메뉴 건너뛰기




Volumn 51, Issue 1, 2015, Pages 313-330

Preconditioning as a Potential Strategy for the Prevention of Parkinson’s Disease

Author keywords

Mitochondrial dysfunction; Oxidative stress; Parkinson s disease; Preconditioning

Indexed keywords

ADAPTATION; ANIMAL; BIOLOGICAL MODEL; HUMAN; METABOLISM; MITOCHONDRION; OXIDATIVE STRESS; PARKINSON DISEASE; PATHOLOGY; PATHOPHYSIOLOGY;

EID: 84897230971     PISSN: 08937648     EISSN: 15591182     Source Type: Journal    
DOI: 10.1007/s12035-014-8689-6     Document Type: Review
Times cited : (24)

References (160)
  • 2
    • 2342538502 scopus 로고    scopus 로고
    • Minocycline reduces the lipopolysaccharide-induced inflammatory reaction, peroxynitrite-mediated nitration of proteins, disruption of the blood–brain barrier, and damage in the nigral dopaminergic system
    • COI: 1:CAS:528:DC%2BD2cXjs1alur4%3D, PID: 15207276
    • Tomas-Camardiel M, Rite I, Herrera AJ, de Pablos RM, Cano J, Machado A, Venero JL (2004) Minocycline reduces the lipopolysaccharide-induced inflammatory reaction, peroxynitrite-mediated nitration of proteins, disruption of the blood–brain barrier, and damage in the nigral dopaminergic system. Neurobiol Dis 16(1):190–201. doi:10.1016/j.nbd.2004.01.010
    • (2004) Neurobiol. Dis. , vol.16 , Issue.1 , pp. 190-201
    • Tomas-Camardiel, M.1    Rite, I.2    Herrera, A.J.3    de Pablos, R.M.4    Cano, J.5    Machado, A.6    Venero, J.L.7
  • 3
    • 84876351189 scopus 로고    scopus 로고
    • Modeling early Parkinson’s disease pathology with chronic low dose MPTP treatment
    • COI: 1:CAS:528:DC%2BC3sXksFWhu7g%3D, PID: 23271419
    • Korecka JA, Eggers R, Swaab DF, Bossers K, Verhaagen J (2013) Modeling early Parkinson’s disease pathology with chronic low dose MPTP treatment. Restor Neurol Neurosci 31(2):155–167
    • (2013) Restor. Neurol. Neurosci. , vol.31 , Issue.2 , pp. 155-167
    • Korecka, J.A.1    Eggers, R.2    Swaab, D.F.3    Bossers, K.4    Verhaagen, J.5
  • 4
    • 2442636348 scopus 로고    scopus 로고
    • Parkinson’s: divergent causes, convergent mechanisms
    • COI: 1:CAS:528:DC%2BD2cXkt1Kjt78%3D, PID: 15155938
    • Greenamyre JT, Hastings TG (2004) Parkinson’s: divergent causes, convergent mechanisms. Science 304(5674):1120–1122
    • (2004) Science , vol.304 , Issue.5674 , pp. 1120-1122
    • Greenamyre, J.T.1    Hastings, T.G.2
  • 5
    • 79955695012 scopus 로고    scopus 로고
    • Life span and locomotor activity modification by glucose and polyphenols in Drosophila melanogaster chronically exposed to oxidative stress-stimuli: implications in Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC3MXjvVWrsb8%3D, PID: 21442225
    • Ortega-Arellano HF, Jimenez-Del-Rio M, Velez-Pardo C (2011) Life span and locomotor activity modification by glucose and polyphenols in Drosophila melanogaster chronically exposed to oxidative stress-stimuli: implications in Parkinson’s disease. Neurochem Res 36(6):1073–1086
    • (2011) Neurochem. Res. , vol.36 , Issue.6 , pp. 1073-1086
    • Ortega-Arellano, H.F.1    Jimenez-Del-Rio, M.2    Velez-Pardo, C.3
  • 6
    • 38349124184 scopus 로고    scopus 로고
    • Molecular physiology of preconditioning-induced brain tolerance to ischemia
    • COI: 1:CAS:528:DC%2BD1cXitVKgtr0%3D, PID: 18195087
    • Obrenovitch TP (2008) Molecular physiology of preconditioning-induced brain tolerance to ischemia. Physiol Rev 88(1):211–247
    • (2008) Physiol. Rev. , vol.88 , Issue.1 , pp. 211-247
    • Obrenovitch, T.P.1
  • 8
    • 1642387600 scopus 로고    scopus 로고
    • The perioperative management of Parkinson’s disease revisited
    • PID: 15062517
    • Gálvez-Jiménez N, Lang AE (2004) The perioperative management of Parkinson’s disease revisited. Neurol Clin 22(2):367–377
    • (2004) Neurol. Clin. , vol.22 , Issue.2 , pp. 367-377
    • Gálvez-Jiménez, N.1    Lang, A.E.2
  • 9
    • 59849125684 scopus 로고    scopus 로고
    • Parkinson disease: not just a movement disorder
    • PID: 19088004
    • Pandya M, Kubu CS, Giroux ML (2008) Parkinson disease: not just a movement disorder. Cleve Clin J Med 75(12):856–864
    • (2008) Cleve Clin J Med , vol.75 , Issue.12 , pp. 856-864
    • Pandya, M.1    Kubu, C.S.2    Giroux, M.L.3
  • 10
    • 41049106214 scopus 로고    scopus 로고
    • Non‐motor symptoms in Parkinson’s disease
    • PID: 18353132
    • Poewe W (2008) Non‐motor symptoms in Parkinson’s disease. Eur J Neurol 15(s1):14–20
    • (2008) Eur J Neurol , vol.15 , Issue.s1 , pp. 14-20
    • Poewe, W.1
  • 11
    • 41149163183 scopus 로고    scopus 로고
    • Parkinson’s disease: clinical features and diagnosis
    • COI: 1:STN:280:DC%2BD1c7oslamtA%3D%3D, PID: 18344392
    • Jankovic J (2008) Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry 79(4):368–376
    • (2008) J Neurol Neurosurg Psychiatry , vol.79 , Issue.4 , pp. 368-376
    • Jankovic, J.1
  • 12
    • 44649170283 scopus 로고    scopus 로고
    • Acetaminophen attenuates dopamine neuron degeneration in animal models of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD1cXmvVKksrg%3D, PID: 18514411
    • Locke CJ, Fox SA, Caldwell GA, Caldwell KA (2008) Acetaminophen attenuates dopamine neuron degeneration in animal models of Parkinson’s disease. Neurosci Lett 439(2):129–133. doi:10.1016/j.neulet.2008.05.003
    • (2008) Neurosci. Lett. , vol.439 , Issue.2 , pp. 129-133
    • Locke, C.J.1    Fox, S.A.2    Caldwell, G.A.3    Caldwell, K.A.4
  • 13
    • 0033681149 scopus 로고    scopus 로고
    • Chronic systemic pesticide exposure reproduces features of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD3cXosVGgsbg%3D, PID: 11100151
    • Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT (2000) Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci 3(12):1301–1306
    • (2000) Nat. Neurosci. , vol.3 , Issue.12 , pp. 1301-1306
    • Betarbet, R.1    Sherer, T.B.2    MacKenzie, G.3    Garcia-Osuna, M.4    Panov, A.V.5    Greenamyre, J.T.6
  • 14
    • 84877636727 scopus 로고    scopus 로고
    • Differentiation between idiopathic and atypical parkinsonian syndromes using three-dimensional magnetic resonance spectroscopic imaging
    • PID: 23334525
    • Gröger A, Bender B, Wurster I, Chadzynski GL, Klose U, Berg D (2013) Differentiation between idiopathic and atypical parkinsonian syndromes using three-dimensional magnetic resonance spectroscopic imaging. J Neurol Neurosurg Psychiatry 84(6):644–649
    • (2013) J Neurol Neurosurg Psychiatry , vol.84 , Issue.6 , pp. 644-649
    • Gröger, A.1    Bender, B.2    Wurster, I.3    Chadzynski, G.L.4    Klose, U.5    Berg, D.6
  • 15
    • 36249014794 scopus 로고    scopus 로고
    • Cholinergic and other neurotransmitter mechanisms in Parkinson’s disease, Parkinson’s disease dementia, and dementia with Lewy bodies
    • PID: 18175396
    • Francis PT, Perry EK (2007) Cholinergic and other neurotransmitter mechanisms in Parkinson’s disease, Parkinson’s disease dementia, and dementia with Lewy bodies. Mov Disord 22(S17):S351–S357
    • (2007) Mov Disord , vol.22 , Issue.S17 , pp. S351-S357
    • Francis, P.T.1    Perry, E.K.2
  • 16
    • 0037337815 scopus 로고    scopus 로고
    • Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases
    • PID: 12633144
    • Zarow C, Lyness SA, Mortimer JA, Chui HC (2003) Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases. Arch Neurol 60(3):337
    • (2003) Arch. Neurol , vol.60 , Issue.3 , pp. 337
    • Zarow, C.1    Lyness, S.A.2    Mortimer, J.A.3    Chui, H.C.4
  • 17
    • 62949177919 scopus 로고    scopus 로고
    • Dopaminergic and serotoninergic deficiencies in young adult rats prenatally exposed to the bacterial lipopolysaccharide
    • COI: 1:CAS:528:DC%2BD1MXjslOkt7o%3D, PID: 19236855
    • Wang S, Yan J-Y, Lo Y-K, Carvey PM, Ling Z (2009) Dopaminergic and serotoninergic deficiencies in young adult rats prenatally exposed to the bacterial lipopolysaccharide. Brain Res 1265:196–204
    • (2009) Brain Res. , vol.1265 , pp. 196-204
    • Wang, S.1    Yan, J.-Y.2    Lo, Y.-K.3    Carvey, P.M.4    Ling, Z.5
  • 19
    • 79953294305 scopus 로고    scopus 로고
    • Oxidative stress and aminopeptidases in Parkinson’s disease patients with and without treatment
    • PID: 20714110
    • Duran R, Barrero FJ, Morales B, Luna JD, Ramirez M, Vives F (2010) Oxidative stress and aminopeptidases in Parkinson’s disease patients with and without treatment. Neurodegener Dis 8(3):109–116
    • (2010) Neurodegener Dis , vol.8 , Issue.3 , pp. 109-116
    • Duran, R.1    Barrero, F.J.2    Morales, B.3    Luna, J.D.4    Ramirez, M.5    Vives, F.6
  • 20
    • 84859107546 scopus 로고    scopus 로고
    • Contributions of central and systemic inflammation to the pathophysiology of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC38XjtF2itb4%3D, PID: 22361232
    • Collins LM, Toulouse A, Connor TJ, Nolan YM (2012) Contributions of central and systemic inflammation to the pathophysiology of Parkinson’s disease. Neuropharmacology 62(7):2154–2168. doi:10.1016/j.neuropharm.2012.01.028
    • (2012) Neuropharmacology , vol.62 , Issue.7 , pp. 2154-2168
    • Collins, L.M.1    Toulouse, A.2    Connor, T.J.3    Nolan, Y.M.4
  • 21
    • 72649106290 scopus 로고    scopus 로고
    • Phosphorus and proton magnetic resonance spectroscopy demonstrates mitochondrial dysfunction in early and advanced Parkinson’s disease
    • PID: 19952056
    • Hattingen E, Magerkurth J, Pilatus U, Mozer A, Seifried C, Steinmetz H, Zanella F, Hilker R (2009) Phosphorus and proton magnetic resonance spectroscopy demonstrates mitochondrial dysfunction in early and advanced Parkinson’s disease. Brain 132(12):3285–3297
    • (2009) Brain , vol.132 , Issue.12 , pp. 3285-3297
    • Hattingen, E.1    Magerkurth, J.2    Pilatus, U.3    Mozer, A.4    Seifried, C.5    Steinmetz, H.6    Zanella, F.7    Hilker, R.8
  • 22
    • 84867516589 scopus 로고    scopus 로고
    • Mitochondrial biogenesis contributes to ischemic neuroprotection afforded by LPS preconditioning
    • COI: 1:CAS:528:DC%2BC38XhsVyksbnL, PID: 23050650
    • Anne Stetler R, Leak RK, Yin W, Zhang L, Wang S, Gao Y, Chen J (2012) Mitochondrial biogenesis contributes to ischemic neuroprotection afforded by LPS preconditioning. J Neurochem 123(s2):125–137
    • (2012) J. Neurochem. , vol.123 , Issue.s2 , pp. 125-137
    • Anne Stetler, R.1    Leak, R.K.2    Yin, W.3    Zhang, L.4    Wang, S.5    Gao, Y.6    Chen, J.7
  • 23
    • 14744289621 scopus 로고    scopus 로고
    • Expression time course and spatial distribution of activated caspase-3 after experimental status epilepticus: contribution of delayed neuronal cell death to seizure-induced neuronal injury
    • COI: 1:CAS:528:DC%2BD2MXhvFynsLc%3D, PID: 15755684
    • Weise J, Engelhorn T, Dörfler A, Aker S, Bähr M, Hufnagel A (2005) Expression time course and spatial distribution of activated caspase-3 after experimental status epilepticus: contribution of delayed neuronal cell death to seizure-induced neuronal injury. Neurobiol Dis 18(3):582–590
    • (2005) Neurobiol. Dis. , vol.18 , Issue.3 , pp. 582-590
    • Weise, J.1    Engelhorn, T.2    Dörfler, A.3    Aker, S.4    Bähr, M.5    Hufnagel, A.6
  • 24
    • 77049108173 scopus 로고    scopus 로고
    • Sustained deficiency of mitochondrial complex I activity during long periods of survival after seizures induced in immature rats by homocysteic acid
    • PID: 19931336
    • Folbergrová J, Ješina P, Haugvicová R, Lisý V, Houštěk J (2010) Sustained deficiency of mitochondrial complex I activity during long periods of survival after seizures induced in immature rats by homocysteic acid. Neurochem Int 56(3):394–403
    • (2010) Neurochem. Int. , vol.56 , Issue.3 , pp. 394-403
    • Folbergrová, J.1    Ješina, P.2    Haugvicová, R.3    Lisý, V.4    Houštěk, J.5
  • 25
    • 0037025256 scopus 로고    scopus 로고
    • Mitochondria, Ca(2+) and neurodegenerative disease
    • COI: 1:CAS:528:DC%2BD38XlvVWltb8%3D, PID: 12151010
    • Krieger C, Duchen MR (2002) Mitochondria, Ca(2+) and neurodegenerative disease. Eur J Pharmacol 447(2–3):177–188. doi:10.1016/s0014-2999(02)01842-3
    • (2002) Eur J Pharmacol , vol.447 , Issue.2-3 , pp. 177-188
    • Krieger, C.1    Duchen, M.R.2
  • 27
    • 84864340428 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptors γ/mitochondrial uncoupling protein 2 signaling protects against seizure-induced neuronal cell death in the hippocampus following experimental status epilepticus
    • Chuang Y-C, Lin T-K, Huang H-Y, Chang W-N, Liou C-W, Chen S-D, Chang AY, Chan SH (2012) Peroxisome proliferator-activated receptors γ/mitochondrial uncoupling protein 2 signaling protects against seizure-induced neuronal cell death in the hippocampus following experimental status epilepticus. J Neuroinflammation 9(1):1–18
    • (2012) J. Neuroinflammation , vol.9 , Issue.1 , pp. 1-18
    • Chuang, Y.-C.1    Lin, T.-K.2    Huang, H.-Y.3    Chang, W.-N.4    Liou, C.-W.5    Chen, S.-D.6    Chang, A.Y.7    Chan, S.H.8
  • 28
    • 33947625069 scopus 로고    scopus 로고
    • Mitochondrial complex I inhibition in cerebral cortex of immature rats following homocysteic acid-induced seizures
    • PID: 17270175
    • Folbergrová J, Ješina P, Drahota Z, Lisý V, Haugvicová R, Vojtíšková A, Houštěk J (2007) Mitochondrial complex I inhibition in cerebral cortex of immature rats following homocysteic acid-induced seizures. Exp Neurol 204(2):597–609
    • (2007) Exp. Neurol. , vol.204 , Issue.2 , pp. 597-609
    • Folbergrová, J.1    Ješina, P.2    Drahota, Z.3    Lisý, V.4    Haugvicová, R.5    Vojtíšková, A.6    Houštěk, J.7
  • 30
    • 0035233083 scopus 로고    scopus 로고
    • Causes of neuronal death in Parkinson’s disease
    • Calne D, Calne SM, (eds), Advances in neurology, 86, Lippincott Williams & Wilkins, Philadelphia:
    • Schapira AHV (2001) Causes of neuronal death in Parkinson’s disease. In: Calne D, Calne SM (eds) Parkinson’s disease, vol 86, Advances in neurology. Lippincott Williams & Wilkins, Philadelphia, pp 155–162
    • (2001) Parkinson’s disease , pp. 155-162
    • Schapira, A.H.V.1
  • 31
    • 77951950272 scopus 로고    scopus 로고
    • Combined R‐α–lipoic acid and acetyl‐L‐carnitine exerts efficient preventative effects in a cellular model of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC3cXlt1Oqsrc%3D, PID: 20414966
    • Zhang H, Jia H, Liu J, Ao N, Yan B, Shen W, Wang X, Li X, Luo C, Liu J (2010) Combined R‐α–lipoic acid and acetyl‐L‐carnitine exerts efficient preventative effects in a cellular model of Parkinson’s disease. J Cell Mol Med 14(1–2):215–225
    • (2010) J. Cell Mol. Med. , vol.14 , Issue.1-2 , pp. 215-225
    • Zhang, H.1    Jia, H.2    Liu, J.3    Ao, N.4    Yan, B.5    Shen, W.6    Wang, X.7    Li, X.8    Luo, C.9    Liu, J.10
  • 32
    • 61349156769 scopus 로고    scopus 로고
    • Mutant Pink1 induces mitochondrial dysfunction in a neuronal cell model of Parkinson’s disease by disturbing calcium flux
    • COI: 1:CAS:528:DC%2BD1MXjsFWgsbs%3D, PID: 19166511
    • Marongiu R, Spencer B, Crews L, Adame A, Patrick C, Trejo M, Dallapiccola B, Valente EM, Masliah E (2009) Mutant Pink1 induces mitochondrial dysfunction in a neuronal cell model of Parkinson’s disease by disturbing calcium flux. J Neurochem 108(6):1561–1574
    • (2009) J. Neurochem. , vol.108 , Issue.6 , pp. 1561-1574
    • Marongiu, R.1    Spencer, B.2    Crews, L.3    Adame, A.4    Patrick, C.5    Trejo, M.6    Dallapiccola, B.7    Valente, E.M.8    Masliah, E.9
  • 33
    • 84859345475 scopus 로고    scopus 로고
    • Impaired complex-I mitochondrial biogenesis in Parkinson disease frontal cortex
    • COI: 1:CAS:528:DC%2BC38XpsFOltro%3D, PID: 23939409
    • Thomas RR, Keeney PM, Bennett JP (2012) Impaired complex-I mitochondrial biogenesis in Parkinson disease frontal cortex. J Parkinsons Dis 2(1):67–76
    • (2012) J Parkinsons Dis , vol.2 , Issue.1 , pp. 67-76
    • Thomas, R.R.1    Keeney, P.M.2    Bennett, J.P.3
  • 34
    • 84867269951 scopus 로고    scopus 로고
    • AMP kinase activation mitigates dopaminergic dysfunction and mitochondrial abnormalities in Drosophila models of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC38XhsFGlsbrM, PID: 23055502
    • Ng C-H, Guan MS, Koh C, Ouyang X, Yu F, Tan E-K, O’Neill SP, Zhang X, Chung J, Lim K-L (2012) AMP kinase activation mitigates dopaminergic dysfunction and mitochondrial abnormalities in Drosophila models of Parkinson’s disease. J Neurosci 32(41):14311–14317
    • (2012) J. Neurosci. , vol.32 , Issue.41 , pp. 14311-14317
    • Ng, C.-H.1    Guan, M.S.2    Koh, C.3    Ouyang, X.4    Yu, F.5    Tan, E.-K.6    O’Neill, S.P.7    Zhang, X.8    Chung, J.9    Lim, K.-L.10
  • 35
    • 31744432453 scopus 로고    scopus 로고
    • Tetrahydrobiopterin causes mitochondrial dysfunction in dopaminergic cells: implications for Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD28XhtV2ntr0%3D, PID: 16343695
    • Choi HJ, Lee SY, Cho Y, No H, Kim SW, Hwang O (2006) Tetrahydrobiopterin causes mitochondrial dysfunction in dopaminergic cells: implications for Parkinson’s disease. Neurochem Int 48(4):255–262
    • (2006) Neurochem. Int. , vol.48 , Issue.4 , pp. 255-262
    • Choi, H.J.1    Lee, S.Y.2    Cho, Y.3    No, H.4    Kim, S.W.5    Hwang, O.6
  • 36
    • 80052002645 scopus 로고    scopus 로고
    • Transient receptor potential melastatin 4 inhibition prevents lipopolysaccharide-induced endothelial cell death
    • COI: 1:CAS:528:DC%2BC3MXhtVOgu7bN, PID: 21565835
    • Becerra A, Echeverría C, Varela D, Sarmiento D, Armisén R, Nuñez-Villena F, Montecinos M, Simon F (2011) Transient receptor potential melastatin 4 inhibition prevents lipopolysaccharide-induced endothelial cell death. Cardiovasc Res 91(4):677–684
    • (2011) Cardiovasc. Res. , vol.91 , Issue.4 , pp. 677-684
    • Becerra, A.1    Echeverría, C.2    Varela, D.3    Sarmiento, D.4    Armisén, R.5    Nuñez-Villena, F.6    Montecinos, M.7    Simon, F.8
  • 37
    • 84861456441 scopus 로고    scopus 로고
    • Rutin protects dopaminergic neurons from oxidative stress in an animal model of Parkinson’s disease
    • PID: 22194158
    • Khan MM, Raza SS, Javed H, Ahmad A, Khan A, Islam F, Safhi MM, Islam F (2012) Rutin protects dopaminergic neurons from oxidative stress in an animal model of Parkinson’s disease. Neurotox res 22(1):1–15
    • (2012) Neurotox res , vol.22 , Issue.1 , pp. 1-15
    • Khan, M.M.1    Raza, S.S.2    Javed, H.3    Ahmad, A.4    Khan, A.5    Islam, F.6    Safhi, M.M.7    Islam, F.8
  • 38
    • 79955129251 scopus 로고    scopus 로고
    • Diazoxide preconditioning against seizure-induced oxidative injury is via the PI3K/Akt pathway in epileptic rat
    • COI: 1:CAS:528:DC%2BC3MXltFaiu78%3D, PID: 21440599
    • Xue Y, Xie N, Cao L, Zhao X, Jiang H, Chi Z (2011) Diazoxide preconditioning against seizure-induced oxidative injury is via the PI3K/Akt pathway in epileptic rat. Neurosci Lett 495(2):130–134
    • (2011) Neurosci. Lett. , vol.495 , Issue.2 , pp. 130-134
    • Xue, Y.1    Xie, N.2    Cao, L.3    Zhao, X.4    Jiang, H.5    Chi, Z.6
  • 39
    • 67650094606 scopus 로고    scopus 로고
    • Effect of centrophenoxine against rotenone-induced oxidative stress in an animal model of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD1MXoslSju7w%3D, PID: 19375462
    • Verma R, Nehru B (2009) Effect of centrophenoxine against rotenone-induced oxidative stress in an animal model of Parkinson’s disease. Neurochem Int 55(6):369–375
    • (2009) Neurochem. Int. , vol.55 , Issue.6 , pp. 369-375
    • Verma, R.1    Nehru, B.2
  • 40
    • 0034624017 scopus 로고    scopus 로고
    • The parkinsonism-inducing drug 1-methyl-4-phenylpyridinium triggers intracellular dopamine oxidation—a novel mechanism of toxicity
    • COI: 1:CAS:528:DC%2BD3cXoslymt7c%3D, PID: 10969076
    • Lotharius J, O’Malley KL (2000) The parkinsonism-inducing drug 1-methyl-4-phenylpyridinium triggers intracellular dopamine oxidation—a novel mechanism of toxicity. J Biol Chem 275(49):38581–38588. doi:10.1074/jbc.M005385200
    • (2000) J Biol Chem , vol.275 , Issue.49 , pp. 38581-38588
    • Lotharius, J.1    O’Malley, K.L.2
  • 41
    • 78650510736 scopus 로고    scopus 로고
    • Proteomic insights into the protective mechanisms of an in vitro oxidative stress model of early stage Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC3cXhs1ajsbzJ, PID: 21056633
    • Bauereis B, Haskins WE, LeBaron RG, Renthal R (2011) Proteomic insights into the protective mechanisms of an in vitro oxidative stress model of early stage Parkinson’s disease. Neurosci Lett 488(1):11–16
    • (2011) Neurosci. Lett. , vol.488 , Issue.1 , pp. 11-16
    • Bauereis, B.1    Haskins, W.E.2    LeBaron, R.G.3    Renthal, R.4
  • 42
    • 79961207643 scopus 로고    scopus 로고
    • Evaluation of markers of oxidative stress, antioxidant function and astrocytic proliferation in the striatum and frontal cortex of Parkinson’s disease brains
    • COI: 1:CAS:528:DC%2BC3MXksFWntbg%3D, PID: 21484266
    • Mythri RB, Venkateshappa C, Harish G, Mahadevan A, Muthane UB, Yasha T, Bharath MS, Shankar S (2011) Evaluation of markers of oxidative stress, antioxidant function and astrocytic proliferation in the striatum and frontal cortex of Parkinson’s disease brains. Neurochem Res 36(8):1452–1463
    • (2011) Neurochem. Res. , vol.36 , Issue.8 , pp. 1452-1463
    • Mythri, R.B.1    Venkateshappa, C.2    Harish, G.3    Mahadevan, A.4    Muthane, U.B.5    Yasha, T.6    Bharath, M.S.7    Shankar, S.8
  • 43
    • 60549100886 scopus 로고    scopus 로고
    • Early stages for Parkinson’s development: α-Synuclein misfolding and aggregation
    • PID: 18633713
    • Yu J, Lyubchenko YL (2009) Early stages for Parkinson’s development: α-Synuclein misfolding and aggregation. J Neuroimmune Pharmacol 4(1):10–16
    • (2009) J Neuroimmune Pharmacol , vol.4 , Issue.1 , pp. 10-16
    • Yu, J.1    Lyubchenko, Y.L.2
  • 44
    • 77951885732 scopus 로고    scopus 로고
    • Non‐classical exocytosis of α‐synuclein is sensitive to folding states and promoted under stress conditions
    • COI: 1:CAS:528:DC%2BC3cXnt1Gnsbo%3D, PID: 20345754
    • Jang A, Lee HJ, Suk JE, Jung JW, Kim KP, Lee SJ (2010) Non‐classical exocytosis of α‐synuclein is sensitive to folding states and promoted under stress conditions. J Neurochem 113(5):1263–1274
    • (2010) J. Neurochem. , vol.113 , Issue.5 , pp. 1263-1274
    • Jang, A.1    Lee, H.J.2    Suk, J.E.3    Jung, J.W.4    Kim, K.P.5    Lee, S.J.6
  • 45
    • 67649515087 scopus 로고    scopus 로고
    • The Parkinson disease‐associated A30P mutation stabilizes α‐synuclein against proteasomal degradation triggered by heme oxygenase‐1 over‐expression in human neuroblastoma cells
    • COI: 1:CAS:528:DC%2BD1MXovV2mtbk%3D, PID: 19457084
    • Song W, Patel A, Qureshi HY, Han D, Schipper HM, Paudel HK (2009) The Parkinson disease‐associated A30P mutation stabilizes α‐synuclein against proteasomal degradation triggered by heme oxygenase‐1 over‐expression in human neuroblastoma cells. J Neurochem 110(2):719–733
    • (2009) J. Neurochem. , vol.110 , Issue.2 , pp. 719-733
    • Song, W.1    Patel, A.2    Qureshi, H.Y.3    Han, D.4    Schipper, H.M.5    Paudel, H.K.6
  • 46
    • 84864318812 scopus 로고    scopus 로고
    • A model of nitric oxide induced alpha-synuclein misfolding in Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC38XhtVymt73M, PID: 22776646
    • Stone DK, Kiyota T, Mosley RL, Gendelman HE (2012) A model of nitric oxide induced alpha-synuclein misfolding in Parkinson’s disease. Neurosci Lett 523(2):167–173. doi:10.1016/j.neulet.2012.06.070
    • (2012) Neurosci. Lett. , vol.523 , Issue.2 , pp. 167-173
    • Stone, D.K.1    Kiyota, T.2    Mosley, R.L.3    Gendelman, H.E.4
  • 47
    • 77955085868 scopus 로고    scopus 로고
    • Redox reactions induced by nitrosative stress mediate protein misfolding and mitochondrial dysfunction in neurodegenerative diseases
    • COI: 1:CAS:528:DC%2BC3cXmsFSqsbo%3D, PID: 20333559
    • Gu Z, Nakamura T, Lipton SA (2010) Redox reactions induced by nitrosative stress mediate protein misfolding and mitochondrial dysfunction in neurodegenerative diseases. Mol Neurobiol 41(2–3):55–72
    • (2010) Mol. Neurobiol. , vol.41 , Issue.2-3 , pp. 55-72
    • Gu, Z.1    Nakamura, T.2    Lipton, S.A.3
  • 48
    • 39749144048 scopus 로고    scopus 로고
    • Mitochondrial dysfunction, oxidative stress, and apoptosis revealed by proteomic and transcriptomic analyses of the striata in two mouse models of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD1cXitlahsA%3D%3D, PID: 18173235
    • Chin MH, Qian W-J, Wang H, Petyuk VA, Bloom JS, Sforza DM, Laćan G, Liu D, Khan AH, Cantor RM (2008) Mitochondrial dysfunction, oxidative stress, and apoptosis revealed by proteomic and transcriptomic analyses of the striata in two mouse models of Parkinson’s disease. J Proteome Res 7(2):666–677
    • (2008) J. Proteome Res. , vol.7 , Issue.2 , pp. 666-677
    • Chin, M.H.1    Qian, W.-J.2    Wang, H.3    Petyuk, V.A.4    Bloom, J.S.5    Sforza, D.M.6    Laćan, G.7    Liu, D.8    Khan, A.H.9    Cantor, R.M.10
  • 49
    • 84866314902 scopus 로고    scopus 로고
    • Tang X: Downregulation of Pael-R expression in a Parkinson’s disease cell model reduces apoptosis. Journal of Clinical Neuroscience
    • Zou T, Xiao B, Tang J, Zhang H, Tang X (2012) Downregulation of Pael-R expression in a Parkinson’s disease cell model reduces apoptosis. Journal of Clinical Neuroscience
    • (2012) Zhang H
    • Zou, T.1    Xiao, B.2    Tang, J.3
  • 50
    • 68749114987 scopus 로고    scopus 로고
    • Impaired glutamate homeostasis and programmed cell death in a chronic MPTP mouse model of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD1MXpvF2mur4%3D, PID: 19523952
    • Meredith G, Totterdell S, Beales M, Meshul C (2009) Impaired glutamate homeostasis and programmed cell death in a chronic MPTP mouse model of Parkinson’s disease. Exp Neurol 219(1):334–340
    • (2009) Exp. Neurol. , vol.219 , Issue.1 , pp. 334-340
    • Meredith, G.1    Totterdell, S.2    Beales, M.3    Meshul, C.4
  • 51
    • 59649089328 scopus 로고    scopus 로고
    • The Parkinson disease protein leucine-rich repeat kinase 2 transduces death signals via Fas-associated protein with death domain and caspase-8 in a cellular model of neurodegeneration
    • COI: 1:CAS:528:DC%2BD1MXhs1KqsbY%3D, PID: 19176810
    • Ho CC-Y, Rideout HJ, Ribe E, Troy CM, Dauer WT (2009) The Parkinson disease protein leucine-rich repeat kinase 2 transduces death signals via Fas-associated protein with death domain and caspase-8 in a cellular model of neurodegeneration. J Neurosci 29(4):1011–1016
    • (2009) J. Neurosci. , vol.29 , Issue.4 , pp. 1011-1016
    • Ho, C.C.-Y.1    Rideout, H.J.2    Ribe, E.3    Troy, C.M.4    Dauer, W.T.5
  • 53
    • 77049122576 scopus 로고    scopus 로고
    • +-induced oxidative stress act by modulating markers of apoptotic death in dopaminergic neurons
    • COI: 1:CAS:528:DC%2BD1MXhsFKgur%2FI, PID: 19466539
    • +-induced oxidative stress act by modulating markers of apoptotic death in dopaminergic neurons. Cell Mol Neurobiol 29(8):1169–1180
    • (2009) Cell. Mol. Neurobiol. , vol.29 , Issue.8 , pp. 1169-1180
    • Bournival, J.1    Quessy, P.2    Martinoli, M.-G.3
  • 54
    • 79961208135 scopus 로고    scopus 로고
    • Protective effect of lycopene on oxidative stress and cognitive decline in rotenone induced model of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC3MXksFWns7s%3D, PID: 21484267
    • Kaur H, Chauhan S, Sandhir R (2011) Protective effect of lycopene on oxidative stress and cognitive decline in rotenone induced model of Parkinson’s disease. Neurochem Res 36(8):1435–1443
    • (2011) Neurochem. Res. , vol.36 , Issue.8 , pp. 1435-1443
    • Kaur, H.1    Chauhan, S.2    Sandhir, R.3
  • 55
    • 78349304719 scopus 로고    scopus 로고
    • Apoptosis-inducing factor mediates dopaminergic cell death in response to LPS-induced inflammatory stimulus: evidence in Parkinson’s disease patients
    • COI: 1:CAS:528:DC%2BC3cXhsVWqsLnF, PID: 20850531
    • Burguillos M, Hajji N, Englund E, Persson A, Cenci A, Machado A, Cano J, Joseph B, Venero J (2011) Apoptosis-inducing factor mediates dopaminergic cell death in response to LPS-induced inflammatory stimulus: evidence in Parkinson’s disease patients. Neurobiol Dis 41(1):177–188
    • (2011) Neurobiol. Dis. , vol.41 , Issue.1 , pp. 177-188
    • Burguillos, M.1    Hajji, N.2    Englund, E.3    Persson, A.4    Cenci, A.5    Machado, A.6    Cano, J.7    Joseph, B.8    Venero, J.9
  • 56
    • 84878486068 scopus 로고    scopus 로고
    • Therapeutic attenuation of neuroinflammation and apoptosis by black tea theaflavin in chronic MPTP/probenecid model of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC3sXitlWr, PID: 22669749
    • Anandhan A, Essa MM, Manivasagam T (2013) Therapeutic attenuation of neuroinflammation and apoptosis by black tea theaflavin in chronic MPTP/probenecid model of Parkinson’s disease. Neurotox res 23(2):166–173
    • (2013) Neurotox res , vol.23 , Issue.2 , pp. 166-173
    • Anandhan, A.1    Essa, M.M.2    Manivasagam, T.3
  • 57
    • 6344275803 scopus 로고    scopus 로고
    • Activation of chaperone-mediated autophagy during oxidative stress
    • COI: 1:CAS:528:DC%2BD2cXpslKgu7w%3D, PID: 15331765
    • Kiffin R, Christian C, Knecht E, Cuervo AM (2004) Activation of chaperone-mediated autophagy during oxidative stress. Mol Biol Cell 15(11):4829–4840. doi:10.1091/mbc.E04-06-0477
    • (2004) Mol Biol Cell , vol.15 , Issue.11 , pp. 4829-4840
    • Kiffin, R.1    Christian, C.2    Knecht, E.3    Cuervo, A.M.4
  • 58
    • 0037417254 scopus 로고    scopus 로고
    • Alzheimer’s disease and Parkinson’s disease
    • Nussbaum RL, Ellis CE (2003) Alzheimer’s disease and Parkinson’s disease. N Engl J Med 348(14):1356–1364
    • (2003) N Engl J Med , vol.348 , Issue.14 , pp. 1356-1364
    • Nussbaum, R.L.1    Ellis, C.E.2
  • 59
    • 0034980546 scopus 로고    scopus 로고
    • Prevalence, clinical manifestations, etiology, and treatment of depression in Parkinson’s disease
    • COI: 1:STN:280:DC%2BD3Mzps1emug%3D%3D, PID: 11449025
    • Slaughter JR, Slaughter KA, Nichols D, Holmes SE, Martens MP (2001) Prevalence, clinical manifestations, etiology, and treatment of depression in Parkinson’s disease. J Neuropsychiatry Clin Neurosci 13(2):187–196
    • (2001) J. Neuropsychiatry Clin. Neurosci. , vol.13 , Issue.2 , pp. 187-196
    • Slaughter, J.R.1    Slaughter, K.A.2    Nichols, D.3    Holmes, S.E.4    Martens, M.P.5
  • 60
    • 33750482613 scopus 로고    scopus 로고
    • Effect of sublethal 6-hydroxydopamine on the response to subsequent oxidative stress in dopaminergic cells: evidence for preconditioning
    • COI: 1:CAS:528:DC%2BD28Xht1yks7vE, PID: 16956375
    • Leak RK, Liou AK, Zigmond MJ (2006) Effect of sublethal 6-hydroxydopamine on the response to subsequent oxidative stress in dopaminergic cells: evidence for preconditioning. J Neurochem 99(4):1151–1163
    • (2006) J. Neurochem. , vol.99 , Issue.4 , pp. 1151-1163
    • Leak, R.K.1    Liou, A.K.2    Zigmond, M.J.3
  • 61
    • 0347133331 scopus 로고    scopus 로고
    • Neuroprotection in Parkinson disease—mysteries, myths, and misconceptions
    • COI: 1:CAS:528:DC%2BD2cXmtlelsg%3D%3D
    • Schapira AHV, Olanow CW (2004) Neuroprotection in Parkinson disease—mysteries, myths, and misconceptions. Jama-J Am Med Assoc 291(3):358–364. doi:10.1001/jama.291.3.358
    • (2004) Jama-J. Am. Med. Assoc. , vol.291 , Issue.3 , pp. 358-364
    • Schapira, A.H.V.1    Olanow, C.W.2
  • 62
    • 71949098755 scopus 로고    scopus 로고
    • Induction of adult human bone marrow mesenchymal stromal cells into functional astrocyte-like cells: potential for restorative treatment in Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD1MXhtFaktr7M, PID: 19127447
    • Bahat-Stroomza M, Barhum Y, Levy YS, Karpov O, Bulvik S, Melamed E, Offen D (2009) Induction of adult human bone marrow mesenchymal stromal cells into functional astrocyte-like cells: potential for restorative treatment in Parkinson’s disease. J Mol Neurosci 39(1–2):199–210. doi:10.1007/s12031-008-9166-3
    • (2009) J Mol Neurosci , vol.39 , Issue.1-2 , pp. 199-210
    • Bahat-Stroomza, M.1    Barhum, Y.2    Levy, Y.S.3    Karpov, O.4    Bulvik, S.5    Melamed, E.6    Offen, D.7
  • 63
    • 46149088212 scopus 로고    scopus 로고
    • Effects of coincident 5-HT1A receptor stimulation and NMDA receptor antagonism on L-DOPA-induced dyskinesia and rotational behaviors in the hemi-parkinsonian rat
    • COI: 1:CAS:528:DC%2BD1cXnvVKhu7c%3D, PID: 18545986
    • Dupre KB, Eskow KL, Steiniger A, Klioueva A, Negron GE, Lormand L, Park JY, Bishop C (2008) Effects of coincident 5-HT1A receptor stimulation and NMDA receptor antagonism on L-DOPA-induced dyskinesia and rotational behaviors in the hemi-parkinsonian rat. Psychopharmacology 199(1):99–108
    • (2008) Psychopharmacology , vol.199 , Issue.1 , pp. 99-108
    • Dupre, K.B.1    Eskow, K.L.2    Steiniger, A.3    Klioueva, A.4    Negron, G.E.5    Lormand, L.6    Park, J.Y.7    Bishop, C.8
  • 64
    • 0033768218 scopus 로고    scopus 로고
    • Dyskinesias and motor fluctuations in Parkinson’s disease. A community-based study
    • PID: 11050029
    • Schrag A, Quinn N (2000) Dyskinesias and motor fluctuations in Parkinson’s disease. A community-based study. Brain 123(11):2297–2305
    • (2000) Brain , vol.123 , Issue.11 , pp. 2297-2305
    • Schrag, A.1    Quinn, N.2
  • 66
    • 0037381274 scopus 로고    scopus 로고
    • Deep brain stimulation for movement disorders: morbidity and mortality in 109 patients
    • PID: 12691402
    • Umemura A, Jaggi JL, Hurtig HI, Siderowf AD, Colcher A, Stern MB, Baltuch GH (2003) Deep brain stimulation for movement disorders: morbidity and mortality in 109 patients. J Neurosurg 98(4):779–784. doi:10.3171/jns.2003.98.4.0779
    • (2003) J Neurosurg , vol.98 , Issue.4 , pp. 779-784
    • Umemura, A.1    Jaggi, J.L.2    Hurtig, H.I.3    Siderowf, A.D.4    Colcher, A.5    Stern, M.B.6    Baltuch, G.H.7
  • 68
    • 72949111183 scopus 로고    scopus 로고
    • Dopaminergic transplantation for Parkinson’s disease: current status and future prospects
    • COI: 1:CAS:528:DC%2BC3cXjslChsQ%3D%3D, PID: 19938101
    • Olanow CW, Kordower JH, Lang AE, Obeso JA (2009) Dopaminergic transplantation for Parkinson’s disease: current status and future prospects. Ann Neurol 66(5):591–596. doi:10.1002/ana.21778
    • (2009) Ann. Neurol. , vol.66 , Issue.5 , pp. 591-596
    • Olanow, C.W.1    Kordower, J.H.2    Lang, A.E.3    Obeso, J.A.4
  • 69
    • 39749132884 scopus 로고    scopus 로고
    • Induction of A9 dopaminergic neurons from neural stem cells improves motor function in an animal model of Parkinson’s disease
    • PID: 18202103
    • O’Keeffe FE, Scott SA, Tyers P, O’Keeffe GW, Dalley JW, Zufferey R, Caldwell MA (2008) Induction of A9 dopaminergic neurons from neural stem cells improves motor function in an animal model of Parkinson’s disease. Brain 131(3):630–641
    • (2008) Brain , vol.131 , Issue.3 , pp. 630-641
    • O’Keeffe, F.E.1    Scott, S.A.2    Tyers, P.3    O’Keeffe, G.W.4    Dalley, J.W.5    Zufferey, R.6    Caldwell, M.A.7
  • 70
    • 74849115546 scopus 로고    scopus 로고
    • Gene therapy using lactoferrin-modified nanoparticles in a rotenone-induced chronic Parkinson model
    • COI: 1:CAS:528:DC%2BC3cXhtlOlsbs%3D, PID: 19909981
    • Huang RQ, Ke WL, Liu Y, Wu DD, Feng LY, Jiang C, Pei YY (2010) Gene therapy using lactoferrin-modified nanoparticles in a rotenone-induced chronic Parkinson model. J Neurol Sci 290(1–2):123–130. doi:10.1016/j.jns.2009.09.032
    • (2010) J Neurol Sci , vol.290 , Issue.1-2 , pp. 123-130
    • Huang, R.Q.1    Ke, W.L.2    Liu, Y.3    Wu, D.D.4    Feng, L.Y.5    Jiang, C.6    Pei, Y.Y.7
  • 72
    • 76949097385 scopus 로고    scopus 로고
    • Gene therapy in Parkinson’s disease rationale and current status
    • COI: 1:CAS:528:DC%2BC3cXlslCis70%3D, PID: 20155994
    • Feng LR, Maguire-Zeiss KA (2010) Gene therapy in Parkinson’s disease rationale and current status. Cns Drugs 24(3):177–192
    • (2010) Cns Drugs , vol.24 , Issue.3 , pp. 177-192
    • Feng, L.R.1    Maguire-Zeiss, K.A.2
  • 73
    • 84883356229 scopus 로고    scopus 로고
    • Amelioration of CNS toxicities of L-dopa in experimental models of Parkinson’s disease by concurrent treatment with Tinospora cordifolia
    • Antony S, DebRoy P, Vadivelan R, Jaysankar K, Vikram M, Nandini S, Sundeep M, Elango K, Suresh B (2010) Amelioration of CNS toxicities of L-dopa in experimental models of Parkinson’s disease by concurrent treatment with Tinospora cordifolia. Hygeia J D Med 2(1):28–37
    • (2010) Hygeia J D Med , vol.2 , Issue.1 , pp. 28-37
    • Antony, S.1    DebRoy, P.2    Vadivelan, R.3    Jaysankar, K.4    Vikram, M.5    Nandini, S.6    Sundeep, M.7    Elango, K.8    Suresh, B.9
  • 74
    • 84863483641 scopus 로고    scopus 로고
    • Microglia-inhibiting activity of Parkinson’s disease drug amantadine
    • COI: 1:CAS:528:DC%2BC38XhtVSgt7jF, PID: 22035588
    • Kim JH, Lee HW, Hwang J, Kim J, Lee MJ, Han HS, Lee WH, Suk K (2012) Microglia-inhibiting activity of Parkinson’s disease drug amantadine. Neurobiol Aging 33(9):2145–2159. doi:10.1016/j.neurobiolaging.2011.08.011
    • (2012) Neurobiol Aging , vol.33 , Issue.9 , pp. 2145-2159
    • Kim, J.H.1    Lee, H.W.2    Hwang, J.3    Kim, J.4    Lee, M.J.5    Han, H.S.6    Lee, W.H.7    Suk, K.8
  • 76
    • 0034769415 scopus 로고    scopus 로고
    • Amantadine in Parkinson’s disease: lymphocyte subsets and IL-2 secreting T cell precursor frequencies
    • COI: 1:CAS:528:DC%2BD3MXnslWquro%3D, PID: 11672995
    • Tribl GG, Wober C, Schonborn V, Brucke T, Deecke L, Panzer S (2001) Amantadine in Parkinson’s disease: lymphocyte subsets and IL-2 secreting T cell precursor frequencies. Exp Gerontol 36(10):1761–1771. doi:10.1016/s0531-5565(01)00128-0
    • (2001) Exp Gerontol , vol.36 , Issue.10 , pp. 1761-1771
    • Tribl, G.G.1    Wober, C.2    Schonborn, V.3    Brucke, T.4    Deecke, L.5    Panzer, S.6
  • 77
    • 78650591431 scopus 로고    scopus 로고
    • Chronic L-DOPA therapy alters central serotonergic function and L-DOPA-induced dopamine release in a region-dependent manner in a rat model of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC3MXovFGi, PID: 21092759
    • Navailles S, Bioulac B, Gross C, De Deurwaerdère P (2011) Chronic L-DOPA therapy alters central serotonergic function and L-DOPA-induced dopamine release in a region-dependent manner in a rat model of Parkinson’s disease. Neurobiol Dis 41(2):585–590
    • (2011) Neurobiol. Dis. , vol.41 , Issue.2 , pp. 585-590
    • Navailles, S.1    Bioulac, B.2    Gross, C.3    De Deurwaerdère, P.4
  • 78
    • 33744990845 scopus 로고    scopus 로고
    • The need for neuroprotective therapies in Parkinson’s disease A clinical perspective
    • PID: 16717249
    • Poewe W (2006) The need for neuroprotective therapies in Parkinson’s disease A clinical perspective. Neurology 66(10 suppl 4):S2–S9
    • (2006) Neurology , vol.66 , Issue.10 suppl 4 , pp. S2-S9
    • Poewe, W.1
  • 79
    • 33645128679 scopus 로고    scopus 로고
    • Intrastriatal infusion of liver growth factor stimulates dopamine terminal sprouting and partially restores motor function in 6-hydroxydopamine-lesioned rats
    • COI: 1:CAS:528:DC%2BD28XivFGmtL8%3D, PID: 16344326
    • Reimers D, Herranz AS, Diaz-Gil JJ, Lobo MVT, Paíno CL, Alonso R, Asensio MJ, Gonzalo-Gobernado R, Bazán E (2006) Intrastriatal infusion of liver growth factor stimulates dopamine terminal sprouting and partially restores motor function in 6-hydroxydopamine-lesioned rats. J Histochem Cytochem 54(4):457–465
    • (2006) J. Histochem. Cytochem. , vol.54 , Issue.4 , pp. 457-465
    • Reimers, D.1    Herranz, A.S.2    Diaz-Gil, J.J.3    Lobo, M.V.T.4    Paíno, C.L.5    Alonso, R.6    Asensio, M.J.7    Gonzalo-Gobernado, R.8    Bazán, E.9
  • 80
    • 79958110929 scopus 로고    scopus 로고
    • Targeting NGF-pathway for developing neuroprotective therapies for multiple sclerosis and other neurological diseases
    • PID: 21701990
    • Colafrancesco V, Villoslada P (2011) Targeting NGF-pathway for developing neuroprotective therapies for multiple sclerosis and other neurological diseases. Arch Ital Biol 149(2):183–192
    • (2011) Arch. Ital. Biol. , vol.149 , Issue.2 , pp. 183-192
    • Colafrancesco, V.1    Villoslada, P.2
  • 81
    • 56149119401 scopus 로고    scopus 로고
    • Neuroprotective effect of resveratrol on 6-OHDA-induced Parkinson’s disease in rats
    • COI: 1:CAS:528:DC%2BD1cXhtlyltLvJ, PID: 18940189
    • Jin F, Wu Q, Lu Y-F, Gong Q-H, Shi J-S (2008) Neuroprotective effect of resveratrol on 6-OHDA-induced Parkinson’s disease in rats. Eur J Pharmacol 600(1):78–82
    • (2008) Eur. J. Pharmacol. , vol.600 , Issue.1 , pp. 78-82
    • Jin, F.1    Wu, Q.2    Lu, Y.-F.3    Gong, Q.-H.4    Shi, J.-S.5
  • 82
    • 0034637089 scopus 로고    scopus 로고
    • Ibuprofen protects dopaminergic neurons against glutamate toxicity in vitro
    • COI: 1:CAS:528:DC%2BD3cXmtVCqurw%3D, PID: 10961664
    • Casper D, Yaparpalvi U, Rempel N, Werner P (2000) Ibuprofen protects dopaminergic neurons against glutamate toxicity in vitro. Neurosci Lett 289(3):201–204
    • (2000) Neurosci. Lett. , vol.289 , Issue.3 , pp. 201-204
    • Casper, D.1    Yaparpalvi, U.2    Rempel, N.3    Werner, P.4
  • 83
    • 0029823573 scopus 로고    scopus 로고
    • Surgical treatment of Parkinson’s disease
    • COI: 1:STN:280:DyaK2s%2FhtVOmsw%3D%3D, PID: 8857784
    • Krauss JK, Jankovic J (1996) Surgical treatment of Parkinson’s disease. Am Fam Physician 54(5):1621–1629
    • (1996) Am Fam Physician , vol.54 , Issue.5 , pp. 1621-1629
    • Krauss, J.K.1    Jankovic, J.2
  • 84
    • 53149122871 scopus 로고    scopus 로고
    • Levodopa/dopamine replacement strategies in Parkinson’s disease—Future directions
    • PID: 18781663
    • Olanow CW (2008) Levodopa/dopamine replacement strategies in Parkinson’s disease—Future directions. Mov Disord 23:S613–S622. doi:10.1002/mds.22061
    • (2008) Mov Disord , vol.23 , pp. S613-S622
    • Olanow, C.W.1
  • 85
    • 33847105184 scopus 로고    scopus 로고
    • Advances in the treatment of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD2sXitFOrsLg%3D, PID: 17258379
    • Singh N, Pillay V, Choonara YE (2007) Advances in the treatment of Parkinson’s disease. Prog Neurobiol 81(1):29–44
    • (2007) Prog. Neurobiol. , vol.81 , Issue.1 , pp. 29-44
    • Singh, N.1    Pillay, V.2    Choonara, Y.E.3
  • 86
    • 0043127471 scopus 로고    scopus 로고
    • Future and current surgical therapies in Parkinson’s disease
    • PID: 12869808
    • Betchen SA, Kaplitt M (2003) Future and current surgical therapies in Parkinson’s disease. Curr Opin Neurol 16(4):487–493
    • (2003) Curr. Opin. Neurol. , vol.16 , Issue.4 , pp. 487-493
    • Betchen, S.A.1    Kaplitt, M.2
  • 88
    • 39749115543 scopus 로고    scopus 로고
    • Surgical management of parkinson’s disease: update and review
    • COI: 1:STN:280:DC%2BC3crjt1eqtg%3D%3D, PID: 20566105
    • Chao Y, Gang L, Na Z, Ming W, Zhong W, Mian W (2007) Surgical management of parkinson’s disease: update and review. Interv Neuroradiol 13(4):359
    • (2007) Interv Neuroradiol , vol.13 , Issue.4 , pp. 359
    • Chao, Y.1    Gang, L.2    Na, Z.3    Ming, W.4    Zhong, W.5    Mian, W.6
  • 89
    • 0023188416 scopus 로고    scopus 로고
    • Kelly PJ, Ahlskog J, Goerss SJ, Daube JR, Duffy JR, Kall BA Computer-assisted stereotactic ventralis lateralis thalamotomy with microelectrode recording control in patients with Parkinson’s disease. In: Mayo Clinic Proceedings, 1987. vol 8. Elsevier, pp 655–664
    • Kelly PJ, Ahlskog J, Goerss SJ, Daube JR, Duffy JR, Kall BA Computer-assisted stereotactic ventralis lateralis thalamotomy with microelectrode recording control in patients with Parkinson’s disease. In: Mayo Clinic Proceedings, 1987. vol 8. Elsevier, pp 655–664
  • 92
    • 3342984956 scopus 로고    scopus 로고
    • Cell transplantation for Parkinson’s disease: present status
    • PID: 15206816
    • Drucker-Colín R, Verdugo-Díaz L (2004) Cell transplantation for Parkinson’s disease: present status. Cell Mol Neurobiol 24(3):301–316
    • (2004) Cell. Mol. Neurobiol. , vol.24 , Issue.3 , pp. 301-316
    • Drucker-Colín, R.1    Verdugo-Díaz, L.2
  • 93
    • 21844473567 scopus 로고    scopus 로고
    • Cell therapy in Parkinson’s disease
    • PID: 15717042
    • Lindvall O, Björklund A (2004) Cell therapy in Parkinson’s disease. NeuroRx 1(4):382–393
    • (2004) NeuroRx , vol.1 , Issue.4 , pp. 382-393
    • Lindvall, O.1    Björklund, A.2
  • 95
    • 34547885762 scopus 로고    scopus 로고
    • Regeneration of the central nervous system using endogenous repair mechanisms
    • COI: 1:CAS:528:DC%2BD2sXhtVWnt7rM, PID: 17697047
    • Okano H, Sakaguchi M, Ohki K, Suzuki N, Sawamoto K (2007) Regeneration of the central nervous system using endogenous repair mechanisms. J Neurochem 102(5):1459–1465
    • (2007) J. Neurochem. , vol.102 , Issue.5 , pp. 1459-1465
    • Okano, H.1    Sakaguchi, M.2    Ohki, K.3    Suzuki, N.4    Sawamoto, K.5
  • 96
    • 84885128563 scopus 로고    scopus 로고
    • Autologous transplantation of GDNF-expressing mesenchymal stem cells protects against MPTP-induced damage in cynomolgus monkeys
    • Ren Z, Wang J, Wang S, Zou C, Li X, Guan Y, Chen Z, Zhang YA (2013) Autologous transplantation of GDNF-expressing mesenchymal stem cells protects against MPTP-induced damage in cynomolgus monkeys. Sci rep 3
    • (2013) Sci rep , pp. 3
    • Ren, Z.1    Wang, J.2    Wang, S.3    Zou, C.4    Li, X.5    Guan, Y.6    Chen, Z.7    Zhang, Y.A.8
  • 97
    • 84873124605 scopus 로고    scopus 로고
    • Neural stem cells transplanted in a mouse model of Parkinson’s disease differentiate to neuronal phenotypes and reduce rotational deficit
    • COI: 1:CAS:528:DC%2BC3sXhslGis78%3D, PID: 23131156
    • Ziavra D, Makri G, Giompres P, Taraviras S, Thomaidou D, Matsas R, Mitsacos A, Kouvelas ED (2012) Neural stem cells transplanted in a mouse model of Parkinson’s disease differentiate to neuronal phenotypes and reduce rotational deficit. CNS Neurol Disord Drug Targets 11(7):829–835
    • (2012) CNS Neurol Disord Drug Targets , vol.11 , Issue.7 , pp. 829-835
    • Ziavra, D.1    Makri, G.2    Giompres, P.3    Taraviras, S.4    Thomaidou, D.5    Matsas, R.6    Mitsacos, A.7    Kouvelas, E.D.8
  • 98
  • 99
    • 85019342064 scopus 로고    scopus 로고
    • Andersson KE: Effects of allogeneic bone marrow-derived mesenchymal stromal cell therapy on voiding function in a rat model of Parkinson’s disease. The Journal of urology
    • Campeau L, Soler R, Sittadjody S, Pareta R, Nomiya M, Zarifpour M, Opara EC, Yoo JJ, Andersson KE (2013) Effects of allogeneic bone marrow-derived mesenchymal stromal cell therapy on voiding function in a rat model of Parkinson’s disease. The Journal of urology
    • (2013) Yoo JJ
    • Campeau, L.1    Soler, R.2    Sittadjody, S.3    Pareta, R.4    Nomiya, M.5    Zarifpour, M.6    Opara, E.C.7
  • 100
    • 77952310482 scopus 로고    scopus 로고
    • Birth, Life and Death of Dopaminergic Neurons in the Substantia Nigra. Springer, In:
    • Yasuhara T, Date I (2009) Gene therapy for Parkinson’s disease. Birth, Life and Death of Dopaminergic Neurons in the Substantia Nigra. Springer, In, pp 301–309
    • (2009) Gene therapy for Parkinson’s disease , pp. 301-309
    • Yasuhara, T.1    Date, I.2
  • 101
    • 84879093348 scopus 로고    scopus 로고
    • Gene therapy for Parkinson’s disease: state-of-the-art treatments for neurodegenerative disease
    • COI: 1:CAS:528:DC%2BC3sXosl2rsLc%3D, PID: 23739006
    • Douglas MR (2013) Gene therapy for Parkinson’s disease: state-of-the-art treatments for neurodegenerative disease. Expert Rev Neurother 13(6):695–705
    • (2013) Expert. Rev. Neurother. , vol.13 , Issue.6 , pp. 695-705
    • Douglas, M.R.1
  • 102
    • 84861191461 scopus 로고    scopus 로고
    • Herpes simplex virus type 1-based amplicon vectors for fundamental research in neurosciences and gene therapy of neurological diseases
    • PID: 22108428
    • Jerusalinsky D, Baez MV, Epstein AL (2012) Herpes simplex virus type 1-based amplicon vectors for fundamental research in neurosciences and gene therapy of neurological diseases. J Physiol Paris 106(1–2):2–11. doi:10.1016/j.jphysparis.2011.11.003
    • (2012) J. Physiol. Paris , vol.106 , Issue.1-2 , pp. 2-11
    • Jerusalinsky, D.1    Baez, M.V.2    Epstein, A.L.3
  • 103
    • 59649116335 scopus 로고    scopus 로고
    • Positron emission tomography imaging demonstrates correlation between behavioral recovery and correction of dopamine neurotransmission after gene therapy
    • COI: 1:CAS:528:DC%2BD1MXhvFCru7k%3D, PID: 19193901
    • Leriche L, Bjorklund T, Breysse N, Besret L, Gregoire MC, Carlsson T, Dolle F, Mandel RJ, Deglon N, Hantraye P, Kirik D (2009) Positron emission tomography imaging demonstrates correlation between behavioral recovery and correction of dopamine neurotransmission after gene therapy. J Neurosci 29(5):1544–1553. doi:10.1523/jneurosci.4491-08.2009
    • (2009) J Neurosci , vol.29 , Issue.5 , pp. 1544-1553
    • Leriche, L.1    Bjorklund, T.2    Breysse, N.3    Besret, L.4    Gregoire, M.C.5    Carlsson, T.6    Dolle, F.7    Mandel, R.J.8    Deglon, N.9    Hantraye, P.10    Kirik, D.11
  • 104
    • 84880006379 scopus 로고    scopus 로고
    • AAV2-mediated striatum delivery of human CDNF prevents the deterioration of midbrain dopamine neurons in a 6-hydroxydopamine induced parkinsonian rat model
    • Ren X, Zhang T, Gong X, Hu G, Ding W, Wang X (2013) AAV2-mediated striatum delivery of human CDNF prevents the deterioration of midbrain dopamine neurons in a 6-hydroxydopamine induced parkinsonian rat model. Experimental neurology
    • (2013) Experimental neurology
    • Ren, X.1    Zhang, T.2    Gong, X.3    Hu, G.4    Ding, W.5    Wang, X.6
  • 107
    • 84856398475 scopus 로고    scopus 로고
    • Lifestyle and cognition. What do we know from the aging and neurodegenerative brain?
    • COI: 1:STN:280:DC%2BC38%2FlvV2gtQ%3D%3D, PID: 21842331
    • Steiner B, Witte V, Floel A (2011) Lifestyle and cognition. What do we know from the aging and neurodegenerative brain? Nervenarzt 82(12):1566–1577. doi:10.1007/s00115-011-3353-0
    • (2011) Nervenarzt , vol.82 , Issue.12 , pp. 1566-1577
    • Steiner, B.1    Witte, V.2    Floel, A.3
  • 108
    • 79955586938 scopus 로고    scopus 로고
    • Pilot dietary study with normoproteic protein-redistributed plant-food diet and motor performance in patients with Parkinson’s disease
    • COI: 1:CAS:528:DC%2BC3MXmslSru7k%3D, PID: 21535916
    • Baroni L, Bonetto C, Tessan F, Goldin D, Cenci L, Magnanini P, Zuliani G (2011) Pilot dietary study with normoproteic protein-redistributed plant-food diet and motor performance in patients with Parkinson’s disease. Nutr Neurosci 14(1):1–9. doi:10.1179/174313211x12966635733231
    • (2011) Nutr Neurosci , vol.14 , Issue.1 , pp. 1-9
    • Baroni, L.1    Bonetto, C.2    Tessan, F.3    Goldin, D.4    Cenci, L.5    Magnanini, P.6    Zuliani, G.7
  • 109
    • 67849116702 scopus 로고    scopus 로고
    • Ketogenic diet protects dopaminergic neurons against 6-OHDA neurotoxicity via up-regulating glutathione in a rat model of Parkinson’s disease
    • COI: 1:CAS:528:DC%2BD1MXps1Wlt78%3D, PID: 19559687
    • Cheng BH, Yang XX, An LX, Gao B, Liu X, Liu SW (2009) Ketogenic diet protects dopaminergic neurons against 6-OHDA neurotoxicity via up-regulating glutathione in a rat model of Parkinson’s disease. Brain Res 1286:25–31. doi:10.1016/j.brainres.2009.06.060
    • (2009) Brain Res. , vol.1286 , pp. 25-31
    • Cheng, B.H.1    Yang, X.X.2    An, L.X.3    Gao, B.4    Liu, X.5    Liu, S.W.6
  • 111
    • 0345227298 scopus 로고    scopus 로고
    • Motor training effects on recovery of function after striatal lesions and striatal grafts
    • PID: 14637098
    • Döbrössy MD, Dunnett SB (2003) Motor training effects on recovery of function after striatal lesions and striatal grafts. Exp Neurol 184(1):274–284
    • (2003) Exp. Neurol. , vol.184 , Issue.1 , pp. 274-284
    • Döbrössy, M.D.1    Dunnett, S.B.2
  • 112
    • 15544374721 scopus 로고    scopus 로고
    • Environmental enrichment in adulthood eliminates neuronal death in experimental Parkinsonism
    • COI: 1:CAS:528:DC%2BD2MXisFGmurk%3D, PID: 15790541
    • Faherty CJ, Raviie Shepherd K, Herasimtschuk A, Smeyne RJ (2005) Environmental enrichment in adulthood eliminates neuronal death in experimental Parkinsonism. Mol Brain Res 134(1):170–179
    • (2005) Mol. Brain Res. , vol.134 , Issue.1 , pp. 170-179
    • Faherty, C.J.1    Raviie Shepherd, K.2    Herasimtschuk, A.3    Smeyne, R.J.4
  • 113
    • 78649318267 scopus 로고    scopus 로고
    • Running exercise protects the substantia nigra dopaminergic neurons against inflammation-induced degeneration via the activation of BDNF signaling pathway
    • COI: 1:CAS:528:DC%2BC3cXhsVGrsLbP, PID: 20851176
    • Wu SY, Wang TF, Yu L, Jen CJ, Chuang JI, Wu FS, Wu CW, Kuo YM (2011) Running exercise protects the substantia nigra dopaminergic neurons against inflammation-induced degeneration via the activation of BDNF signaling pathway. Brain Behav Immun 25(1):135–146. doi:10.1016/j.bbi.2010.09.006
    • (2011) Brain Behav Immun , vol.25 , Issue.1 , pp. 135-146
    • Wu, S.Y.1    Wang, T.F.2    Yu, L.3    Jen, C.J.4    Chuang, J.I.5    Wu, F.S.6    Wu, C.W.7    Kuo, Y.M.8
  • 114
    • 0036602967 scopus 로고    scopus 로고
    • Exercise: a behavioral intervention to enhance brain health and plasticity
    • COI: 1:CAS:528:DC%2BD38Xks1ymtL8%3D, PID: 12086747
    • Cotman CW, Berchtold NC (2002) Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci 25(6):295–301
    • (2002) Trends Neurosci. , vol.25 , Issue.6 , pp. 295-301
    • Cotman, C.W.1    Berchtold, N.C.2
  • 115
    • 18144446836 scopus 로고    scopus 로고
    • Circulating insulin-like growth factor I mediates effects of exercise on the brain
    • COI: 1:CAS:528:DC%2BD3cXisVertb8%3D, PID: 10751445
    • Carro E, Nuñez A, Busiguina S, Torres-Aleman I (2000) Circulating insulin-like growth factor I mediates effects of exercise on the brain. J Neurosci 20(8):2926–2933
    • (2000) J. Neurosci. , vol.20 , Issue.8 , pp. 2926-2933
    • Carro, E.1    Nuñez, A.2    Busiguina, S.3    Torres-Aleman, I.4
  • 116
    • 79952087921 scopus 로고    scopus 로고
    • 9 −tetrahydrocannabinol (THC) protects against pentylenetetrazole (PTZ)-induced cognitive damage
    • COI: 1:CAS:528:DC%2BC3MXjvVyju7g%3D, PID: 21315768
    • 9 −tetrahydrocannabinol (THC) protects against pentylenetetrazole (PTZ)-induced cognitive damage. Behav Brain Res 220(1):194–201
    • (2011) Behav. Brain Res. , vol.220 , Issue.1 , pp. 194-201
    • Assaf, F.1    Fishbein, M.2    Gafni, M.3    Keren, O.4    Sarne, Y.5
  • 117
    • 36348984521 scopus 로고    scopus 로고
    • Evaluation of glutathione metabolism in NMDA preconditioning against quinolinic acid-induced seizures in mice cerebral cortex and hippocampus
    • COI: 1:CAS:528:DC%2BD2sXhtlyrs7bL, PID: 17980354
    • Vandresen-Filho S, de Araújo HB, Franco JL, Boeck CR, Dafre AL, Tasca CI (2007) Evaluation of glutathione metabolism in NMDA preconditioning against quinolinic acid-induced seizures in mice cerebral cortex and hippocampus. Brain Res 1184:38–45
    • (2007) Brain Res , vol.1184 , pp. 38-45
    • Vandresen-Filho, S.1    de Araújo, H.B.2    Franco, J.L.3    Boeck, C.R.4    Dafre, A.L.5    Tasca, C.I.6
  • 118
    • 78651390307 scopus 로고    scopus 로고
    • NMDA preconditioning protects against quinolinic acid-induced seizures via PKA, PI3K and MAPK/ERK signaling pathways
    • de Araújo HB, Vandresen-Filho S, Martins WC, Boeck CR, Tasca CI (2011) NMDA preconditioning protects against quinolinic acid-induced seizures via PKA, PI3K and MAPK/ERK signaling pathways. Behav Brain Res 219(1):92–97
    • (2011) Behav. Brain Res. , vol.219 , Issue.1 , pp. 92-97
    • de Araújo, H.B.1    Vandresen-Filho, S.2    Martins, W.C.3    Boeck, C.R.4    Tasca, C.I.5
  • 119
    • 76849083197 scopus 로고    scopus 로고
    • Behavioural and histological effects of preconditioning with lipopolysaccharide in epileptic rats
    • COI: 1:CAS:528:DC%2BD1MXhtVOmu7fK, PID: 19728087
    • Dmowska M, Cybulska R, Schoenborn R, Piersiak T, Jaworska-Adamu J, Gawron A (2010) Behavioural and histological effects of preconditioning with lipopolysaccharide in epileptic rats. Neurochem Res 35(2):262–272
    • (2010) Neurochem. Res. , vol.35 , Issue.2 , pp. 262-272
    • Dmowska, M.1    Cybulska, R.2    Schoenborn, R.3    Piersiak, T.4    Jaworska-Adamu, J.5    Gawron, A.6
  • 120
    • 79960286634 scopus 로고    scopus 로고
    • Lipopolysaccharide-induced preconditioning against ischemic injury is associated with changes in Toll-like receptor 4 expression in the rat developing brain
    • COI: 1:CAS:528:DC%2BC3MXntVygurs%3D, PID: 21659958
    • Hickey E, Shi H, Van Arsdell G, Askalan R (2011) Lipopolysaccharide-induced preconditioning against ischemic injury is associated with changes in Toll-like receptor 4 expression in the rat developing brain. Pediatr Res 70(1):10–14
    • (2011) Pediatr. Res. , vol.70 , Issue.1 , pp. 10-14
    • Hickey, E.1    Shi, H.2    Van Arsdell, G.3    Askalan, R.4
  • 121
    • 56349130733 scopus 로고    scopus 로고
    • Hippocampal transcriptome after status epilepticus in mice rendered seizure damage-tolerant by epileptic preconditioning features suppressed calcium and neuronal excitability pathways
    • COI: 1:CAS:528:DC%2BD1cXhsVCms7jE, PID: 18804535
    • Jimenez-Mateos EM, Hatazaki S, Johnson MB, Bellver-Estelles C, Mouri G, Bonner C, Prehn JH, Meller R, Simon RP, Henshall DC (2008) Hippocampal transcriptome after status epilepticus in mice rendered seizure damage-tolerant by epileptic preconditioning features suppressed calcium and neuronal excitability pathways. Neurobiol Dis 32(3):442–453
    • (2008) Neurobiol. Dis. , vol.32 , Issue.3 , pp. 442-453
    • Jimenez-Mateos, E.M.1    Hatazaki, S.2    Johnson, M.B.3    Bellver-Estelles, C.4    Mouri, G.5    Bonner, C.6    Prehn, J.H.7    Meller, R.8    Simon, R.P.9    Henshall, D.C.10
  • 122
    • 84865238053 scopus 로고    scopus 로고
    • Lipopolysaccharide-induced microglial activation and neuroprotection against experimental brain injury is independent of hematogenous TLR4
    • COI: 1:CAS:528:DC%2BC38Xht1Ortr%2FN, PID: 22915113
    • Chen Z, Jalabi W, Shpargel KB, Farabaugh KT, Dutta R, Yin X, Kidd GJ, Bergmann CC, Stohlman SA, Trapp BD (2012) Lipopolysaccharide-induced microglial activation and neuroprotection against experimental brain injury is independent of hematogenous TLR4. J Neurosci 32(34):11706–11715
    • (2012) J. Neurosci. , vol.32 , Issue.34 , pp. 11706-11715
    • Chen, Z.1    Jalabi, W.2    Shpargel, K.B.3    Farabaugh, K.T.4    Dutta, R.5    Yin, X.6    Kidd, G.J.7    Bergmann, C.C.8    Stohlman, S.A.9    Trapp, B.D.10
  • 123
    • 77955095337 scopus 로고    scopus 로고
    • Role of autophagy in protection afforded by hypoxic preconditioning against MPP + −induced neurotoxicity in SH-SY5Y cells
    • COI: 1:CAS:528:DC%2BC3cXptlCkt7w%3D, PID: 20541008
    • Tzeng YW, Lee LY, Chao PL, Lee HC, Wu RT, Lin AMY (2010) Role of autophagy in protection afforded by hypoxic preconditioning against MPP + −induced neurotoxicity in SH-SY5Y cells. Free Radic Biol Med 49(5):839–846. doi:10.1016/j.freeradbiomed.2010.06.004
    • (2010) Free Radic Biol Med , vol.49 , Issue.5 , pp. 839-846
    • Tzeng, Y.W.1    Lee, L.Y.2    Chao, P.L.3    Lee, H.C.4    Wu, R.T.5    Lin, A.M.Y.6
  • 124
    • 34447135619 scopus 로고    scopus 로고
    • Lipopolysaccharide induces early tolerance to excitotoxicity via nitric oxide and cGMP
    • COI: 1:CAS:528:DC%2BD2sXhtVKhtr3N, PID: 17761917
    • Orio M, Kunz A, Kawano T, Anrather J, Zhou P, Iadecola C (2007) Lipopolysaccharide induces early tolerance to excitotoxicity via nitric oxide and cGMP. Stroke 38(10):2812–2817. doi:10.1161/strokeaha.107.486837
    • (2007) Stroke , vol.38 , Issue.10 , pp. 2812-2817
    • Orio, M.1    Kunz, A.2    Kawano, T.3    Anrather, J.4    Zhou, P.5    Iadecola, C.6
  • 125
    • 68149180674 scopus 로고    scopus 로고
    • Ischemia-induced mitochondrial apoptosis is significantly attenuated by ischemic preconditioning
    • COI: 1:CAS:528:DC%2BD1MXptFCksrw%3D, PID: 19283470
    • Racay P, Chomova M, Tatarkova Z, Kaplan P, Hatok J, Dobrota D (2009) Ischemia-induced mitochondrial apoptosis is significantly attenuated by ischemic preconditioning. Cell Mol Neurobiol 29(6–7):901–908
    • (2009) Cell. Mol. Neurobiol. , vol.29 , Issue.6-7 , pp. 901-908
    • Racay, P.1    Chomova, M.2    Tatarkova, Z.3    Kaplan, P.4    Hatok, J.5    Dobrota, D.6
  • 126
    • 33846635975 scopus 로고    scopus 로고
    • Preconditioning reprograms the response to ischemic injury and primes the emergence of unique endogenous neuroprotective phenotypes a speculative synthesis
    • PID: 17261715
    • Stenzel-Poore MP, Stevens SL, King JS, Simon RP (2007) Preconditioning reprograms the response to ischemic injury and primes the emergence of unique endogenous neuroprotective phenotypes a speculative synthesis. Stroke 38(2):680–685
    • (2007) Stroke , vol.38 , Issue.2 , pp. 680-685
    • Stenzel-Poore, M.P.1    Stevens, S.L.2    King, J.S.3    Simon, R.P.4
  • 127
    • 59349116736 scopus 로고    scopus 로고
    • Toll-like receptor signaling in endogenous neuroprotection and stroke
    • COI: 1:CAS:528:DC%2BD1MXhsFKrt7c%3D, PID: 18809468
    • Marsh BJ, Williams-Karnesky RL, Stenzel-Poore MP (2009) Toll-like receptor signaling in endogenous neuroprotection and stroke. Neuroscience 158(3):1007–1020. doi:10.1016/j.neuroscience.2008.07.067
    • (2009) Neuroscience , vol.158 , Issue.3 , pp. 1007-1020
    • Marsh, B.J.1    Williams-Karnesky, R.L.2    Stenzel-Poore, M.P.3
  • 128
    • 79958248587 scopus 로고    scopus 로고
    • Multiple preconditioning paradigms converge on interferon regulatory factor-dependent signaling to promote tolerance to ischemic brain injury
    • COI: 1:CAS:528:DC%2BC3MXnsl2muro%3D, PID: 21653850
    • Stevens SL, Leung PY, Vartanian KB, Gopalan B, Yang T, Simon RP, Stenzel-Poore MP (2011) Multiple preconditioning paradigms converge on interferon regulatory factor-dependent signaling to promote tolerance to ischemic brain injury. J Neurosci 31(23):8456–8463. doi:10.1523/jneurosci.0821-11.2011
    • (2011) J Neurosci , vol.31 , Issue.23 , pp. 8456-8463
    • Stevens, S.L.1    Leung, P.Y.2    Vartanian, K.B.3    Gopalan, B.4    Yang, T.5    Simon, R.P.6    Stenzel-Poore, M.P.7
  • 129
    • 68549089184 scopus 로고    scopus 로고
    • Systemic lipopolysaccharide protects the brain from ischemic injury by reprogramming the response of the brain to stroke: a critical role for IRF3
    • COI: 1:CAS:528:DC%2BD1MXpvFOmurg%3D, PID: 19657036
    • Marsh B, Stevens SL, Packard AE, Gopalan B, Hunter B, Leung PY, Harrington CA, Stenzel-Poore MP (2009) Systemic lipopolysaccharide protects the brain from ischemic injury by reprogramming the response of the brain to stroke: a critical role for IRF3. J Neurosci 29(31):9839–9849
    • (2009) J. Neurosci. , vol.29 , Issue.31 , pp. 9839-9849
    • Marsh, B.1    Stevens, S.L.2    Packard, A.E.3    Gopalan, B.4    Hunter, B.5    Leung, P.Y.6    Harrington, C.A.7    Stenzel-Poore, M.P.8
  • 130
    • 80053974958 scopus 로고    scopus 로고
    • LPS preconditioning redirects TLR signaling following stroke: TRIF-IRF3 plays a seminal role in mediating tolerance to ischemic injury
    • COI: 1:CAS:528:DC%2BC3MXhs1OqtbbI, PID: 21999375
    • Vartanian KB, Stevens SL, Marsh BJ, Williams-Karnesky R, Lessov NS, Stenzel-Poore MP (2011) LPS preconditioning redirects TLR signaling following stroke: TRIF-IRF3 plays a seminal role in mediating tolerance to ischemic injury. J Neuroinflammation 8(1):140
    • (2011) J. Neuroinflammation , vol.8 , Issue.1 , pp. 140
    • Vartanian, K.B.1    Stevens, S.L.2    Marsh, B.J.3    Williams-Karnesky, R.4    Lessov, N.S.5    Stenzel-Poore, M.P.6
  • 132
    • 78349313801 scopus 로고    scopus 로고
    • Integrating multiple aspects of mitochondrial dynamics in neurons: age-related differences and dynamic changes in a chronic rotenone model
    • COI: 1:CAS:528:DC%2BC3cXhsVWqsLbM, PID: 20850532
    • Arnold B, Cassady SJ, VanLaar VS, Berman SB (2011) Integrating multiple aspects of mitochondrial dynamics in neurons: age-related differences and dynamic changes in a chronic rotenone model. Neurobiol Dis 41(1):189–200
    • (2011) Neurobiol. Dis. , vol.41 , Issue.1 , pp. 189-200
    • Arnold, B.1    Cassady, S.J.2    VanLaar, V.S.3    Berman, S.B.4
  • 133
    • 84855687153 scopus 로고    scopus 로고
    • Impaired mitochondrial biogenesis contributes to mitochondrial dysfunction in Alzheimer’s disease
    • COI: 1:CAS:528:DC%2BC38XitVKksbk%3D, PID: 22077634
    • Sheng B, Wang X, Su B, Hg L, Casadesus G, Perry G, Zhu X (2012) Impaired mitochondrial biogenesis contributes to mitochondrial dysfunction in Alzheimer’s disease. J Neurochem 120(3):419–429
    • (2012) J. Neurochem. , vol.120 , Issue.3 , pp. 419-429
    • Sheng, B.1    Wang, X.2    Su, B.3    Hg, L.4    Casadesus, G.5    Perry, G.6    Zhu, X.7
  • 134
    • 79952369773 scopus 로고    scopus 로고
    • Glycogen synthase kinase‐3 inhibition reduces ischemic cerebral damage, restores impaired mitochondrial biogenesis and prevents ROS production
    • COI: 1:CAS:528:DC%2BC3MXjvFWntL0%3D, PID: 21210815
    • Valerio A, Bertolotti P, Delbarba A, Perego C, Dossena M, Ragni M, Spano P, Carruba MO, De Simoni MG, Nisoli E (2011) Glycogen synthase kinase‐3 inhibition reduces ischemic cerebral damage, restores impaired mitochondrial biogenesis and prevents ROS production. J Neurochem 116(6):1148–1159
    • (2011) J. Neurochem. , vol.116 , Issue.6 , pp. 1148-1159
    • Valerio, A.1    Bertolotti, P.2    Delbarba, A.3    Perego, C.4    Dossena, M.5    Ragni, M.6    Spano, P.7    Carruba, M.O.8    De Simoni, M.G.9    Nisoli, E.10
  • 135
    • 84872790484 scopus 로고    scopus 로고
    • Mitochondrial quality, dynamics and functional capacity in Parkinson’s disease cybrid cell lines selected for Lewy body expression
    • COI: 1:CAS:528:DC%2BC3sXjsFyqt74%3D, PID: 23351342
    • Cronin-Furman EN, Borland MK, Bergquist KE, Bennett JP Jr, Trimmer PA (2013) Mitochondrial quality, dynamics and functional capacity in Parkinson’s disease cybrid cell lines selected for Lewy body expression. Mol Neurodegener 8(1):6
    • (2013) Mol. Neurodegener. , vol.8 , Issue.1 , pp. 6
    • Cronin-Furman, E.N.1    Borland, M.K.2    Bergquist, K.E.3    Bennett, J.P.4    Trimmer, P.A.5
  • 136
    • 71549164305 scopus 로고    scopus 로고
    • Cybrid models of Parkinson’s disease show variable mitochondrial biogenesis and genotype-respiration relationships
    • COI: 1:CAS:528:DC%2BD1MXhsVGnt73L, PID: 19815014
    • Keeney PM, Dunham LD, Quigley CK, Morton SL, Bergquist KE, Bennett JP (2009) Cybrid models of Parkinson’s disease show variable mitochondrial biogenesis and genotype-respiration relationships. Exp Neurol 220(2):374–382
    • (2009) Exp. Neurol. , vol.220 , Issue.2 , pp. 374-382
    • Keeney, P.M.1    Dunham, L.D.2    Quigley, C.K.3    Morton, S.L.4    Bergquist, K.E.5    Bennett, J.P.6
  • 137
    • 39849094082 scopus 로고    scopus 로고
    • Transient hypoxia stimulates mitochondrial biogenesis in brain subcortex by a neuronal nitric oxide synthase dependent mechanism
    • COI: 1:CAS:528:DC%2BD1cXjt1Cqt7o%3D, PID: 18305236
    • Gutsaeva DR, Carraway MS, Suliman HB, Demchenko IT, Shitara H, Yonekawa H, Piantadosi CA (2008) Transient hypoxia stimulates mitochondrial biogenesis in brain subcortex by a neuronal nitric oxide synthase dependent mechanism. J Neurosci 28(9):2015–2024. doi:10.1523/jneurosci.5654-07.2008
    • (2008) J Neurosci , vol.28 , Issue.9 , pp. 2015-2024
    • Gutsaeva, D.R.1    Carraway, M.S.2    Suliman, H.B.3    Demchenko, I.T.4    Shitara, H.5    Yonekawa, H.6    Piantadosi, C.A.7
  • 138
    • 84862782562 scopus 로고    scopus 로고
    • Exercise induces mitochondrial biogenesis after brain ischemia in rats
    • COI: 1:CAS:528:DC%2BC38XjsFKmu7o%3D, PID: 22266265
    • Zhang Q, Wu Y, Zhang P, Sha H, Jia J, Hu Y, Zhu J (2012) Exercise induces mitochondrial biogenesis after brain ischemia in rats. Neuroscience 205:10–17
    • (2012) Neuroscience , vol.205 , pp. 10-17
    • Zhang, Q.1    Wu, Y.2    Zhang, P.3    Sha, H.4    Jia, J.5    Hu, Y.6    Zhu, J.7
  • 139
    • 80053099024 scopus 로고    scopus 로고
    • Impaired mitochondrial biogenesis in hippocampi of rats with chronic seizures
    • COI: 1:CAS:528:DC%2BC3MXht1eitrnM, PID: 21854834
    • Han Y, Lin Y, Xie N, Xue Y, Tao H, Rui C, Xu J, Cao L, Liu X, Jiang H (2011) Impaired mitochondrial biogenesis in hippocampi of rats with chronic seizures. Neuroscience 194:234–240
    • (2011) Neuroscience , vol.194 , pp. 234-240
    • Han, Y.1    Lin, Y.2    Xie, N.3    Xue, Y.4    Tao, H.5    Rui, C.6    Xu, J.7    Cao, L.8    Liu, X.9    Jiang, H.10
  • 141
    • 55749114205 scopus 로고    scopus 로고
    • Rapidly increased neuronal mitochondrial biogenesis after hypoxic-ischemic brain injury
    • PID: 18723421
    • Yin W, Signore AP, Iwai M, Cao GD, Gao YQ, Chen J (2008) Rapidly increased neuronal mitochondrial biogenesis after hypoxic-ischemic brain injury. Stroke 39(11):3057–3063. doi:10.1161/strokeaha.108.520114
    • (2008) Stroke , vol.39 , Issue.11 , pp. 3057-3063
    • Yin, W.1    Signore, A.P.2    Iwai, M.3    Cao, G.D.4    Gao, Y.Q.5    Chen, J.6
  • 142
    • 33747587373 scopus 로고    scopus 로고
    • Resveratrol mimics ischemic preconditioning in the brain
    • COI: 1:CAS:528:DC%2BD28XptlCitrY%3D, PID: 16395277
    • Raval AP, Dave KR, Perez-Pinzon MA (2006) Resveratrol mimics ischemic preconditioning in the brain. J Cereb Blood Flow Metab 26(9):1141–1147. doi:10.1038/sj.jcbfm.9600262
    • (2006) J Cereb Blood Flow Metab , vol.26 , Issue.9 , pp. 1141-1147
    • Raval, A.P.1    Dave, K.R.2    Perez-Pinzon, M.A.3
  • 143
    • 48749087518 scopus 로고    scopus 로고
    • Lipopolysaccharide preconditioning induces protection against lipopolysaccharide-induced neurotoxicity in organotypic midbrain slice culture
    • COI: 1:CAS:528:DC%2BD1MXntFOmtr8%3D, PID: 18668149
    • Ding Y, Li L (2008) Lipopolysaccharide preconditioning induces protection against lipopolysaccharide-induced neurotoxicity in organotypic midbrain slice culture. Neurosci bull 24(4):209–218
    • (2008) Neurosci bull , vol.24 , Issue.4 , pp. 209-218
    • Ding, Y.1    Li, L.2
  • 144
    • 16444369209 scopus 로고    scopus 로고
    • Lipopolysaccharide induces both a primary and a secondary phase of sensitization in the developing rat brain
    • COI: 1:CAS:528:DC%2BD2MXlsVyjsLk%3D, PID: 15879289
    • Eklind S, Mallard C, Arvidsson P, Hagberg H (2005) Lipopolysaccharide induces both a primary and a secondary phase of sensitization in the developing rat brain. Pediatr Res 58(1):112–116
    • (2005) Pediatr. Res. , vol.58 , Issue.1 , pp. 112-116
    • Eklind, S.1    Mallard, C.2    Arvidsson, P.3    Hagberg, H.4
  • 145
    • 2342430991 scopus 로고    scopus 로고
    • Molecular mechanisms of endotoxin tolerance
    • COI: 1:CAS:528:DC%2BD2cXlvFWrur0%3D, PID: 15119998
    • Fan HK, Cook JA (2004) Molecular mechanisms of endotoxin tolerance. J Endotoxin Res 10(2):71–84. doi:10.1179/096805104225003997
    • (2004) J Endotoxin Res , vol.10 , Issue.2 , pp. 71-84
    • Fan, H.K.1    Cook, J.A.2
  • 148
    • 84869493412 scopus 로고    scopus 로고
    • Lipopolysaccharide-preconditioning protects against endotoxin-induced white matter injury in the neonatal rat brain
    • COI: 1:CAS:528:DC%2BC38XhsFymu7nN, PID: 23063716
    • Kumral A, Tuzun F, Ozbal S, Ergur BU, Yilmaz O, Duman N, Ozkan H (2012) Lipopolysaccharide-preconditioning protects against endotoxin-induced white matter injury in the neonatal rat brain. Brain Res 1489:81–89. doi:10.1016/j.brainres.2012.10.015
    • (2012) Brain Res. , vol.1489 , pp. 81-89
    • Kumral, A.1    Tuzun, F.2    Ozbal, S.3    Ergur, B.U.4    Yilmaz, O.5    Duman, N.6    Ozkan, H.7
  • 149
    • 77956873174 scopus 로고    scopus 로고
    • Maintenance of anti-inflammatory cytokines and reduction of glial activation in the ischemic hippocampal CA1 region preconditioned with lipopolysaccharide
    • COI: 1:CAS:528:DC%2BC3cXpsFOltL8%3D, PID: 20580380
    • Yu JT, Lee CH, Yoo KY, Choi JH, Li H, Park OK, Yan B, Hwang IK, Kwon YG, Kim YM, Won MH (2010) Maintenance of anti-inflammatory cytokines and reduction of glial activation in the ischemic hippocampal CA1 region preconditioned with lipopolysaccharide. J Neurol Sci 296(1–2):69–78. doi:10.1016/j.jns.2010.06.004
    • (2010) J Neurol Sci , vol.296 , Issue.1-2 , pp. 69-78
    • Yu, J.T.1    Lee, C.H.2    Yoo, K.Y.3    Choi, J.H.4    Li, H.5    Park, O.K.6    Yan, B.7    Hwang, I.K.8    Kwon, Y.G.9    Kim, Y.M.10    Won, M.H.11
  • 150
    • 0034768584 scopus 로고    scopus 로고
    • Ischemic preconditioning and lipopolysaccharide attenuate nuclear factor-kappa B activation and gene expression of inflammatory cytokines in the ischemia-reperfused rat heart
    • COI: 1:CAS:528:DC%2BD3MXosFCnsrg%3D
    • Hiasa G, Hamada M, Ikeda S, Hiwada K (2001) Ischemic preconditioning and lipopolysaccharide attenuate nuclear factor-kappa B activation and gene expression of inflammatory cytokines in the ischemia-reperfused rat heart. Jpn Circ J-English Edition 65(11):984–990. doi:10.1253/jcj.65.984
    • (2001) Jpn. Circ. J.-English Edition , vol.65 , Issue.11 , pp. 984-990
    • Hiasa, G.1    Hamada, M.2    Ikeda, S.3    Hiwada, K.4
  • 151
    • 77954177568 scopus 로고    scopus 로고
    • The Akt-endothelial nitric oxide synthase pathway in lipopolysaccharide preconditioning-induced hypoxic-ischemic tolerance in the neonatal rat brain
    • COI: 1:CAS:528:DC%2BC3cXot1WltL0%3D, PID: 20508195
    • Lin HY, Wu CL, Huang CC (2010) The Akt-endothelial nitric oxide synthase pathway in lipopolysaccharide preconditioning-induced hypoxic-ischemic tolerance in the neonatal rat brain. Stroke 41(7):1543–1551. doi:10.1161/strokeaha.109.574004
    • (2010) Stroke , vol.41 , Issue.7 , pp. 1543-1551
    • Lin, H.Y.1    Wu, C.L.2    Huang, C.C.3
  • 152
    • 77952956708 scopus 로고    scopus 로고
    • Microglial ablation and lipopolysaccharide preconditioning affects pilocarpine-induced seizures in mice
    • COI: 1:CAS:528:DC%2BC3cXmtFahtLY%3D, PID: 20382223
    • Mirrione MM, Konomos DK, Gravanis I, Dewey SL, Aguzzi A, Heppner FL, Tsirka SE (2010) Microglial ablation and lipopolysaccharide preconditioning affects pilocarpine-induced seizures in mice. Neurobiol Dis 39(1):85–97
    • (2010) Neurobiol. Dis. , vol.39 , Issue.1 , pp. 85-97
    • Mirrione, M.M.1    Konomos, D.K.2    Gravanis, I.3    Dewey, S.L.4    Aguzzi, A.5    Heppner, F.L.6    Tsirka, S.E.7
  • 154
    • 78650413857 scopus 로고    scopus 로고
    • Endoplasmic reticulum stress inducers provide protection against 6-hydroxydopamine-induced cytotoxicity
    • COI: 1:CAS:528:DC%2BC3cXhsF2jt73K, PID: 20974203
    • Hara H, Kamiya T, Adachi T (2011) Endoplasmic reticulum stress inducers provide protection against 6-hydroxydopamine-induced cytotoxicity. Neurochem Int 58(1):35–43
    • (2011) Neurochem. Int. , vol.58 , Issue.1 , pp. 35-43
    • Hara, H.1    Kamiya, T.2    Adachi, T.3
  • 155
    • 34548620779 scopus 로고    scopus 로고
    • Mechanism of 6-hydroxydopamine neurotoxicity: the role of NADPH oxidase and microglial activation in 6-hydroxydopamine-induced degeneration of dopaminergic neurons
    • COI: 1:CAS:528:DC%2BD2sXhtF2qu7bN, PID: 17573824
    • Rodriguez-Pallares J, Parga JA, Munoz A, Rey P, Guerra AJ, Labandeira-Garcia JL (2007) Mechanism of 6-hydroxydopamine neurotoxicity: the role of NADPH oxidase and microglial activation in 6-hydroxydopamine-induced degeneration of dopaminergic neurons. J Neurochem 103(1):145–156. doi:10.1111/j.1471-4159.2007.04699.x
    • (2007) J. Neurochem. , vol.103 , Issue.1 , pp. 145-156
    • Rodriguez-Pallares, J.1    Parga, J.A.2    Munoz, A.3    Rey, P.4    Guerra, A.J.5    Labandeira-Garcia, J.L.6
  • 156
    • 20444415298 scopus 로고    scopus 로고
    • Adaptation to hydrogen peroxide enhances PC12 cell tolerance against oxidative damage
    • COI: 1:CAS:528:DC%2BD2MXlt1Kisbs%3D, PID: 15878237
    • Chen ZH, Yoshida Y, Saito Y, Niki E (2005) Adaptation to hydrogen peroxide enhances PC12 cell tolerance against oxidative damage. Neurosci Lett 383(3):256–259. doi:10.1016/j.neulet.2005.04.022
    • (2005) Neurosci. Lett. , vol.383 , Issue.3 , pp. 256-259
    • Chen, Z.H.1    Yoshida, Y.2    Saito, Y.3    Niki, E.4
  • 157
    • 84865118338 scopus 로고    scopus 로고
    • Prolonged hyperoxia preconditioning attenuates behavioral symptoms of 6-hydroxydopamine-induced Parkinsonism
    • COI: 1:CAS:528:DC%2BC3sXhs1ertr0%3D, PID: 22781835
    • Hamidi GA, Faraji A, Zarmehri HA, Haghdoost-Yazdi H (2012) Prolonged hyperoxia preconditioning attenuates behavioral symptoms of 6-hydroxydopamine-induced Parkinsonism. Neurol Res 34(7):636–642. doi:10.1179/1743132812y.0000000056
    • (2012) Neurol Res , vol.34 , Issue.7 , pp. 636-642
    • Hamidi, G.A.1    Faraji, A.2    Zarmehri, H.A.3    Haghdoost-Yazdi, H.4
  • 158
    • 16644370665 scopus 로고    scopus 로고
    • Thrombin preconditioning provides protection in a 6-hydroxydopamine Parkinson’s disease model
    • COI: 1:CAS:528:DC%2BD2cXhtFGgs7zN, PID: 15619541
    • Cannon JR, Keep RF, Hua Y, Richardson RJ, Schallert T, Xi GH (2005) Thrombin preconditioning provides protection in a 6-hydroxydopamine Parkinson’s disease model. Neurosci Lett 373(3):189–194. doi:10.1016/j.neulet.2004.10.089
    • (2005) Neurosci. Lett. , vol.373 , Issue.3 , pp. 189-194
    • Cannon, J.R.1    Keep, R.F.2    Hua, Y.3    Richardson, R.J.4    Schallert, T.5    Xi, G.H.6
  • 159
    • 0036151857 scopus 로고    scopus 로고
    • Neuroprotection and neurodegenerative disease
    • PID: 11749009
    • Vajda FJ (2002) Neuroprotection and neurodegenerative disease. J Clin Neurosci 9(1):4–8
    • (2002) J Clin Neurosci , vol.9 , Issue.1 , pp. 4-8
    • Vajda, F.J.1
  • 160
    • 84856583697 scopus 로고    scopus 로고
    • The promise of neuroprotective agents in Parkinson’s disease
    • Seidl SE, Potashkin JA (2011) The promise of neuroprotective agents in Parkinson’s disease. Front Neurol 2
    • (2011) Front Neurol , pp. 2
    • Seidl, S.E.1    Potashkin, J.A.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.