Skip to main content

Advertisement

Log in

Defective Autophagy in Parkinson’s Disease: Role of Oxidative Stress

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Parkinson’s disease (PD) is a paradigmatic example of neurodegenerative disorder with a critical role of oxidative stress in its etiopathogenesis. Genetic susceptibility factors of PD, such as mutations in Parkin, PTEN-induced kinase 1, and DJ-1 as well as the exposure to pesticides and heavy metals, both contribute to altered redox balance and degeneration of dopaminergic neurons in the substantia nigra. Dysregulation of autophagy, a lysosomal-driven process of self degradation of cellular organelles and protein aggregates, is also implicated in PD and PD-related mutations, and environmental toxins deregulate autophagy. However, experimental evidence suggests a complex and ambiguous role of autophagy in PD since either impaired or abnormally upregulated autophagic flux has been shown to cause neuronal loss. Finally, it is generally believed that oxidative stress is a strong proautophagic stimulus. However, some evidence coming from neurobiology as well as from other fields indicate an inhibitory role of reactive oxygen species and reactive nitrogen species on the autophagic machinery. This review examines the scientific evidence supporting different concepts on how autophagy is dysregulated in PD and attempts to reconcile apparently contradictory views on the role of oxidative stress in autophagy regulation. The complex relationship between autophagy and oxidative stress is also considered in the context of the ongoing search for a novel PD therapy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Berry C, La Vecchia C, Nicotera P (2011) Paraquat and Parkinson’s disease. Cell Death Differ 17:1115–1125

    Article  CAS  Google Scholar 

  2. Lees AJ, Hardy J, Revesz T (2009) Parkinson’s disease. Lancet 373:2055–2066

    Article  PubMed  CAS  Google Scholar 

  3. Lipski J, Nistico R, Berretta N, Guatteo E, Bernardi G, Mercuri NB (2011) l-DOPA: a scapegoat for accelerated neurodegeneration in Parkinson’s disease? Prog Neurobiol 94:389–407

    Article  PubMed  CAS  Google Scholar 

  4. Pacher P, Beckman JS, Liaudet L (2007) Nitric oxide and peroxynitrite in health and disease. Physiol Rev 87:315–424

    Article  PubMed  CAS  Google Scholar 

  5. Malkus KA, Tsika E, Ischiropoulos H (2009) Oxidative modifications, mitochondrial dysfunction, and impaired protein degradation in Parkinson’s disease: how neurons are lost in the Bermuda triangle. Mol Neurodegener 4:24

    Article  PubMed  CAS  Google Scholar 

  6. Crifo C, Capuozzo E, Siems W, Salerno C (2005) Inhibition of ion transport ATPases by HNE. Biofactors 24:137–140

    Article  PubMed  CAS  Google Scholar 

  7. Yoritaka A, Hattori N, Uchida K, Tanaka M, Stadtman ER, Mizuno Y (1996) Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease. Proc Natl Acad Sci USA 93:2696–2701

    Article  PubMed  CAS  Google Scholar 

  8. Wong AS, Lee RH, Cheung AY, Yeung PK, Chung SK, Cheung ZH, Ip NY (2011) Cdk5-mediated phosphorylation of endophilin B1 is required for induced autophagy in models of Parkinson’s disease. Nat Cell Biol 13:568–579

    Article  PubMed  CAS  Google Scholar 

  9. Zhu JH, Horbinski C, Guo F, Watkins S, Uchiyama Y, Chu CT (2007) Regulation of autophagy by extracellular signal-regulated protein kinases during 1-methyl-4-phenylpyridinium-induced cell death. Am J Pathol 170:75–86

    Article  PubMed  CAS  Google Scholar 

  10. Castino R, Lazzeri G, Lenzi P, Bellio N, Follo C, Ferrucci M, Fornai F, Isidoro C (2008) Suppression of autophagy precipitates neuronal cell death following low doses of methamphetamine. J Neurochem 106:1426–1439

    Article  PubMed  CAS  Google Scholar 

  11. Castino R, Bellio N, Follo C, Murphy D, Isidoro C (2010) Inhibition of PI3k class III-dependent autophagy prevents apoptosis and necrosis by oxidative stress in dopaminergic neuroblastoma cells. Toxicol Sci 117:152–162

    Article  PubMed  CAS  Google Scholar 

  12. Cheung ZH, Ip NY (2011) Autophagy deregulation in neurodegenerative diseases—recent advances and future perspectives. J Neurochem 118:317–325

    Article  PubMed  CAS  Google Scholar 

  13. Nistico R, Mehdawy B, Piccirilli S, Mercuri N (2011) Paraquat- and rotenone-induced models of Parkinson’s disease. Int J Immunopathol Pharmacol 24:313–322

    PubMed  CAS  Google Scholar 

  14. He C, Klionsky DJ (2009) Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43:67–93

    Article  PubMed  CAS  Google Scholar 

  15. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, Tanaka K, Cuervo AM, Czaja MJ (2009) Autophagy regulates lipid metabolism. Nature 458:1131–1135

    Article  PubMed  CAS  Google Scholar 

  16. Harris H, Rubinsztein DC (2011) Control of autophagy as a therapy for neurodegenerative disease. Nat Rev Neurol 8:108–117

    Article  PubMed  CAS  Google Scholar 

  17. Chen N, Karantza V (2011) Autophagy as a therapeutic target in cancer. Cancer Biol Ther 11:157–168

    Article  PubMed  CAS  Google Scholar 

  18. Levine B, Mizushima N, Virgin HW (2011) Autophagy in immunity and inflammation. Nature 469:323–335

    Article  PubMed  CAS  Google Scholar 

  19. Dong H, Czaja MJ (2011) Regulation of lipid droplets by autophagy. Trends Endocrinol Metab 22:234–240

    Article  PubMed  CAS  Google Scholar 

  20. Li L, Zhang X, Le W (2010) Autophagy dysfunction in Alzheimer’s disease. Neurodegener Dis 7:265–271

    PubMed  Google Scholar 

  21. Chen S, Zhang X, Song L, Le W (2012) Autophagy dysregulation in amyotrophic lateral sclerosis. Brain Pathol 22:110–116

    Article  PubMed  CAS  Google Scholar 

  22. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E, Tanaka K (2006) Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 441:880–884

    Article  PubMed  CAS  Google Scholar 

  23. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N (2006) Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441:885–889

    Article  PubMed  CAS  Google Scholar 

  24. Hayashi-Nishino M, Fujita N, Noda T, Yamaguchi A, Yoshimori T, Yamamoto A (2009) A subdomain of the endoplasmic reticulum forms a cradle for autophagosome formation. Nat Cell Biol 11:1433–1437

    Article  PubMed  CAS  Google Scholar 

  25. Matsunaga K, Saitoh T, Tabata K, Omori H, Satoh T, Kurotori N, Maejima I, Shirahama-Noda K, Ichimura T, Isobe T, Akira S, Noda T, Yoshimori T (2009) Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat Cell Biol 11:385–396

    Article  PubMed  CAS  Google Scholar 

  26. Yang Z, Klionsky DJ (2010) Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol 22:124–131

    Article  PubMed  CAS  Google Scholar 

  27. Koga H, Cuervo AM (2011) Chaperone-mediated autophagy dysfunction in the pathogenesis of neurodegeneration. Neurobiol Dis 43:29–37

    Article  PubMed  CAS  Google Scholar 

  28. Arias E, Cuervo AM (2011) Chaperone-mediated autophagy in protein quality control. Curr Opin Cell Biol 23:184–189

    Article  PubMed  CAS  Google Scholar 

  29. Mijaljica D, Prescott M, Devenish RJ (2011) Microautophagy in mammalian cells: revisiting a 40-year-old conundrum. Autophagy 7:673–682

    Article  PubMed  CAS  Google Scholar 

  30. Perry TL, Yong VW (1986) Idiopathic Parkinson’s disease, progressive supranuclear palsy and glutathione metabolism in the substantia nigra of patients. Neurosci Lett 67:269–274

    Article  PubMed  CAS  Google Scholar 

  31. Sofic E, Sapcanin A, Tahirovic I, Gavrankapetanovic I, Jellinger K, Reynolds GP, Tatschner T, Riederer P (2006) Antioxidant capacity in postmortem brain tissues of Parkinson’s and Alzheimer’s diseases. J Neural Transm Suppl (71):39–43

  32. Saggu H, Cooksey J, Dexter D, Wells FR, Lees A, Jenner P, Marsden CD (1989) A selective increase in particulate superoxide dismutase activity in parkinsonian substantia nigra. J Neurochem 53:692–697

    Article  PubMed  CAS  Google Scholar 

  33. Yoritaka A, Hattori N, Mori H, Kato K, Mizuno Y (1997) An immunohistochemical study on manganese superoxide dismutase in Parkinson’s disease. J Neurol Sci 148:181–186

    Article  PubMed  CAS  Google Scholar 

  34. Schapira AH, Mann VM, Cooper JM, Dexter D, Daniel SE, Jenner P, Clark JB, Marsden CD (1990) Anatomic and disease specificity of NADH CoQ1 reductase (complex I) deficiency in Parkinson’s disease. J Neurochem 55:2142–2145

    Article  PubMed  CAS  Google Scholar 

  35. Schapira AH, Cooper JM, Dexter D, Clark JB, Jenner P, Marsden CD (1990) Mitochondrial complex I deficiency in Parkinson’s disease. J Neurochem 54:823–827

    Article  PubMed  CAS  Google Scholar 

  36. Dexter DT, Carter CJ, Wells FR, Javoy-Agid F, Agid Y, Lees A, Jenner P, Marsden CD (1989) Basal lipid peroxidation in substantia nigra is increased in Parkinson’s disease. J Neurochem 52:381–389

    Article  PubMed  CAS  Google Scholar 

  37. Alam ZI, Jenner A, Daniel SE, Lees AJ, Cairns N, Marsden CD, Jenner P, Halliwell B (1997) Oxidative DNA damage in the parkinsonian brain: an apparent selective increase in 8-hydroxyguanine levels in substantia nigra. J Neurochem 69:1196–1203

    Article  PubMed  CAS  Google Scholar 

  38. Hastings TG (2009) The role of dopamine oxidation in mitochondrial dysfunction: implications for Parkinson’s disease. J Bioenerg Biomembr 41:469–472

    Article  PubMed  CAS  Google Scholar 

  39. Jinsmaa Y, Florang VR, Rees JN, Mexas LM, Eckert LL, Allen EM, Anderson DG, Doorn JA (2011) Dopamine-derived biological reactive intermediates and protein modifications: implications for Parkinson’s disease. Chem Biol Interact 192:118–121

    Article  PubMed  CAS  Google Scholar 

  40. Haavik J, Almas B, Flatmark T (1997) Generation of reactive oxygen species by tyrosine hydroxylase: a possible contribution to the degeneration of dopaminergic neurons? J Neurochem 68:328–332

    Article  PubMed  CAS  Google Scholar 

  41. Gluck MR, Zeevalk GD (2004) Inhibition of brain mitochondrial respiration by dopamine and its metabolites: implications for Parkinson’s disease and catecholamine-associated diseases. J Neurochem 91:788–795

    Article  PubMed  CAS  Google Scholar 

  42. Guzman JN, Sanchez-Padilla J, Wokosin D, Kondapalli J, Ilijic E, Schumacker PT, Surmeier DJ (2010) Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature 468:696–700

    Article  PubMed  CAS  Google Scholar 

  43. McCoy MK, Cookson MR (2011) DJ-1 regulation of mitochondrial function and autophagy through oxidative stress. Autophagy 7:531–532

    Article  PubMed  CAS  Google Scholar 

  44. Surmeier DJ, Guzman JN, Sanchez-Padilla J, Goldberg JA (2011) The origins of oxidant stress in Parkinson’s disease and therapeutic strategies. Antioxid Redox Signal 14:1289–1301

    Article  PubMed  CAS  Google Scholar 

  45. Tansey MG, Goldberg MS (2011) Neuroinflammation in Parkinson’s disease: its role in neuronal death and implications for therapeutic intervention. Neurobiol Dis 37:510–518

    Article  CAS  Google Scholar 

  46. Janda E, Visalli V, Colica C, Aprigliano S, Musolino V, Vadala N, Muscoli C, Sacco I, Iannone M, Rotiroti D, Spedding M, Mollace V (2011) The protective effect of tianeptine on Gp120-induced apoptosis in astroglial cells: role of GS and NOS, and NF-kappaB suppression. Br J Pharmacol 164:1590–1599

    Article  PubMed  CAS  Google Scholar 

  47. Mollace V, Iannone M, Muscoli C, Palma E, Granato T, Rispoli V, Nistico R, Rotiroti D, Salvemini D (2003) The role of oxidative stress in paraquat-induced neurotoxicity in rats: protection by non peptidyl superoxide dismutase mimetic. Neurosci Lett 335:163–166

    Article  PubMed  CAS  Google Scholar 

  48. Gao HM, Zhou H, Zhang F, Wilson BC, Kam W, Hong JS (2011) HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration. J Neurosci 31:1081–1092

    Article  PubMed  CAS  Google Scholar 

  49. Mollace V, Colasanti M, Rodino P, Massoud R, Lauro GM, Nistico G (1993) Cytokine-induced nitric oxide generation by cultured astrocytoma cells involves Ca(++)-calmodulin-independent NO-synthase. Biochem Biophys Res Commun 191:327–334

    Article  PubMed  CAS  Google Scholar 

  50. Muscoli C, Visalli V, Colica C, Nistico R, Palma E, Costa N, Rotiroti D, Nistico G, Mollace V (2005) The effect of inflammatory stimuli on NMDA-related activation of glutamine synthase in human cultured astroglial cells. Neurosci Lett 373:184–188

    Article  PubMed  CAS  Google Scholar 

  51. Terada S, Ishizu H, Yokota O, Tsuchiya K, Nakashima H, Ishihara T, Fujita D, Ueda K, Ikeda K, Kuroda S (2003) Glial involvement in diffuse Lewy body disease. Acta Neuropathol 105:163–169

    PubMed  CAS  Google Scholar 

  52. Piao YS, Wakabayashi K, Hayashi S, Yoshimoto M, Takahashi H (2000) Aggregation of alpha-synuclein/NACP in the neuronal and glial cells in diffuse Lewy body disease: a survey of six patients. Clin Neuropathol 19:163–169

    PubMed  CAS  Google Scholar 

  53. Wakabayashi K, Hayashi S, Yoshimoto M, Kudo H, Takahashi H (2000) NACP/alpha-synuclein-positive filamentous inclusions in astrocytes and oligodendrocytes of Parkinson’s disease brains. Acta Neuropathol 99:14–20

    Article  PubMed  CAS  Google Scholar 

  54. Gu XL, Long CX, Sun L, Xie C, Lin X, Cai H (2010) Astrocytic expression of Parkinson’s disease-related A53T alpha-synuclein causes neurodegeneration in mice. Mol Brain 3:12

    Article  PubMed  CAS  Google Scholar 

  55. Sidoryk-Wegrzynowicz M, Wegrzynowicz M, Lee E, Bowman AB, Aschner M (2011) Role of astrocytes in brain function and disease. Toxicol Pathol 39:115–123

    Article  PubMed  Google Scholar 

  56. Miyazaki I, Asanuma M, Kikkawa Y, Takeshima M, Murakami S, Miyoshi K, Sogawa N, Kita T (2011) Astrocyte-derived metallothionein protects dopaminergic neurons from dopamine quinone toxicity. Glia 59:435–451

    Article  PubMed  Google Scholar 

  57. Drechsel DA, Patel M (2009) Differential contribution of the mitochondrial respiratory chain complexes to reactive oxygen species production by redox cycling agents implicated in parkinsonism. Toxicol Sci 112:427–434

    Article  PubMed  CAS  Google Scholar 

  58. Crifo C, Siems W, Soro S, Salerno C (2005) Inhibition of defective adenylosuccinate lyase by HNE: a neurological disease that may be affected by oxidative stress. Biofactors 24:131–136

    Article  PubMed  CAS  Google Scholar 

  59. Danielson SR, Andersen JK (2008) Oxidative and nitrative protein modifications in Parkinson’s disease. Free Radic Biol Med 44:1787–1794

    Article  PubMed  CAS  Google Scholar 

  60. Janda E, Parafati M, Aprigliano S, Carresi C, Visalli V, Sacco I, Ventrice D, Mega T, Vadalá N, Rinaldi S, Musolino V, Palma E, Gratteri S, Rotiroti D, Mollace V (2012) The antidote effect of quinone oxidoreductase 2 (QR2) inhibitor on paraquat-induced toxicity in vitro and in vivo. Br J Pharmacol. Epub 2012 Jan 31

  61. Fu Y, Buryanovskyy L, Zhang Z (2008) Quinone reductase 2 is a catechol quinone reductase. J Biol Chem 283:23829–23835

    Article  PubMed  CAS  Google Scholar 

  62. Nuytemans K, Theuns J, Cruts M, Van Broeckhoven C (2010) Genetic etiology of Parkinson disease associated with mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation update. Hum Mutat 31:763–780

    Article  PubMed  CAS  Google Scholar 

  63. Isidoro C, Biagioni F, Giorgi FS, Fulceri F, Paparelli A, Fornai F (2009) The role of autophagy on the survival of dopamine neurons. Curr Top Med Chem 9:869–879

    PubMed  CAS  Google Scholar 

  64. Hardy J (2010) Genetic analysis of pathways to Parkinson disease. Neuron 68:201–206

    Article  PubMed  CAS  Google Scholar 

  65. Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioannidis JP, Kruger R, Rocca WA, Schneider NK, Lesnick TG, Lincoln SJ, Hulihan MM, Aasly JO, Ashizawa T, Chartier-Harlin MC, Checkoway H, Ferrarese C, Hadjigeorgiou G, Hattori N, Kawakami H, Lambert JC, Lynch T, Mellick GD, Papapetropoulos S, Parsian A, Quattrone A, Riess O, Tan EK, Van Broeckhoven C (2006) Collaborative analysis of alpha-synuclein gene promoter variability and Parkinson disease. JAMA 296:661–670

    Article  PubMed  CAS  Google Scholar 

  66. Bartels T, Choi JG, Selkoe DJ (2011) alpha-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature 477:107–110

    Article  PubMed  CAS  Google Scholar 

  67. Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D (2004) Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 305:1292–1295

    Article  PubMed  CAS  Google Scholar 

  68. Winslow AR, Chen CW, Corrochano S, Acevedo-Arozena A, Gordon DE, Peden AA, Lichtenberg M, Menzies FM, Ravikumar B, Imarisio S, Brown S, O’Kane CJ, Rubinsztein DC (2011) Alpha-synuclein impairs macroautophagy: implications for Parkinson’s disease. J Cell Biol 190:1023–1037

    Article  CAS  Google Scholar 

  69. Xilouri M, Vogiatzi T, Vekrellis K, Park D, Stefanis L (2009) Abberant alpha-synuclein confers toxicity to neurons in part through inhibition of chaperone-mediated autophagy. PLoS One 4:e5515

    Article  PubMed  CAS  Google Scholar 

  70. Outeiro TF, Klucken J, Bercury K, Tetzlaff J, Putcha P, Oliveira LM, Quintas A, McLean PJ, Hyman BT (2009) Dopamine-induced conformational changes in alpha-synuclein. PLoS One 4:e6906

    Article  PubMed  CAS  Google Scholar 

  71. Martinez-Vicente M, Talloczy Z, Kaushik S, Massey AC, Mazzulli J, Mosharov EV, Hodara R, Fredenburg R, Wu DC, Follenzi A, Dauer W, Przedborski S, Ischiropoulos H, Lansbury PT, Sulzer D, Cuervo AM (2008) Dopamine-modified alpha-synuclein blocks chaperone-mediated autophagy. J Clin Invest 118:777–788

    PubMed  CAS  Google Scholar 

  72. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N (1998) Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 392:605–608

    Article  PubMed  CAS  Google Scholar 

  73. Narendra D, Tanaka A, Suen DF, Youle RJ (2008) Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol 183:795–803

    Article  PubMed  CAS  Google Scholar 

  74. Vives-Bauza C, Zhou C, Huang Y, Cui M, de Vries RL, Kim J, May J, Tocilescu MA, Liu W, Ko HS, Magrane J, Moore DJ, Dawson VL, Grailhe R, Dawson TM, Li C, Tieu K, Przedborski S (2010) PINK1-dependent recruitment of Parkin to mitochondria in mitophagy. Proc Natl Acad Sci USA 107:378–383

    Article  PubMed  CAS  Google Scholar 

  75. Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier CA, Sou YS, Saiki S, Kawajiri S, Sato F, Kimura M, Komatsu M, Hattori N, Tanaka K (2010) PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol 189:211–221

    Article  PubMed  CAS  Google Scholar 

  76. Narendra DP, SM J, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, Youle RJ (2010) PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol 8:e1000298

    Article  PubMed  CAS  Google Scholar 

  77. Kitada T, Asakawa S, Minoshima S, Mizuno Y, Shimizu N (2000) Molecular cloning, gene expression, and identification of a splicing variant of the mouse parkin gene. Mamm Genome 11:417–421

    Article  PubMed  CAS  Google Scholar 

  78. Gegg ME, Cooper JM, Chau KY, Rojo M, Schapira AH, Taanman JW (2010) Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/Parkin-dependent manner upon induction of mitophagy. Hum Mol Genet 19:4861–4870

    Article  PubMed  CAS  Google Scholar 

  79. Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, Springer W (2010) PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol 12:119–131

    Article  PubMed  CAS  Google Scholar 

  80. Poole AC, Thomas RE, Yu S, Vincow ES, Pallanck L (2010) The mitochondrial fusion-promoting factor mitofusin is a substrate of the PINK1/parkin pathway. PLoS One 5:e10054

    Article  PubMed  CAS  Google Scholar 

  81. Ziviani E, Tao RN, Whitworth AJ (2010) Drosophila parkin requires PINK1 for mitochondrial translocation and ubiquitinates mitofusin. Proc Natl Acad Sci USA 107:5018–5023

    Article  PubMed  CAS  Google Scholar 

  82. Michiorri S, Gelmetti V, Giarda E, Lombardi F, Romano F, Marongiu R, Nerini-Molteni S, Sale P, Vago R, Arena G, Torosantucci L, Cassina L, Russo MA, Dallapiccola B, Valente EM, Casari G (2010) The Parkinson-associated protein PINK1 interacts with Beclin1 and promotes autophagy. Cell Death Differ 17:962–974

    Article  PubMed  CAS  Google Scholar 

  83. Khandelwal PJ, Herman AM, Hoe HS, Rebeck GW, Moussa CE (2011) Parkin mediates Beclin-dependent autophagic clearance of defective mitochondria and ubiquitinated Abeta in AD models. Hum Mol Genet 20:2091–2102

    Article  PubMed  CAS  Google Scholar 

  84. Palacino JJ, Sagi D, Goldberg MS, Krauss S, Motz C, Wacker M, Klose J, Shen J (2004) Mitochondrial dysfunction and oxidative damage in Parkin-deficient mice. J Biol Chem 279:18614–18622

    Article  PubMed  CAS  Google Scholar 

  85. Pesah Y, Pham T, Burgess H, Middlebrooks B, Verstreken P, Zhou Y, Harding M, Bellen H, Mardon G (2004) Drosophila parkin mutants have decreased mass and cell size and increased sensitivity to oxygen radical stress. Development 131:2183–2194

    Article  PubMed  CAS  Google Scholar 

  86. Kabuta T, Furuta A, Aoki S, Furuta K, Wada K (2008) Aberrant interaction between Parkinson disease-associated mutant UCH-L1 and the lysosomal receptor for chaperone-mediated autophagy. J Biol Chem 283:23731–23738

    Article  PubMed  CAS  Google Scholar 

  87. Leroy E, Boyer R, Auburger G, Leube B, Ulm G, Mezey E, Harta G, Brownstein MJ, Jonnalagada S, Chernova T, Dehejia A, Lavedan C, Gasser T, Steinbach PJ, Wilkinson KD, Polymeropoulos MH (1998) The ubiquitin pathway in Parkinson’s disease. Nature 395:451–452

    Article  PubMed  CAS  Google Scholar 

  88. Setsuie R, Wang YL, Mochizuki H, Osaka H, Hayakawa H, Ichihara N, Li H, Furuta A, Sano Y, Sun YJ, Kwon J, Kabuta T, Yoshimi K, Aoki S, Mizuno Y, Noda M, Wada K (2007) Dopaminergic neuronal loss in transgenic mice expressing the Parkinson’s disease-associated UCH-L1 I93M mutant. Neurochem Int 50:119–129

    Article  PubMed  CAS  Google Scholar 

  89. Kabuta T, Setsuie R, Mitsui T, Kinugawa A, Sakurai M, Aoki S, Uchida K, Wada K (2008) Aberrant molecular properties shared by familial Parkinson’s disease-associated mutant UCH-L1 and carbonyl-modified UCH-L1. Hum Mol Genet 17:1482–1496

    Article  PubMed  CAS  Google Scholar 

  90. Liu Y, Fallon L, Lashuel HA, Liu Z, Lansbury PT Jr (2002) The UCH-L1 gene encodes two opposing enzymatic activities that affect alpha-synuclein degradation and Parkinson’s disease susceptibility. Cell 111:209–218

    Article  PubMed  CAS  Google Scholar 

  91. Choi J, Levey AI, Weintraub ST, Rees HD, Gearing M, Chin LS, Li L (2004) Oxidative modifications and down-regulation of ubiquitin carboxyl-terminal hydrolase L1 associated with idiopathic Parkinson’s and Alzheimer’s diseases. J Biol Chem 279:13256–13264

    Article  PubMed  CAS  Google Scholar 

  92. Kahle PJ, Waak J, Gasser T (2009) DJ-1 and prevention of oxidative stress in Parkinson’s disease and other age-related disorders. Free Radic Biol Med 47:1354–1361

    Article  PubMed  CAS  Google Scholar 

  93. Krebiehl G, Ruckerbauer S, Burbulla LF, Kieper N, Maurer B, Waak J, Wolburg H, Gizatullina Z, Gellerich FN, Woitalla D, Riess O, Kahle PJ, Proikas-Cezanne T, Kruger R (2010) Reduced basal autophagy and impaired mitochondrial dynamics due to loss of Parkinson’s disease-associated protein DJ-1. PLoS One 5:e9367

    Article  PubMed  CAS  Google Scholar 

  94. Irrcher I, Aleyasin H, Seifert EL, Hewitt SJ, Chhabra S, Phillips M, Lutz AK, Rousseaux MW, Bevilacqua L, Jahani-Asl A, Callaghan S, MacLaurin JG, Winklhofer KF, Rizzu P, Rippstein P, Kim RH, Chen CX, Fon EA, Slack RS, Harper ME, McBride HM, Mak TW, Park DS (2010) Loss of the Parkinson’s disease-linked gene DJ-1 perturbs mitochondrial dynamics. Hum Mol Genet 19:3734–3746

    Article  PubMed  CAS  Google Scholar 

  95. MacLeod D, Dowman J, Hammond R, Leete T, Inoue K, Abeliovich A (2006) The familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron 52:587–593

    Article  PubMed  CAS  Google Scholar 

  96. Alegre-Abarrategui J, Christian H, Lufino MM, Mutihac R, Venda LL, Ansorge O, Wade-Martins R (2009) LRRK2 regulates autophagic activity and localizes to specific membrane microdomains in a novel human genomic reporter cellular model. Hum Mol Genet 18:4022–4034

    Article  PubMed  CAS  Google Scholar 

  97. Plowey ED, Chu CT (2010) Synaptic dysfunction in genetic models of Parkinson’s disease: a role for autophagy? Neurobiol Dis 43:60–67

    Article  PubMed  CAS  Google Scholar 

  98. Cherra SJ 3rd, Kulich SM, Uechi G, Balasubramani M, Mountzouris J, Day BW, Chu CT (2011) Regulation of the autophagy protein LC3 by phosphorylation. J Cell Biol 190:533–539

    Article  CAS  Google Scholar 

  99. Pienaar IS, Burn D, Morris C, Dexter D (2011) Synaptic protein alterations in Parkinson’s disease. Mol Neurobiol 45:126–143

    Article  PubMed  CAS  Google Scholar 

  100. Chan D, Citro A, Cordy JM, Shen GC, Wolozin B (2011) Rac1 protein rescues neurite retraction caused by G2019S leucine-rich repeat kinase 2 (LRRK2). J Biol Chem 286:16140–16149

    Article  PubMed  CAS  Google Scholar 

  101. Wang A, Costello S, Cockburn M, Zhang X, Bronstein J, Ritz B (2011) Parkinson’s disease risk from ambient exposure to pesticides. Eur J Epidemiol 26:547–555

    Article  PubMed  CAS  Google Scholar 

  102. Cicchetti F, Drouin-Ouellet J, Gross RE (2009) Environmental toxins and Parkinson’s disease: what have we learned from pesticide-induced animal models? Trends Pharmacol Sci 30:475–483

    Article  PubMed  CAS  Google Scholar 

  103. Duty S, Jenner P (2011) Animal models of Parkinson’s disease: a source of novel treatments and clues to the cause of the disease. Br J Pharmacol 164:1357–1391

    Article  PubMed  CAS  Google Scholar 

  104. Nicklas WJ, Vyas I, Heikkila RE (1985) Inhibition of NADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life Sci 36:2503–2508

    Article  PubMed  CAS  Google Scholar 

  105. Dehay B, Bove J, Rodriguez-Muela N, Perier C, Recasens A, Boya P, Vila M (2010) Pathogenic lysosomal depletion in Parkinson’s disease. J Neurosci 30:12535–12544

    Article  PubMed  CAS  Google Scholar 

  106. Cai ZL, Shi JJ, Yang YP, Cao BY, Wang F, Huang JZ, Yang F, Zhang P, Liu CF (2009) MPP+ impairs autophagic clearance of alpha-synuclein by impairing the activity of dynein. Neuroreport 20:569–573

    Article  PubMed  CAS  Google Scholar 

  107. Wong AS, Cheung ZH, Ip NY (2011) Molecular machinery of macroautophagy and its deregulation in diseases. Biochim Biophys Acta 1812:1490–1497

    Article  PubMed  CAS  Google Scholar 

  108. Chen Y, McMillan-Ward E, Kong J, Israels SJ, Gibson SB (2008) Oxidative stress induces autophagic cell death independent of apoptosis in transformed and cancer cells. Cell Death Differ 15:171–182

    Article  PubMed  CAS  Google Scholar 

  109. Chu CT, Zhu J, Dagda R (2007) Beclin 1-independent pathway of damage-induced mitophagy and autophagic stress: implications for neurodegeneration and cell death. Autophagy 3:663–666

    PubMed  CAS  Google Scholar 

  110. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B (2005) Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 122:927–939

    Article  PubMed  CAS  Google Scholar 

  111. Bove J, Martinez-Vicente M, Vila M (2011) Fighting neurodegeneration with rapamycin: mechanistic insights. Nat Rev Neurosci 12:437–452

    Article  PubMed  CAS  Google Scholar 

  112. Lim J, Kim HW, Youdim MB, Rhyu IJ, Choe KM, Oh YJ (2011) Binding preference of p62 towards LC3-ll during dopaminergic neurotoxin-induced impairment of autophagic flux. Autophagy 7:51–60

    Article  PubMed  CAS  Google Scholar 

  113. Drechsel DA, Patel M (2008) Role of reactive oxygen species in the neurotoxicity of environmental agents implicated in Parkinson’s disease. Free Radic Biol Med 44:1873–1886

    Article  PubMed  CAS  Google Scholar 

  114. 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 3:1301–1306

    Article  PubMed  CAS  Google Scholar 

  115. Betarbet R, Canet-Aviles RM, Sherer TB, Mastroberardino PG, McLendon C, Kim JH, Lund S, Na HM, Taylor G, Bence NF, Kopito R, Seo BB, Yagi T, Yagi A, Klinefelter G, Cookson MR, Greenamyre JT (2006) Intersecting pathways to neurodegeneration in Parkinson’s disease: effects of the pesticide rotenone on DJ-1, alpha-synuclein, and the ubiquitin-proteasome system. Neurobiol Dis 22:404–420

    Article  PubMed  CAS  Google Scholar 

  116. Chen Y, McMillan-Ward E, Kong J, Israels SJ, Gibson SB (2007) Mitochondrial electron-transport-chain inhibitors of complexes I and II induce autophagic cell death mediated by reactive oxygen species. J Cell Sci 120:4155–4166

    Article  PubMed  CAS  Google Scholar 

  117. Pan T, Rawal P, Wu Y, Xie W, Jankovic J, Le W (2009) Rapamycin protects against rotenone-induced apoptosis through autophagy induction. Neuroscience 164:541–551

    Article  PubMed  CAS  Google Scholar 

  118. Dadakhujaev S, Noh HS, Jung EJ, Cha JY, Baek SM, Ha JH, Kim DR (2011) Autophagy protects the rotenone-induced cell death in alpha-synuclein overexpressing SH-SY5Y cells. Neurosci Lett 472:47–52

    Article  CAS  Google Scholar 

  119. Yu WH, Dorado B, Figueroa HY, Wang L, Planel E, Cookson MR, Clark LN, Duff KE (2009) Metabolic activity determines efficacy of macroautophagic clearance of pathological oligomeric alpha-synuclein. Am J Pathol 175:736–747

    Article  PubMed  CAS  Google Scholar 

  120. Wu Y, Li X, Xie W, Jankovic J, Le W, Pan T (2011) Neuroprotection of deferoxamine on rotenone-induced injury via accumulation of HIF-1 alpha and induction of autophagy in SH-SY5Y cells. Neurochem Int 57:198–205

    Article  CAS  Google Scholar 

  121. Xiong N, Jia M, Chen C, Xiong J, Zhang Z, Huang J, Hou L, Yang H, Cao X, Liang Z, Sun S, Lin Z, Wang T (2011) Potential autophagy enhancers attenuate rotenone-induced toxicity in SH-SY5Y. Neuroscience 199:292–302

    Article  PubMed  CAS  Google Scholar 

  122. Dinis-Oliveira RJ, de Pinho PG, Santos L, Teixeira H, Magalhaes T, Santos A, de Lourdes Bastos M, Remiao F, Duarte JA, Carvalho F (2009) Postmortem analyses unveil the poor efficacy of decontamination, anti-inflammatory and immunosuppressive therapies in paraquat human intoxications. PLoS One 4:e7149

    Article  PubMed  CAS  Google Scholar 

  123. Hatcher JM, Pennell KD, Miller GW (2008) Parkinson’s disease and pesticides: a toxicological perspective. Trends Pharmacol Sci 29:322–329

    Article  PubMed  CAS  Google Scholar 

  124. Mak SK, McCormack AL, Manning-Bog AB, Cuervo AM, Di Monte DA (2010) Lysosomal degradation of alpha-synuclein in vivo. J Biol Chem 285:13621–13629

    Article  PubMed  CAS  Google Scholar 

  125. Manning-Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Di Monte DA (2002) The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein. J Biol Chem 277:1641–1644

    Article  PubMed  CAS  Google Scholar 

  126. Wills J, Credle J, Oaks AW, Duka V, Lee JH, Jones J, Sidhu A (2012) Paraquat, but not maneb, induces synucleinopathy and tauopathy in striata of mice through inhibition of proteasomal and autophagic pathways. PLoS One 7:e30745

    Article  PubMed  CAS  Google Scholar 

  127. Gonzalez-Polo RA, Niso-Santano M, Ortiz-Ortiz MA, Gomez-Martin A, Moran JM, Garcia-Rubio L, Francisco-Morcillo J, Zaragoza C, Soler G, Fuentes JM (2007) Inhibition of paraquat-induced autophagy accelerates the apoptotic cell death in neuroblastoma SH-SY5Y cells. Toxicol Sci 97:448–458

    Article  PubMed  CAS  Google Scholar 

  128. Niso-Santano M, Bravo-San Pedro JM, Gomez-Sanchez R, Climent V, Soler G, Fuentes JM, Gonzalez-Polo RA (2011) ASK1 overexpression accelerates paraquat-induced autophagy via endoplasmic reticulum stress. Toxicol Sci 119:156–168

    Article  PubMed  CAS  Google Scholar 

  129. Gonzalez-Polo R, Niso-Santano M, Moran JM, Ortiz-Ortiz MA, Bravo-San Pedro JM, Soler G, Fuentes JM (2009) Silencing DJ-1 reveals its contribution in paraquat-induced autophagy. J Neurochem 109:889–898

    Article  PubMed  CAS  Google Scholar 

  130. Adam-Vizi V, Chinopoulos C (2006) Bioenergetics and the formation of mitochondrial reactive oxygen species. Trends Pharmacol Sci 27:639–645

    Article  PubMed  CAS  Google Scholar 

  131. Chen Y, Azad MB, Gibson SB (2009) Superoxide is the major reactive oxygen species regulating autophagy. Cell Death Differ 16:1040–1052

    Article  PubMed  CAS  Google Scholar 

  132. Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L, Elazar Z (2007) Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J 26:1749–1760

    Article  PubMed  CAS  Google Scholar 

  133. Circu ML, Aw TY (2010) Reactive oxygen species, cellular redox systems, and apoptosis. Free Radic Biol Med 48:749–762

    Article  PubMed  CAS  Google Scholar 

  134. Choi KC, Kim SH, Ha JY, Kim ST, Son JH (2010) A novel mTOR activating protein protects dopamine neurons against oxidative stress by repressing autophagy related cell death. J Neurochem 112:366–376

    Article  PubMed  CAS  Google Scholar 

  135. Higgins GC, Devenish RJ, Beart PM, Nagley P (2011) Autophagic activity in cortical neurons under acute oxidative stress directly contributes to cell death. Cell Mol Life Sci 68:3725–3740

    Article  PubMed  CAS  Google Scholar 

  136. Castino R, Bellio N, Nicotra G, Follo C, Trincheri NF, Isidoro C (2007) Cathepsin D-Bax death pathway in oxidative stressed neuroblastoma cells. Free Radic Biol Med 42:1305–1316

    Article  PubMed  CAS  Google Scholar 

  137. Castino R, Fiorentino I, Cagnin M, Giovia A, Isidoro C (2011) Chelation of lysosomal iron protects dopaminergic SH-SY5Y neuroblastoma cells from hydrogen peroxide toxicity by precluding autophagy and Akt dephosphorylation. Toxicol Sci 123:523–541

    Article  PubMed  CAS  Google Scholar 

  138. Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, Bray K, Reddy A, Bhanot G, Gelinas C, Dipaola RS, Karantza-Wadsworth V, White E (2009) Autophagy suppresses tumorigenesis through elimination of p62. Cell 137:1062–1075

    Article  PubMed  CAS  Google Scholar 

  139. Narendra D, Kane LA, Hauser DN, Fearnley IM, Youle RJ (2010) p62/SQSTM1 is required for Parkin-induced mitochondrial clustering but not mitophagy; VDAC1 is dispensable for both. Autophagy 6:1090–1106

    Article  PubMed  CAS  Google Scholar 

  140. Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, Asara JM, Fitzpatrick J, Dillin A, Viollet B, Kundu M, Hansen M, Shaw RJ (2011) Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331:456–461

    Article  PubMed  CAS  Google Scholar 

  141. Liu F, Guan JL (2011) FIP200, an essential component of mammalian autophagy is indispensible for fetal hematopoiesis. Autophagy 7:229–230

    Article  PubMed  CAS  Google Scholar 

  142. Wu Y, Li X, Zhu JX, Xie W, Le W, Fan Z, Jankovic J, Pan T (2011) Resveratrol-activated AMPK/SIRT1/autophagy in cellular models of Parkinson’s disease. Neurosignals 19:163–174

    Article  PubMed  CAS  Google Scholar 

  143. Yamasaki R, Zhang J, Koshiishi I, Sastradipura Suniarti DF, Wu Z, Peters C, Schwake M, Uchiyama Y, Kira J, Saftig P, Utsumi H, Nakanishi H (2007) Involvement of lysosomal storage-induced p38 MAP kinase activation in the overproduction of nitric oxide by microglia in cathepsin D-deficient mice. Mol Cell Neurosci 35:573–584

    Article  PubMed  CAS  Google Scholar 

  144. Park J, Choi K, Jeong E, Kwon D, Benveniste EN, Choi C (2004) Reactive oxygen species mediate chloroquine-induced expression of chemokines by human astroglial cells. Glia 47:9–20

    Article  PubMed  Google Scholar 

  145. Park BC, Park SH, Paek SH, Park SY, Kwak MK, Choi HG, Yong CS, Yoo BK, Kim JA (2008) Chloroquine-induced nitric oxide increase and cell death is dependent on cellular GSH depletion in A172 human glioblastoma cells. Toxicol Lett 178:52–60

    Article  PubMed  CAS  Google Scholar 

  146. Meulener M, Whitworth AJ, Armstrong-Gold CE, Rizzu P, Heutink P, Wes PD, Pallanck LJ, Bonini NM (2005) Drosophila DJ-1 mutants are selectively sensitive to environmental toxins associated with Parkinson’s disease. Curr Biol 15:1572–1577

    Article  PubMed  CAS  Google Scholar 

  147. Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, Wakeham A, You-Ten AJ, Kalia SK, Horne P, Westaway D, Lozano AM, Anisman H, Park DS, Mak TW (2005) Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci USA 102:5215–5220

    Article  PubMed  CAS  Google Scholar 

  148. Inden M, Taira T, Kitamura Y, Yanagida T, Tsuchiya D, Takata K, Yanagisawa D, Nishimura K, Taniguchi T, Kiso Y, Yoshimoto K, Agatsuma T, Koide-Yoshida S, Iguchi-Ariga SM, Shimohama S, Ariga H (2006) PARK7 DJ-1 protects against degeneration of nigral dopaminergic neurons in Parkinson’s disease rat model. Neurobiol Dis 24:144–158

    Article  PubMed  CAS  Google Scholar 

  149. Zhou W, Freed CR (2005) DJ-1 up-regulates glutathione synthesis during oxidative stress and inhibits A53T alpha-synuclein toxicity. J Biol Chem 280:43150–43158

    Article  PubMed  CAS  Google Scholar 

  150. Paterna JC, Leng A, Weber E, Feldon J, Bueler H (2007) DJ-1 and Parkin modulate dopamine-dependent behavior and inhibit MPTP-induced nigral dopamine neuron loss in mice. Mol Ther 15:698–704

    Article  PubMed  CAS  Google Scholar 

  151. Inden M, Kitamura Y, Takahashi K, Takata K, Ito N, Niwa R, Funayama R, Nishimura K, Taniguchi T, Honda T, Taira T, Ariga H (2011) Protection against dopaminergic neurodegeneration in Parkinson’s disease-model animals by a modulator of the oxidized form of DJ-1, a wild-type of familial Parkinson’s disease-linked PARK7. J Pharmacol Sci 117:189–203

    Article  PubMed  CAS  Google Scholar 

  152. Greene JC, Whitworth AJ, Kuo I, Andrews LA, Feany MB, Pallanck LJ (2003) Mitochondrial pathology and apoptotic muscle degeneration in Drosophila Parkin mutants. Proc Natl Acad Sci USA 100:4078–4083

    Article  PubMed  CAS  Google Scholar 

  153. Greene JC, Whitworth AJ, Andrews LA, Parker TJ, Pallanck LJ (2005) Genetic and genomic studies of Drosophila Parkin mutants implicate oxidative stress and innate immune responses in pathogenesis. Hum Mol Genet 14:799–811

    Article  PubMed  CAS  Google Scholar 

  154. Goldberg MS, Fleming SM, Palacino JJ, Cepeda C, Lam HA, Bhatnagar A, Meloni EG, Wu N, Ackerson LC, Klapstein GJ, Gajendiran M, Roth BL, Chesselet MF, Maidment NT, Levine MS, Shen J (2003) Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons. J Biol Chem 278:43628–43635

    Article  PubMed  CAS  Google Scholar 

  155. Bensaad K, Cheung EC, Vousden KH (2009) Modulation of intracellular ROS levels by TIGAR controls autophagy. EMBO J 28:3015–3026

    Article  PubMed  CAS  Google Scholar 

  156. Crighton D, Wilkinson S, O’Prey J, Syed N, Smith P, Harrison PR, Gasco M, Garrone O, Crook T, Ryan KM (2006) DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 126:121–134

    Article  PubMed  CAS  Google Scholar 

  157. Galluzzi L, Morselli E, Kepp O, Maiuri MC, Kroemer G (2011) Defective autophagy control by the p53 rheostat in cancer. Cell Cycle 9:250–255

    Article  Google Scholar 

  158. Bridges KR (1987) Ascorbic acid inhibits lysosomal autophagy of ferritin. J Biol Chem 262:14773–14778

    PubMed  CAS  Google Scholar 

  159. Castino R, Isidoro C, Murphy D (2005) Autophagy-dependent cell survival and cell death in an autosomal dominant familial neurohypophyseal diabetes insipidus in vitro model. FASEB J 19:1024–1026

    PubMed  CAS  Google Scholar 

  160. Gomez-Santos C, Ferrer I, Santidrian AF, Barrachina M, Gil J, Ambrosio S (2003) Dopamine induces autophagic cell death and alpha-synuclein increase in human neuroblastoma SH-SY5Y cells. J Neurosci Res 73:341–350

    Article  PubMed  CAS  Google Scholar 

  161. Fei XF, Qin ZH, Xiang B, Li LY, Han F, Fukunaga K, Liang ZQ (2009) Olomoucine inhibits cathepsin L nuclear translocation, activates autophagy and attenuates toxicity of 6-hydroxydopamine. Brain Res 1264:85–97

    Article  PubMed  CAS  Google Scholar 

  162. Marin C, Aguilar E (2011) In vivo 6-OHDA-induced neurodegeneration and nigral autophagic markers expression. Neurochem Int 58:521–526

    Article  PubMed  CAS  Google Scholar 

  163. Underwood BR, Imarisio S, Fleming A, Rose C, Krishna G, Heard P, Quick M, Korolchuk VI, Renna M, Sarkar S, Garcia-Arencibia M, O’Kane CJ, Murphy MP, Rubinsztein DC (2010) Antioxidants can inhibit basal autophagy and enhance neurodegeneration in models of polyglutamine disease. Hum Mol Genet 19:3413–3429

    Article  PubMed  CAS  Google Scholar 

  164. Shen W, Ganetzky B (2009) Autophagy promotes synapse development in Drosophila. J Cell Biol 187:71–79

    Article  PubMed  CAS  Google Scholar 

  165. Milton VJ, Jarrett HE, Gowers K, Chalak S, Briggs L, Robinson IM, Sweeney ST (2011) Oxidative stress induces overgrowth of the Drosophila neuromuscular junction. Proc Natl Acad Sci USA 108:17521–17526

    Article  PubMed  CAS  Google Scholar 

  166. West RJ, Sweeney ST (2012) Oxidative stress and autophagy: mediators of synapse growth? Autophagy 8:284–285

    Article  PubMed  CAS  Google Scholar 

  167. Hill BG, Haberzettl P, Ahmed Y, Srivastava S, Bhatnagar A (2008) Unsaturated lipid peroxidation-derived aldehydes activate autophagy in vascular smooth-muscle cells. Biochem J 410:525–534

    Article  PubMed  CAS  Google Scholar 

  168. Barsoum MJ, Yuan H, Gerencser AA, Liot G, Kushnareva Y, Graber S, Kovacs I, Lee WD, Waggoner J, Cui J, White AD, Bossy B, Martinou JC, Youle RJ, Lipton SA, Ellisman MH, Perkins GA, Bossy-Wetzel E (2006) Nitric oxide-induced mitochondrial fission is regulated by dynamin-related GTPases in neurons. EMBO J 25:3900–3911

    Article  PubMed  CAS  Google Scholar 

  169. Janjetovic K, Misirkic M, Vucicevic L, Harhaji L, Trajkovic V (2008) Synergistic antiglioma action of hyperthermia and nitric oxide. Eur J Pharmacol 583:1–10

    Article  PubMed  CAS  Google Scholar 

  170. Sarkar S, Korolchuk VI, Renna M, Imarisio S, Fleming A, Williams A, Garcia-Arencibia M, Rose C, Luo S, Underwood BR, Kroemer G, O’Kane CJ, Rubinsztein DC (2011) Complex inhibitory effects of nitric oxide on autophagy. Mol Cell 43:19–32

    Article  PubMed  CAS  Google Scholar 

  171. Wei Y, Pattingre S, Sinha S, Bassik M, Levine B (2008) JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol Cell 30:678–688

    Article  PubMed  CAS  Google Scholar 

  172. Wang Q, Liang B, Shirwany NA, Zou MH (2010) 2-deoxy-d-glucose treatment of endothelial cells induces autophagy by reactive oxygen species-mediated activation of the AMP-activated protein kinase. PLoS One 6:e17234

    Article  CAS  Google Scholar 

  173. Zhang J, Xie Z, Dong Y, Wang S, Liu C, Zou MH (2008) Identification of nitric oxide as an endogenous activator of the AMP-activated protein kinase in vascular endothelial cells. J Biol Chem 283:27452–27461

    Article  PubMed  CAS  Google Scholar 

  174. Zou MH, Kirkpatrick SS, Davis BJ, Nelson JS, Wiles WG, Schlattner U, Neumann D, Brownlee M, Freeman MB, Goldman MH (2004) Activation of the AMP-activated protein kinase by the anti-diabetic drug metformin in vivo. Role of mitochondrial reactive nitrogen species. J Biol Chem 279:43940–43951

    Article  PubMed  CAS  Google Scholar 

  175. Alves da Costa C, Checler F (2011) Apoptosis in Parkinson’s disease: is p53 the missing link between genetic and sporadic Parkinsonism? Cell Signal 23:963–968

    Article  PubMed  CAS  Google Scholar 

  176. Cuadrado A, Moreno-Murciano P, Pedraza-Chaverri J (2009) The transcription factor Nrf2 as a new therapeutic target in Parkinson’s disease. Expert Opin Ther Targets 13:319–329

    Article  PubMed  CAS  Google Scholar 

  177. van der Vos KE, Coffer PJ (2011) The extending network of FOXO transcriptional target genes. Antioxid Redox Signal 14:579–592

    Article  PubMed  CAS  Google Scholar 

  178. Smith PD, Crocker SJ, Jackson-Lewis V, Jordan-Sciutto KL, Hayley S, Mount MP, O’Hare MJ, Callaghan S, Slack RS, Przedborski S, Anisman H, Park DS (2003) Cyclin-dependent kinase 5 is a mediator of dopaminergic neuron loss in a mouse model of Parkinson’s disease. Proc Natl Acad Sci USA 100:13650–13655

    Article  PubMed  CAS  Google Scholar 

  179. Smith PD, Mount MP, Shree R, Callaghan S, Slack RS, Anisman H, Vincent I, Wang X, Mao Z, Park DS (2006) Calpain-regulated p35/cdk5 plays a central role in dopaminergic neuron death through modulation of the transcription factor myocyte enhancer factor 2. J Neurosci 26:440–447

    Article  PubMed  CAS  Google Scholar 

  180. Qu D, Rashidian J, Mount MP, Aleyasin H, Parsanejad M, Lira A, Haque E, Zhang Y, Callaghan S, Daigle M, Rousseaux MW, Slack RS, Albert PR, Vincent I, Woulfe JM, Park DS (2007) Role of Cdk5-mediated phosphorylation of Prx2 in MPTP toxicity and Parkinson’s disease. Neuron 55:37–52

    Article  PubMed  CAS  Google Scholar 

  181. Takahashi Y, Coppola D, Matsushita N, Cualing HD, Sun M, Sato Y, Liang C, Jung JU, Cheng JQ, Mule JJ, Pledger WJ, Wang HG (2007) Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis. Nat Cell Biol 9:1142–1151

    Article  PubMed  CAS  Google Scholar 

  182. Moran JM, Gonzalez-Polo RA, Ortiz-Ortiz MA, Niso-Santano M, Soler G, Fuentes JM (2008) Identification of genes associated with paraquat-induced toxicity in SH-SY5Y cells by PCR array focused on apoptotic pathways. J Toxicol Environ Health A 71:1457–1467

    Article  PubMed  CAS  Google Scholar 

  183. Eisenberg-Lerner A, Kimchi A (2011) PKD is a kinase of Vps34 that mediates ROS-induced autophagy downstream of DAPk. Cell Death Differ 19:788–797

    Article  PubMed  CAS  Google Scholar 

  184. Lotze MT, Tracey KJ (2005) High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol 5:331–342

    Article  PubMed  CAS  Google Scholar 

  185. Tang D, Kang R, Livesey KM, Zeh HJ 3rd, Lotze MT (2011) High mobility group box 1 (HMGB1) activates an autophagic response to oxidative stress. Antioxid Redox Signal 15:2185–2195

    Article  PubMed  CAS  Google Scholar 

  186. Tang D, Kang R, Livesey KM, Cheh CW, Farkas A, Loughran P, Hoppe G, Bianchi ME, Tracey KJ, Zeh HJ 3rd, Lotze MT (2011) Endogenous HMGB1 regulates autophagy. J Cell Biol 190:881–892

    Article  CAS  Google Scholar 

  187. Tang D, Kang R, Livesey KM, Kroemer G, Billiar TR, Van Houten B, Zeh HJ 3rd, Lotze MT (2011) High-mobility group box 1 is essential for mitochondrial quality control. Cell Metab 13:701–711

    Article  PubMed  CAS  Google Scholar 

  188. Gu Z, Nakamura T, Yao D, Shi ZQ, Lipton SA (2005) Nitrosative and oxidative stress links dysfunctional ubiquitination to Parkinson’s disease. Cell Death Differ 12:1202–1204

    Article  PubMed  CAS  Google Scholar 

  189. Yao D, Gu Z, Nakamura T, Shi ZQ, Ma Y, Gaston B, Palmer LA, Rockenstein EM, Zhang Z, Masliah E, Uehara T, Lipton SA (2004) Nitrosative stress linked to sporadic Parkinson’s disease: S-nitrosylation of parkin regulates its E3 ubiquitin ligase activity. Proc Natl Acad Sci USA 101:10810–10814

    Article  PubMed  CAS  Google Scholar 

  190. LaVoie MJ, Ostaszewski BL, Weihofen A, Schlossmacher MG, Selkoe DJ (2005) Dopamine covalently modifies and functionally inactivates parkin. Nat Med 11:1214–1221

    Article  PubMed  CAS  Google Scholar 

  191. Meng F, Yao D, Shi Y, Kabakoff J, Wu W, Reicher J, Ma Y, Moosmann B, Masliah E, Lipton SA, Gu Z (2011) Oxidation of the cysteine-rich regions of parkin perturbs its E3 ligase activity and contributes to protein aggregation. Mol Neurodegener 6:34

    Article  PubMed  CAS  Google Scholar 

  192. Luciani A, Villella VR, Esposito S, Brunetti-Pierri N, Medina DL, Settembre C, Gavina M, Raia V, Ballabio A, Maiuri L (2010) Cystic fibrosis: a disorder with defective autophagy. Autophagy 7:104–106

    Article  Google Scholar 

  193. Luciani A, Villella VR, Esposito S, Brunetti-Pierri N, Medina D, Settembre C, Gavina M, Pulze L, Giardino I, Pettoello-Mantovani M, D’Apolito M, Guido S, Masliah E, Spencer B, Quaratino S, Raia V, Ballabio A, Maiuri L (2010) Defective CFTR induces aggresome formation and lung inflammation in cystic fibrosis through ROS-mediated autophagy inhibition. Nat Cell Biol 12:863–875

    Article  PubMed  CAS  Google Scholar 

  194. D C, Curro M, Ferlazzo N, Condello S, Ientile R (2012) Monitoring of transglutaminase2 under different oxidative stress conditions. Amino Acids 42:1037–1043

    Article  CAS  Google Scholar 

  195. Mazzio EA, Close F, Soliman KF (2011) The biochemical and cellular basis for nutraceutical strategies to attenuate neurodegeneration in Parkinson’s disease. Int J Mol Sci 12:506–569

    Article  PubMed  CAS  Google Scholar 

  196. LeWitt PA, Taylor DC (2008) Protection against Parkinson’s disease progression: clinical experience. Neurotherapeutics 5:210–225

    Article  PubMed  CAS  Google Scholar 

  197. Clark J, Clore EL, Zheng K, Adame A, Masliah E, Simon DK (2010) Oral N-acetyl-cysteine attenuates loss of dopaminergic terminals in alpha-synuclein overexpressing mice. PLoS One 5:e12333

    Article  PubMed  CAS  Google Scholar 

  198. Chen CM, Yin MC, Hsu CC, Liu TC (2007) Antioxidative and anti-inflammatory effects of four cysteine-containing agents in striatum of MPTP-treated mice. Nutrition 23:589–597

    Article  PubMed  CAS  Google Scholar 

  199. Sun L, Xu S, Zhou M, Wang C, Wu Y, Chan P (2010) Effects of cysteamine on MPTP-induced dopaminergic neurodegeneration in mice. Brain Res 1335:74–82

    Article  PubMed  CAS  Google Scholar 

  200. Sun AY, Wang Q, Simonyi A, Sun GY (2008) Botanical phenolics and brain health. Neuromolecular Med 10:259–274

    Article  PubMed  CAS  Google Scholar 

  201. Albani D, Polito L, Signorini A, Forloni G (2010) Neuroprotective properties of resveratrol in different neurodegenerative disorders. Biofactors 36:370–376

    Article  PubMed  CAS  Google Scholar 

  202. Mythri RB, Bharath MS (2012) Curcumin: a potential neuroprotective agent in Parkinson’s disease. Curr Pharm Des 18:91–99

    Article  PubMed  CAS  Google Scholar 

  203. Scapagnini G, Vasto S, Abraham NG, Caruso C, Zella D, Fabio G (2011) Modulation of Nrf2/ARE pathway by food polyphenols: a nutritional neuroprotective strategy for cognitive and neurodegenerative disorders. Mol Neurobiol 44:192–201

    Article  PubMed  CAS  Google Scholar 

  204. Zhang X, Li L, Chen S, Yang D, Wang Y, Zhang X, Wang Z, Le W (2011) Rapamycin treatment augments motor neuron degeneration in SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Autophagy 7:412–425

    Article  PubMed  CAS  Google Scholar 

  205. Tain LS, Mortiboys H, Tao RN, Ziviani E, Bandmann O, Whitworth AJ (2009) Rapamycin activation of 4E-BP prevents parkinsonian dopaminergic neuron loss. Nat Neurosci 12:1129–1135

    Article  PubMed  CAS  Google Scholar 

  206. Paris I, Munoz P, Huenchuguala S, Couve E, Sanders LH, Greenamyre JT, Caviedes P, Segura-Aguilar J (2011) Autophagy protects against aminochrome-induced cell death in substantia nigra-derived cell line. Toxicol Sci 121:376–388

    Article  PubMed  CAS  Google Scholar 

  207. Li W, Zhu S, Li J, Assa A, Jundoria A, Xu J, Fan S, Eissa NT, Tracey KJ, Sama AE, Wang H (2011) EGCG stimulates autophagy and reduces cytoplasmic HMGB1 levels in endotoxin-stimulated macrophages. Biochem Pharmacol 81:1152–1163

    Article  PubMed  CAS  Google Scholar 

  208. Wang K, Liu R, Li J, Mao J, Lei Y, Wu J, Zeng J, Zhang T, Wu H, Chen L, Huang C, Wei Y (2011) Quercetin induces protective autophagy in gastric cancer cells: involvement of Akt-mTOR- and hypoxia-induced factor 1alpha-mediated signaling. Autophagy 7:966–978

    Article  PubMed  CAS  Google Scholar 

  209. Filomeni G, Graziani I, De Zio D, Dini L, Centonze D, Rotilio G, Ciriolo MR (2012) Neuroprotection of kaempferol by autophagy in models of rotenone-mediated acute toxicity: possible implications for Parkinson’s disease. Neurobiol Aging 33:767–785

    Article  PubMed  CAS  Google Scholar 

  210. Di Zanni E, Bachetti T, Parodi S, Bocca P, Prigione I, Di Lascio S, Fornasari D, Ravazzolo R, Ceccherini I (2012) In vitro drug treatments reduce the deleterious effects of aggregates containing polyAla expanded PHOX2B proteins. Neurobiol Dis 45:508–518

    Article  PubMed  CAS  Google Scholar 

  211. Scarlatti F, Maffei R, Beau I, Codogno P, Ghidoni R (2008) Role of non-canonical Beclin 1-independent autophagy in cell death induced by resveratrol in human breast cancer cells. Cell Death Differ 15:1318–1329

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

E. Janda, V. Mollace, and C. Carresi acknowledge Environmental Protection Agency (ARPACal) for research funding. Research on autophagy and neurodegeneration in the laboratory of C. Isidoro were funded by Compagnia S. Paolo di Torino (Neuroscience Project 2009).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Elzbieta Janda.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Janda, E., Isidoro, C., Carresi, C. et al. Defective Autophagy in Parkinson’s Disease: Role of Oxidative Stress. Mol Neurobiol 46, 639–661 (2012). https://doi.org/10.1007/s12035-012-8318-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-012-8318-1

Keywords

Navigation