Basic mechanisms of neurodegeneration: a critical update

Kurt A Jellinger, Kurt A Jellinger

Abstract

Neurodegenerative diseases are characterized by progressive dysfunction of specific populations of neurons, determining clinical presentation. Neuronal loss is associated with extra and intracellular accumulation of misfolded proteins, the hallmarks of many neurodegenerative proteinopathies. Major basic processes include abnormal protein dynamics due to deficiency of the ubiquitin-proteosome-autophagy system, oxidative stress and free radical formation, mitochondrial dysfunction, impaired bioenergetics, dysfunction of neurotrophins, 'neuroinflammatory' processes and (secondary) disruptions of neuronal Golgi apparatus and axonal transport. These interrelated mechanisms lead to programmed cell death is a long run over many years. Neurodegenerative disorders are classified according to known genetic mechanisms or to major components of protein deposits, but recent studies showed both overlap and intraindividual diversities between different phenotypes. Synergistic mechanisms between pathological proteins suggest common pathogenic mechanisms. Animal models and other studies have provided insight into the basic neurodegeneration and cell death programs, offering new ways for future prevention/treatment strategies.

Figures

Fig 1
Fig 1
Algorithm for classification of neurodegenerative diseases with protein deposits (proteinopathies) (from [1]).
Fig 2
Fig 2
Cascade of neurotoxic effects of protein oligomeres leading to neuronal dysfunction/ND; illustrated by the suggested relationship between Aβ and αSyn oligomers.
Fig 3
Fig 3
Model of protein misfolding and fibrillation leading to deposition of aggregated proteins in cells and extracellular spaces via actions of the UPS, phagosomes and aggresomes, either causing cell death or cytoprotection.
Fig 4
Fig 4
Schematic interaction of proteins in neurodegenerative diseases and mitochondria. Accumulation of mitochondrial DNA mutations may induce ROS production and cause oxidative damage in aging. In AD, ROS production and decreased ATP may contribute to production of Aβ peptides that may enter mitochondria, induce free radicals, decrease cytochrome oxidase activity and inhibit ATP generation. APP is transported to outer mitochondrial membranes, blocks import of nuclear cytchrome oxidase proteins to mitchondria, and may decrease cytochrome oxidase activity. In AD and models, Aβ is found in mitochondrial matrix, produces free radicals and causes mitochondrial dysfunction. N-terminal portion of ApoE4, γ-secretase complex proteins, e.g. presenilins and nicastrin, associated with mitochondria, may contribute to Aβ production and cause oxidative damage. In HD neurons, Htt binds to outer mitochondrial membrane und induces free radical production; H2O2 may also interrupt calcium uptake. In PD neurons, mutant proteins of αSyn, Parkin, PINK1 and DJ1 are associated with mitochondria and cause their dysfunction. In ALS, mutant SOD1, localized in inner and outer mitochondrial membranes and matrix, induces oxidative damage; associated with impairment of complexes II and IV. Frataxin, a gene product in FRA, is a mitochondrial protein responsible for heme biosynthesis and formation of iron-sulfur clusters, facilitating the accumulation of iron and inducing free radicals (modified from [257])
Fig 5
Fig 5
Overview of apoptotic signalling through the receptor-mediated (‘extrinsic’) and mitochondria bases (‘intrinsic’) pathways, showing all key molecular players of apoptosis, the importance of the caspase cascade via interaction with different death domaines, and the role of effective caspase driving the execution of the cell death program.
Fig 6
Fig 6
Diverse pathways leading to cell death, illustrated by the concept of the apoptosis-necrosis continuum that integrates the various death pathways and subsequent intracellular signalling pathways (ER stress, UPS, ATP loss, etc.) to help explain the complex patterns of neuronal death (mix of PCD-types I, II and/or III) (modified from [245]).

References

    1. Jellinger KA. Recent advances in our understanding of neurodegeneration. J Neural Transm. 2009;116:1111–62.
    1. Skovronsky DM, Lee VM-Y, Trojanowski JQ. Neurodegenerative diseases: new concepts of pathogenesis and their therapeutic implications. Annu Rev Pathol Mech Dis. 2006;1:151–70.
    1. Golde TE, Miller VM. Proteinopathy-induced neuronal senescence: a hypothesis for brain failure in Alzheimer’s and other neurodegenerative diseases. Alzheimers Res Ther. 2009;1:5.
    1. Uversky VN. Intrinsic disorder in proteins associated with neurodegenerative diseases. Front Biosci. 2009;14:5188–238.
    1. Herczenik E, Gebbink MF. Molecular and cellular aspects of protein misfolding and disease. FASEB J. 2008;22:2115–33.
    1. Ovádi J, Orosz F. Protein folding and misfolding: neurodegenerative diseases. Netherlands: Springer; 2009. p. 274.
    1. Dickson DW. Neuropathology of non-Alzheimer degenerative disorders. Int J Clin Exp Pathol. 2010;3:1–22.
    1. Kovacs GG, Budka H. Protein-based neuropathology and molecular classification of human neurodegenerative diseases. In: Ovádi J, Orosz F, editors. Protein folding and misfolding: neurodegenerative diseases. Netherlands: Springer; 2009. pp. 251–72.
    1. Migliore L, Coppede F. Genetics, environmental factors and the emerging role of epigenetics in neurodegenerative diseases. Mutat Res. 2009;667:82–97.
    1. Palop JJ, Chin J, Mucke L. A network dysfunction perspective on neurodegenerative diseases. Nature. 2006;443:768–73.
    1. Seeley WW, Crawford RK, Zhou J, et al. Neurodegenerative diseases target large-scale human brain networks. Neuron. 2009;62:42–52.
    1. Ecroyd H, Carver JA. Unraveling the mysteries of protein folding and misfolding. IUBMB Life. 2008;60:769–74.
    1. Morris AM, Watzky MA, Finke RG. Protein aggregation kinetics, mechanism, and curve-fitting: a review of the literature. Biochim Biophys Acta. 2009;1794:375–97.
    1. Israeli E, Sharon R. Beta-synuclein occurs in vivo in lipid-associated oligomers and forms hetero-oligomers with alpha-synuclein. J Neurochem. 2009;108:465–74.
    1. Selkoe DJ. Soluble oligomers of the amyloid beta-protein impair synaptic plasticity and behavior. Behav Brain Res. 2008;192:106–13.
    1. Soto C, Estrada LD. Protein misfolding and neurodegeneration. Arch Neurol. 2008;65:184–9.
    1. Aguzzi A, Sigurdson C, Heikenwaelder M. Molecular mechanisms of prion pathogenesis. Annu Rev Pathol. 2008;3:11–40.
    1. Lafaye P, Achour I, England P, et al. Single-domain antibodies recognize selectively small oligomeric forms of amyloid beta, prevent Abeta-induced neurotoxicity and inhibit fibril formation. Mol Immunol. 2009;46:695–704.
    1. Moore RA, Taubner LM, Priola SA. Prion protein misfolding and disease. Curr Opin Struct Biol. 2009;19:14–22.
    1. Williams AJ, Paulson HL. Polyglutamine neurodegeneration: protein misfolding revisited. Trends Neurosci. 2008;31:521–8.
    1. Xia W, Yang T, Shankar G, et al. A specific enzyme-linked immunosorbent assay for measuring beta-amyloid protein oligomers in human plasma and brain tissue of patients with Alzheimer disease. Arch Neurol. 2009;66:190–9.
    1. Tomic JL, Pensalfini A, Head E, et al. Soluble fibrillar oligomer levels are elevated in Alzheimer’s disease brain and correlate with cognitive dysfunction. Neurobiol Dis. 2009;35:352–8.
    1. Harmeier A, Wozny C, Rost BR, et al. Role of amyloid-beta glycine 33 in oligomerization, toxicity, and neuronal plasticity. J Neurosci. 2009;29:7582–90.
    1. Kim J, Onstead L, Randle S, et al. Abeta40 inhibits amyloid deposition in vivo. J Neurosci. 2007;27:627–33.
    1. Schmidt M, Sachse C, Richter W, et al. Comparison of Alzheimer Abeta(1–40) and Abeta(1–42) amyloid fibrils reveals similar protofilament structures. Proc Natl Acad Sci USA. 2009;106:19813–8.
    1. Lauren J, Gimbel DA, Nygaard HB, et al. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457:1128–32.
    1. Shankar GM, Li S, Mehta TH, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14:837–42.
    1. Abramov E, Dolev I, Fogel H, et al. Amyloid-beta as a positive endogenous regulator of release probability at hippocampal synapses. Nat Neurosci. 2009;12:1567–76.
    1. Nygaard HB, Strittmatter SM. Cellular prion protein mediates the toxicity of beta-amyloid oligomers: implications for Alzheimer disease. Arch Neurol. 2009;66:1325–8.
    1. Nimmrich V, Ebert U. Is Alzheimer’s disease a result of presynaptic failure? Synaptic dysfunctions induced by oligomeric beta-amyloid. Rev Neurosci. 2009;20:1–12.
    1. Dinamarca MC, Colombres M, Cerpa W, et al. Beta-amyloid oligomers affect the structure and function of the postsynaptic region: role of the Wnt signaling pathway. Neurodegener Dis. 2008;5:149–52.
    1. Moreno H, Yu E, Pigino G, et al. Synaptic transmission block by presynaptic injection of oligomeric amyloid beta. Proc Natl Acad Sci USA. 2009;106:5901–6.
    1. Deshpande A, Kawai H, Metherate R, et al. A role for synaptic zinc in activity-dependent Abeta oligomer formation and accumulation at excitatory synapses. J Neurosci. 2009;29:4004–15.
    1. Wang Z, Wang B, Yang L, et al. Presynaptic and postsynaptic interaction of the amyloid precursor protein promotes peripheral and central synaptogenesis. J Neurosci. 2009;29:10788–801.
    1. Hoe HS, Fu Z, Makarova A, et al. The effects of amyloid precursor protein on postsynaptic composition and activity. J Biol Chem. 2009;284:8495–506.
    1. Noguchi A, Matsumura S, Dezawa M, et al. Isolation and characterization of patient-derived, toxic, high mass amyloid beta-protein (Abeta) assembly from Alzheimer disease brains. J Biol Chem. 2009;284:32895–905.
    1. Gunzburg MJ, Perugini MA, Howlett GJ. Structural basis for the recognition and cross-linking of amyloid fibrils by human apolipoprotein E. J Biol Chem. 2007;282:35831–41.
    1. Vintem AP, Henriques AG, Da Cruz ESOA, et al. PP1 inhibition by Abeta peptide as a potential pathological mechanism in Alzheimer’s disease. Neurotoxicol Teratol. 2009;31:85–8.
    1. Karlnoski RA, Rosenthal A, Kobayashi D, et al. Suppression of amyloid deposition leads to long-term reductions in Alzheimer’s pathologies in Tg2576 mice. J Neurosci. 2009;29:4964–71.
    1. Huang HC, Jiang ZF. Accumulated amyloid-beta peptide and hyperphosphorylated tau protein: relationship and links in Alzheimer’s disease. J Alzheimers Dis. 2009;16:15–27.
    1. De Felice FG, Wu D, Lambert MP, et al. Alzheimer’s disease-type neuronal tau hyperphosphorylation induced by A beta oligomers. Neurobiol Aging. 2008;29:1334–47.
    1. Kellner A, Matschke J, Bernreuther C, et al. Autoantibodies against beta-amyloid are common in Alzheimer’s disease and help control plaque burden. Ann Neurol. 2009;65:24–31.
    1. Meli G, Visintin M, Cannistraci I, et al. Direct in vivo intracellular selection of conformation-sensitive antibody domains targeting Alzheimer’s amyloid-beta oligomers. J Mol Biol. 2009;387:584–606.
    1. Relini A, Cavalleri O, Rolandi R, et al. The two-fold aspect of the interplay of amyloidogenic proteins with lipid membranes. Chem Phys Lipids. 2009;158:1–9.
    1. Oddo S, Caccamo A, Smith IF, et al. A dynamic relationship between intracellular and extracellular pools of Abeta. Am J Pathol. 2006;168:184–94.
    1. LaFerla FM, Green KN, Oddo S. Intracellular amyloid-beta in Alzheimer’s disease. Nat Rev Neurosci. 2007;8:499–509.
    1. Savioz A, Leuba G, Vallet PG, et al. Contribution of neural networks to Alzheimer disease’s progression. Brain Res Bull. 2009;80:309–14.
    1. Sahara N, Maeda S, Takashima A. Tau oligomerization: a role for tau aggregation intermediates linked to neurodegeneration. Curr Alzheimer Res. 2008;5:591–8.
    1. Brunden KR, Trojanowski JQ, Lee VM. Evidence that non-fibrillar tau causes pathology linked to neurodegeneration and behavioral impairments. J Alzheimers Dis. 2008;14:393–9.
    1. Small SA, Duff K. Linking Abeta and tau in late-onset Alzheimer’s disease: a dual pathway hypothesis. Neuron. 2008;60:534–42.
    1. Barrantes A, Sotres J, Hernando-Perez M, et al. Tau aggregation followed by atomic force microscopy and surface plasmon resonance, and single molecule tau-tau interaction probed by atomic force spectroscopy. J Alzheimers Dis. 2009;18:141–51.
    1. Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem. 2009;284:12845–52.
    1. Ren PH, Lauckner JE, Kachirskaia I, et al. Cytoplasmic penetration and persistent infection of mammalian cells by polyglutamine aggregates. Nat Cell Biol. 2009;11:219–25.
    1. Clavaguera F, Bolmont T, Crowther RA, et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat Cell Biol. 2009;11:909–13.
    1. Jeganathan S, Von Bergen M, Mandelkow EM, et al. The natively unfolded character of tau and its aggregation to Alzheimer-like paired helical filaments. Biochemistry. 2008;47:10526–39.
    1. Kazantsev AG, Kolchinsky AM. Central role of alpha-synuclein oligomers in neurodegeneration in Parkinson disease. Arch Neurol. 2008;65:1577–81.
    1. Uversky VN. Neuropathology, biochemistry, and biophysics of alpha-synuclein aggregation. J Neurochem. 2007;103:17–37.
    1. Waxman EA, Giasson BI. Molecular mechanisms of alpha-synuclein neurodegeneration. Biochim Biophys Acta. 2009;1792:616–24.
    1. Crews L, Tsigelny I, Hashimoto M, et al. Role of synucleins in Alzheimer’s disease. Neurotox Res. 2009;16:306–17.
    1. Cullen V, Lindfors M, Ng J, et al. Cathepsin D expression level affects alpha-synuclein processing, aggregation, and toxicity in vivo. Mol Brain. 2009 Doi: .
    1. Duka T, Duka V, Joyce JN, et al. Alpha-Synuclein contributes to GSK-3beta-catalyzed Tau phosphorylation in Parkinson’s disease models. FASEB J. 2009;23:2820–30.
    1. Paleologou KE, Kragh CL, Mann DM, et al. Detection of elevated levels of soluble alpha-synuclein oligomers in post-mortem brain extracts from patients with dementia with Lewy bodies. Brain. 2009;132:1093–101.
    1. Danzer KM, Krebs SK, Wolff M, et al. Seeding induced by alpha-synuclein oligomers provides evidence for spreading of alpha-synuclein pathology. J Neurochem. 2009;111:192–203.
    1. Luk KC, Song C, O’Brien P, et al. Exogenous alpha-synuclein fibrils seed the formation of Lewy body-like intracellular inclusions in cultured cells. Proc Natl Acad Sci USA. 2009;106:20051–6.
    1. Desplats P, Lee HJ, Bae EJ, et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci USA. 2009;106:13010–5.
    1. Li C, Lutz EA, Slade KM, et al. 19F NMR studies of alpha-synuclein conformation and fibrillation. Biochemistry. 2009;48:8578–84.
    1. Emadi S, Kasturirangan S, Wang MS, et al. Detecting morphologically distinct oligomeric forms of alpha-synuclein. J Biol Chem. 2009;284:11048–58.
    1. Banci L, Bertini I, Boca M, et al. Structural and dynamic aspects related to oligomerization of apo SOD1 and its mutants. Proc Natl Acad Sci USA. 2009;106:6980–5.
    1. Kwiatkowski TJ, Jr, Bosco DA, Leclerc AL, et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323:1205–8.
    1. Neumann M, Sampathu DM, Kwong LK, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.
    1. Liu HN, Sanelli T, Horne P, et al. Lack of evidence of monomer/misfolded superoxide dismutase-1 in sporadic amyotrophic lateral sclerosis. Ann Neurol. 2009;66:75–80.
    1. Alberti S, Halfmann R, King O, et al. A systematic survey identifies prions and illuminates sequence features of prionogenic proteins. Cell. 2009;137:146–58.
    1. Fontaine SN, Brown DR. Mechanisms of prion protein aggregation. Protein Pept Lett. 2009;16:14–26.
    1. Kellett KA, Hooper NM. Prion protein and Alzheimer disease. Prion. 2009;3:190–4.
    1. Matilla-Duenas A, Sanchez I, Corral-Juan M, et al. Cellular and molecular pathways triggering neurodegeneration in the spinocerebellar ataxias. Cerebellum. 2009 Doi: .
    1. Schwartz AL, Ciechanover A. Targeting proteins for destruction by the ubiquitin system: implications for human pathobiology. Ann Rev Pharmacol Toxicol. 2009;49:73–96.
    1. Muchowski PJ, Wacker JL. Modulation of neurodegeneration by molecular chaperones. Nat Rev Neurosci. 2005;6:11–22.
    1. Hartl FU, Hayer-Hartl M. Converging concepts of protein folding in vitro and in vivo. Nat Struct Mol Biol. 2009;16:574–81.
    1. Kubota H. Quality control against misfolded proteins in the cytosol: a network for cell survival. J Biochem. 2009;146:609–16.
    1. Scheper W, Hoozemans JJ. Endoplasmic reticulum protein quality control in neurodegenerative disease: the good, the bad and the therapy. Curr Med Chem. 2009;16:615–26.
    1. Eliezer D. Biophysical characterization of intrinsically disordered proteins. Curr Opin Struct Biol. 2009;19:23–30.
    1. Mendoza-Espinosa P, Garcia-Gonzalez V, Moreno A, et al. Disorder-to-order conformational transitions in protein structure and its relationship to disease. Mol Cell Biochem. 2009;330:105–20.
    1. Balch WE, Morimoto RI, Dillin A, et al. Adapting proteostasis for disease intervention. Science. 2008;319:916–9.
    1. Winklhofer KF, Tatzelt J, Haass C. The two faces of protein misfolding: gain- and loss-of-function in neurodegenerative diseases. EMBO J. 2008;27:336–49.
    1. Mirzaei H, Regnier F. Protein:protein aggregation induced by protein oxidation. J Chromatogr B Analyt Technol Biomed Life Sci. 2008;873:8–14.
    1. Hutt DM, Powers ET, Balch WE. The proteostasis boundary in misfolding diseases of membrane traffic. FEBS Lett. 2009;583:2639–46.
    1. Powers ET, Morimoto RI, Dillin A, et al. Biological and chemical approaches to diseases of proteostasis deficiency. Annu Rev Biochem. 2009;78:959–91.
    1. Arndt V, Rogon C, Hohfeld J. To be, or not to be–molecular chaperones in protein degradation. Cell Mol Life Sci. 2007;64:2525–41.
    1. Broadley SA, Hartl FU. The role of molecular chaperones in human misfolding diseases. FEBS Lett. 2009;583:2647–53.
    1. Roelofs J, Park S, Haas W, et al. Chaperone-mediated pathway of proteasome regulatory particle assembly. Nature. 2009;459:861–5.
    1. Kalmar B, Greensmith L. Induction of heat shock proteins for protection against oxidative stress. Adv Drug Deliv Rev. 2009;61:310–8.
    1. Calderwood SK, Murshid A, Prince T. The shock of aging: molecular chaperones and the heat shock response in longevity and aging–a mini-review. Gerontology. 2009;55:550–8.
    1. Goldbaum O, Riedel M, Stahnke T, et al. The small heat shock protein HSP25 protects astrocytes against stress induced by proteasomal inhibition. Glia. 2009;57:1566–77.
    1. Roodveldt C, Bertoncini CW, Andersson A, et al. Chaperone proteostasis in Parkinson’s disease: stabilization of the Hsp70/alpha-synuclein complex by Hip. EMBO J. 2009;28:3758–70.
    1. Malhotra JD, Kaufman RJ. The endoplasmic reticulum and the unfolded protein response. Semin Cell Dev Biol. 2007;18:716–31.
    1. Lindholm D, Wootz H, Korhonen L. ER stress and neurodegenerative diseases. Cell Death Differ. 2006;13:385–92.
    1. Uversky VN. Alpha-synuclein misfolding and neurodegenerative diseases. Curr Protein Pept Sci. 2008;9:507–40.
    1. Tan JM, Wong ES, Lim KL. Protein misfolding and aggregation in Parkinson disease. Antioxid Redox Signal. 2009;11:2119–34.
    1. Mondragon-Rodriguez S, Basurto-Islas G, Binder LI, et al. Conformational changes and cleavage; are these responsible for the tau aggregation in Alzheimers disease. Future Neurol. 2009;4:39–53.
    1. Takashima A. Hyperphosphorylated tau is a cause of neuronal dysfunction in tauopathy. J Alzheimers Dis. 2008;14:371–5.
    1. Spires-Jones TL, Stoothoff WH, De Calignon A, et al. Tau pathophysiology in neurodegeneration: a tangled issue. Trends Neurosci. 2009;32:150–9.
    1. Hatters DM. Protein misfolding inside cells: the case of huntingtin and Huntington’s disease. IUBMB Life. 2008;60:724–8.
    1. Okamoto S, Pouladi MA, Talantova M, et al. Balance between synaptic versus extrasynaptic NMDA receptor activity influences inclusions and neurotoxicity of mutant huntingtin. Nat Med. 2009;15:1407–13.
    1. Reijonen S, Putkonen N, Norremolle A, et al. Inhibition of endoplasmic reticulum stress counteracts neuronal cell death and protein aggregation caused by N-terminal mutant huntingtin proteins. Exp Cell Res. 2008;314:950–60.
    1. Woulfe J. Nuclear bodies in neurodegenerative disease. Biochim Biophys Acta. 2008;1783:2195–206.
    1. Castellani RJ, Zhu X, Lee H-G, et al. Molecular pathogenesis of Alzheimer disease: reductionist versus expansionist approaches. Int J Mol Sci. 2009;10:1386–406.
    1. Austin RC. The unfolded protein response in health and disease. Antioxid Redox Signal. 2009;11:2279–87.
    1. Schroder M. Endoplasmic reticulum stress responses. FASEB J. 2008;65:862–94.
    1. Bezprozvanny I. Calcium signaling and neurodegenerative diseases. Trends Mol Med. 2009;15:89–100.
    1. Marambaud P, Dreses-Werringloer U, Vingtdeux V. Calcium signaling in neurodegeneration. Mol Neurodegener. 2009;4:20.
    1. Hoozemans JJ, Van Haastert ES, Nijholt DA, et al. The unfolded protein response is activated in pretangle neurons in Alzheimer’s disease hippocampus. Am J Pathol. 2009;174:1241–51.
    1. Hoozemans JJM, Scheper W. Endoplasmic reticulum stress in neurodegeneration. In: Ovádi J, Orosz F, editors. Protein folding and misfolding: neurodegenerative diseases. Netherlands: Springer; 2009. pp. 111–32.
    1. Inoue H, Kondo T, Lin L. Protein misfolding and axonal protection in neurodegenerative diseases. In: Ovádi J, Orosz F, et al., editors. Protein folding and misfolding: neurodegenerative diseases. Netherlands: Springer; 2009. pp. 97–110.
    1. Leverenz JB, Umar I, Wang Q, et al. Proteomic identification of novel proteins in cortical lewy bodies. Brain Pathol. 2007;17:139–45.
    1. Burns SA, Moran CR, Pingerelli PL, et al. Redefining the Lewy body: evidence of distinct protein composition and nonrandom assembly process (abstract) Mov Disord. 2008;23:S22.
    1. Tai HC, Schuman EM. Ubiquitin, the proteasome and protein degradation in neuronal function and dysfunction. Nat Rev Neurosci. 2008;9:826–38.
    1. Strobel G. The spectrum series: grappling with the overlap between Alzheimer’s and Parkinson’s diseases. J Alzheimers Dis. 2009;18:625–40.
    1. Arai T, Mackenzie IR, Hasegawa M, et al. Phosphorylated TDP-43 in Alzheimer’s disease and dementia with Lewy bodies. Acta Neuropathol. 2009;117:125–36.
    1. Fujishiro H, Tsuboi Y, Lin WL, et al. Co-localization of tau and alpha-synuclein in the olfactory bulb in Alzheimer’s disease with amygdala Lewy bodies. Acta Neuropathol. 2008;116:17–24.
    1. Tsigelny IF, Crews L, Desplats P, et al. Mechanisms of hybrid oligomer formation in the pathogenesis of combined Alzheimer’s and Parkinson’s diseases. PLoS One. 2008;3:e3135.
    1. Clarimon J, Molina-Porcel L, Gomez-Isla T, et al. Early-onset familial lewy body dementia with extensive tauopathy: a clinical, genetic, and neuropathological study. J Neuropathol Exp Neurol. 2009;68:73–82.
    1. Takahashi RH, Capetillo-Zarate E, Lin MT, et al. Co-occurrence of Alzheimer’s disease beta-amyloid and tau pathologies at synapses. Neurobiol Aging. 2008 ; in press: doi: .
    1. Muntane G, Dalfo E, Martinez A, et al. Phosphorylation of tau and alpha-synuclein in synaptic-enriched fractions of the frontal cortex in Alzheimer’s disease, and in Parkinson’s disease and related alpha-synucleinopathies. Neuroscience. 2008;152:913–23.
    1. Duka T, Rusnak M, Drolet RE, et al. Alpha-synuclein induces hyperphosphorylation of Tau in the MPTP model of parkinsonism. FASEB J. 2006;20:2302–12.
    1. Shao CY, Crary JF, Rao C, et al. Atypical protein kinase C in neurodegenerative disease II: PKCiota/lambda in tauopathies and alpha-synucleinopathies. J Neuropathol Exp Neurol. 2006;65:327–35.
    1. Higashi S, Iseki E, Yamamoto R, et al. Concurrence of TDP-43, tau and alpha-synuclein pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res. 2007;1184:284–94.
    1. Riedel M, Goldbaum O, Richter-Landsberg C. alpha-Synuclein promotes the recruitment of tau to protein inclusions in oligodendroglial cells: effects of oxidative and proteolytic stress. J Mol Neurosci. 2009;39:226–34.
    1. Obi K, Akiyama H, Kondo H, et al. Relationship of phosphorylated alpha-synuclein and tau accumulation to Abeta deposition in the cerebral cortex of dementia with Lewy bodies. Exp Neurol. 2008;210:409–20.
    1. Mandal PK, Pettegrew JW, Masliah E, et al. Interaction between Abeta peptide and alpha synuclein: molecular mechanisms in overlapping pathology of Alzheimer’s and Parkinson’s in dementia with Lewy body disease. Neurochem Res. 2006;31:1153–62.
    1. Fein JA, Sokolow S, Miller CA, et al. Co-localization of amyloid beta and tau pathology in Alzheimer’s disease synaptosomes. Am J Pathol. 2008;172:1683–92.
    1. Rhein V, Song X, Wiesner A, et al. Amyloid-beta and tau synergistically impair the oxidative phosphorylation system in triple transgenic Alzheimer’s disease mice. Proc Natl Acad Sci USA. 2009;106:20057–62.
    1. Kwong LK, Uryu K, Trojanowski JQ, et al. TDP-43 proteinopathies: neurodegenerative protein misfolding diseases without amyloidosis. Neurosignals. 2008;16:41–51.
    1. Lehman NL. The ubiquitin proteasome system in neuropathology. Acta Neuropathol. 2009;118:329–47.
    1. Hirsch C, Gauss R, Horn SC, et al. The ubiquitylation machinery of the endoplasmic reticulum. Nature. 2009;458:453–60.
    1. Ciechanover A. Tracing the history of the ubiquitin proteolytic system: the pioneering article. Biochem Biophys Res Commun. 2009;387:1–10.
    1. Huang L, Chen CH. Proteasome regulators: activators and inhibitors. Curr Med Chem. 2009;16:931–9.
    1. Schulman BA, Harper JW. Ubiquitin-like protein activation by E1 enzymes: the apex for downstream signalling pathways. Nat Rev Mol Cell Biol. 2009;10:319–31.
    1. Nedelsky NB, Todd PK, Taylor JP. Autophagy and the ubiquitin-proteasome system: collaborators in neuroprotection. Biochim Biophys Acta. 2008;1782:691–9.
    1. Kirkin V, McEwan DG, Novak I, et al. A role for ubiquitin in selective autophagy. Mol Cell. 2009;34:259–69.
    1. Leon S, Haguenauer-Tsapis R. Ubiquitin ligase adaptors: regulators of ubiquitylation and endocytosis of plasma membrane proteins. Exp Cell Res. 2009;315:1574–83.
    1. Eisele F, Wolf DH. Degradation of misfolded protein in the cytoplasm is mediated by the ubiquitin ligase Ubr1. FEBS Lett. 2008;582:4143–6.
    1. Finley D. Recognition and processing of ubiquitin-protein conjugates by the proteasome. Annu Rev Biochem. 2009;78:477–513.
    1. Ye Y, Rape M. Building ubiquitin chains: E2 enzymes at work. Nat Rev Mol Cell Biol. 2009;10:755–64.
    1. Setsuie R, Sakurai M, Sakaguchi Y, et al. Ubiquitin dimers control the hydrolase activity of UCH-L3. Neurochem Int. 2009;54:314–21.
    1. Su V, Lau AF. Ubiquitin-like and ubiquitin-associated domain proteins: significance in proteasomal degradation. Cell Mol Life Sci. 2009;66:2819–33.
    1. Vashisht AA, Zumbrennen KB, Huang X, et al. Control of iron homeostasis by an iron-regulated ubiquitin ligase. Science. 2009;326:718–21.
    1. Bergink S, Jentsch S. Principles of ubiquitin and SUMO modifications in DNA repair. Nature. 2009;458:461–7.
    1. Stehmeier P, Muller S. Regulation of p53 family members by the ubiquitin-like SUMO system. DNA Repair. 2009;8:491–8.
    1. Komander D. The emerging complexity of protein ubiquitination. Biochem Soc Trans. 2009;37:937–53.
    1. Xu P, Duong DM, Seyfried NT, et al. Quantitative proteomics reveals the function of unconventional ubiquitin chains in proteasomal degradation. Cell. 2009;137:133–45.
    1. Qiao L, Zhang J. Inhibition of lysosomal functions reduces proteasomal activity. Neurosci Lett. 2009;456:15–9.
    1. Wong ESP, Tan JMM, Lim K-L. Dynamic role of ubiquitination in the management of misfolded proteins associated with neurdegenerative diseases. In: Ovádi J, Orosz F, editors. Protein folding and misfolding: neurodegenerative diseases. Netherlands: Springer: 2009. pp. 77–95.
    1. Tydlacka S, Wang CE, Wang X, et al. Differential activities of the ubiquitin-proteasome system in neurons versus glia may account for the preferential accumulation of misfolded proteins in neurons. J Neurosci. 2008;28:13285–95.
    1. Martinet W, Agostinis P, Vanhoecke B, et al. Autophagy in disease: a double-edged sword with therapeutic potential. Clin Sci. 2009;116:697–712.
    1. Galluzzi L, Vicencio JM, Kepp O, et al. To die or not to die: that is the autophagic question. Curr Mol Med. 2008;8:78–91.
    1. Lee JA. Autophagy in neurodegeneration: two sides of the same coin. BMB Rep. 2009;42:324–30.
    1. Jaeger PA, Wyss-Coray T. All-you-can-eat: autophagy in neurodegeneration and neuroprotection. Mol Neurodegener. 2009;4:16.
    1. Eskelinen EL, Saftig P. Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta. 2009;1793:664–73.
    1. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132:27–42.
    1. Todde V, Veenhuis M, Van Der Klei IJ. Autophagy: principles and significance in health and disease. Biochim Biophys Acta. 2009;1792:3–13.
    1. Yue Z, Friedman L, Komatsu M, et al. The cellular pathways of neuronal autophagy and their implication in neurodegenerative diseases. Biochim Biophys Acta. 2009;1793:1496–507.
    1. He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 2009
    1. Chu CT, Plowey ED, Dagda RK, et al. Autophagy in neurite injury and neurodegeneration: in vitro and in vivo models. Methods Enzymol. 2009;453:217–49.
    1. Chen Y, Azad MB, Gibson SB. Superoxide is the major reactive oxygen species regulating autophagy. Cell Death Differ. 2009;16:1040–52.
    1. Puyal J, Ginet V, Vaslin A, et al. [The two faces of autophagy in the nervous system] Med Sci. 2009;25:383–90.
    1. Tolkovsky AM. Mitophagy. Biochim Biophys Acta. 2009;1793:1508–15.
    1. Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature. 2006;443:780–6.
    1. Cuervo AM, Stefanis L, Fredenburg R, et al. Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science. 2004;305:1292–5.
    1. Martinez-Vicente M, Talloczy Z, Kaushik S, et al. Dopamine-modified alpha-synuclein blocks chaperone-mediated autophagy. J Clin Invest. 2008;118:777–88.
    1. Spencer B, Potkar R, Trejo M, et al. Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson’s and Lewy body diseases. J Neurosci. 2009;29:13578–88.
    1. Pickford F, Masliah E, Britschgi M, et al. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest. 2008;118:2190–9.
    1. Hetz C, Thielen P, Matus S, et al. XBP-1 deficiency in the nervous system protects against amyotrophic lateral sclerosis by increasing autophagy. Genes Dev. 2009;23:2294–306.
    1. Uchiyama Y, Shibata M, Koike M, et al. Autophagy-physiology and pathophysiology. Histochem Cell Biol. 2008;129:407–20.
    1. Korolchuk VI, Mansilla A, Menzies FM, et al. Autophagy inhibition compromises degradation of ubiquitin-proteasome pathway substrates. Mol Cell. 2009;33:517–27.
    1. Cherra SJ, Chu CT. Autophagy in neuroprotection and neurodegeneration: a question of balance. Future Neurol. 2008;3:309–23.
    1. Tooze SA, Schiavo G. Liaisons dangereuses: autophagy, neuronal survival and neurodegeneration. Curr Opin Neurobiol. 2008;18:504–15.
    1. Rami A. Review: autophagy in neurodegeneration: firefighter and/or incendiarist. Neuropathol Appl Neurobiol. 2009;35:449–61.
    1. Li H, Guo M. Protein degradation in Parkinson disease revisited: it’s complex. J Clin Invest. 2009;119:442–5.
    1. Lim KL. Ubiquitin-proteasome system dysfunction in Parkinson’s disease: current evidence and controversies. Expert Rev Proteomics. 2007;4:769–81.
    1. Pan T, Kondo S, Le W, et al. The role of autophagy-lysosome pathway in neurodegeneration associated with Parkinson’s disease. Brain. 2008;131:1969–78.
    1. Eliezer D. Synuclein structure and function in Parkinson’s disease. In: Ovádi J, Orosz F, editors. Protein folding and misfolding: neurodegenerative diseases. Netherlands: Springer; 2009. pp. 159–74.
    1. Schlossmacher MG, Frosch MP, Gai WP, et al. Parkin localizes to the Lewy bodies of Parkinson disease and dementia with Lewy bodies. Am J Pathol. 2002;160:1655–67.
    1. Xiong H, Wang D, Chen L, et al. Parkin, PINK1, and DJ-1 form a ubiquitin E3 ligase complex promoting unfolded protein degradation. J Clin Invest. 2009;119:650–60.
    1. Schiesling C, Kieper N, Seidel K, et al. Review: familial Parkinson’s disease–genetics, clinical phenotype and neuropathology in relation to the common sporadic form of the disease. Neuropathol Appl Neurobiol. 2008;34:255–71.
    1. Haugarvoll K, Rademakers R, Kachergus JM, et al. LRRK2 R1441C parkinsonism is clinically similar to sporadic Parkinson disease. Neurology. 2008;70:1456–60.
    1. Zimprich A, Biskup S, Leitner P, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601–7.
    1. Whaley NR, Uitti RJ, Dickson DW, et al. Clinical and pathologic features of families with LRRK2-associated Parkinson’s disease. J Neural Transm Suppl. 2006:221–9.
    1. Santpere G, Ferrer I. LRRK2 and neurodegeneration. Acta Neuropathol. 2009;117:227–46.
    1. Oddo S. The ubiquitin-proteasome system in Alzheimer’s disease. J Cell Mol Med. 2008;12:363–73.
    1. Tseng BP, Green KN, Chan JL, et al. Abeta inhibits the proteasome and enhances amyloid and tau accumulation. Neurobiol Aging. 2008;29:1607–18.
    1. Liu YH, Wei W, Yin J, et al. Proteasome inhibition increases tau accumulation independent of phosphorylation. Neurobiol Aging. 2009;30:1949–61.
    1. Zouambia M, Fischer DF, Hobo B, et al. Proteasome subunit proteins and neuropathology in tauopathies and synucleinopathies: consequences for proteomic analyses. Proteomics. 2008;8:1221–36.
    1. Gil JM, Rego AC. Mechanisms of neurodegeneration in Huntington’s disease. Eur J Neurosci. 2008;27:2803–20.
    1. Iwata A, Nagashima Y, Matsumoto L, et al. Intranuclear degradation of polyglutamine aggregates by the ubiquitin-proteasome system. J Biol Chem. 2009;284:9796–803.
    1. Mitra S, Tsvetkov AS, Finkbeiner S. Single neuron ubiquitin-proteasome dynamics accompanying inclusion body formation in Huntington disease. J Biol Chem. 2009;284:4398–403.
    1. Wang J, Wang CE, Orr A, et al. Impaired ubiquitin-proteasome system activity in the synapses of Huntington’s disease mice. J Cell Biol. 2008;180:1177–89.
    1. Davies JE, Sarkar S, Rubinsztein DC. The ubiquitin proteasome system in Huntington’s disease and the spinocerebellar ataxias. BMC Biochem. 2007;8:S2.
    1. Olzmann JA, Li L, Chin LS. Aggresome formation and neurodegenerative diseases: therapeutic implications. Curr Med Chem. 2008;15:47–60.
    1. Chu Y, Dodiya H, Aebischer P, et al. Alterations in lysosomal and proteasomal markers in Parkinson’s disease: relationship to alpha-synuclein inclusions. Neurobiol Dis. 2009;35:385–98.
    1. Mishra A, Godavarthi SK, Maheshwari M, et al. The ubiquitin ligase E6-AP is induced and recruited to aggresomes in response to proteasome inhibition and may be involved in the ubiquitination of Hsp70-bound misfolded proteins. J Biol Chem. 2009;284:10537–45.
    1. Gardiner J, Barton D, Overall R, Marc J. Neurotrophic support and oxidative stress: converging effects in the normal and diseased nervous system. Neuroscientist. 2009;15:47–61.
    1. Sultana R, Butterfield DA. Oxidatively modified, mitochondria-relevant brain proteins in subjects with Alzheimer disease and mild cognitive impairment. J Bioenerg Biomembr. 2009;41:441–6.
    1. Halliwell B. Oxidative stress and neurodegeneration: where are we now. J Neurochem. 2006;97:1634–58.
    1. Nunomura A, Hofer T, Moreira PI, et al. RNA oxidation in Alzheimer disease and related neurodegenerative disorders. Acta Neuropathol. 2009;118:151–66.
    1. Martinez A, Portero-Otin M, Pamplona R, et al. Protein targets of oxidative damage in human neurodegenerative diseases with abnormal protein aggregates. Brain Pathol. 2009 DOI: .
    1. Nakamura T, Lipton SA. Cell death: protein misfolding and neurodegenerative diseases. Apoptosis. 2009;14:455–68.
    1. Bond CE, Greenfield SA. Multiple cascade effects of oxidative stress on astroglia. Glia. 2007;55:1348–61.
    1. Ballatori N, Krance SM, Notenboom S, et al. Glutathione dysregulation and the etiology and progression of human diseases. Biol Chem. 2009;390:191–214.
    1. Castellani RJ, Nunomura A, Rolston RK, et al. Sublethal RNA oxidation as a mechanism for neurodegenerative disease. Int J Mol Sci. 2008;9:789–806.
    1. Migliore L, Coppede F. Environmental-induced oxidative stress in neurodegenerative disorders and aging. Mutat Res. 2009;674:73–84.
    1. Browne SE, Beal MF. Oxidative damage in Huntington’s disease pathogenesis. Antioxid Redox Signal. 2006;8:2061–73.
    1. Wang J, Markesbery WR, Lovell MA. Increased oxidative damage in nuclear and mitochondrial DNA in mild cognitive impairment. J Neurochem. 2006;96:825–32.
    1. Pratico D, Sung S. Lipid peroxidation and oxidative imbalance: early functional events in Alzheimer’s disease. J Alzheimers Dis. 2004;6:171–5.
    1. Mancuso M, Coppede F, Migliore L, et al. Mitochondrial dysfunction, oxidative stress and neurodegeneration. J Alzheimers Dis. 2006;10:59–73.
    1. Sayre LM, Perry G, Smith MA. Oxidative stress and neurotoxicity. Chem Res Toxicol. 2008;21:172–88.
    1. Ward RJ, Crichton RR. Iron and its role in neurodegenerative diseases. In: Sigel A, Sigel H, Sigel RKO, editors. Metal ions in life sciences. Vol. 1. New York: Wiley & Sons; 2006. pp. 227–79.
    1. Mancuso M, Davidzon G, Kurlan RM, et al. Hereditary ferritinopathy: a novel mutation, its cellular pathology, and pathogenetic insights. J Neuropathol Exp Neurol. 2005;64:280–94.
    1. Sparaco M, Gaeta LM, Santorelli FM, et al. Friedreich’s ataxia: oxidative stress and cytoskeletal abnormalities. J Neurol Sci. 2009;287:111–8.
    1. Altamura S, Muckenthaler MU. Iron toxicity in diseases of aging: Alzheimer’s disease, Parkinson’s disease and atherosclerosis. J Alzheimers Dis. 2009;16:879–95.
    1. Arreguin S, Nelson P, Padway S, et al. Dopamine complexes of iron in the etiology and pathogenesis of Parkinson’s disease. J Inorg Biochem. 2009;103:87–93.
    1. Sultana R, Perluigi M, Butterfield DA. Oxidatively modified proteins in Alzheimer’s disease (AD), mild cognitive impairment and animal models of AD: role of Abeta in pathogenesis. Acta Neuropathol. 2009 ; in press: doi. .
    1. Sonnen JA, Breitner JC, Lovell MA, et al. Free radical-mediated damage to brain in Alzheimer’s disease and its transgenic mouse models. Free Radic Biol Med. 2008;45:219–30.
    1. Zheng L, Kagedal K, Dehvari N, et al. Oxidative stress induces macroautophagy of amyloid beta-protein and ensuing apoptosis. Free Radic Biol Med. 2009;46:422–9.
    1. Reddy PH, Beal MF. Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med. 2008;14:45–53.
    1. Yan SD, Bierhaus A, Nawroth PP, et al. RAGE and Alzheimer’s disease: a progression factor for amyloid-beta-induced cellular perturbation. J Alzheimers Dis. 2009;16:833–43.
    1. Origlia N, Righi M, Capsoni S, et al. Receptor for advanced glycation end product-dependent activation of p38 mitogen-activated protein kinase contributes to amyloid-beta-mediated cortical synaptic dysfunction. J Neurosci. 2008;28:3521–30.
    1. Wang MY, Ross-Cisneros FN, Aggarwal D, et al. Receptor for advanced glycation end products is upregulated in optic neuropathy of Alzheimer’s disease. Acta Neuropathol. 2009;118:381–9.
    1. Takuma K, Fang F, Zhang W, et al. RAGE-mediated signaling contributes to intraneuronal transport of amyloid-beta and neuronal dysfunction. Proc Natl Acad Sci USA. 2009;106:20021–6.
    1. Pedersen PL. Mitochondrial matters of the brain: amyloid formation and Alzheimer’s disease introduction. J Bioenerg Biomembr. 2009;41:403–5.
    1. Reed TT, Pierce WM, Jr, Turner DM, et al. Proteomic identification of nitrated brain proteins in early Alzheimer’s disease inferior parietal lobule. J Cell Mol Med. 2008;13:2019–29.
    1. Sun KH, De Pablo Y, Vincent F, et al. Deregulated Cdk5 promotes oxidative stress and mitochondrial dysfunction. J Neurochem. 2008;107:265–78.
    1. Cho DH, Nakamura T, Fang J, et al. S-nitrosylation of Drp1 mediates beta-amyloid-related mitochondrial fission and neuronal injury. Science. 2009;324:102–5.
    1. Zhao B. Natural antioxidants protect neurons in Alzheimer’s disease and Parkinson’s disease. Neurochem Res. 2009;34:630–8.
    1. Miller RL, James-Kracke M, Sun GY, et al. Oxidative and inflammatory pathways in Parkinson’s disease. Neurochem Res. 2009;34:55–65.
    1. Jenner P. Oxidative stress and Parkinson’s disease. Handb Clin Neurol. 2007;83:507–20.
    1. Tsang AH, Chung KK. Oxidative and nitrosative stress in Parkinson’s disease. Biochim Biophys Acta. 2008;1792:643–50.
    1. Olivares D, Huang X, Branden L, et al. Physiological and pathological role of alpha-synuclein in Parkinson’s disease through iron mediated oxidative stress; the role of a putative iron-responsive element. Int J Mol Sci. 2009;10:1226–60.
    1. Tribl F, Asan E, Arzberger T, et al. Identification of L-ferritin in neuromelanin granules of the human substantia nigra: a targeted proteomics approach. Mol Cell Proteomics. 2009;8:1832–8.
    1. Martin HL, Teismann P. Glutathione–a review on its role and significance in Parkinson’s disease. FASEB J. 2009;23:3263–72.
    1. Shaikh S, Nicholson LF. Advanced glycation end products induce in vitro cross-linking of alpha-synuclein and accelerate the process of intracellular inclusion body formation. J Neurosci Res. 2008;86:2071–82.
    1. Power JH, Blumbergs PC. Cellular glutathione peroxidase in human brain: cellular distribution, and its potential role in the degradation of Lewy bodies in Parkinson’s disease and dementia with Lewy bodies. Acta Neuropathol. 2009;117:63–73.
    1. Rochet J-C, Liu F. Inhibition of alpha-synuclein aggregation by antioxidants and chaperones in Parkinson’s disease. In: Ovádi J, Orosz F, editors. Protein folding and misfolding: neurodegenerative diseases. Netherlands: Springer; 2009. pp. 175–206.
    1. Przedborski S. Moving Along (Newsletter of the Movement Disorder Soc. 2009;13:1.
    1. Levy OA, Malagelada C, Greene LA. Cell death pathways in Parkinson’s disease: proximal triggers, distal effectors, and final steps. Apoptosis. 2009;14:478–500.
    1. Nagley P, Higgins GC, Atkin JD, et al. Multifaceted deaths orchestrated by mitochondria in neurones. BBA – Molec Basis Dis. 2010;1802:167–85.
    1. Sorolla MA, Reverter-Branchat G, Tamarit J, et al. Proteomic and oxidative stress analysis in human brain samples of Huntington disease. Free Radic Biol Med. 2008;45:667–78.
    1. Martinez A, Dalfo E, Muntane G, et al. Glycolitic enzymes are targets of oxidation in aged human frontal cortex and oxidative damage of these proteins is increased in progressive supranuclear palsy. J Neural Transm. 2008;115:59–66.
    1. Santpere G, Ferrer I. Delineation of early changes in cases with progressive supranuclear palsy-like pathology. Astrocytes in striatum are primary targets of tau phosphorylation and GFAP oxidation. Brain Pathol. 2009;19:177–87.
    1. Pasinelli P, Belford ME, Lennon N, et al. Amyotrophic lateral sclerosis-associated SOD1 mutant proteins bind and aggregate with Bcl-2 in spinal cord mitochondria. Neuron. 2004;43:19–30.
    1. Magrane J, Hervias I, Henning MS, et al. Mutant SOD1 in neuronal mitochondria causes toxicity and mitochondrial dynamics abnormalities. Hum Mol Genet. 2009;18:4552–64.
    1. Barber SC, Mead RJ, Shaw PJ. Oxidative stress in ALS: a mechanism of neurodegeneration and a therapeutic target. Biochim Biophys Acta. 2006;1762:1051–67.
    1. Baumer D, Parkinson N, Talbot K. TARDBP in amyotrophic lateral sclerosis: identification of a novel variant but absence of copy number variation. Journal of Neurology Neurosurgery and Psychiatry. 2009;80:1283–5.
    1. Yang JL, Weissman L, Bohr VA, et al. Mitochondrial DNA damage and repair in neurodegenerative disorders. DNA Repair. 2008;7:1110–20.
    1. Gupta S, Kass GE, Szegezdi E, et al. The mitochondrial death pathway: a promising therapeutic target in diseases. J Cell Mol Med. 2009;13:1004–33.
    1. Perkins G, Bossy-Wetzel E, Ellisman MH. New insights into mitochondrial structure during cell death. Exp Neurol. 2009;218:183–92.
    1. Dubinsky JM. Heterogeneity of nervous system mitochondria: location, location, location. Exp Neurol. 2009;218:293–307.
    1. Reddy PH. Mitochondrial medicine for aging and neurodegenerative diseases. Neuromolecular Med. 2008;10:291–315.
    1. Chen H, Chan DC. Mitochondrial dynamics–fusion, fission, movement, and mitophagy–in neurodegenerative diseases. Hum Mol Genet. 2009;18:R169–76.
    1. Celsi F, Pizzo P, Brini M, et al. Mitochondria, calcium and cell death: a deadly triad in neurodegeneration. Biochim Biophys Acta. 2009;1787:335–44.
    1. Schapira AH. Mitochondria in the aetiology and pathogenesis of Parkinson’s disease. Lancet Neurol. 2008;7:97–109.
    1. Banoei MM, Houshmand M, Panahi MS, et al. Huntington’s disease and mitochondrial DNA deletions: event or regular mechanism for mutant huntingtin protein and CAG repeats expansion. Cell Mol Neurobiol. 2007;27:867–75.
    1. Quintanilla RA, Johnson GV. Role of mitochondrial dysfunction in the pathogenesis of Huntington’s disease. Brain Res Bull. 2009;80:242–7.
    1. Distelmaier F, Koopman WJ, Van Den Heuvel LP, et al. Mitochondrial complex I deficiency: from organelle dysfunction to clinical disease. Brain. 2009;132:833–42.
    1. Armstrong JS. Mitochondrial membrane permeabilization: the sine qua non for cell death. Bioessays. 2006;28:253–60.
    1. Jeyaraju DV, Cisbani G, Pellegrini L. Calcium regulation of mitochondria motility and morphology. Biochim Biophys Acta. 2009;1787:1363–73.
    1. Nicholls DG. Mitochondrial calcium function and dysfunction in the central nervous system. Biochim Biophys Acta. 2009;1787:1416–24.
    1. Bueler H. Impaired mitochondrial dynamics and function in the pathogenesis of Parkinson’s disease. Exp Neurol. 2009;218:235–46.
    1. Martin LJ. DNA damage and repair: relevance to mechanisms of neurodegeneration. J Neuropathol Exp Neurol. 2008;67:377–87.
    1. Katyal S, McKinnon PJ. DNA repair deficiency and neurodegeneration. Cell Cycle. 2007;6:2360–5.
    1. Coppede F, Migliore L. DNA damage and repair in Alzheimer’s disease. Curr Alzheimer Res. 2009;6:36–47.
    1. Lu B. Mitochondrial dynamics and neurodegeneration. Curr Neurol Neurosci Rep. 2009;9:212–9.
    1. Cogswell JP, Ward J, Taylor IA, et al. Identification of miRNA changes in Alzheimer’s disease brain and CSF yields putative biomarkers and insights into disease pathways. J Alzheimers Dis. 2008;14:27–41.
    1. Saba R, Goodman CD, Huzarewich RL, et al. A miRNA signature of prion induced neurodegeneration. PLoS One. 2008;3:e3652.
    1. Hebert SS, De Strooper B. Alterations of the microRNA network cause neurodegenerative disease. Trends Neurosci. 2009;32:199–206.
    1. Nelson PT, Wang WX, Rajeev BW. MicroRNAs (miRNAs) in neurodegenerative diseases. Brain Pathol. 2008;18:130–8.
    1. Bushati N, Cohen SM. MicroRNAs in neurodegeneration. Curr Opin Neurobiol. 2008;18:292–6.
    1. Swerdlow RH, Khan SM. The Alzheimer’s disease mitochondrial cascade hypothesis: an update. Exp Neurol. 2009;218:308–15.
    1. Mancuso M, Calsolaro V, Orsucci D, et al. Is there a primary role of the mitochondrial genome in Alzheimer’s disease. J Bioenerg Biomembr. 2009;41:411–6.
    1. Wang X, Su B, Zheng L, et al. The role of abnormal mitochondrial dynamics in the pathogenesis of Alzheimer’s disease. J Neurochem. 2009;109:153–9.
    1. Hauptmann S, Scherping I, Drose S, et al. Mitochondrial dysfunction: an early event in Alzheimer pathology accumulates with age in AD transgenic mice. Neurobiol Aging. 2009;30:1574–86.
    1. Melov S, Adlard PA, Morten K, et al. Mitochondrial oxidative stress causes hyperphosphorylation of tau. PLoS One. 2007;2:e536.
    1. De Vos KJ, Grierson AJ, Ackerley S, et al. Role of axonal transport in neurodegenerative diseases. Annu Rev Neurosci. 2008;31:151–73.
    1. Quintanilla RA, Matthews-Roberson TA, Dolan PJ, et al. Caspase-cleaved tau expression results in mitochondrial dysfunction in cortical neurons. Implications for the pathogenesis of Alzheimer disease. J Biol Chem. 2009;284:18754–66.
    1. Terni B, Boada J, Portero-Otin M, et al. Mitochondrial ATP-Synthase in the entorhinal cortex is a target of oxidative stress at stages I/II of Alzheimer’s disease pathology. Brain Pathol. 2009 Doi: .
    1. Shapira IL. The Nun study. J Am Dent Assoc. 2008;139:130–1.
    1. Onyango IG. Mitochondrial dysfunction and oxidative stress in Parkinson’s disease. Neurochem Res. 2008;33:589–97.
    1. Vila M, Ramonet D, Perier C. Mitochondrial alterations in Parkinson’s disease: new clues. J Neurochem. 2008;107:317–28.
    1. Yao Z, Wood NW. Cell death pathways in Parkinson’s disease: role of mitochondria. Antioxid Redox Signal. 2009;11:2135–49.
    1. Davison EJ, Pennington K, Hung CC, et al. Proteomic analysis of increased Parkin expression and its interactants provides evidence for a role in modulation of mitochondrial function. Proteomics. 2009;9:4284–97.
    1. Van Laar VS, Berman SB. Mitochondrial dynamics in Parkinson’s disease. Exp Neurol. 2009;218:247–56.
    1. Berger AK, Cortese GP, Amodeo KD, et al. Parkin selectively alters the intrinsic threshold for mitochondrial cytochrome c release. Hum Mol Genet. 2009;18:4317–28.
    1. Burns MP, Zhang L, Rebeck GW, et al. Parkin promotes intracellular Abeta1-42 clearance. Hum Mol Genet. 2009;18:3206–16.
    1. Park J, Lee G, Chung J. The PINK1-Parkin pathway is involved in the regulation of mitochondrial remodeling process. Biochem Biophys Res Commun. 2009;378:518–23.
    1. Kao SY. Regulation of DNA repair by parkin. Biochem Biophys Res Commun. 2009;382:321–5.
    1. Yang Y, Lu B. Mitochondrial morphogenesis, distribution, and Parkinson disease: insights from PINK1. J Neuropathol Exp Neurol. 2009;68:953–63.
    1. Gupta A, Dawson VL, Dawson TM. What causes cell death in Parkinson’s disease. Ann Neurol. 2008;64:S3–15.
    1. Freestone PS, Chung KK, Guatteo E, et al. Acute action of rotenone on nigral dopaminergic neurons – involvement of reactive oxygen species and disruption of Ca homeostasis. Eur J Neurosci. 2009;30:1849–59.
    1. Sherer TB, Betarbet R, Testa CM, et al. Mechanism of toxicity in rotenone models of Parkinson’s disease. J Neurosci. 2003;23:10756–64.
    1. Drechsel DA, Patel M. Role of reactive oxygen species in the neurotoxicity of environmental agents implicated in Parkinson’s disease. Free Radic Biol Med. 2008;44:1873–86.
    1. Alvarez-Fischer D, Guerreiro S, Hunot S, et al. Modelling Parkinson-like neurodegeneration via osmotic minipump delivery of MPTP and probenecid. J Neurochem. 2008;107:701–11.
    1. Halliday G, Herrero MT, Murphy K, et al. No Lewy pathology in monkeys with over 10 years of severe MPTP Parkinsonism. Mov Disord. 2009;24:1519–23.
    1. Choi WS, Kruse SE, Palmiter RD, et al. Mitochondrial complex I inhibition is not required for dopaminergic neuron death induced by rotenone, MPP+, or paraquat. Proc Natl Acad Sci USA. 2008;105:15136–41.
    1. Borland MK, Trimmer PA, Rubinstein JD, et al. Chronic, low-dose rotenone reproduces Lewy neurites found in early stages of Parkinson’s disease, reduces mitochondrial movement and slowly kills differentiated SH-SY5Y neural cells. Mol Neurodegener. 2008;3:21.
    1. Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443:787–95.
    1. Lim D, Fedrizzi L, Tartari M, et al. Calcium homeostasis and mitochondrial dysfunction in striatal neurons of Huntington disease. J Biol Chem. 2008;283:5780–9.
    1. Estrada Sanchez AM, Mejia-Toiber J, Massieu L. Excitotoxic neuronal death and the pathogenesis of Huntington’s disease. Arch Med Res. 2008;39:265–76.
    1. Chang WH, Tien CL, Chen TJ, et al. Decreased protein synthesis of Hsp27 associated with cellular toxicity in a cell model of Machado-Joseph disease. Neuroscience Letters. 2009;454:152–6.
    1. Yu YC, Kuo CL, Cheng WL, et al. Decreased antioxidant enzyme activity and increased mitochondrial DNA damage in cellular models of Machado-Joseph disease. J Neurosci Res. 2009;87:1884–91.
    1. Fan J, Hu Z, Zeng L, et al. Golgi apparatus and neurodegenerative diseases. Int J Dev Neurosci. 2008;26:523–34.
    1. Gonatas NK, Stieber A, Gonatas JO. Fragmentation of the Golgi apparatus in neurodegenerative diseases and cell death. J Neurol Sci. 2006;246:21–30.
    1. Fujita Y, Ohama E, Takatama M, et al. Fragmentation of Golgi apparatus of nigral neurons with alpha-synuclein-positive inclusions in patients with Parkinson’s disease. Acta Neuropathol. 2006;112:261–5.
    1. Vickers JC, King AE, Woodhouse A, et al. Axonopathy and cytoskeletal disruption in degenerative diseases of the central nervous system. Brain Res Bull. 2009;80:217–23.
    1. Morfini GA, Burns M, Binder LI, et al. Axonal transport defects in neurodegenerative diseases. J Neurosci. 2009;29:12776–86.
    1. Prins KW, Humston JL, Mehta A, et al. Dystrophin is a microtubule-associated protein. J Cell Biol. 2009;186:363–9.
    1. Yates DM, Manser C, De Vos KJ, et al. Neurofilament subunit (NFL) head domain phosphorylation regulates axonal transport of neurofilaments. Eur J Cell Biol. 2009;88:193–202.
    1. Goldstein AY, Wang X, Schwarz TL. Axonal transport and the delivery of pre-synaptic components. Curr Opin Neurobiol. 2008;18:495–503.
    1. Barry DM, Millecamps S, Julien JP, et al. New movements in neurofilament transport, turnover and disease. Exp Cell Res. 2007;313:2110–20.
    1. Yuan A, Rao MV, Kumar A, et al. Neurofilament transport in vivo minimally requires hetero-oligomer formation. J Neurosci. 2003;23:9452–8.
    1. Hirokawa N, Noda Y, Tanaka Y, et al. Kinesin superfamily motor proteins and intracellular transport. Nat Rev Mol Cell Biol. 2009;10:682–96.
    1. Roy S, Zhang B, Lee VM, et al. Axonal transport defects: a common theme in neurodegenerative diseases. Acta Neuropathol. 2005;109:5–13.
    1. Schindowski K, Belarbi K, Buee L. Neurotrophic factors in Alzheimer’s disease: role of axonal transport. Genes Brain Behav. 2008;7:43–56.
    1. Stokin GB, Goldstein LS. Axonal transport and Alzheimer’s disease. Annu Rev Biochem. 2006;75:607–27.
    1. Muresan V, Varvel NH, Lamb BT, et al. The cleavage products of amyloid-beta precursor protein are sorted to distinct carrier vesicles that are independently transported within neurites. J Neurosci. 2009;29:3565–78.
    1. Pigino G, Morfini G, Atagi Y, et al. Disruption of fast axonal transport is a pathogenic mechanism for intraneuronal amyloid beta. Proc Natl Acad Sci USA. 2009;106:5907–12.
    1. Shah SB, Nolan R, Davis E, et al. Examination of potential mechanisms of amyloid-induced defects in neuronal transport. Neurobiol Dis. 2009;36:11–25.
    1. Stoothoff W, Jones PB, Spires-Jones TL, et al. Differential effect of three-repeat and four-repeat tau on mitochondrial axonal transport. J Neurochem. 2009;111:417–27.
    1. Falzone TL, Stokin GB, Lillo C, et al. Axonal stress kinase activation and tau misbehavior induced by kinesin-1 transport defects. J Neurosci. 2009;29:5758–67.
    1. Yuan A, Kumar A, Peterhoff C, et al. Axonal transport rates in vivo are unaffected by tau deletion or overexpression in mice. J Neurosci. 2008;28:1682–7.
    1. Li W, Hoffman PN, Stirling W, et al. Axonal transport of human alpha-synuclein slows with aging but is not affected by familial Parkinson’s disease-linked mutations. J Neurochem. 2004;88:401–10.
    1. Kramer ML, Schulz-Schaeffer WJ. Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci. 2007;27:1405–10.
    1. Rothstein JD. Current hypotheses for the underlying biology of amyotrophic lateral sclerosis. Ann Neurol. 2009;65:S3–9.
    1. Strom AL, Gal J, Shi P, et al. Retrograde axonal transport and motor neuron disease. J Neurochem. 2008;106:495–505.
    1. Her LS, Goldstein LS. Enhanced sensitivity of striatal neurons to axonal transport defects induced by mutant huntingtin. J Neurosci. 2008;28:13662–72.
    1. Bossers K, Meerhoff G, Balesar R, et al. Analysis of gene expression in Parkinson’s disease: possible involvement of neurotrophic support and axon guidance in dopaminergic cell death. Brain Pathol. 2009;19:91–107.
    1. Peng S, Garzon DJ, Marchese M, et al. Decreased brain-derived neurotrophic factor depends on amyloid aggregation state in transgenic mouse models of Alzheimer’s disease. J Neurosci. 2009;29:9321–9.
    1. Tapia-Arancibia L, Aliaga E, Silhol M, et al. New insights into brain BDNF function in normal aging and Alzheimer disease. Brain Res Rev. 2008;59:201–20.
    1. Bulbarelli A, Lonati E, Cazzaniga E, et al. TrkA pathway activation induced by amyloid-beta (Abeta) Mol Cell Neurosci. 2009;40:365–73.
    1. Arevalo MA, Roldan PM, Chacon PJ, et al. Amyloid beta serves as an NGF-like neurotrophic factor or acts as a NGF antagonist depending on its concentration. Journal of Neurochemistry. 2009;111:1425–33.
    1. Sotthibundhu A, Sykes AM, Fox B, et al. Beta-amyloid(1–42) induces neuronal death through the p75 neurotrophin receptor. J Neurosci. 2008;28:3941–6.
    1. Schulte-Herbruggen O, Jockers-Scherubl MC, Hellweg R. Neurotrophins: from pathophysiology to treatment in Alzheimer’s disease. Curr Alzheimer Res. 2008;5:38–44.
    1. Fombonne J, Rabizadeh S, Banwait S, et al. Selective vulnerability in Alzheimer’s disease: amyloid precursor protein and p75(NTR) interaction. Ann Neurol. 2009;65:294–303.
    1. McAlpine FE, Lee JK, Harms AS, et al. Inhibition of soluble TNF signaling in a mouse model of Alzheimer’s disease prevents pre-plaque amyloid-associated neuropathology. Neurobiol Dis. 2009;34:163–77.
    1. Nikolaev A, McLaughlin T, O’Leary DD, et al. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature. 2009;457:981–9.
    1. Bruno MA, Leon WC, Fragoso G, et al. Amyloid beta-induced nerve growth factor dysmetabolism in Alzheimer disease. J Neuropathol Exp Neurol. 2009;68:857–69.
    1. Nagahara AH, Merrill DA, Coppola G, et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat Med. 2009;15:331–7.
    1. Zuccato C, Cattaneo E. Brain-derived neurotrophic factor in neurodegenerative diseases. Nat Rev Neurol. 2009;5:311–22.
    1. Zuccato C, Marullo M, Conforti P, et al. Systematic assessment of BDNF and its receptor levels in human cortices affected by Huntington’s disease. Brain Pathol. 2008;18:225–38.
    1. Stolp HB, Dziegielewska KM. Review: role of developmental inflammation and blood-brain barrier dysfunction in neurodevelopmental and neurodegenerative diseases. Neuropathol Appl Neurobiol. 2009;35:132–46.
    1. Kell DB. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med Genomics. 2009;2:2.
    1. Galimberti D, Fenoglio C, Scarpini E. Inflammation in neurodegenerative disorders: friend or foe. Curr Aging Sci. 2008;1:30–41.
    1. Piehl F, Olsson T. Inflammation and susceptibility to neurodegeneration: the use of unbiased genetics to decipher critical regulatory pathways. Neuroscience. 2009;158:1143–50.
    1. Graber JJ, Dhib-Jalbut S. Protective autoimmunity in the nervous system. Pharmacol Ther. 2009;121:147–59.
    1. Salminen A, Ojala J, Kauppinen A, et al. Inflammation in Alzheimer’s disease: amyloid-ß oligomers trigger innate immunity defence via pattern recognition receptors. Progr Neurobiol. 2009;87:181–94.
    1. Lucin KM, Wyss-Coray T. Immune activation in brain aging and neurodegeneration: too much or too little. Neuron. 2009;64:110–22.
    1. Sawada M, Sawada H, Nagatsu T. Effects of aging on neuroprotective and neurotoxic properties of microglia in neurodegenerative diseases. Neurodegener Dis. 2008;5:254–6.
    1. Neumann H, Kotter MR, Franklin RJ. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain. 2009;132:288–95.
    1. Lee M, Schwab C, Yu S, et al. Astrocytes produce the antiinflammatory and neuroprotective agent hydrogen sulfide. Neurobiol Aging. 2009;30:1523–34.
    1. Sondag CM, Dhawan G, Combs CK. Beta amyloid oligomers and fibrils stimulate differential activation of primary microglia. J Neuroinflammation. 2009;6:1–13.
    1. Reale M, Iarlori C, Feliciani C, et al. Peripheral chemokine receptors, their ligands, cytokines and Alzheimer’s disease. J Alzheimers Dis. 2008;14:147–59.
    1. Maccioni RB, Rojo LE, Fernandez JA, et al. The role of neuroimmunomodulation in Alzheimer’s disease. Ann N Y Acad Sci. 2009;1153:240–6.
    1. Holmes C, Cunningham C, Zotova E, et al. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009;73:768–74.
    1. Jaeger LB, Dohgu S, Sultana R, et al. Lipopolysaccharide alters the blood-brain barrier transport of amyloid beta protein: a mechanism for inflammation in the progression of Alzheimer’s disease. Brain Behavior and Immunity. 2009;23:507–17.
    1. Vukic V, Callaghan D, Walker D, et al. Expression of inflammatory genes induced by beta-amyloid peptides in human brain endothelial cells and in Alzheimer’s brain is mediated by the JNK-AP1 signaling pathway. Neurobiol Dis. 2009;34:95–106.
    1. Di Bona D, Candore G, Franceschi C, et al. Systematic review by meta-analyses on the possible role of TNF-alpha polymorphisms in association with Alzheimer’s disease. Brain Res Rev. 2009;61:60–8.
    1. McCoy MK, Tansey MG. TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease. J Neuroinflammation. 2008;5:45.
    1. Foster JK, Verdile G, Bates KA, et al. Immunization in Alzheimer’s disease: naive hope or realistic clinical potential. Mol Psychiatry. 2009;14:239–51.
    1. Holmes C, Boche D, Wilkinson D, et al. Long-term effects of Abeta42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet. 2008;372:216–23.
    1. Deane R, Bell RD, Sagare A, et al. Clearance of amyloid-beta peptide across the blood-brain barrier: implication for therapies in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2009;8:16–30.
    1. Streit WJ, Braak H, Xue QS, et al. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 2009;118:475–85.
    1. Chakrabarty P, Jansen-West K, Beccard A, et al. Massive gliosis induced by interleukin-6 suppresses A{beta} deposition in vivo: evidence against inflammation as a driving force for amyloid deposition. FASEB J. 2009 .
    1. Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection. Lancet Neurol. 2009;8:382–97.
    1. Gao HM, Kotzbauer PT, Uryu K, et al. Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J Neurosci. 2008;28:7687–98.
    1. Zecca L, Wilms H, Geick S, et al. Human neuromelanin induces neuroinflammation and neurodegeneration in the rat substantia nigra: implications for Parkinson’s disease. Acta Neuropathol. 2008;116:47–55.
    1. Brochard V, Combadiere B, Prigent A, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119:182–92.
    1. Hunter RL, Dragicevic N, Seifert K, et al. Inflammation induces mitochondrial dysfunction and dopaminergic neurodegeneration in the nigrostriatal system. J Neurochem. 2007;100:1375–86.
    1. Kim YS, Joh TH. Microglia, major player in the brain inflammation: their roles in the pathogenesis of Parkinson’s disease. Exp Mol Med. 2006;38:333–47.
    1. Pieper HC, Evert BO, Kaut O, et al. Different methylation of the TNF-alpha promoter in cortex and substantia nigra: implications for selective neuronal vulnerability. Neurobiol Dis. 2008;32:521–7.
    1. Pavese N, Gerhard A, Tai YF, et al. Microglial activation correlates with severity in Huntington disease: a clinical and PET study. Neurology. 2006;66:1638–43.
    1. Tai YF, Pavese N, Gerhard A, et al. Microglial activation in presymptomatic Huntington’s disease gene carriers. Brain. 2007;130:1759–66.
    1. Mantovani S, Garbelli S, Pasini A, et al. Immune system alterations in sporadic amyotrophic lateral sclerosis patients suggest an ongoing neuroinflammatory process. J Neuroimmunol. 2009;210:73–9.
    1. Schwab C, Klegeris A, McGeer PL. Inflammation in transgenic mouse models of neurodegenerative disorders. Biochim Biophys Acta. 2009 Doi: .
    1. Fendrick SE, Xue QS, Streit WJ. Formation of multinucleated giant cells and microglial degeneration in rats expressing a mutant Cu/Zn superoxide dismutase gene. J Neuroinflammation. 2007;4:9.
    1. Simmons DA, Casale M, Alcon B, et al. Ferritin accumulation in dystrophic microglia is an early event in the development of Huntington’s disease. Glia. 2007;55:1074–84.
    1. Rogers J, Mastroeni D, Leonard B, et al. Neuroinflammation in Alzheimer’s disease and Parkinson’s disease: are microglia pathogenic in either disorder. Int Rev Neurobiol. 2007;82:235–46.
    1. Kroemer G, Galluzzi L, Vandenabeele P, et al. Classification of cell death: recommendations of the nomenclature committee on cell death 2009. Cell Death Differ. 2009;16:3–11.
    1. Bredesen DE. Programmed cell death mechanisms in neurological disease. Curr Mol Med. 2008;8:173–86.
    1. Gorman AM. Neuronal cell death in neurodegenerative diseases: recurring themes around protein handling. J Cell Mol Med. 2008;12:2263–80.
    1. Higgins GC, Beart PM, Nagley P. Oxidative stress triggers neuronal caspase-independent death: endonuclease G involvement in programmed cell death-type III. Cell Mol Life Sci. 2009;66:2773–87.
    1. Henriquez M, Armisen R, Stutzin A, et al. Cell death by necrosis, a regulated way to go. Curr Mol Med. 2008;8:187–206.
    1. Bulat N, Widmann C. Caspase substrates and neurodegenerative diseases. Brain Res Bull. 2009;80:251–67.
    1. Kroemer G, Levine B. Autophagic cell death: the story of a misnomer. Nat Rev Mol Cell Biol. 2008;9:1004–10.
    1. Winslow AR, Rubinsztein DC. Autophagy in neurodegeneration and development. Biochim Biophys Acta. 2008;1782:723–9.
    1. Klionsky DJ, Abeliovich H, Agostinis P, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 2008;4:151–75.
    1. Uchiyama Y, Koike M, Shibata M, et al. Autophagic neuron death. Methods Enzymol. 2009;453:33–51.
    1. Franco R, Sanchez-Olea R, Reyes-Reyes EM, et al. Environmental toxicity, oxidative stress and apoptosis: menage a Trois. Mutat Res. 2009;674:3–22.
    1. Ribe EM, Serrano-Saiz E, Akpan N, et al. Mechanisms of neuronal death in disease: defining the models and the players. Biochem J. 2008;415:165–82.
    1. Woodhouse A, Dickson TC, West AK, et al. No difference in expression of apoptosis-related proteins and apoptotic morphology in control, pathologically aged and Alzheimer’s disease cases. Neurobiol Dis. 2006;22:323–33.
    1. Louneva N, Cohen JW, Han LY, et al. Caspase-3 is enriched in postsynaptic densities and increased in Alzheimer’s disease. Am J Pathol. 2008;173:1488–95.
    1. Miyoshi K, Ohyagi Y, Sakae N, et al. Enhancement of activation of caspases by presenilin 1 gene mutations and its inhibition by secretase inhibitors. J Alzheimers Dis. 2009;16:551–64.
    1. Raina AK, Sayre LM, Atwood CS. Apoptotic and oxidative indicators in Alzheimer disease. In: LeBlanc AC, editor. Neuromethods, Vol. 37: apoptosis: tech niques and protocols. 2nd ed. Totowa, NJ: Humana Press; 2002. pp. 225–46.
    1. Schwab C, Arai T, Hasegawa M, et al. TDP-43 pathology in familial British dementia. Acta Neuropathol. 2009;118:303–11.
    1. SantaCruz K, Lewis J, Spires T, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309:476–81.
    1. Katsuse O, Lin WL, Lewis J, et al. Neurofibrillary tangle-related synaptic alterations of spinal motor neurons of P301L tau transgenic mice. Neurosci Lett. 2006;409:95–9.
    1. Cotman CW, Poon WW, Rissman RA, et al. The role of caspase cleavage of tau in Alzheimer disease neuropathology. J Neuropathol Exp Neurol. 2005;64:104–12.
    1. Rohn TT, Head E. Caspases as therapeutic targets in Alzheimer’s disease: is it time to “cut” to the chase. Int J Clin Exp Pathol. 2009;2:108–18.
    1. Schindowski K, Belarbi K, Bretteville A, et al. Neurogenesis and cell cycle-reactivated neuronal death during pathogenic tau aggregation. Genes Brain Behav. 2008;7:92–100.
    1. Yang DS, Kumar A, Stavrides P, et al. Neuronal apoptosis and autophagy cross talk in aging PS/APP mice, a model of Alzheimer’s disease. Am J Pathol. 2008;173:665–81.
    1. Cookson MR. alpha-Synuclein and neuronal cell death. Mol Neurodegener. 2009;4:9.
    1. Shibata N, Kakita A, Takahashi H, et al. Persistent cleavage and nuclear translocation of apoptosis-inducing factor in motor neurons in the spinal cord of sporadic amyotrophic lateral sclerosis patients. Acta Neuropathol. 2009;118:755–62.
    1. Riederer IM, Schiffrin M, Kovari E, et al. Ubiquitination and cysteine nitrosylation during aging and Alzheimer’s disease. Brain Res Bull. 2009;80:233–41.
    1. Rossi D, Volterra A. Astrocytic dysfunction: insights on the role in neurodegeneration. Brain Res Bull. 2009;80:224–32.
    1. Chadi G, Maximino JR, De Oliveira GP. The importance of molecular histology to study glial influence on neurodegenerative disorders. Focus on recent developed single cell laser microdissection. J Mol Histol. 2009;40:241–50.
    1. Lobsiger CS, Cleveland DW. Glial cells as intrinsic components of non-cell-autonomous neurodegenerative disease. Nat Neurosci. 2007;10:1355–60.
    1. Song YJ, Halliday GM, Holton JL, et al. Degeneration in different parkinsonian syndromes relates to astrocyte type and astrocyte protein expression. J Neuropathol Exp Neurol. 2009;68:1073–83.
    1. Wenning GK, Stefanova N, Jellinger KA, et al. Multiple system atrophy: a primary oligodendrogliopathy. Ann Neurol. 2008;64:239–46.
    1. Uyama N, Uchihara T, Mochizuki Y, et al. Selective nuclear shrinkage of oligodendrocytes lacking glial cytoplasmic inclusions in multiple system atrophy: a 3-dimensional volumetric study. J Neuropathol Exp Neurol. 2009;68:1084–91.
    1. Simpson CL, Lemmens R, Miskiewicz K, et al. Variants of the elongator protein 3 (ELP3) gene are associated with motor neuron degeneration. Hum Mol Genet. 2009;18:472–81.
    1. McGeer PL, McGeer EG. Glial reactions in Parkinson’s disease. Mov Disord. 2008;23:474–83.
    1. Nagai M, Re DB, Nagata T, et al. Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat Neurosci. 2007;10:615–22.
    1. Maragakis NJ, Rothstein JD. Mechanisms of disease: astrocytes in neurodegenerative disease. Nat Clin Pract Neurol. 2006;2:679–89.
    1. Parpura V, Haydon PG. Astrocytes in pathophysiology of the nervous system. New York: Springer; 2009.
    1. Zurich MG, Monnet-Tschudi F. Contribution of in vitro neurotoxicology studies to the elucidation of neurodegenerative processes. Brain Res Bull. 2009;80:211–6.
    1. Choonara YE, Pillay V, Du Toit LC, et al. Trends in the molecular pathogenesis and clinical therapeutics of common neurodegenerative disorders. Int J Mol Sci. 2009;10:2510–57.
    1. Kelly BL, Vassar R, Ferreira A. Beta-amyloid-induced dynamin 1 depletion in hippocampal neurons. A potential mechanism for early cognitive decline in Alzheimer disease. J Biol Chem. 2005;280:31746–53.
    1. Koffie RM, Meyer-Luehmann M, Hashimoto T, et al. Oligomeric amyloid beta associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc Natl Acad Sci U S A. 2009;106:4012–7.
    1. Lacor PN, Buniel MC, Chang L, et al. Synaptic targeting by Alzheimer’s-related amyloid beta oligomers. J Neurosci. 2004;24:10191–200.
    1. Meyer-Luehmann M, Coomaraswamy J, Bolmont T, et al. Exogenous induction of cerebral beta-amyloidogenesis is governed by agent and host. Science. 2006;313:1781–4.
    1. Hu X, Crick SL, Bu G, et al. Amyloid seeds formed by cellular uptake, concentration, and aggregation of the amyloid-beta peptide. Proc Natl Acad Sci U S A. 2009;106:20324–9.
    1. Xie YY, Zhou CJ, Zhou ZR, et al. Interaction with synphilin-1 promotes inclusion formation of alpha-synuclein: mechanistic insights and pathological implication. FASEB J. 2010;24:196–205.
    1. Desplats P, Lee HJ, Bae EJ, et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci U S A. 2009;106:13010–5.
    1. Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem. 2009;284:12845–52.
    1. Ren PH, Lauckner JE, Kachirskaia I, et al. Cytoplasmic penetration and persistent infection of mammalian cells by polyglutamine aggregates. Nat Cell Biol. 2009;11:219–25.
    1. Naiki H, Nagai Y. Molecular pathogenesis of protein misfolding diseases: pathological molecular environments versus quality control systems against misfolded proteins. J Biochem. 2009;146:751–6.
    1. Bandopadhyay R, De Belleroche J. Pathogenesis of Parkinson’s disease: emerging role of molecular chaperones. Trends Mol Med. 2010;16:27–36.
    1. Wertz IE, Dixit VM. Regulation of death receptor signaling by the ubiquitin system. Cell Death Differ. 2010;17:14–24.
    1. Tsvetkov P, Reuven N, Shaul Y. Ubiquitin-independent p53 proteasomal degradation. Cell Death Differ. 2010;17:103–8.
    1. Bader M, Steller H. Regulation of cell death by the ubiquitin-proteasome system. Curr Opin Cell Biol. 2009;21:878–84.
    1. Michiorri S, Gelmetti V, Giarda E, et al. The Parkinson-associated protein PINK1 interacts with Beclin1 and promotes autophagy. Cell Death Differ. 2010 Doi: .
    1. Rosen KM, Moussa CE, Lee HK, et al. Parkin reverses intracellular beta-amyloid accumulation and its negative effects on proteasome function. J Neurosci Res. 2010;88:167–78.
    1. Smith MA, Zhu X, Tabaton M, et al. Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment. J Alzheimers Dis. 2010;19:363–72.
    1. Cozzi A, Rovelli E, Frizzale G, et al. Oxidative stress and cell death in cells expressing L-ferritin variants causing neuroferritinopathy. Neurobiol Dis. 2010;37:77–85.
    1. Zhao W, Beers DR, Henkel JS, et al. Extracellular mutant SOD1 induces microglial-mediated motoneuron injury. Glia. 2010;58:231–43.
    1. Boumezbeur F, Mason GF, De Graaf RA, et al. Altered brain mitochondrial metabolism in healthy aging as assessed by in vivo magnetic resonance spectroscopy. J Cereb Blood Flow Metab. 2010;30:211–21.
    1. Martinelli P, Rugarli EI. Emerging roles of mitochondrial proteases in neurodegeneration. Biochim Biophys Acta. 2010;1797:1–10.
    1. Gibson GE, Starkov A, Blass JP, et al. Cause and consequence: mitochondrial dysfunction initiates and propagates neuronal dysfunction, neuronal death and behavioral abnormalities in age-associated neurodegenerative diseases. Biochim Biophys Acta. 2010;1802:122–34.
    1. Du H, Yan SS. Mitochondrial permeability transition pore in Alzheimer’s disease: cyclophilin D and amyloid beta. Biochim Biophys Acta. 2010;1802:198–204.
    1. Moreira PI, Carvalho C, Zhu X, et al. Mitochondrial dysfunction is a trigger of Alzheimer’s disease pathophysiology. Biochim Biophys Acta. 2010;1802:2–10.
    1. Lynn BC, Wang J, Markesbery WR, Lovell MA. Quantitative changes in the mitochondrial proteome from subjects with mild cognitive impairment, early stage, and late stage Alzheimer’s disease. J Alzheimers Dis. 2010;19:325–39.
    1. Wang X, Su B, Perry G, et al. Insights into amyloid-beta-induced mitochondrial dysfunction in Alzheimer disease. Free Radic Biol Med. 2007;43:1569–73.
    1. Yu JT, Chang RC, Tan L. Calcium dysregulation in Alzheimer’s disease: from mechanisms to therapeutic opportunities. Prog Neurobiol. 2009;89:240–55.
    1. Green KN. Calcium in the initiation, progression and as an effector of Alzheimer’s disease pathology. J Cell Mol Med. 2009;13:2787–99.
    1. Winklhofer KF, Haass C. Mitochondrial dysfunction in Parkinson’s disease. Biochim Biophys Acta. 2010;1802:29–44.
    1. Estevez AR, Arduino DM, Swerdlow RH, et al. Microtubule depolymerization potentiates alpha-synuclein oligomerization. Front Ag Neurosci. 2010:1. : doi: .
    1. Gandhi S, Wood-Kaczmar A, Yao Z, et al. PINK1-associated Parkinson’s disease is caused by neuronal vulnerability to calcium-induced cell death. Mol Cell. 2009;33:627–38.
    1. Gegg ME, Cooper JM, Schapira AH, et al. Silencing of PINK1 expression affects mitochondrial DNA and oxidative phosphorylation in dopaminergic cells. PLoS One. 2009;4:e4756.
    1. Damiano M, Galvan L, Deglon N, et al. Mitochondria in Huntington’s disease. Biochim Biophys Acta. 2010;1802:52–61.
    1. Shi P, Gal J, Kwinter DM, et al. Mitochondrial dysfunction in amyotrophic lateral sclerosis. Biochim Biophys Acta. 2010;1802:45–51.
    1. Martin LJ. The mitochondrial permeability transition pore: a molecular target for amyotrophic lateral sclerosis therapy. Biochim Biophys Acta. 2010;1802:186–97.
    1. Kichev A, Ilieva EV, Pinol-Ripoll G, et al. Cell death and learning impairment in mice caused by in vitro modified pro-NGF can be related to its increased oxidative modifications in Alzheimer disease. Am J Pathol. 2009;175:2574–85.
    1. Lynch MA. Age-related neuroinflammatory changes negatively impact on neuronal function. Front Ag Neurosci. 2010;1 : Doi: .
    1. Ryu JK, McLarnon JG. A leaky blood-brain barrier, fibrinogen infiltration and microglial reactivity in inflamed Alzheimer’s disease brain. J Cell Mol Med. 2009;13:2911–25.
    1. Davenport CM, Sevastou IG, Hooper C, et al. Inhibiting p53 pathways in microglia attenuates microglial-evoked neurotoxicity following exposure to Alzheimer peptides. J Neurochem. 2009;112:552–63.
    1. Long-Smith CM, Sullivan AM, Nolan YM. The influence of microglia on the pathogenesis of Parkinson’s disease. Prog Neurobiol. 2009;89:277–87.

Source: PubMed

Подписаться