Kalonia H (2013) Synergistical neuroprotection of rofecoxib and statins against malonic acid induced Huntington’s disease like symptoms and related cognitive dysfunction in rats. Eur J Pharmacol 709(1–3):1–12PubMedCrossRef Kumar A, Sharma N, Mishra J, Kalonia H (2013) Synergistical neuroprotection of rofecoxib and statins against malonic acid induced Huntington’s disease like symptoms and related cognitive dysfunction in rats. Eur J Pharmacol 709(1–3):1–12PubMedCrossRef
go back to reference Kwan P, Brodie MJ (2001) Neuropsychological effects of epilepsy and antiepileptic drugs. Lancet 357(9251):216–222PubMedCrossRef Kwan P, Brodie MJ (2001) Neuropsychological effects of epilepsy and antiepileptic drugs. Lancet 357(9251):216–222PubMedCrossRef
go back to reference Lampen A, Carlberg C, Nau H (2001) Peroxisome proliferator-activated receptor delta is a specific sensor for teratogenic valproic acid derivatives. Eur J Pharmacol 431(1):25–33PubMedCrossRef Lampen A, Carlberg C, Nau H (2001) Peroxisome proliferator-activated receptor delta is a specific sensor for teratogenic valproic acid derivatives. Eur J Pharmacol 431(1):25–33PubMedCrossRef
go back to reference Langley B, Gensert JM, Beal MF, Ratan RR (2005) Remodeling chromatin and stress resistance in the central nervous system: histone deacetylase inhibitors as novel and broadly effective neuroprotective agents. Curr Drug Targets CNS Neurol Disord 4(1):41–50PubMedCrossRef Langley B, Gensert JM, Beal MF, Ratan RR (2005) Remodeling chromatin and stress resistance in the central nervous system: histone deacetylase inhibitors as novel and broadly effective neuroprotective agents. Curr Drug Targets CNS Neurol Disord 4(1):41–50PubMedCrossRef
go back to reference Lee EY, Lee JE, Park JH, Shin IC, Koh HC (2012) Rosiglitazone, a PPAR-gamma agonist, protects against striatal dopaminergic neurodegeneration induced by 6-OHDA lesions in the substantia nigra of rats. Toxicol Lett 213(3):332–344PubMedCrossRef Lee EY, Lee JE, Park JH, Shin IC, Koh HC (2012) Rosiglitazone, a PPAR-gamma agonist, protects against striatal dopaminergic neurodegeneration induced by 6-OHDA lesions in the substantia nigra of rats. Toxicol Lett 213(3):332–344PubMedCrossRef
go back to reference Liu Y, Peterson DA, Kimura H, Schubert D (1997) Mechanism of cellular 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction. J Neurochem 69(2):581–593PubMedCrossRef Liu Y, Peterson DA, Kimura H, Schubert D (1997) Mechanism of cellular 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction. J Neurochem 69(2):581–593PubMedCrossRef
go back to reference Luck H (1965) Catalase. In: Hans UB (ed) Methods of enzymatic analysis, 2nd edn. Academic Press, New York, pp 885–894CrossRef Luck H (1965) Catalase. In: Hans UB (ed) Methods of enzymatic analysis, 2nd edn. Academic Press, New York, pp 885–894CrossRef
go back to reference Mark RJ, Ashford JW, Goodman Y, Mattson MP (1995) Anticonvulsants attenuate amyloid beta-peptide neurotoxicity, Ca2+ deregulation, and cytoskeletal pathology. Neurobiol Aging 16:187–198PubMedCrossRef Mark RJ, Ashford JW, Goodman Y, Mattson MP (1995) Anticonvulsants attenuate amyloid beta-peptide neurotoxicity, Ca2+ deregulation, and cytoskeletal pathology. Neurobiol Aging 16:187–198PubMedCrossRef
go back to reference Morris R (1984) Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods 11(1):47–60PubMedCrossRef Morris R (1984) Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods 11(1):47–60PubMedCrossRef
go back to reference Park SW, Yi JH, Miranpuri G, Satriotomo I, Bowen K, Resnick DK, Vemuganti R (2007) Thiazolidinedione class of peroxisome proliferator-activated receptor gamma agonists prevents neuronal damage, motor dysfunction, myelin loss, neuropathic pain, and inflammation after spinal cord injury in adult rats. J Pharmacol Exp Ther 320:1002–1012PubMedCrossRef Park SW, Yi JH, Miranpuri G, Satriotomo I, Bowen K, Resnick DK, Vemuganti R (2007) Thiazolidinedione class of peroxisome proliferator-activated receptor gamma agonists prevents neuronal damage, motor dysfunction, myelin loss, neuropathic pain, and inflammation after spinal cord injury in adult rats. J Pharmacol Exp Ther 320:1002–1012PubMedCrossRef
go back to reference Paxinos G, Watson C (2007) The rat brain in stereotaxic coordinates, 6th edn. Academic Press, San Diego Paxinos G, Watson C (2007) The rat brain in stereotaxic coordinates, 6th edn. Academic Press, San Diego
go back to reference Pedersen WA, McMillan PJ, Kulstad JJ, Leverenz JB, Craft S, Haynatzki GR (2006) Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Exp Neurol 199:265–273PubMedCrossRef Pedersen WA, McMillan PJ, Kulstad JJ, Leverenz JB, Craft S, Haynatzki GR (2006) Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Exp Neurol 199:265–273PubMedCrossRef
go back to reference Pemberton LA, Kerr SJ, Smythe G, Brew BJ (1997) Quinolinic acid production by macrophages stimulated with IFN-gamma, TNF-alpha, and IFN-alpha. J Interf Cytokine Res 17:589–595CrossRef Pemberton LA, Kerr SJ, Smythe G, Brew BJ (1997) Quinolinic acid production by macrophages stimulated with IFN-gamma, TNF-alpha, and IFN-alpha. J Interf Cytokine Res 17:589–595CrossRef
go back to reference Peng GS, Li G, Tzeng NS, Chen PS, Chuang DM, Hsu YD, Yang S, Hong JS (2005) Valproate pretreatment protects dopaminergic neurons from LPS-induced neurotoxicity in rat primary midbrain cultures: role of microglia. Brain Res 134(1):162–169CrossRef Peng GS, Li G, Tzeng NS, Chen PS, Chuang DM, Hsu YD, Yang S, Hong JS (2005) Valproate pretreatment protects dopaminergic neurons from LPS-induced neurotoxicity in rat primary midbrain cultures: role of microglia. Brain Res 134(1):162–169CrossRef
go back to reference Perez-De La Cruz V, Carrillo-Mora P, Santamaria A (2012) Quinolinic acid, an endogenous molecule combining excitotoxicity, oxidative stress and other toxic mechanisms. Int J Tryptophan Res 5:1–8PubMedCentralPubMed Perez-De La Cruz V, Carrillo-Mora P, Santamaria A (2012) Quinolinic acid, an endogenous molecule combining excitotoxicity, oxidative stress and other toxic mechanisms. Int J Tryptophan Res 5:1–8PubMedCentralPubMed
go back to reference Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS (2001) Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem 276:36734–36741PubMedCrossRef Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS (2001) Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem 276:36734–36741PubMedCrossRef
go back to reference Poeggeler B, Rassoulpour A, Wu HQ, Guidetti P, Roberts RC, Schwarcz R (2007) Dopamine receptor activation reveals a novel, kynurenate-sensitive component of striatal N-methyl-d-aspartate neurotoxicity. Neuroscience 148:188–197PubMedCentralPubMedCrossRef Poeggeler B, Rassoulpour A, Wu HQ, Guidetti P, Roberts RC, Schwarcz R (2007) Dopamine receptor activation reveals a novel, kynurenate-sensitive component of striatal N-methyl-d-aspartate neurotoxicity. Neuroscience 148:188–197PubMedCentralPubMedCrossRef
go back to reference Ponchaut S, Draye JP, Veitch K, Van Hoof F (1991) Influence of chronic administration of valproate on ultrastructure and enzyme content of peroxisomes in rat liver and kidney. Oxidation of valproate by liver peroxisomes. Biochem Pharmacol 41:1419–1428PubMedCrossRef Ponchaut S, Draye JP, Veitch K, Van Hoof F (1991) Influence of chronic administration of valproate on ultrastructure and enzyme content of peroxisomes in rat liver and kidney. Oxidation of valproate by liver peroxisomes. Biochem Pharmacol 41:1419–1428PubMedCrossRef
go back to reference Quintanilla RA, Jin YN, Fuenzalida K, Bronfman M, Johnson GV (2008) Rosiglitazone treatment prevents mitochondrial dysfunction in mutant huntingtin-expressing cells: possible role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in the pathogenesis of Huntington disease. J Biol Chem 283:25628–25637PubMedCentralPubMedCrossRef Quintanilla RA, Jin YN, Fuenzalida K, Bronfman M, Johnson GV (2008) Rosiglitazone treatment prevents mitochondrial dysfunction in mutant huntingtin-expressing cells: possible role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in the pathogenesis of Huntington disease. J Biol Chem 283:25628–25637PubMedCentralPubMedCrossRef
go back to reference Reiner A, Wang HB, Del Mar N, Sakata K, Yoo W, Deng YP (2012) BDNF may play a differential role in the protective effect of the mGluR2/3 agonist LY379268 on striatal projection neurons in R6/2 Huntington’s disease mice. Brain Res 1473:161–172PubMedCentralPubMedCrossRef Reiner A, Wang HB, Del Mar N, Sakata K, Yoo W, Deng YP (2012) BDNF may play a differential role in the protective effect of the mGluR2/3 agonist LY379268 on striatal projection neurons in R6/2 Huntington’s disease mice. Brain Res 1473:161–172PubMedCentralPubMedCrossRef
go back to reference Rossato JI, Zeni G, Mello CF, Rubin MA, Rocha JB (2002) Ebselen blocks the quinolinic acid-induced production of thiobarbituric acid reactive species but does not prevent the behavioral alterations produced by intra-striatal quinolinic acid administration in the rat. Neurosci Lett 318:137–140PubMedCrossRef Rossato JI, Zeni G, Mello CF, Rubin MA, Rocha JB (2002) Ebselen blocks the quinolinic acid-induced production of thiobarbituric acid reactive species but does not prevent the behavioral alterations produced by intra-striatal quinolinic acid administration in the rat. Neurosci Lett 318:137–140PubMedCrossRef
go back to reference Sadri-Vakili G, Cha JH (2006) Histone deacetylase inhibitors: a novel therapeutic approach to Huntington’s disease (complex mechanism of neuronal death). Curr Alzheimer Res 3(4):403–408PubMedCrossRef Sadri-Vakili G, Cha JH (2006) Histone deacetylase inhibitors: a novel therapeutic approach to Huntington’s disease (complex mechanism of neuronal death). Curr Alzheimer Res 3(4):403–408PubMedCrossRef
go back to reference Samadi P, Boutet A, Rymar VV, Rawal K, Maheux J, Kvann JC, Tomaszewski M, Beaubien F, Cloutier JF, Levesque D, Sadikot AF (2013) Relationship between BDNF expression in major striatal afferents, striatum morphology and motor behavior in the R6/2 mouse model of Huntington’s disease. Genes Brain Behav 12(1):108–124PubMedCrossRef Samadi P, Boutet A, Rymar VV, Rawal K, Maheux J, Kvann JC, Tomaszewski M, Beaubien F, Cloutier JF, Levesque D, Sadikot AF (2013) Relationship between BDNF expression in major striatal afferents, striatum morphology and motor behavior in the R6/2 mouse model of Huntington’s disease. Genes Brain Behav 12(1):108–124PubMedCrossRef
go back to reference Sanberg PR, Calderon SF, Giordano M, Tew JM, Norman AB (1989) The quinolinic acid model of Huntington’s disease: locomotor abnormalities. Exp Neurol 105(1):45–53PubMedCrossRef Sanberg PR, Calderon SF, Giordano M, Tew JM, Norman AB (1989) The quinolinic acid model of Huntington’s disease: locomotor abnormalities. Exp Neurol 105(1):45–53PubMedCrossRef
go back to reference Santamaria A, Rios C (1993) MK-801, an N-methyl-d-aspartate receptor antagonist, blocks quinolinic acid-induced lipid peroxidation in rat corpus striatum. Neurosci Lett 159(1–2):51–54PubMedCrossRef Santamaria A, Rios C (1993) MK-801, an N-methyl-d-aspartate receptor antagonist, blocks quinolinic acid-induced lipid peroxidation in rat corpus striatum. Neurosci Lett 159(1–2):51–54PubMedCrossRef
go back to reference Schwarcz R, Whetsell WO Jr, Mangano RM (1983) Quinolinic acid: an endogenous metabolite that produces axon-sparing lesions in rat brain. Science 219(4582):316–318PubMedCrossRef Schwarcz R, Whetsell WO Jr, Mangano RM (1983) Quinolinic acid: an endogenous metabolite that produces axon-sparing lesions in rat brain. Science 219(4582):316–318PubMedCrossRef
go back to reference Schwarcz R, Guidetti P, Sathyasaikumar KV, Muchowski PJ (2010) Of mice, rats and men: revisiting the quinolinic acid hypothesis of Huntington’s disease. Prog Neurobiol 90(2):230–245PubMedCentralPubMedCrossRef Schwarcz R, Guidetti P, Sathyasaikumar KV, Muchowski PJ (2010) Of mice, rats and men: revisiting the quinolinic acid hypothesis of Huntington’s disease. Prog Neurobiol 90(2):230–245PubMedCentralPubMedCrossRef
go back to reference Shear DA, Dong J, Gundy CD, Haik-Creguer KL, Dunbar GL (1998) Comparison of intrastriatal injections of quinolinic acid and 3-nitropropionic acid for use in animal models of Huntington’s disease. Prog Neuro-psychopharmacol Biol Psychiatry 22(7):1217–1240CrossRef Shear DA, Dong J, Gundy CD, Haik-Creguer KL, Dunbar GL (1998) Comparison of intrastriatal injections of quinolinic acid and 3-nitropropionic acid for use in animal models of Huntington’s disease. Prog Neuro-psychopharmacol Biol Psychiatry 22(7):1217–1240CrossRef
go back to reference Shehadeh J, Fernandes HB, Mullins MMZ, Graham RK, Leavitt BR, Hayden MR, Raymond LA (2006) Striatal neuronal apoptosis is preferentially enhanced by NMDA receptor activation in YAC transgenic mouse model of Huntington disease. Neurobiol Dis 21(2):392–403PubMedCrossRef Shehadeh J, Fernandes HB, Mullins MMZ, Graham RK, Leavitt BR, Hayden MR, Raymond LA (2006) Striatal neuronal apoptosis is preferentially enhanced by NMDA receptor activation in YAC transgenic mouse model of Huntington disease. Neurobiol Dis 21(2):392–403PubMedCrossRef
go back to reference Simonian NA, Getz RL, Leveque JC, Konradi C, Coyle JT (1996) Kainic acid induces apoptosis in neurons. Neuroscience 75:1047–1055PubMedCrossRef Simonian NA, Getz RL, Leveque JC, Konradi C, Coyle JT (1996) Kainic acid induces apoptosis in neurons. Neuroscience 75:1047–1055PubMedCrossRef
go back to reference Sottocasa GL, Kuylenstierna B, Ernster L, Bergstrand A (1967) An electron-transport system associated with the outer membrane of liver mitochondria. A biochemical and morphological study. J Cell Biol 32:415–438PubMedCentralPubMedCrossRef Sottocasa GL, Kuylenstierna B, Ernster L, Bergstrand A (1967) An electron-transport system associated with the outer membrane of liver mitochondria. A biochemical and morphological study. J Cell Biol 32:415–438PubMedCentralPubMedCrossRef
go back to reference Sudha S, Lakshmana MK, Pradhan N (1995) Chronic phenytoin induced impairment of learning and memory with associated changes in brain acetylcholine esterase activity and monoamine levels. Pharmacol Biochem Behav 52(1):119–124PubMedCrossRef Sudha S, Lakshmana MK, Pradhan N (1995) Chronic phenytoin induced impairment of learning and memory with associated changes in brain acetylcholine esterase activity and monoamine levels. Pharmacol Biochem Behav 52(1):119–124PubMedCrossRef
go back to reference Tuzcu M, Baydas G (2006) Effect of melatonin and vitamin E on diabetes-induced learning and memory impairment in rats. Eur J Pharmacol 537(1–3):106–110PubMedCrossRef Tuzcu M, Baydas G (2006) Effect of melatonin and vitamin E on diabetes-induced learning and memory impairment in rats. Eur J Pharmacol 537(1–3):106–110PubMedCrossRef
go back to reference Valvassori SS, Rezin GT, Ferreira CL, Moretti M, Goncalves CL, Cardoso MR, Streck EL, Kapczinski F, Quevedo J (2010) Effects of mood stabilizers on mitochondrial respiratory chain activity in brain of rats treated with d-amphetamine. J Psychiatr Res 44(14):903–909PubMedCrossRef Valvassori SS, Rezin GT, Ferreira CL, Moretti M, Goncalves CL, Cardoso MR, Streck EL, Kapczinski F, Quevedo J (2010) Effects of mood stabilizers on mitochondrial respiratory chain activity in brain of rats treated with d-amphetamine. J Psychiatr Res 44(14):903–909PubMedCrossRef
go back to reference Van den Branden C, Roels F (1985) Peroxisomal beta-oxidation and sodium valproate. Biochem Pharmacol 34(12):2147–2149PubMedCrossRef Van den Branden C, Roels F (1985) Peroxisomal beta-oxidation and sodium valproate. Biochem Pharmacol 34(12):2147–2149PubMedCrossRef
go back to reference Verma R, Mishra V, Gupta K, Sasmal D, Raghubir R (2011) Neuroprotection by rosiglitazone in transient focal cerebral ischemia might not be mediated by glutamate transporter-1. J Neurosci Res 89:1849–1858PubMedCrossRef Verma R, Mishra V, Gupta K, Sasmal D, Raghubir R (2011) Neuroprotection by rosiglitazone in transient focal cerebral ischemia might not be mediated by glutamate transporter-1. J Neurosci Res 89:1849–1858PubMedCrossRef
go back to reference Wang L, Ashley-Koch A, Steffens DC, Krishnan KR, Taylor WD (2012) Impact of BDNF Val66Met and 5-HTTLPR polymorphism variants on neural substrates related to sadness and executive function. Genes Brain Behav 11(3):352–359PubMedCentralPubMedCrossRef Wang L, Ashley-Koch A, Steffens DC, Krishnan KR, Taylor WD (2012) Impact of BDNF Val66Met and 5-HTTLPR polymorphism variants on neural substrates related to sadness and executive function. Genes Brain Behav 11(3):352–359PubMedCentralPubMedCrossRef
go back to reference Yang Y, Qin X, Liu S, Li J, Zhu X, Gao T, Wang X (2011) Peroxisome proliferator-activated receptor γ is inhibited by histone deacetylase 4 in cortical neurons under oxidative stress. J Neurochem 118(3):429–439PubMedCrossRef Yang Y, Qin X, Liu S, Li J, Zhu X, Gao T, Wang X (2011) Peroxisome proliferator-activated receptor γ is inhibited by histone deacetylase 4 in cortical neurons under oxidative stress. J Neurochem 118(3):429–439PubMedCrossRef
go back to reference Zadori D, Geisz A, Vamos E, Vecsei L, Klivenyi P (2009) Valproate ameliorates the survival and the motor performance in a transgenic mouse model of Huntington’s disease. Pharmacol Biochem Behav 94(1):148–153PubMedCrossRef Zadori D, Geisz A, Vamos E, Vecsei L, Klivenyi P (2009) Valproate ameliorates the survival and the motor performance in a transgenic mouse model of Huntington’s disease. Pharmacol Biochem Behav 94(1):148–153PubMedCrossRef
go back to reference Zahler WL, Cleland WW (1968) A specific and sensitive assay for disulfides. J Biol Chem 243(4):716–719PubMed Zahler WL, Cleland WW (1968) A specific and sensitive assay for disulfides. J Biol Chem 243(4):716–719PubMed
go back to reference Zuccato C, Cattaneo E (2007) Role of brain-derived neurotrophic factor in Huntington’s disease. Prog Neurobiol 81(5–6):294–330PubMedCrossRef Zuccato C, Cattaneo E (2007) Role of brain-derived neurotrophic factor in Huntington’s disease. Prog Neurobiol 81(5–6):294–330PubMedCrossRef
Metadata
Title
Rosiglitazone Synergizes the Neuroprotective Effects of Valproic Acid Against Quinolinic Acid-Induced Neurotoxicity in Rats: Targeting PPARγ and HDAC Pathways
Authors
Jitendriya Mishra
Tanya Chaudhary
Anil Kumar
Publication date
01-08-2014
Publisher
Springer US
Published in
Neurotoxicity Research / Issue 2/2014
Print ISSN: 1029-8428
Electronic ISSN: 1476-3524
DOI
https://doi.org/10.1007/s12640-014-9458-z

Other articles of this Issue 2/2014

Neurotoxicity Research 2/2014 Go to the issue