메뉴 건너뛰기




Volumn 13, Issue 10, 2014, Pages 1045-1060

The role of iron in brain ageing and neurodegenerative disorders

Author keywords

[No Author keywords available]

Indexed keywords

5,7 DICHLORO 2 [(DIMETHYLAMINO)METHYL] 8 HYDROXYQUINOLINE; CARBOHYDRATE; CERULOPLASMIN; DEFERASIROX; DEFERIPRONE; DEFEROXAMINE; DNA; FERRITIN; FERROPORTIN; FERROUS ION; HEPCIDIN; HEPHAESTIN; IRON; IRON CHELATING AGENT; IRON REGULATORY FACTOR; LIPID; NATURAL RESISTANCE ASSOCIATED MACROPHAGE PROTEIN 2; NEUROMELANIN; PROTEIN; QUINOLINE DERIVATIVE; TRANSFERRIN; TRANSFERRIN RECEPTOR; UNCLASSIFIED DRUG; BIOLOGICAL MARKER; NEUROPROTECTIVE AGENT; NONHEME IRON PROTEIN;

EID: 84907999177     PISSN: 14744422     EISSN: 14744465     Source Type: Journal    
DOI: 10.1016/S1474-4422(14)70117-6     Document Type: Review
Times cited : (1300)

References (224)
  • 3
    • 79952162002 scopus 로고    scopus 로고
    • Regulation of cellular iron metabolism
    • Wang J, Pantopoulos K Regulation of cellular iron metabolism. Biochem J 2011, 434:365-381.
    • (2011) Biochem J , vol.434 , pp. 365-381
    • Wang, J.1    Pantopoulos, K.2
  • 4
    • 0344154421 scopus 로고    scopus 로고
    • Labile iron pool: the main determinant of cellular response to oxidative stress
    • Kruszewski M Labile iron pool: the main determinant of cellular response to oxidative stress. Mutat Res 2003, 531:81-92.
    • (2003) Mutat Res , vol.531 , pp. 81-92
    • Kruszewski, M.1
  • 5
    • 0035103932 scopus 로고    scopus 로고
    • Iron, Neuromelanin and ferritin content in the substantia nigra of normal subjects at different ages: consequences for iron storage and neurodegenerative processes
    • Zecca L, Gallorini M, Schünemann V, et al. Iron, Neuromelanin and ferritin content in the substantia nigra of normal subjects at different ages: consequences for iron storage and neurodegenerative processes. J Neurochem 2001, 76:1766-1773.
    • (2001) J Neurochem , vol.76 , pp. 1766-1773
    • Zecca, L.1    Gallorini, M.2    Schünemann, V.3
  • 6
    • 3042793602 scopus 로고    scopus 로고
    • The role of iron and copper molecules in the neuronal vulnerability of locus coeruleus and substantia nigra during aging
    • Zecca L, Stroppolo A, Gatti A, et al. The role of iron and copper molecules in the neuronal vulnerability of locus coeruleus and substantia nigra during aging. Proc Natl Acad Sci USA 2004, 101:9843-9848.
    • (2004) Proc Natl Acad Sci USA , vol.101 , pp. 9843-9848
    • Zecca, L.1    Stroppolo, A.2    Gatti, A.3
  • 8
    • 77952672976 scopus 로고    scopus 로고
    • Mechanisms of brain iron transport: insight into neurodegeneration and CNS disorders
    • Mills E, Dong XP, Wang F, Xu H Mechanisms of brain iron transport: insight into neurodegeneration and CNS disorders. Future Med Chem 2010, 2:51-64.
    • (2010) Future Med Chem , vol.2 , pp. 51-64
    • Mills, E.1    Dong, X.P.2    Wang, F.3    Xu, H.4
  • 9
    • 47649126246 scopus 로고    scopus 로고
    • Forging a field: the golden age of iron biology
    • Andrews NC Forging a field: the golden age of iron biology. Blood 2008, 112:219-230.
    • (2008) Blood , vol.112 , pp. 219-230
    • Andrews, N.C.1
  • 11
    • 50949102412 scopus 로고    scopus 로고
    • Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network
    • Muckenthaler MU, Galy B, Hentze MW Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu Rev Nutr 2008, 28:197-213.
    • (2008) Annu Rev Nutr , vol.28 , pp. 197-213
    • Muckenthaler, M.U.1    Galy, B.2    Hentze, M.W.3
  • 12
    • 33746864096 scopus 로고    scopus 로고
    • Molecular control of vertebrate iron homeostasis by iron regulatory proteins
    • Wallander ML, Leibold EA, Eisenstein RS Molecular control of vertebrate iron homeostasis by iron regulatory proteins. Biochim Biophys Acta 2006, 1763:668-689.
    • (2006) Biochim Biophys Acta , vol.1763 , pp. 668-689
    • Wallander, M.L.1    Leibold, E.A.2    Eisenstein, R.S.3
  • 13
    • 0033961913 scopus 로고    scopus 로고
    • Transferrin and transferrin receptor function in brain barrier systems
    • Moos T, Morgan EH Transferrin and transferrin receptor function in brain barrier systems. Cell Mol Neurobiol 2000, 20:77-95.
    • (2000) Cell Mol Neurobiol , vol.20 , pp. 77-95
    • Moos, T.1    Morgan, E.H.2
  • 15
    • 34748899552 scopus 로고    scopus 로고
    • Brain iron metabolism: neurobiology and neurochemistry
    • Ke Y, Qian ZM Brain iron metabolism: neurobiology and neurochemistry. Prog Neurobiol 2007, 83:149-173.
    • (2007) Prog Neurobiol , vol.83 , pp. 149-173
    • Ke, Y.1    Qian, Z.M.2
  • 16
    • 84857373169 scopus 로고    scopus 로고
    • Functional roles of transferrin in the brain
    • Leitner DF, Connor JR Functional roles of transferrin in the brain. Biochim Biophys Acta 2012, 1820:393-402.
    • (2012) Biochim Biophys Acta , vol.1820 , pp. 393-402
    • Leitner, D.F.1    Connor, J.R.2
  • 17
    • 0038711587 scopus 로고    scopus 로고
    • Glycosylphosphatidylinositol-anchored ceruloplasmin is required for iron efflux from cells in the central nervous system
    • Jeong SY, David S Glycosylphosphatidylinositol-anchored ceruloplasmin is required for iron efflux from cells in the central nervous system. J Biol Chem 2003, 278:27144-27148.
    • (2003) J Biol Chem , vol.278 , pp. 27144-27148
    • Jeong, S.Y.1    David, S.2
  • 19
    • 33645460545 scopus 로고    scopus 로고
    • Upregulation of iron regulatory proteins and divalent metal transporter-1 isoforms in the rat hippocampus after kainate induced neuronal injury
    • Huang E, Ong WY, Go ML, Connor JR Upregulation of iron regulatory proteins and divalent metal transporter-1 isoforms in the rat hippocampus after kainate induced neuronal injury. Exp Brain Res 2006, 170:376-386.
    • (2006) Exp Brain Res , vol.170 , pp. 376-386
    • Huang, E.1    Ong, W.Y.2    Go, M.L.3    Connor, J.R.4
  • 22
    • 79959528020 scopus 로고    scopus 로고
    • Heterogenous distribution of ferroportin-containing neurons in mouse brain
    • Boserup MW, Lichota J, Haile D, Moos T Heterogenous distribution of ferroportin-containing neurons in mouse brain. Biometals 2011, 24:357-375.
    • (2011) Biometals , vol.24 , pp. 357-375
    • Boserup, M.W.1    Lichota, J.2    Haile, D.3    Moos, T.4
  • 23
    • 79951858362 scopus 로고    scopus 로고
    • Developmental iron uptake and axonal transport in the retina of the rat
    • Moos T, Bernth N, Courtois Y, Morgan EH Developmental iron uptake and axonal transport in the retina of the rat. Mol Cell Neurosci 2011, 46:607-613.
    • (2011) Mol Cell Neurosci , vol.46 , pp. 607-613
    • Moos, T.1    Bernth, N.2    Courtois, Y.3    Morgan, E.H.4
  • 24
    • 23044503950 scopus 로고    scopus 로고
    • Microcytic anemia, erythropoietic protoporphyria, and neurodegeneration in mice with targeted deletion of iron-regulatory protein 2
    • Cooperman SS, Meyron-Holtz EG, Olivierre-Wilson H, Ghosh MC, McConnell JP, Rouault TA Microcytic anemia, erythropoietic protoporphyria, and neurodegeneration in mice with targeted deletion of iron-regulatory protein 2. Blood 2005, 106:1084-1091.
    • (2005) Blood , vol.106 , pp. 1084-1091
    • Cooperman, S.S.1    Meyron-Holtz, E.G.2    Olivierre-Wilson, H.3    Ghosh, M.C.4    McConnell, J.P.5    Rouault, T.A.6
  • 25
    • 29244463365 scopus 로고    scopus 로고
    • Astrocyte-endothelial interactions at the blood-brain barrier
    • Abbott NJ, Rönnbäck L, Hansson E Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci 2006, 7:41-53.
    • (2006) Nat Rev Neurosci , vol.7 , pp. 41-53
    • Abbott, N.J.1    Rönnbäck, L.2    Hansson, E.3
  • 26
    • 0028326826 scopus 로고
    • Studies of the ultrastructure and permeability of the blood-brain barrier in the developing corpus callosum in postnatal rat brain using electron dense tracers
    • Xu J, Ling EA Studies of the ultrastructure and permeability of the blood-brain barrier in the developing corpus callosum in postnatal rat brain using electron dense tracers. J Anat 1994, 184:227-237.
    • (1994) J Anat , vol.184 , pp. 227-237
    • Xu, J.1    Ling, E.A.2
  • 27
    • 10744219518 scopus 로고    scopus 로고
    • Expression of the iron transporter ferroportin in synaptic vesicles and the blood-brain barrier
    • Wu LJ, Leenders AG, Cooperman S, et al. Expression of the iron transporter ferroportin in synaptic vesicles and the blood-brain barrier. Brain Res 2004, 1001:108-117.
    • (2004) Brain Res , vol.1001 , pp. 108-117
    • Wu, L.J.1    Leenders, A.G.2    Cooperman, S.3
  • 28
    • 0025648166 scopus 로고
    • Cellular distribution of transferrin, ferritin, and iron in normal and aged human brains
    • Connor JR, Menzies SL, St Martin SM, Mufson EJ Cellular distribution of transferrin, ferritin, and iron in normal and aged human brains. J Neurosci Res 1990, 27:595-611.
    • (1990) J Neurosci Res , vol.27 , pp. 595-611
    • Connor, J.R.1    Menzies, S.L.2    St Martin, S.M.3    Mufson, E.J.4
  • 30
    • 0030174825 scopus 로고    scopus 로고
    • Relationship of iron to oligodendrocytes and myelination
    • Connor JR, Menzies SL Relationship of iron to oligodendrocytes and myelination. Glia 1996, 17:83-93.
    • (1996) Glia , vol.17 , pp. 83-93
    • Connor, J.R.1    Menzies, S.L.2
  • 32
    • 84858078783 scopus 로고    scopus 로고
    • Iron homeostasis in astrocytes and microglia is differentially regulated by TNF-α and TGF-β1
    • Rathore KI, Redensek A, David S Iron homeostasis in astrocytes and microglia is differentially regulated by TNF-α and TGF-β1. Glia 2012, 60:738-750.
    • (2012) Glia , vol.60 , pp. 738-750
    • Rathore, K.I.1    Redensek, A.2    David, S.3
  • 33
    • 0029433318 scopus 로고
    • Cellular management of iron in the brain
    • Connor JR, Menzies SL Cellular management of iron in the brain. J Neurol Sci 1995, 134(suppl):33-44.
    • (1995) J Neurol Sci , vol.134 , pp. 33-44
    • Connor, J.R.1    Menzies, S.L.2
  • 34
    • 3042636759 scopus 로고    scopus 로고
    • The IL-6- and lipopolysaccharide-induced transcription of hepcidin in HFE-, transferrin receptor 2-, and beta 2-microglobulin-deficient hepatocytes
    • Lee P, Peng H, Gelbart T, Beutler E The IL-6- and lipopolysaccharide-induced transcription of hepcidin in HFE-, transferrin receptor 2-, and beta 2-microglobulin-deficient hepatocytes. Proc Natl Acad Sci USA 2004, 101:9263-9265.
    • (2004) Proc Natl Acad Sci USA , vol.101 , pp. 9263-9265
    • Lee, P.1    Peng, H.2    Gelbart, T.3    Beutler, E.4
  • 35
    • 2342510407 scopus 로고    scopus 로고
    • IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin
    • Nemeth E, Rivera S, Gabayan V, et al. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest 2004, 113:1271-1276.
    • (2004) J Clin Invest , vol.113 , pp. 1271-1276
    • Nemeth, E.1    Rivera, S.2    Gabayan, V.3
  • 36
    • 84885953717 scopus 로고    scopus 로고
    • Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous system cells
    • Urrutia P, Aguirre P, Esparza A, et al. Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous system cells. J Neurochem 2013, 126:541-549.
    • (2013) J Neurochem , vol.126 , pp. 541-549
    • Urrutia, P.1    Aguirre, P.2    Esparza, A.3
  • 38
    • 58549098084 scopus 로고    scopus 로고
    • Blood-brain barrier: ageing and microvascular disease--systematic review and meta-analysis
    • Farrall AJ, Wardlaw JM Blood-brain barrier: ageing and microvascular disease--systematic review and meta-analysis. Neurobiol Aging 2009, 30:337-352.
    • (2009) Neurobiol Aging , vol.30 , pp. 337-352
    • Farrall, A.J.1    Wardlaw, J.M.2
  • 40
    • 83855163999 scopus 로고    scopus 로고
    • Pathogenic implications of iron accumulation in multiple sclerosis
    • Williams R, Buchheit CL, Berman NE, LeVine SM Pathogenic implications of iron accumulation in multiple sclerosis. J Neurochem 2012, 120:7-25.
    • (2012) J Neurochem , vol.120 , pp. 7-25
    • Williams, R.1    Buchheit, C.L.2    Berman, N.E.3    LeVine, S.M.4
  • 42
    • 84961042345 scopus 로고
    • The effect of age on the non-haemin iron in the human brain
    • Hallgren B, Sourander P The effect of age on the non-haemin iron in the human brain. J Neurochem 1958, 3:41-51.
    • (1958) J Neurochem , vol.3 , pp. 41-51
    • Hallgren, B.1    Sourander, P.2
  • 43
    • 84890862804 scopus 로고    scopus 로고
    • Iron levels in the human brain: a post-mortem study of anatomical region differences and age-related changes
    • Ramos P, Santos A, Pinto NR, Mendes R, Magalhães T, Almeida A Iron levels in the human brain: a post-mortem study of anatomical region differences and age-related changes. J Trace Elem Med Biol 2014, 28:13-17.
    • (2014) J Trace Elem Med Biol , vol.28 , pp. 13-17
    • Ramos, P.1    Santos, A.2    Pinto, N.R.3    Mendes, R.4    Magalhães, T.5    Almeida, A.6
  • 45
    • 0030012587 scopus 로고    scopus 로고
    • Interaction of Neuromelanin and iron in substantia nigra and other areas of human brain
    • Zecca L, Shima T, Stroppolo A, et al. Interaction of Neuromelanin and iron in substantia nigra and other areas of human brain. Neuroscience 1996, 73:407-415.
    • (1996) Neuroscience , vol.73 , pp. 407-415
    • Zecca, L.1    Shima, T.2    Stroppolo, A.3
  • 46
    • 0035073517 scopus 로고    scopus 로고
    • The determination of iron and other metals by INAA in cortex, cerebellum and putamen of human brain and in their neuromelanins
    • Zecca L, Tampellini D, Rizzio E, Giaveri G, Gallorini M The determination of iron and other metals by INAA in cortex, cerebellum and putamen of human brain and in their neuromelanins. J Radioanal Nucl Chem 2001, 248:129-131.
    • (2001) J Radioanal Nucl Chem , vol.248 , pp. 129-131
    • Zecca, L.1    Tampellini, D.2    Rizzio, E.3    Giaveri, G.4    Gallorini, M.5
  • 47
    • 84855418321 scopus 로고    scopus 로고
    • Aluminium, iron and copper in human brain tissues donated to the Medical Research Council's Cognitive Function and Ageing Study
    • House E, Esiri M, Forster G, Ince PG, Exley C Aluminium, iron and copper in human brain tissues donated to the Medical Research Council's Cognitive Function and Ageing Study. Metallomics 2012, 4:56-65.
    • (2012) Metallomics , vol.4 , pp. 56-65
    • House, E.1    Esiri, M.2    Forster, G.3    Ince, P.G.4    Exley, C.5
  • 48
    • 33846568415 scopus 로고    scopus 로고
    • Brain ferritin iron may influence age- and gender-related risks of neurodegeneration
    • Bartzokis G, Tishler TA, Lu PH, et al. Brain ferritin iron may influence age- and gender-related risks of neurodegeneration. Neurobiol Aging 2007, 28:414-423.
    • (2007) Neurobiol Aging , vol.28 , pp. 414-423
    • Bartzokis, G.1    Tishler, T.A.2    Lu, P.H.3
  • 49
    • 67650091951 scopus 로고    scopus 로고
    • Age-related iron deposition in the basal ganglia: quantitative analysis in healthy subjects
    • Aquino D, Bizzi A, Grisoli M, et al. Age-related iron deposition in the basal ganglia: quantitative analysis in healthy subjects. Radiology 2009, 252:165-172.
    • (2009) Radiology , vol.252 , pp. 165-172
    • Aquino, D.1    Bizzi, A.2    Grisoli, M.3
  • 50
    • 79956032674 scopus 로고    scopus 로고
    • Gender and iron genes may modify associations between brain iron and memory in healthy aging
    • Bartzokis G, Lu PH, Tingus K, et al. Gender and iron genes may modify associations between brain iron and memory in healthy aging. Neuropsychopharmacology 2011, 36:1375-1384.
    • (2011) Neuropsychopharmacology , vol.36 , pp. 1375-1384
    • Bartzokis, G.1    Lu, P.H.2    Tingus, K.3
  • 51
    • 84855461629 scopus 로고    scopus 로고
    • MRI estimates of brain iron concentration in normal aging using quantitative susceptibility mapping
    • Bilgic B, Pfefferbaum A, Rohlfing T, Sullivan EV, Adalsteinsson E MRI estimates of brain iron concentration in normal aging using quantitative susceptibility mapping. Neuroimage 2012, 59:2625-2635.
    • (2012) Neuroimage , vol.59 , pp. 2625-2635
    • Bilgic, B.1    Pfefferbaum, A.2    Rohlfing, T.3    Sullivan, E.V.4    Adalsteinsson, E.5
  • 52
    • 33744931389 scopus 로고    scopus 로고
    • Neuromelanin and iron in human locus coeruleus and substantia nigra during aging: consequences for neuronal vulnerability
    • Zucca FA, Bellei C, Giannelli S, et al. Neuromelanin and iron in human locus coeruleus and substantia nigra during aging: consequences for neuronal vulnerability. J Neural Transm 2006, 113:757-767.
    • (2006) J Neural Transm , vol.113 , pp. 757-767
    • Zucca, F.A.1    Bellei, C.2    Giannelli, S.3
  • 53
    • 56249140814 scopus 로고    scopus 로고
    • New melanic pigments in the human brain that accumulate in aging and block environmental toxic metals
    • Zecca L, Bellei C, Costi P, et al. New melanic pigments in the human brain that accumulate in aging and block environmental toxic metals. Proc Natl Acad Sci USA 2008, 105:17567-17572.
    • (2008) Proc Natl Acad Sci USA , vol.105 , pp. 17567-17572
    • Zecca, L.1    Bellei, C.2    Costi, P.3
  • 54
    • 33845768784 scopus 로고    scopus 로고
    • Microglia-mediated neurotoxicity: uncovering the molecular mechanisms
    • Block ML, Zecca L, Hong JS Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci 2007, 8:57-69.
    • (2007) Nat Rev Neurosci , vol.8 , pp. 57-69
    • Block, M.L.1    Zecca, L.2    Hong, J.S.3
  • 55
    • 51349095669 scopus 로고    scopus 로고
    • Microglial dystrophy in the aged and Alzheimer's disease brain is associated with ferritin immunoreactivity
    • Lopes KO, Sparks DL, Streit WJ Microglial dystrophy in the aged and Alzheimer's disease brain is associated with ferritin immunoreactivity. Glia 2008, 56:1048-1060.
    • (2008) Glia , vol.56 , pp. 1048-1060
    • Lopes, K.O.1    Sparks, D.L.2    Streit, W.J.3
  • 56
  • 58
  • 59
    • 78149433116 scopus 로고    scopus 로고
    • Role of epigenetics in Alzheimer's and Parkinson's disease
    • Kwok JB Role of epigenetics in Alzheimer's and Parkinson's disease. Epigenomics 2010, 2:671-682.
    • (2010) Epigenomics , vol.2 , pp. 671-682
    • Kwok, J.B.1
  • 60
    • 57249095767 scopus 로고    scopus 로고
    • Lipid peroxidation of membrane phospholipids generates hydroxy-alkenals and oxidized phospholipids active in physiological and/or pathological conditions
    • Catalá A Lipid peroxidation of membrane phospholipids generates hydroxy-alkenals and oxidized phospholipids active in physiological and/or pathological conditions. Chem Phys Lipids 2009, 157:1-11.
    • (2009) Chem Phys Lipids , vol.157 , pp. 1-11
    • Catalá, A.1
  • 62
    • 84866848255 scopus 로고    scopus 로고
    • 4-Hydroxy-2-nonenal, a reactive product of lipid peroxidation, and neurodegenerative diseases: a toxic combination illuminated by redox proteomics studies
    • Perluigi M, Coccia R, Butterfield DA 4-Hydroxy-2-nonenal, a reactive product of lipid peroxidation, and neurodegenerative diseases: a toxic combination illuminated by redox proteomics studies. Antioxid Redox Signal 2012, 17:1590-1609.
    • (2012) Antioxid Redox Signal , vol.17 , pp. 1590-1609
    • Perluigi, M.1    Coccia, R.2    Butterfield, D.A.3
  • 63
    • 67349106731 scopus 로고    scopus 로고
    • A novel transferrin/TfR2-mediated mitochondrial iron transport system is disrupted in Parkinson's disease
    • Mastroberardino PG, Hoffman EK, Horowitz MP, et al. A novel transferrin/TfR2-mediated mitochondrial iron transport system is disrupted in Parkinson's disease. Neurobiol Dis 2009, 34:417-431.
    • (2009) Neurobiol Dis , vol.34 , pp. 417-431
    • Mastroberardino, P.G.1    Hoffman, E.K.2    Horowitz, M.P.3
  • 64
    • 77956218282 scopus 로고    scopus 로고
    • Mitochondrial iron metabolism and its role in neurodegeneration
    • Horowitz MP, Greenamyre JT Mitochondrial iron metabolism and its role in neurodegeneration. J Alzheimers Dis 2010, 20(suppl 2):S551-S568.
    • (2010) J Alzheimers Dis , vol.20 , pp. S551-S568
    • Horowitz, M.P.1    Greenamyre, J.T.2
  • 65
    • 0026015052 scopus 로고
    • Catechol oxidation by peroxidase-positive astrocytes in primary culture: an electron spin resonance study
    • Schipper HM, Kotake Y, Janzen EG Catechol oxidation by peroxidase-positive astrocytes in primary culture: an electron spin resonance study. J Neurosci 1991, 11:2170-2176.
    • (1991) J Neurosci , vol.11 , pp. 2170-2176
    • Schipper, H.M.1    Kotake, Y.2    Janzen, E.G.3
  • 66
    • 0001672745 scopus 로고    scopus 로고
    • Intraneuronal dopamine-quinone synthesis: a review
    • Sulzer D, Zecca L Intraneuronal dopamine-quinone synthesis: a review. Neurotox Res 2000, 1:181-195.
    • (2000) Neurotox Res , vol.1 , pp. 181-195
    • Sulzer, D.1    Zecca, L.2
  • 67
    • 15344342576 scopus 로고    scopus 로고
    • Dopamine-dependent iron toxicity in cells derived from rat hypothalamus
    • Paris I, Martinez-Alvarado P, Cárdenas S, et al. Dopamine-dependent iron toxicity in cells derived from rat hypothalamus. Chem Res Toxicol 2005, 18:415-419.
    • (2005) Chem Res Toxicol , vol.18 , pp. 415-419
    • Paris, I.1    Martinez-Alvarado, P.2    Cárdenas, S.3
  • 68
    • 0029384919 scopus 로고
    • Iron-mediated bioactivation of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in glial cultures
    • Di Monte DA, Schipper HM, Hetts S, Langston JW Iron-mediated bioactivation of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in glial cultures. Glia 1995, 15:203-206.
    • (1995) Glia , vol.15 , pp. 203-206
    • Di Monte, D.A.1    Schipper, H.M.2    Hetts, S.3    Langston, J.W.4
  • 69
    • 84879594849 scopus 로고    scopus 로고
    • Metallobiology of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity
    • Hare DJ, Adlard PA, Doble PA, Finkelstein DI Metallobiology of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity. Metallomics 2013, 5:91-109.
    • (2013) Metallomics , vol.5 , pp. 91-109
    • Hare, D.J.1    Adlard, P.A.2    Doble, P.A.3    Finkelstein, D.I.4
  • 70
    • 77955978437 scopus 로고    scopus 로고
    • Dose- and time-dependent alpha-synuclein aggregation induced by ferric iron in SK-N-SH cells
    • Li WJ, Jiang H, Song N, Xie JX Dose- and time-dependent alpha-synuclein aggregation induced by ferric iron in SK-N-SH cells. Neurosci Bull 2010, 26:205-210.
    • (2010) Neurosci Bull , vol.26 , pp. 205-210
    • Li, W.J.1    Jiang, H.2    Song, N.3    Xie, J.X.4
  • 71
    • 0036724204 scopus 로고    scopus 로고
    • Iron (III) induces aggregation of hyperphosphorylated tau and its reduction to iron (II) reverses the aggregation: implications in the formation of neurofibrillary tangles of Alzheimer's disease
    • Yamamoto A, Shin RW, Hasegawa K, et al. Iron (III) induces aggregation of hyperphosphorylated tau and its reduction to iron (II) reverses the aggregation: implications in the formation of neurofibrillary tangles of Alzheimer's disease. J Neurochem 2002, 82:1137-1147.
    • (2002) J Neurochem , vol.82 , pp. 1137-1147
    • Yamamoto, A.1    Shin, R.W.2    Hasegawa, K.3
  • 72
    • 84864913557 scopus 로고    scopus 로고
    • Iron and ER stress in neurodegenerative disease
    • Liu Y, Connor JR Iron and ER stress in neurodegenerative disease. Biometals 2012, 25:837-845.
    • (2012) Biometals , vol.25 , pp. 837-845
    • Liu, Y.1    Connor, J.R.2
  • 74
    • 84861541814 scopus 로고    scopus 로고
    • Ferroptosis: an iron-dependent form of nonapoptotic cell death
    • Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012, 149:1060-1072.
    • (2012) Cell , vol.149 , pp. 1060-1072
    • Dixon, S.J.1    Lemberg, K.M.2    Lamprecht, M.R.3
  • 75
    • 84655164280 scopus 로고    scopus 로고
    • The role of metallobiology and amyloid-β peptides in Alzheimer's disease
    • Roberts BR, Ryan TM, Bush AI, Masters CL, Duce JA The role of metallobiology and amyloid-β peptides in Alzheimer's disease. J Neurochem 2012, 120(suppl 1):149-166.
    • (2012) J Neurochem , vol.120 , pp. 149-166
    • Roberts, B.R.1    Ryan, T.M.2    Bush, A.I.3    Masters, C.L.4    Duce, J.A.5
  • 76
    • 0033964859 scopus 로고    scopus 로고
    • In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer's disease: a central role for bound transition metals
    • Sayre LM, Perry G, Harris PL, Liu Y, Schubert KA, Smith MA In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer's disease: a central role for bound transition metals. J Neurochem 2000, 74:270-279.
    • (2000) J Neurochem , vol.74 , pp. 270-279
    • Sayre, L.M.1    Perry, G.2    Harris, P.L.3    Liu, Y.4    Schubert, K.A.5    Smith, M.A.6
  • 77
    • 18744374615 scopus 로고    scopus 로고
    • Is oxidative damage the fundamental pathogenic mechanism of Alzheimer's and other neurodegenerative diseases?
    • Perry G, Nunomura A, Hirai K, et al. Is oxidative damage the fundamental pathogenic mechanism of Alzheimer's and other neurodegenerative diseases?. Free Radic Biol Med 2002, 33:1475-1479.
    • (2002) Free Radic Biol Med , vol.33 , pp. 1475-1479
    • Perry, G.1    Nunomura, A.2    Hirai, K.3
  • 78
    • 67649739079 scopus 로고    scopus 로고
    • Iron toxicity in diseases of aging: Alzheimer's disease, Parkinson's disease and atherosclerosis
    • Altamura S, Muckenthaler MU Iron toxicity in diseases of aging: Alzheimer's disease, Parkinson's disease and atherosclerosis. J Alzheimers Dis 2009, 16:879-895.
    • (2009) J Alzheimers Dis , vol.16 , pp. 879-895
    • Altamura, S.1    Muckenthaler, M.U.2
  • 79
    • 84879082514 scopus 로고    scopus 로고
    • Implication of the proprotein convertases in iron homeostasis: proprotein convertase 7 sheds human transferrin receptor 1 and furin activates hepcidin
    • Guillemot J, Canuel M, Essalmani R, Prat A, Seidah NG Implication of the proprotein convertases in iron homeostasis: proprotein convertase 7 sheds human transferrin receptor 1 and furin activates hepcidin. Hepatology 2013, 57:2514-2524.
    • (2013) Hepatology , vol.57 , pp. 2514-2524
    • Guillemot, J.1    Canuel, M.2    Essalmani, R.3    Prat, A.4    Seidah, N.G.5
  • 80
    • 54049133647 scopus 로고    scopus 로고
    • A potential pathogenetic role of iron in Alzheimer's disease
    • Silvestri L, Camaschella C A potential pathogenetic role of iron in Alzheimer's disease. J Cell Mol Med 2008, 12:1548-1550.
    • (2008) J Cell Mol Med , vol.12 , pp. 1548-1550
    • Silvestri, L.1    Camaschella, C.2
  • 81
    • 0347928847 scopus 로고    scopus 로고
    • An iron-responsive element type II in the 5'-untranslated region of the Alzheimer's amyloid precursor protein transcript
    • Rogers JT, Randall JD, Cahill CM, et al. An iron-responsive element type II in the 5'-untranslated region of the Alzheimer's amyloid precursor protein transcript. J Biol Chem 2002, 277:45518-45528.
    • (2002) J Biol Chem , vol.277 , pp. 45518-45528
    • Rogers, J.T.1    Randall, J.D.2    Cahill, C.M.3
  • 82
    • 77956647381 scopus 로고    scopus 로고
    • Iron-export ferroxidase activity of β-amyloid precursor protein is inhibited by zinc in Alzheimer's disease
    • Duce JA, Tsatsanis A, Cater MA, et al. Iron-export ferroxidase activity of β-amyloid precursor protein is inhibited by zinc in Alzheimer's disease. Cell 2010, 142:857-867.
    • (2010) Cell , vol.142 , pp. 857-867
    • Duce, J.A.1    Tsatsanis, A.2    Cater, M.A.3
  • 83
    • 84865051177 scopus 로고    scopus 로고
    • A synthetic peptide with the putative iron binding motif of amyloid precursor protein (APP) does not catalytically oxidize iron
    • Ebrahimi KH, Hagedoorn PL, Hagen WR A synthetic peptide with the putative iron binding motif of amyloid precursor protein (APP) does not catalytically oxidize iron. PLoS One 2012, 7:e40287.
    • (2012) PLoS One , vol.7 , pp. e40287
    • Ebrahimi, K.H.1    Hagedoorn, P.L.2    Hagen, W.R.3
  • 85
    • 84856708923 scopus 로고    scopus 로고
    • Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export
    • Lei P, Ayton S, Finkelstein DI, et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nat Med 2012, 18:291-295.
    • (2012) Nat Med , vol.18 , pp. 291-295
    • Lei, P.1    Ayton, S.2    Finkelstein, D.I.3
  • 86
    • 12244310498 scopus 로고    scopus 로고
    • Magnetic resonance imaging of brain iron
    • Schenck JF Magnetic resonance imaging of brain iron. J Neurol Sci 2003, 207:99-102.
    • (2003) J Neurol Sci , vol.207 , pp. 99-102
    • Schenck, J.F.1
  • 87
    • 13944261999 scopus 로고    scopus 로고
    • Imaging iron stores in the brain using magnetic resonance imaging
    • Haacke EM, Cheng NY, House MJ, et al. Imaging iron stores in the brain using magnetic resonance imaging. Magn Reson Imaging 2005, 23:1-25.
    • (2005) Magn Reson Imaging , vol.23 , pp. 1-25
    • Haacke, E.M.1    Cheng, N.Y.2    House, M.J.3
  • 88
    • 27344439224 scopus 로고    scopus 로고
    • In vivo magnetic resonance microimaging of individual amyloid plaques in Alzheimer's transgenic mice
    • Jack CR, Wengenack TM, Reyes DA, et al. In vivo magnetic resonance microimaging of individual amyloid plaques in Alzheimer's transgenic mice. J Neurosci 2005, 25:10041-10048.
    • (2005) J Neurosci , vol.25 , pp. 10041-10048
    • Jack, C.R.1    Wengenack, T.M.2    Reyes, D.A.3
  • 89
    • 79959575876 scopus 로고    scopus 로고
    • High-field MRI of single histological slices using an inductively coupled, self-resonant microcoil: application to ex vivo samples of patients with Alzheimer's disease
    • Nabuurs RJ, Hegeman I, Natté R, et al. High-field MRI of single histological slices using an inductively coupled, self-resonant microcoil: application to ex vivo samples of patients with Alzheimer's disease. NMR Biomed 2011, 24:351-357.
    • (2011) NMR Biomed , vol.24 , pp. 351-357
    • Nabuurs, R.J.1    Hegeman, I.2    Natté, R.3
  • 90
    • 83055194393 scopus 로고    scopus 로고
    • High field magnetic resonance microscopy of the human hippocampus in Alzheimer's disease: quantitative imaging and correlation with iron
    • Antharam V, Collingwood JF, Bullivant JP, et al. High field magnetic resonance microscopy of the human hippocampus in Alzheimer's disease: quantitative imaging and correlation with iron. Neuroimage 2012, 59:1249-1260.
    • (2012) Neuroimage , vol.59 , pp. 1249-1260
    • Antharam, V.1    Collingwood, J.F.2    Bullivant, J.P.3
  • 91
    • 73949159970 scopus 로고    scopus 로고
    • Subregional neuroanatomical change as a biomarker for Alzheimer's disease
    • the Alzheimer's Disease Neuroimaging Initiative
    • Holland D, Brewer JB, Hagler DJ, Fennema-Notestine C, Dale AM Subregional neuroanatomical change as a biomarker for Alzheimer's disease. Proc Natl Acad Sci USA 2009, 106:20954-20959. the Alzheimer's Disease Neuroimaging Initiative.
    • (2009) Proc Natl Acad Sci USA , vol.106 , pp. 20954-20959
    • Holland, D.1    Brewer, J.B.2    Hagler, D.J.3    Fennema-Notestine, C.4    Dale, A.M.5
  • 92
    • 84876361328 scopus 로고    scopus 로고
    • Role of brain iron accumulation in cognitive dysfunction: evidence from animal models and human studies
    • Schröder N, Figueiredo LS, de Lima MN Role of brain iron accumulation in cognitive dysfunction: evidence from animal models and human studies. J Alzheimers Dis 2013, 34:797-812.
    • (2013) J Alzheimers Dis , vol.34 , pp. 797-812
    • Schröder, N.1    Figueiredo, L.S.2    de Lima, M.N.3
  • 93
    • 0031914718 scopus 로고    scopus 로고
    • Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes
    • Holcomb L, Gordon MN, McGowan E, et al. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat Med 1998, 4:97-100.
    • (1998) Nat Med , vol.4 , pp. 97-100
    • Holcomb, L.1    Gordon, M.N.2    McGowan, E.3
  • 94
    • 0031981072 scopus 로고    scopus 로고
    • Amyloid-beta deposition in Alzheimer transgenic mice is associated with oxidative stress
    • Smith MA, Hirai K, Hsiao K, et al. Amyloid-beta deposition in Alzheimer transgenic mice is associated with oxidative stress. J Neurochem 1998, 70:2212-2215.
    • (1998) J Neurochem , vol.70 , pp. 2212-2215
    • Smith, M.A.1    Hirai, K.2    Hsiao, K.3
  • 95
    • 17644397044 scopus 로고    scopus 로고
    • Histological co-localization of iron in Abeta plaques of PS/APP transgenic mice
    • Falangola MF, Lee SP, Nixon RA, Duff K, Helpern JA Histological co-localization of iron in Abeta plaques of PS/APP transgenic mice. Neurochem Res 2005, 30:201-205.
    • (2005) Neurochem Res , vol.30 , pp. 201-205
    • Falangola, M.F.1    Lee, S.P.2    Nixon, R.A.3    Duff, K.4    Helpern, J.A.5
  • 96
    • 78951481093 scopus 로고    scopus 로고
    • Increased brain iron coincides with early plaque formation in a mouse model of Alzheimer's disease
    • Leskovjan AC, Kretlow A, Lanzirotti A, Barrea R, Vogt S, Miller LM Increased brain iron coincides with early plaque formation in a mouse model of Alzheimer's disease. Neuroimage 2011, 55:32-38.
    • (2011) Neuroimage , vol.55 , pp. 32-38
    • Leskovjan, A.C.1    Kretlow, A.2    Lanzirotti, A.3    Barrea, R.4    Vogt, S.5    Miller, L.M.6
  • 97
    • 80052800581 scopus 로고    scopus 로고
    • Long-term overexpression of heme oxygenase 1 promotes tau aggregation in mouse brain by inducing tau phosphorylation
    • Hui Y, Wang D, Li W, et al. Long-term overexpression of heme oxygenase 1 promotes tau aggregation in mouse brain by inducing tau phosphorylation. J Alzheimers Dis 2011, 26:299-313.
    • (2011) J Alzheimers Dis , vol.26 , pp. 299-313
    • Hui, Y.1    Wang, D.2    Li, W.3
  • 98
    • 0029032632 scopus 로고
    • Expression of heme oxygenase-1 in the senescent and Alzheimer-diseased brain
    • Schipper HM, Cissé S, Stopa EG Expression of heme oxygenase-1 in the senescent and Alzheimer-diseased brain. Ann Neurol 1995, 37:758-768.
    • (1995) Ann Neurol , vol.37 , pp. 758-768
    • Schipper, H.M.1    Cissé, S.2    Stopa, E.G.3
  • 99
    • 29844450760 scopus 로고    scopus 로고
    • Glial heme oxygenase-1 expression in Alzheimer disease and mild cognitive impairment
    • Schipper HM, Bennett DA, Liberman A, et al. Glial heme oxygenase-1 expression in Alzheimer disease and mild cognitive impairment. Neurobiol Aging 2006, 27:252-261.
    • (2006) Neurobiol Aging , vol.27 , pp. 252-261
    • Schipper, H.M.1    Bennett, D.A.2    Liberman, A.3
  • 100
    • 0023638828 scopus 로고
    • Increased nigral iron content in postmortem parkinsonian brain
    • Dexter DT, Wells FR, Agid F, et al. Increased nigral iron content in postmortem parkinsonian brain. Lancet 1987, 330:1219-1220.
    • (1987) Lancet , vol.330 , pp. 1219-1220
    • Dexter, D.T.1    Wells, F.R.2    Agid, F.3
  • 101
    • 0024356620 scopus 로고
    • Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson's disease
    • Dexter DT, Wells FR, Lees AJ, et al. Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson's disease. J Neurochem 1989, 52:1830-1836.
    • (1989) J Neurochem , vol.52 , pp. 1830-1836
    • Dexter, D.T.1    Wells, F.R.2    Lees, A.J.3
  • 102
    • 0024557734 scopus 로고
    • Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains
    • Riederer P, Sofic E, Rausch WD, et al. Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. J Neurochem 1989, 52:515-520.
    • (1989) J Neurochem , vol.52 , pp. 515-520
    • Riederer, P.1    Sofic, E.2    Rausch, W.D.3
  • 103
    • 0026034279 scopus 로고
    • Iron and aluminum increase in the substantia nigra of patients with Parkinson's disease: an X-ray microanalysis
    • Hirsch EC, Brandel JP, Galle P, Javoy-Agid F, Agid Y Iron and aluminum increase in the substantia nigra of patients with Parkinson's disease: an X-ray microanalysis. J Neurochem 1991, 56:446-451.
    • (1991) J Neurochem , vol.56 , pp. 446-451
    • Hirsch, E.C.1    Brandel, J.P.2    Galle, P.3    Javoy-Agid, F.4    Agid, Y.5
  • 104
    • 78049460797 scopus 로고    scopus 로고
    • Magnetic resonance imaging markers of Parkinson's disease nigrostriatal signature
    • Péran P, Cherubini A, Assogna F, et al. Magnetic resonance imaging markers of Parkinson's disease nigrostriatal signature. Brain 2010, 133:3423-3433.
    • (2010) Brain , vol.133 , pp. 3423-3433
    • Péran, P.1    Cherubini, A.2    Assogna, F.3
  • 105
    • 84876250024 scopus 로고    scopus 로고
    • Does structural neuroimaging reveal a disturbance of iron metabolism in Parkinson's disease? Implications from MRI and TCS studies
    • Gröger A, Berg D Does structural neuroimaging reveal a disturbance of iron metabolism in Parkinson's disease? Implications from MRI and TCS studies. J Neural Transm 2012, 119:1523-1528.
    • (2012) J Neural Transm , vol.119 , pp. 1523-1528
    • Gröger, A.1    Berg, D.2
  • 106
    • 0033586471 scopus 로고    scopus 로고
    • Blood vessels change in the mesencephalon of patients with Parkinson's disease
    • Faucheux BA, Bonnet AM, Agid Y, Hirsch EC Blood vessels change in the mesencephalon of patients with Parkinson's disease. Lancet 1999, 353:981-982.
    • (1999) Lancet , vol.353 , pp. 981-982
    • Faucheux, B.A.1    Bonnet, A.M.2    Agid, Y.3    Hirsch, E.C.4
  • 107
    • 13144255682 scopus 로고    scopus 로고
    • Blood-brain barrier dysfunction in parkinsonian midbrain in vivo
    • Kortekaas R, Leenders KL, van Oostrom JC, et al. Blood-brain barrier dysfunction in parkinsonian midbrain in vivo. Ann Neurol 2005, 57:176-179.
    • (2005) Ann Neurol , vol.57 , pp. 176-179
    • Kortekaas, R.1    Leenders, K.L.2    van Oostrom, J.C.3
  • 108
    • 0028786932 scopus 로고
    • Expression of lactoferrin receptors is increased in the mesencephalon of patients with Parkinson disease
    • Faucheux BA, Nillesse N, Damier P, et al. Expression of lactoferrin receptors is increased in the mesencephalon of patients with Parkinson disease. Proc Natl Acad Sci USA 1995, 92:9603-9607.
    • (1995) Proc Natl Acad Sci USA , vol.92 , pp. 9603-9607
    • Faucheux, B.A.1    Nillesse, N.2    Damier, P.3
  • 109
    • 57449092273 scopus 로고    scopus 로고
    • Divalent metal transporter 1 (DMT1) contributes to neurodegeneration in animal models of Parkinson's disease
    • Salazar J, Mena N, Hunot S, et al. Divalent metal transporter 1 (DMT1) contributes to neurodegeneration in animal models of Parkinson's disease. Proc Natl Acad Sci USA 2008, 105:18578-18583.
    • (2008) Proc Natl Acad Sci USA , vol.105 , pp. 18578-18583
    • Salazar, J.1    Mena, N.2    Hunot, S.3
  • 110
    • 0036306510 scopus 로고    scopus 로고
    • Association study between iron-related genes polymorphisms and Parkinson's disease
    • the French Parkinson's disease genetic study group
    • Borie C, Gasparini F, Verpillat P, et al. Association study between iron-related genes polymorphisms and Parkinson's disease. J Neurol 2002, 249:801-804. the French Parkinson's disease genetic study group.
    • (2002) J Neurol , vol.249 , pp. 801-804
    • Borie, C.1    Gasparini, F.2    Verpillat, P.3
  • 111
    • 33747179546 scopus 로고    scopus 로고
    • Association of HFE common mutations with Parkinson's disease, Alzheimer's disease and mild cognitive impairment in a Portuguese cohort
    • Guerreiro RJ, Bras JM, Santana I, et al. Association of HFE common mutations with Parkinson's disease, Alzheimer's disease and mild cognitive impairment in a Portuguese cohort. BMC Neurol 2006, 6:24.
    • (2006) BMC Neurol , vol.6 , pp. 24
    • Guerreiro, R.J.1    Bras, J.M.2    Santana, I.3
  • 113
    • 0036670712 scopus 로고    scopus 로고
    • Aggregation and neurotoxicity of alpha-synuclein and related peptides
    • el-Agnaf OM, Irvine GB Aggregation and neurotoxicity of alpha-synuclein and related peptides. Biochem Soc Trans 2002, 30:559-565.
    • (2002) Biochem Soc Trans , vol.30 , pp. 559-565
    • el-Agnaf, O.M.1    Irvine, G.B.2
  • 114
    • 0035941201 scopus 로고    scopus 로고
    • Metal-triggered structural transformations, aggregation, and fibrillation of human alpha-synuclein. A possible molecular NK between Parkinson's disease and heavy metal exposure
    • Uversky VN, Li J, Fink AL Metal-triggered structural transformations, aggregation, and fibrillation of human alpha-synuclein. A possible molecular NK between Parkinson's disease and heavy metal exposure. J Biol Chem 2001, 276:44284-44296.
    • (2001) J Biol Chem , vol.276 , pp. 44284-44296
    • Uversky, V.N.1    Li, J.2    Fink, A.L.3
  • 116
    • 0025374969 scopus 로고
    • Decreased ferritin levels in brain in Parkinson's disease
    • Dexter DT, Carayon A, Vidailhet M, et al. Decreased ferritin levels in brain in Parkinson's disease. J Neurochem 1990, 55:16-20.
    • (1990) J Neurochem , vol.55 , pp. 16-20
    • Dexter, D.T.1    Carayon, A.2    Vidailhet, M.3
  • 117
    • 0025821265 scopus 로고
    • Alterations in the levels of iron, ferritin and other trace metals in Parkinson's disease and other neurodegenerative diseases affecting the basal ganglia
    • Dexter DT, Carayon A, Javoy-Agid F, et al. Alterations in the levels of iron, ferritin and other trace metals in Parkinson's disease and other neurodegenerative diseases affecting the basal ganglia. Brain 1991, 114:1953-1975.
    • (1991) Brain , vol.114 , pp. 1953-1975
    • Dexter, D.T.1    Carayon, A.2    Javoy-Agid, F.3
  • 118
    • 0027995435 scopus 로고
    • Complex I, iron, and ferritin in Parkinson's disease substantia nigra
    • Mann VM, Cooper JM, Daniel SE, et al. Complex I, iron, and ferritin in Parkinson's disease substantia nigra. Ann Neurol 1994, 36:876-881.
    • (1994) Ann Neurol , vol.36 , pp. 876-881
    • Mann, V.M.1    Cooper, J.M.2    Daniel, S.E.3
  • 119
    • 0029092802 scopus 로고
    • A quantitative analysis of isoferritins in select regions of aged, parkinsonian, and Alzheimer's diseased brains
    • Connor JR, Snyder BS, Arosio P, Loeffler DA, LeWitt P A quantitative analysis of isoferritins in select regions of aged, parkinsonian, and Alzheimer's diseased brains. J Neurochem 1995, 65:717-724.
    • (1995) J Neurochem , vol.65 , pp. 717-724
    • Connor, J.R.1    Snyder, B.S.2    Arosio, P.3    Loeffler, D.A.4    LeWitt, P.5
  • 120
    • 0032898122 scopus 로고    scopus 로고
    • Iron in the basal ganglia in Parkinson's disease. An in vitro study using extended X-ray absorption fine structure and cryo-electron microscopy
    • Griffiths PD, Dobson BR, Jones GR, Clarke DT Iron in the basal ganglia in Parkinson's disease. An in vitro study using extended X-ray absorption fine structure and cryo-electron microscopy. Brain 1999, 122:667-673.
    • (1999) Brain , vol.122 , pp. 667-673
    • Griffiths, P.D.1    Dobson, B.R.2    Jones, G.R.3    Clarke, D.T.4
  • 121
    • 0036798434 scopus 로고    scopus 로고
    • Lack of up-regulation of ferritin is associated with sustained iron regulatory protein-1 binding activity in the substantia nigra of patients with Parkinson's disease
    • Faucheux BA, Martin ME, Beaumont C, et al. Lack of up-regulation of ferritin is associated with sustained iron regulatory protein-1 binding activity in the substantia nigra of patients with Parkinson's disease. J Neurochem 2002, 83:320-330.
    • (2002) J Neurochem , vol.83 , pp. 320-330
    • Faucheux, B.A.1    Martin, M.E.2    Beaumont, C.3
  • 122
    • 0026666528 scopus 로고
    • Iron-melanin complex in substantia nigra of parkinsonian brains: an x-ray microanalysis
    • Jellinger K, Kienzl E, Rumpelmair G, et al. Iron-melanin complex in substantia nigra of parkinsonian brains: an x-ray microanalysis. J Neurochem 1992, 59:1168-1171.
    • (1992) J Neurochem , vol.59 , pp. 1168-1171
    • Jellinger, K.1    Kienzl, E.2    Rumpelmair, G.3
  • 123
    • 0026746512 scopus 로고
    • Neuromelanin-containing neurons of the substantia nigra accumulate iron and aluminum in Parkinson's disease: a LAMMA study
    • Good PF, Olanow CW, Perl DP Neuromelanin-containing neurons of the substantia nigra accumulate iron and aluminum in Parkinson's disease: a LAMMA study. Brain Res 1992, 593:343-346.
    • (1992) Brain Res , vol.593 , pp. 343-346
    • Good, P.F.1    Olanow, C.W.2    Perl, D.P.3
  • 124
    • 0042433192 scopus 로고    scopus 로고
    • Neuromelanin associated redox-active iron is increased in the substantia nigra of patients with Parkinson's disease
    • Faucheux BA, Martin ME, Beaumont C, Hauw JJ, Agid Y, Hirsch EC Neuromelanin associated redox-active iron is increased in the substantia nigra of patients with Parkinson's disease. J Neurochem 2003, 86:1142-1148.
    • (2003) J Neurochem , vol.86 , pp. 1142-1148
    • Faucheux, B.A.1    Martin, M.E.2    Beaumont, C.3    Hauw, J.J.4    Agid, Y.5    Hirsch, E.C.6
  • 125
    • 0023789193 scopus 로고
    • Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains
    • McGeer PL, Itagaki S, Boyes BE, McGeer EG Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology 1988, 38:1285-1291.
    • (1988) Neurology , vol.38 , pp. 1285-1291
    • McGeer, P.L.1    Itagaki, S.2    Boyes, B.E.3    McGeer, E.G.4
  • 126
    • 0032829070 scopus 로고    scopus 로고
    • Evidence of active nerve cell degeneration in the substantia nigra of humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure
    • Langston JW, Forno LS, Tetrud J, Reeves AG, Kaplan JA, Karluk D Evidence of active nerve cell degeneration in the substantia nigra of humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure. Ann Neurol 1999, 46:598-605.
    • (1999) Ann Neurol , vol.46 , pp. 598-605
    • Langston, J.W.1    Forno, L.S.2    Tetrud, J.3    Reeves, A.G.4    Kaplan, J.A.5    Karluk, D.6
  • 127
    • 47749110631 scopus 로고    scopus 로고
    • Human Neuromelanin induces neuroinflammation and neurodegeneration in the rat substantia nigra: implications for Parkinson's disease
    • 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.
    • (2008) Acta Neuropathol , vol.116 , pp. 47-55
    • Zecca, L.1    Wilms, H.2    Geick, S.3
  • 128
    • 78650527643 scopus 로고    scopus 로고
    • Neuromelanin activates microglia and induces degeneration of dopaminergic neurons: implications for progression of Parkinson's disease
    • Zhang W, Phillips K, Wielgus AR, et al. Neuromelanin activates microglia and induces degeneration of dopaminergic neurons: implications for progression of Parkinson's disease. Neurotox Res 2011, 19:63-72.
    • (2011) Neurotox Res , vol.19 , pp. 63-72
    • Zhang, W.1    Phillips, K.2    Wielgus, A.R.3
  • 129
    • 84873469778 scopus 로고    scopus 로고
    • Higher iron in the red nucleus marks Parkinson's dyskinesia
    • Lewis MM, Du G, Kidacki M, et al. Higher iron in the red nucleus marks Parkinson's dyskinesia. Neurobiol Aging 2013, 34:1497-1503.
    • (2013) Neurobiol Aging , vol.34 , pp. 1497-1503
    • Lewis, M.M.1    Du, G.2    Kidacki, M.3
  • 130
    • 84873680782 scopus 로고    scopus 로고
    • Decreased iron levels in the temporal cortex in postmortem human brains with Parkinson disease
    • Yu X, Du T, Song N, et al. Decreased iron levels in the temporal cortex in postmortem human brains with Parkinson disease. Neurology 2013, 80:492-495.
    • (2013) Neurology , vol.80 , pp. 492-495
    • Yu, X.1    Du, T.2    Song, N.3
  • 131
    • 0033597299 scopus 로고    scopus 로고
    • Reduced ferroxidase activity in the cerebrospinal fluid from patients with Parkinson's disease
    • Boll MC, Sotelo J, Otero E, Alcaraz-Zubeldia M, Rios C Reduced ferroxidase activity in the cerebrospinal fluid from patients with Parkinson's disease. Neurosci Lett 1999, 265:155-158.
    • (1999) Neurosci Lett , vol.265 , pp. 155-158
    • Boll, M.C.1    Sotelo, J.2    Otero, E.3    Alcaraz-Zubeldia, M.4    Rios, C.5
  • 132
    • 48449094910 scopus 로고    scopus 로고
    • Free copper, ferroxidase and SOD1 activities, lipid peroxidation and NO(x) content in the CSF. A different marker profile in four neurodegenerative diseases
    • Boll MC, Alcaraz-Zubeldia M, Montes S, Rios C Free copper, ferroxidase and SOD1 activities, lipid peroxidation and NO(x) content in the CSF. A different marker profile in four neurodegenerative diseases. Neurochem Res 2008, 33:1717-1723.
    • (2008) Neurochem Res , vol.33 , pp. 1717-1723
    • Boll, M.C.1    Alcaraz-Zubeldia, M.2    Montes, S.3    Rios, C.4
  • 133
    • 80054992416 scopus 로고    scopus 로고
    • Plasma ceruloplasmin ferroxidase activity correlates with the nigral sonographic area in Parkinson's disease patients: a pilot study
    • Martínez-Hernández R, Montes S, Higuera-Calleja J, et al. Plasma ceruloplasmin ferroxidase activity correlates with the nigral sonographic area in Parkinson's disease patients: a pilot study. Neurochem Res 2011, 36:2111-2115.
    • (2011) Neurochem Res , vol.36 , pp. 2111-2115
    • Martínez-Hernández, R.1    Montes, S.2    Higuera-Calleja, J.3
  • 134
    • 83455177604 scopus 로고    scopus 로고
    • Ceruloplasmin oxidation, a feature of Parkinson's disease CSF, inhibits ferroxidase activity and promotes cellular iron retention
    • Olivieri S, Conti A, Iannaccone S, et al. Ceruloplasmin oxidation, a feature of Parkinson's disease CSF, inhibits ferroxidase activity and promotes cellular iron retention. J Neurosci 2011, 31:18568-18577.
    • (2011) J Neurosci , vol.31 , pp. 18568-18577
    • Olivieri, S.1    Conti, A.2    Iannaccone, S.3
  • 135
    • 0036703490 scopus 로고    scopus 로고
    • Ceruloplasmin regulates iron levels in the CNS and prevents free radical injury
    • Patel BN, Dunn RJ, Jeong SY, Zhu Q, Julien JP, David S Ceruloplasmin regulates iron levels in the CNS and prevents free radical injury. J Neurosci 2002, 22:6578-6586.
    • (2002) J Neurosci , vol.22 , pp. 6578-6586
    • Patel, B.N.1    Dunn, R.J.2    Jeong, S.Y.3    Zhu, Q.4    Julien, J.P.5    David, S.6
  • 136
    • 84878398613 scopus 로고    scopus 로고
    • Ceruloplasmin dysfunction and therapeutic potential for Parkinson disease
    • Ayton S, Lei P, Duce JA, et al. Ceruloplasmin dysfunction and therapeutic potential for Parkinson disease. Ann Neurol 2013, 73:554-559.
    • (2013) Ann Neurol , vol.73 , pp. 554-559
    • Ayton, S.1    Lei, P.2    Duce, J.A.3
  • 137
    • 5444246509 scopus 로고    scopus 로고
    • Ceruloplasmin gene variations and substantia nigra hyperechogenicity in Parkinson disease
    • Hochstrasser H, Bauer P, Walter U, et al. Ceruloplasmin gene variations and substantia nigra hyperechogenicity in Parkinson disease. Neurology 2004, 63:1912-1917.
    • (2004) Neurology , vol.63 , pp. 1912-1917
    • Hochstrasser, H.1    Bauer, P.2    Walter, U.3
  • 138
    • 84879529900 scopus 로고    scopus 로고
    • Serum iron levels and the risk of Parkinson disease: a mendelian randomization study
    • the PD GWAS Consortium, the International Parkinson's Disease Genomics Consortiumthe International Parkinson's Disease Genomics Consortium, the Wellcome Trust Case Control Consortium 2the Wellcome Trust Case Control Consortium 2, the Genetics of Iron Status Consortiumthe Genetics of Iron Status Consortium
    • Pichler I, Del Greco M F, Gögele M, et al. Serum iron levels and the risk of Parkinson disease: a mendelian randomization study. PLoS Med 2013, 10:e1001462. the PD GWAS Consortium, the International Parkinson's Disease Genomics Consortiumthe International Parkinson's Disease Genomics Consortium, the Wellcome Trust Case Control Consortium 2the Wellcome Trust Case Control Consortium 2, the Genetics of Iron Status Consortiumthe Genetics of Iron Status Consortium.
    • (2013) PLoS Med , vol.10 , pp. e1001462
    • Pichler, I.1    Del Greco, M.F.2    Gögele, M.3
  • 139
    • 79958773207 scopus 로고    scopus 로고
    • Dietary intake of metals and risk of Parkinson's disease: a case-control study in Japan
    • the Fukuoka Kinki Parkinson's Disease Study Group
    • Miyake Y, Tanaka K, Fukushima W, et al. Dietary intake of metals and risk of Parkinson's disease: a case-control study in Japan. J Neurol Sci 2011, 306:98-102. the Fukuoka Kinki Parkinson's Disease Study Group.
    • (2011) J Neurol Sci , vol.306 , pp. 98-102
    • Miyake, Y.1    Tanaka, K.2    Fukushima, W.3
  • 140
    • 33745809508 scopus 로고    scopus 로고
    • Blood donations, iron stores, and risk of Parkinson's disease
    • Logroscino G, Chen H, Wing A, Ascherio A Blood donations, iron stores, and risk of Parkinson's disease. Mov Disord 2006, 21:835-838.
    • (2006) Mov Disord , vol.21 , pp. 835-838
    • Logroscino, G.1    Chen, H.2    Wing, A.3    Ascherio, A.4
  • 141
    • 70350557479 scopus 로고    scopus 로고
    • Anemia or low hemoglobin levels preceding Parkinson disease: a case-control study
    • Savica R, Grossardt BR, Carlin JM, et al. Anemia or low hemoglobin levels preceding Parkinson disease: a case-control study. Neurology 2009, 73:1381-1387.
    • (2009) Neurology , vol.73 , pp. 1381-1387
    • Savica, R.1    Grossardt, B.R.2    Carlin, J.M.3
  • 143
    • 79955012377 scopus 로고    scopus 로고
    • Direct visualization of Parkinson's disease by in vivo human brain imaging using 7.0T magnetic resonance imaging
    • Cho ZH, Oh SH, Kim JM, et al. Direct visualization of Parkinson's disease by in vivo human brain imaging using 7.0T magnetic resonance imaging. Mov Disord 2011, 26:713-718.
    • (2011) Mov Disord , vol.26 , pp. 713-718
    • Cho, Z.H.1    Oh, S.H.2    Kim, J.M.3
  • 145
    • 42049123661 scopus 로고    scopus 로고
    • Midbrain iron content in early Parkinson disease: a potential biomarker of disease status
    • Martin WR, Wieler M, Gee M Midbrain iron content in early Parkinson disease: a potential biomarker of disease status. Neurology 2008, 70:1411-1417.
    • (2008) Neurology , vol.70 , pp. 1411-1417
    • Martin, W.R.1    Wieler, M.2    Gee, M.3
  • 146
    • 0038107152 scopus 로고    scopus 로고
    • Differential diagnosis of Parkinson's disease and atypical parkinsonian disorders by magnetic resonance imaging
    • Savoiardo M Differential diagnosis of Parkinson's disease and atypical parkinsonian disorders by magnetic resonance imaging. Neurol Sci 2003, 24(suppl 1):S35-S37.
    • (2003) Neurol Sci , vol.24 , pp. S35-S37
    • Savoiardo, M.1
  • 147
    • 84858285794 scopus 로고    scopus 로고
    • Brain iron deposition fingerprints in Parkinson's disease and progressive supranuclear palsy
    • Boelmans K, Holst B, Hackius M, et al. Brain iron deposition fingerprints in Parkinson's disease and progressive supranuclear palsy. Mov Disord 2012, 27:421-427.
    • (2012) Mov Disord , vol.27 , pp. 421-427
    • Boelmans, K.1    Holst, B.2    Hackius, M.3
  • 148
    • 63649106965 scopus 로고    scopus 로고
    • Visualization of neuromelanin in the substantia nigra and locus ceruleus at 1·5T using a 3D-gradient echo sequence with magnetization transfer contrast
    • Nakane T, Nihashi T, Kawai H, Naganawa S Visualization of neuromelanin in the substantia nigra and locus ceruleus at 1·5T using a 3D-gradient echo sequence with magnetization transfer contrast. Magn Reson Med Sci 2008, 7:205-210.
    • (2008) Magn Reson Med Sci , vol.7 , pp. 205-210
    • Nakane, T.1    Nihashi, T.2    Kawai, H.3    Naganawa, S.4
  • 149
    • 56549090225 scopus 로고    scopus 로고
    • Neuromelanin-sensitive MRI. Basics, technique and clinical applications
    • Sasaki M, Shibata E, Kudo K, Tohyama K Neuromelanin-sensitive MRI. Basics, technique and clinical applications. Clin Neuroradiol 2008, 18:147-153.
    • (2008) Clin Neuroradiol , vol.18 , pp. 147-153
    • Sasaki, M.1    Shibata, E.2    Kudo, K.3    Tohyama, K.4
  • 150
    • 33745896818 scopus 로고    scopus 로고
    • Neuromelanin magnetic resonance imaging of locus ceruleus and substantia nigra in Parkinson's disease
    • Sasaki M, Shibata E, Tohyama K, et al. Neuromelanin magnetic resonance imaging of locus ceruleus and substantia nigra in Parkinson's disease. Neuroreport 2006, 17:1215-1218.
    • (2006) Neuroreport , vol.17 , pp. 1215-1218
    • Sasaki, M.1    Shibata, E.2    Tohyama, K.3
  • 151
    • 84876412536 scopus 로고    scopus 로고
    • Changes in substantia nigra and locus coeruleus in patients with early-stage Parkinson's disease using Neuromelanin-sensitive MR imaging
    • Ohtsuka C, Sasaki M, Konno K, et al. Changes in substantia nigra and locus coeruleus in patients with early-stage Parkinson's disease using Neuromelanin-sensitive MR imaging. Neurosci Lett 2013, 541:93-98.
    • (2013) Neurosci Lett , vol.541 , pp. 93-98
    • Ohtsuka, C.1    Sasaki, M.2    Konno, K.3
  • 152
    • 77955351608 scopus 로고    scopus 로고
    • Transcranial sonography for the discrimination of idiopathic Parkinson's disease from the atypical parkinsonian syndromes
    • Bouwmans AE, Vlaar AM, Srulijes K, Mess WH, Weber WE Transcranial sonography for the discrimination of idiopathic Parkinson's disease from the atypical parkinsonian syndromes. Int Rev Neurobiol 2010, 90:121-146.
    • (2010) Int Rev Neurobiol , vol.90 , pp. 121-146
    • Bouwmans, A.E.1    Vlaar, A.M.2    Srulijes, K.3    Mess, W.H.4    Weber, W.E.5
  • 153
    • 27844547163 scopus 로고    scopus 로고
    • In vivo detection of iron and Neuromelanin by transcranial sonography: a new approach for early detection of substantia nigra damage
    • Zecca L, Berg D, Arzberger T, et al. In vivo detection of iron and Neuromelanin by transcranial sonography: a new approach for early detection of substantia nigra damage. Mov Disord 2005, 20:1278-1285.
    • (2005) Mov Disord , vol.20 , pp. 1278-1285
    • Zecca, L.1    Berg, D.2    Arzberger, T.3
  • 154
    • 72649090521 scopus 로고    scopus 로고
    • Ferroportin 1 but not hephaestin contributes to iron accumulation in a cell model of Parkinson's disease
    • Song N, Wang J, Jiang H, Xie J Ferroportin 1 but not hephaestin contributes to iron accumulation in a cell model of Parkinson's disease. Free Radic Biol Med 2010, 48:332-341.
    • (2010) Free Radic Biol Med , vol.48 , pp. 332-341
    • Song, N.1    Wang, J.2    Jiang, H.3    Xie, J.4
  • 155
    • 0027991538 scopus 로고
    • Degeneration of nigrostriatal dopaminergic neurons increases iron within the substantia nigra: a histochemical and neurochemical study
    • Oestreicher E, Sengstock GJ, Riederer P, Olanow CW, Dunn AJ, Arendash GW Degeneration of nigrostriatal dopaminergic neurons increases iron within the substantia nigra: a histochemical and neurochemical study. Brain Res 1994, 660:8-18.
    • (1994) Brain Res , vol.660 , pp. 8-18
    • Oestreicher, E.1    Sengstock, G.J.2    Riederer, P.3    Olanow, C.W.4    Dunn, A.J.5    Arendash, G.W.6
  • 156
    • 58149347604 scopus 로고    scopus 로고
    • Deep gray matter involvement on brain MRI scans is associated with clinical progression in multiple sclerosis
    • Neema M, Arora A, Healy BC, et al. Deep gray matter involvement on brain MRI scans is associated with clinical progression in multiple sclerosis. J Neuroimaging 2009, 19:3-8.
    • (2009) J Neuroimaging , vol.19 , pp. 3-8
    • Neema, M.1    Arora, A.2    Healy, B.C.3
  • 157
    • 79959604087 scopus 로고    scopus 로고
    • MRI assessment of iron deposition in multiple sclerosis
    • Ropele S, de Graaf W, Khalil M, et al. MRI assessment of iron deposition in multiple sclerosis. J Magn Reson Imaging 2011, 34:13-21.
    • (2011) J Magn Reson Imaging , vol.34 , pp. 13-21
    • Ropele, S.1    de Graaf, W.2    Khalil, M.3
  • 158
    • 84868535759 scopus 로고    scopus 로고
    • Progressive multiple sclerosis: pathology and pathogenesis
    • Lassmann H, van Horssen J, Mahad D Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol 2012, 8:647-656.
    • (2012) Nat Rev Neurol , vol.8 , pp. 647-656
    • Lassmann, H.1    van Horssen, J.2    Mahad, D.3
  • 159
    • 58149237714 scopus 로고    scopus 로고
    • Quantitative in vivo magnetic resonance imaging of multiple sclerosis at 7 Tesla with sensitivity to iron
    • Hammond KE, Metcalf M, Carvajal L, et al. Quantitative in vivo magnetic resonance imaging of multiple sclerosis at 7 Tesla with sensitivity to iron. Ann Neurol 2008, 64:707-713.
    • (2008) Ann Neurol , vol.64 , pp. 707-713
    • Hammond, K.E.1    Metcalf, M.2    Carvajal, L.3
  • 160
    • 83755224887 scopus 로고    scopus 로고
    • Tracking iron in multiple sclerosis: a combined imaging and histopathological study at 7 Tesla
    • Bagnato F, Hametner S, Yao B, et al. Tracking iron in multiple sclerosis: a combined imaging and histopathological study at 7 Tesla. Brain 2011, 134:3602-3615.
    • (2011) Brain , vol.134 , pp. 3602-3615
    • Bagnato, F.1    Hametner, S.2    Yao, B.3
  • 161
    • 84455206532 scopus 로고    scopus 로고
    • Chronic multiple sclerosis lesions: characterization with high-field-strength MR imaging
    • Yao B, Bagnato F, Matsuura E, et al. Chronic multiple sclerosis lesions: characterization with high-field-strength MR imaging. Radiology 2012, 262:206-215.
    • (2012) Radiology , vol.262 , pp. 206-215
    • Yao, B.1    Bagnato, F.2    Matsuura, E.3
  • 162
    • 0034999085 scopus 로고    scopus 로고
    • Proinflammatory cytokines promote glial heme oxygenase-1 expression and mitochondrial iron deposition: implications for multiple sclerosis
    • Mehindate K, Sahlas DJ, Frankel D, et al. Proinflammatory cytokines promote glial heme oxygenase-1 expression and mitochondrial iron deposition: implications for multiple sclerosis. J Neurochem 2001, 77:1386-1395.
    • (2001) J Neurochem , vol.77 , pp. 1386-1395
    • Mehindate, K.1    Sahlas, D.J.2    Frankel, D.3
  • 163
    • 13344270899 scopus 로고    scopus 로고
    • Friedreich's ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion
    • Campuzano V, Montermini L, Moltò MD, et al. Friedreich's ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science 1996, 271:1423-1427.
    • (1996) Science , vol.271 , pp. 1423-1427
    • Campuzano, V.1    Montermini, L.2    Moltò, M.D.3
  • 164
    • 84865644479 scopus 로고    scopus 로고
    • Friedreich ataxia: new pathways
    • Pandolfo M Friedreich ataxia: new pathways. J Child Neurol 2012, 27:1204-1211.
    • (2012) J Child Neurol , vol.27 , pp. 1204-1211
    • Pandolfo, M.1
  • 165
    • 77953669993 scopus 로고    scopus 로고
    • Human iron-sulfur cluster assembly, cellular iron homeostasis, and disease
    • Ye H, Rouault TA Human iron-sulfur cluster assembly, cellular iron homeostasis, and disease. Biochemistry 2010, 49:4945-4956.
    • (2010) Biochemistry , vol.49 , pp. 4945-4956
    • Ye, H.1    Rouault, T.A.2
  • 166
    • 0032971328 scopus 로고    scopus 로고
    • Increased iron in the dentate nucleus of patients with Friedrich's ataxia
    • Waldvogel D, van Gelderen P, Hallett M Increased iron in the dentate nucleus of patients with Friedrich's ataxia. Ann Neurol 1999, 46:123-125.
    • (1999) Ann Neurol , vol.46 , pp. 123-125
    • Waldvogel, D.1    van Gelderen, P.2    Hallett, M.3
  • 167
    • 79952814526 scopus 로고    scopus 로고
    • Friedreich's ataxia: pathology, pathogenesis, and molecular genetics
    • Koeppen AH Friedreich's ataxia: pathology, pathogenesis, and molecular genetics. J Neurol Sci 2011, 303:1-12.
    • (2011) J Neurol Sci , vol.303 , pp. 1-12
    • Koeppen, A.H.1
  • 168
    • 84862321330 scopus 로고    scopus 로고
    • Current concepts and controversies in neurodegeneration with brain iron accumulation
    • Keogh MJ, Chinnery PF Current concepts and controversies in neurodegeneration with brain iron accumulation. Semin Pediatr Neurol 2012, 19:51-56.
    • (2012) Semin Pediatr Neurol , vol.19 , pp. 51-56
    • Keogh, M.J.1    Chinnery, P.F.2
  • 169
    • 84856044221 scopus 로고    scopus 로고
    • Neurodegeneration with brain iron accumulation-clinical syndromes and neuroimaging
    • Schipper HM Neurodegeneration with brain iron accumulation-clinical syndromes and neuroimaging. Biochim Biophys Acta 2012, 1822:350-360.
    • (2012) Biochim Biophys Acta , vol.1822 , pp. 350-360
    • Schipper, H.M.1
  • 170
    • 84880805523 scopus 로고    scopus 로고
    • Iron metabolism in the CNS: implications for neurodegenerative diseases
    • Rouault TA Iron metabolism in the CNS: implications for neurodegenerative diseases. Nat Rev Neurosci 2013, 14:551-564.
    • (2013) Nat Rev Neurosci , vol.14 , pp. 551-564
    • Rouault, T.A.1
  • 171
    • 84863725376 scopus 로고    scopus 로고
    • Neurodegeneration with brain iron accumulation
    • Dusek P, Schneider SA Neurodegeneration with brain iron accumulation. Curr Opin Neurol 2012, 25:499-506.
    • (2012) Curr Opin Neurol , vol.25 , pp. 499-506
    • Dusek, P.1    Schneider, S.A.2
  • 172
    • 79961146091 scopus 로고    scopus 로고
    • Genetics of neurodegeneration with brain iron accumulation
    • Gregory A, Hayflick SJ Genetics of neurodegeneration with brain iron accumulation. Curr Neurol Neurosci Rep 2011, 11:254-261.
    • (2011) Curr Neurol Neurosci Rep , vol.11 , pp. 254-261
    • Gregory, A.1    Hayflick, S.J.2
  • 174
    • 79953665042 scopus 로고    scopus 로고
    • Novel histopathologic findings in molecularly-confirmed pantothenate kinase-associated neurodegeneration
    • Kruer MC, Hiken M, Gregory A, et al. Novel histopathologic findings in molecularly-confirmed pantothenate kinase-associated neurodegeneration. Brain 2011, 134:947-958.
    • (2011) Brain , vol.134 , pp. 947-958
    • Kruer, M.C.1    Hiken, M.2    Gregory, A.3
  • 175
    • 84863337915 scopus 로고    scopus 로고
    • Neuroimaging features of neurodegeneration with brain iron accumulation
    • Kruer MC, Boddaert N, Schneider SA, et al. Neuroimaging features of neurodegeneration with brain iron accumulation. AJNR Am J Neuroradiol 2012, 33:407-414.
    • (2012) AJNR Am J Neuroradiol , vol.33 , pp. 407-414
    • Kruer, M.C.1    Boddaert, N.2    Schneider, S.A.3
  • 177
    • 33745553895 scopus 로고    scopus 로고
    • PLA2G6, encoding a phospholipase A2, is mutated in neurodegenerative disorders with high brain iron
    • Morgan NV, Westaway SK, Morton JE, et al. PLA2G6, encoding a phospholipase A2, is mutated in neurodegenerative disorders with high brain iron. Nat Genet 2006, 38:752-754.
    • (2006) Nat Genet , vol.38 , pp. 752-754
    • Morgan, N.V.1    Westaway, S.K.2    Morton, J.E.3
  • 178
    • 42949158281 scopus 로고    scopus 로고
    • Phenotypic spectrum of neurodegeneration associated with mutations in the PLA2G6 gene (PLAN)
    • Kurian MA, Morgan NV, MacPherson L, et al. Phenotypic spectrum of neurodegeneration associated with mutations in the PLA2G6 gene (PLAN). Neurology 2008, 70:1623-1629.
    • (2008) Neurology , vol.70 , pp. 1623-1629
    • Kurian, M.A.1    Morgan, N.V.2    MacPherson, L.3
  • 179
    • 78249252333 scopus 로고    scopus 로고
    • Defective FA2H leads to a novel form of neurodegeneration with brain iron accumulation (NBIA)
    • Kruer MC, Paisán-Ruiz C, Boddaert N, et al. Defective FA2H leads to a novel form of neurodegeneration with brain iron accumulation (NBIA). Ann Neurol 2010, 68:611-618.
    • (2010) Ann Neurol , vol.68 , pp. 611-618
    • Kruer, M.C.1    Paisán-Ruiz, C.2    Boddaert, N.3
  • 180
    • 33749133430 scopus 로고    scopus 로고
    • Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase
    • Ramirez A, Heimbach A, Gründemann J, et al. Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat Genet 2006, 38:1184-1191.
    • (2006) Nat Genet , vol.38 , pp. 1184-1191
    • Ramirez, A.1    Heimbach, A.2    Gründemann, J.3
  • 181
    • 77953338445 scopus 로고    scopus 로고
    • ATP13A2 mutations (PARK9) cause neurodegeneration with brain iron accumulation
    • Schneider SA, Paisan-Ruiz C, Quinn NP, et al. ATP13A2 mutations (PARK9) cause neurodegeneration with brain iron accumulation. Mov Disord 2010, 25:979-984.
    • (2010) Mov Disord , vol.25 , pp. 979-984
    • Schneider, S.A.1    Paisan-Ruiz, C.2    Quinn, N.P.3
  • 182
    • 79959328761 scopus 로고    scopus 로고
    • ATP13A2-related neurodegeneration (PARK9) without evidence of brain iron accumulation
    • Chien HF, Bonifati V, Barbosa ER ATP13A2-related neurodegeneration (PARK9) without evidence of brain iron accumulation. Mov Disord 2011, 26:1364-1365.
    • (2011) Mov Disord , vol.26 , pp. 1364-1365
    • Chien, H.F.1    Bonifati, V.2    Barbosa, E.R.3
  • 183
    • 84878841473 scopus 로고    scopus 로고
    • β-Propeller protein-associated neurodegeneration: a new X-linked dominant disorder with brain iron accumulation
    • Hayflick SJ, Kruer MC, Gregory A, et al. β-Propeller protein-associated neurodegeneration: a new X-linked dominant disorder with brain iron accumulation. Brain 2013, 136:1708-1717.
    • (2013) Brain , vol.136 , pp. 1708-1717
    • Hayflick, S.J.1    Kruer, M.C.2    Gregory, A.3
  • 184
    • 34547782810 scopus 로고    scopus 로고
    • Copper and iron disorders of the brain
    • Madsen E, Gitlin JD Copper and iron disorders of the brain. Annu Rev Neurosci 2007, 30:317-337.
    • (2007) Annu Rev Neurosci , vol.30 , pp. 317-337
    • Madsen, E.1    Gitlin, J.D.2
  • 185
    • 33845899114 scopus 로고    scopus 로고
    • Clinical features and natural history of neuroferritinopathy caused by the FTL1 460InsA mutation
    • Chinnery PF, Crompton DE, Birchall D, et al. Clinical features and natural history of neuroferritinopathy caused by the FTL1 460InsA mutation. Brain 2007, 130:110-119.
    • (2007) Brain , vol.130 , pp. 110-119
    • Chinnery, P.F.1    Crompton, D.E.2    Birchall, D.3
  • 187
    • 38149140232 scopus 로고    scopus 로고
    • Expression of a mutant form of the ferritin light chain gene induces neurodegeneration and iron overload in transgenic mice
    • Vidal R, Miravalle L, Gao X, et al. Expression of a mutant form of the ferritin light chain gene induces neurodegeneration and iron overload in transgenic mice. J Neurosci 2008, 28:60-67.
    • (2008) J Neurosci , vol.28 , pp. 60-67
    • Vidal, R.1    Miravalle, L.2    Gao, X.3
  • 188
    • 77953713610 scopus 로고    scopus 로고
    • Accumulation of oxidative DNA damage in brain mitochondria in mouse model of hereditary ferritinopathy
    • Deng X, Vidal R, Englander EW Accumulation of oxidative DNA damage in brain mitochondria in mouse model of hereditary ferritinopathy. Neurosci Lett 2010, 479:44-48.
    • (2010) Neurosci Lett , vol.479 , pp. 44-48
    • Deng, X.1    Vidal, R.2    Englander, E.W.3
  • 189
    • 84865601614 scopus 로고    scopus 로고
    • MRI findings in neuroferritinopathy
    • Ohta E, Takiyama Y MRI findings in neuroferritinopathy. Neurol Res Int 2012, 2012:197438.
    • (2012) Neurol Res Int , vol.2012 , pp. 197438
    • Ohta, E.1    Takiyama, Y.2
  • 190
    • 84867801862 scopus 로고    scopus 로고
    • Progressive brain iron accumulation in neuroferritinopathy measured by the thalamic T2* relaxation rate
    • McNeill A, Gorman G, Khan A, Horvath R, Blamire AM, Chinnery PF Progressive brain iron accumulation in neuroferritinopathy measured by the thalamic T2* relaxation rate. AJNR Am J Neuroradiol 2012, 33:1810-1813.
    • (2012) AJNR Am J Neuroradiol , vol.33 , pp. 1810-1813
    • McNeill, A.1    Gorman, G.2    Khan, A.3    Horvath, R.4    Blamire, A.M.5    Chinnery, P.F.6
  • 192
    • 33846225133 scopus 로고    scopus 로고
    • Huntington's disease
    • Walker FO Huntington's disease. Lancet 2007, 369:218-228.
    • (2007) Lancet , vol.369 , pp. 218-228
    • Walker, F.O.1
  • 194
    • 34548790267 scopus 로고    scopus 로고
    • Myelin breakdown and iron changes in Huntington's disease: pathogenesis and treatment implications
    • Bartzokis G, Lu PH, Tishler TA, et al. Myelin breakdown and iron changes in Huntington's disease: pathogenesis and treatment implications. Neurochem Res 2007, 32:1655-1664.
    • (2007) Neurochem Res , vol.32 , pp. 1655-1664
    • Bartzokis, G.1    Lu, P.H.2    Tishler, T.A.3
  • 196
    • 84873461127 scopus 로고    scopus 로고
    • MRI T2 Hypointensities in basal ganglia of premanifest Huntington's disease
    • RRN1173
    • Jurgens CK, Jasinschi R, Ekin A, et al. MRI T2 Hypointensities in basal ganglia of premanifest Huntington's disease. PLoS Curr 2010, 8:RRN1173.
    • (2010) PLoS Curr , vol.8
    • Jurgens, C.K.1    Jasinschi, R.2    Ekin, A.3
  • 197
    • 0035936633 scopus 로고    scopus 로고
    • MRI measurement of brain iron in patients with restless legs syndrome
    • Allen RP, Barker PB, Wehrl F, Song HK, Earley CJ MRI measurement of brain iron in patients with restless legs syndrome. Neurology 2001, 56:263-265.
    • (2001) Neurology , vol.56 , pp. 263-265
    • Allen, R.P.1    Barker, P.B.2    Wehrl, F.3    Song, H.K.4    Earley, C.J.5
  • 198
    • 38049072727 scopus 로고    scopus 로고
    • The role of iron in restless legs syndrome
    • Allen RP, Earley CJ The role of iron in restless legs syndrome. Mov Disord 2007, 22(suppl 18):S440-S448.
    • (2007) Mov Disord , vol.22 , pp. S440-S448
    • Allen, R.P.1    Earley, C.J.2
  • 199
    • 0042626103 scopus 로고    scopus 로고
    • Neuropathological examination suggests impaired brain iron acquisition in restless legs syndrome
    • Connor JR, Boyer PJ, Menzies SL, et al. Neuropathological examination suggests impaired brain iron acquisition in restless legs syndrome. Neurology 2003, 61:304-309.
    • (2003) Neurology , vol.61 , pp. 304-309
    • Connor, J.R.1    Boyer, P.J.2    Menzies, S.L.3
  • 200
    • 2342554293 scopus 로고    scopus 로고
    • Decreased transferrin receptor expression by Neuromelanin cells in restless legs syndrome
    • Connor JR, Wang XS, Patton SM, et al. Decreased transferrin receptor expression by Neuromelanin cells in restless legs syndrome. Neurology 2004, 62:1563-1567.
    • (2004) Neurology , vol.62 , pp. 1563-1567
    • Connor, J.R.1    Wang, X.S.2    Patton, S.M.3
  • 201
    • 77951651688 scopus 로고    scopus 로고
    • Intravenous iron dextran for severe refractory restless legs syndrome
    • Ondo WG Intravenous iron dextran for severe refractory restless legs syndrome. Sleep Med 2010, 11:494-496.
    • (2010) Sleep Med , vol.11 , pp. 494-496
    • Ondo, W.G.1
  • 202
    • 84862987741 scopus 로고    scopus 로고
    • The future of ultra-high field MRI and fMRI for study of the human brain
    • Duyn JH The future of ultra-high field MRI and fMRI for study of the human brain. Neuroimage 2012, 62:1241-1248.
    • (2012) Neuroimage , vol.62 , pp. 1241-1248
    • Duyn, J.H.1
  • 203
    • 84863522265 scopus 로고    scopus 로고
    • Quantitative susceptibility mapping (QSM) as a means to measure brain iron? A post mortem validation study
    • Langkammer C, Schweser F, Krebs N, et al. Quantitative susceptibility mapping (QSM) as a means to measure brain iron? A post mortem validation study. Neuroimage 2012, 62:1593-1599.
    • (2012) Neuroimage , vol.62 , pp. 1593-1599
    • Langkammer, C.1    Schweser, F.2    Krebs, N.3
  • 204
    • 84875216484 scopus 로고    scopus 로고
    • MR susceptibility imaging
    • Duyn J MR susceptibility imaging. J Magn Reson 2013, 229:198-207.
    • (2013) J Magn Reson , vol.229 , pp. 198-207
    • Duyn, J.1
  • 205
    • 77649257562 scopus 로고    scopus 로고
    • Layer-specific variation of iron content in cerebral cortex as a source of MRI contrast
    • Fukunaga M, Li TQ, van Gelderen P, et al. Layer-specific variation of iron content in cerebral cortex as a source of MRI contrast. Proc Natl Acad Sci USA 2010, 107:3834-3839.
    • (2010) Proc Natl Acad Sci USA , vol.107 , pp. 3834-3839
    • Fukunaga, M.1    Li, T.Q.2    van Gelderen, P.3
  • 206
    • 84872336461 scopus 로고    scopus 로고
    • A serial in vivo 7T magnetic resonance phase imaging study of white matter lesions in multiple sclerosis
    • Bian W, Harter K, Hammond-Rosenbluth KE, et al. A serial in vivo 7T magnetic resonance phase imaging study of white matter lesions in multiple sclerosis. Mult Scler 2013, 19:69-75.
    • (2013) Mult Scler , vol.19 , pp. 69-75
    • Bian, W.1    Harter, K.2    Hammond-Rosenbluth, K.E.3
  • 207
    • 84859880865 scopus 로고    scopus 로고
    • Iron accumulation in deep cortical layers accounts for MRI signal abnormalities in ALS: correlating 7 tesla MRI and pathology
    • Kwan JY, Jeong SY, Van Gelderen P, et al. Iron accumulation in deep cortical layers accounts for MRI signal abnormalities in ALS: correlating 7 tesla MRI and pathology. PLoS One 2012, 7:e35241.
    • (2012) PLoS One , vol.7 , pp. e35241
    • Kwan, J.Y.1    Jeong, S.Y.2    Van Gelderen, P.3
  • 209
    • 84877044901 scopus 로고    scopus 로고
    • Micro-compartment specific T2* relaxation in the brain
    • Sati P, van Gelderen P, Silva AC, et al. Micro-compartment specific T2* relaxation in the brain. Neuroimage 2013, 77:268-278.
    • (2013) Neuroimage , vol.77 , pp. 268-278
    • Sati, P.1    van Gelderen, P.2    Silva, A.C.3
  • 210
    • 34347370842 scopus 로고    scopus 로고
    • Selective iron chelation in Friedreich ataxia: biologic and clinical implications
    • Boddaert N, Le Quan Sang KH, Rötig A, et al. Selective iron chelation in Friedreich ataxia: biologic and clinical implications. Blood 2007, 110:401-408.
    • (2007) Blood , vol.110 , pp. 401-408
    • Boddaert, N.1    Le Quan Sang, K.H.2    Rötig, A.3
  • 212
    • 0242684415 scopus 로고    scopus 로고
    • Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: a novel therapy for Parkinson's disease
    • Kaur D, Yantiri F, Rajagopalan S, et al. Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: a novel therapy for Parkinson's disease. Neuron 2003, 37:899-909.
    • (2003) Neuron , vol.37 , pp. 899-909
    • Kaur, D.1    Yantiri, F.2    Rajagopalan, S.3
  • 213
    • 84655167560 scopus 로고    scopus 로고
    • Long-term improvement under deferiprone in a case of neurodegeneration with brain iron accumulation
    • Kwiatkowski A, Ryckewaert G, Jissendi Tchofo P, et al. Long-term improvement under deferiprone in a case of neurodegeneration with brain iron accumulation. Parkinsonism Relat Disord 2012, 18:110-112.
    • (2012) Parkinsonism Relat Disord , vol.18 , pp. 110-112
    • Kwiatkowski, A.1    Ryckewaert, G.2    Jissendi Tchofo, P.3
  • 214
    • 84872063167 scopus 로고    scopus 로고
    • Disease modifying strategy based upon iron chelation in Parkinson's disease: a translational study
    • Devos D, Moreau C, Kluza V, et al. Disease modifying strategy based upon iron chelation in Parkinson's disease: a translational study. Neurology 2012, 78(suppl):P02.240.
    • (2012) Neurology , vol.78 , pp. P02.240
    • Devos, D.1    Moreau, C.2    Kluza, V.3
  • 215
    • 84907964767 scopus 로고    scopus 로고
    • Targeting brain chelatable iron as a therapeutic strategy for Parkinson's disease. Translational and clinical studies
    • (abstr).
    • Moreau C, Devedjian J-C, Kluza J, et al. Targeting brain chelatable iron as a therapeutic strategy for Parkinson's disease. Translational and clinical studies. Am J Hematol 2013, 88:e36. (abstr).
    • (2013) Am J Hematol , vol.88 , pp. e36
    • Moreau, C.1    Devedjian, J.-C.2    Kluza, J.3
  • 216
    • 84908007961 scopus 로고    scopus 로고
    • Altered brain iron homeostasis in Parkinson's disease and potential for iron chelation therapy
    • (abstr).
    • Dexter D, Ward R, Crichton R, Kallo V, Srai SK Altered brain iron homeostasis in Parkinson's disease and potential for iron chelation therapy. Am J Hematol 2013, 88:e163. (abstr).
    • (2013) Am J Hematol , vol.88 , pp. e163
    • Dexter, D.1    Ward, R.2    Crichton, R.3    Kallo, V.4    Srai, S.K.5
  • 217
    • 0025726462 scopus 로고
    • Intramuscular desferrioxamine in patients with Alzheimer's disease
    • Crapper McLachlan DR, Kruck TP, Kalow W, et al. Intramuscular desferrioxamine in patients with Alzheimer's disease. Lancet 1991, 337:1304-1308.
    • (1991) Lancet , vol.337 , pp. 1304-1308
    • Crapper McLachlan, D.R.1    Kruck, T.P.2    Kalow, W.3
  • 218
    • 80052684579 scopus 로고    scopus 로고
    • The Alzheimer's therapeutic PBT2 promotes amyloid-β degradation and GSK3 phosphorylation via a metal chaperone activity
    • Crouch PJ, Savva MS, Hung LW, et al. The Alzheimer's therapeutic PBT2 promotes amyloid-β degradation and GSK3 phosphorylation via a metal chaperone activity. J Neurochem 2011, 119:220-230.
    • (2011) J Neurochem , vol.119 , pp. 220-230
    • Crouch, P.J.1    Savva, M.S.2    Hung, L.W.3
  • 219
    • 48949098573 scopus 로고    scopus 로고
    • Safety, efficacy, and biomarker findings of PBT2 in targeting Aβ as a modifying therapy for Alzheimer's disease: a phase IIa, double-blind, randomised, placebo-controlled trial
    • on behalf of the PBT2-201-EURO study group
    • Lannfelt L, Blennow K, Zetterberg H, et al. Safety, efficacy, and biomarker findings of PBT2 in targeting Aβ as a modifying therapy for Alzheimer's disease: a phase IIa, double-blind, randomised, placebo-controlled trial. Lancet Neurol 2008, 7:779-786. on behalf of the PBT2-201-EURO study group.
    • (2008) Lancet Neurol , vol.7 , pp. 779-786
    • Lannfelt, L.1    Blennow, K.2    Zetterberg, H.3
  • 220
    • 79952694447 scopus 로고    scopus 로고
    • Combined therapy with idebenone and deferiprone in patients with Friedreich's ataxia
    • Velasco-Sánchez D, Aracil A, Montero R, et al. Combined therapy with idebenone and deferiprone in patients with Friedreich's ataxia. Cerebellum 2011, 10:1-8.
    • (2011) Cerebellum , vol.10 , pp. 1-8
    • Velasco-Sánchez, D.1    Aracil, A.2    Montero, R.3
  • 221
    • 0036623360 scopus 로고    scopus 로고
    • Aceruloplasminemia: new clinical, pathophysiological and therapeutic insights
    • Loréal O, Turlin B, Pigeon C, et al. Aceruloplasminemia: new clinical, pathophysiological and therapeutic insights. J Hepatol 2002, 36:851-856.
    • (2002) J Hepatol , vol.36 , pp. 851-856
    • Loréal, O.1    Turlin, B.2    Pigeon, C.3
  • 223
    • 84879628275 scopus 로고    scopus 로고
    • Effectiveness of oral iron chelator treatment with deferasirox in an aceruloplasminemia patient with a novel ceruloplasmin gene mutation
    • Suzuki Y, Yoshida K, Aburakawa Y, et al. Effectiveness of oral iron chelator treatment with deferasirox in an aceruloplasminemia patient with a novel ceruloplasmin gene mutation. Intern Med 2013, 52:1527-1530.
    • (2013) Intern Med , vol.52 , pp. 1527-1530
    • Suzuki, Y.1    Yoshida, K.2    Aburakawa, Y.3
  • 224
    • 2342434172 scopus 로고    scopus 로고
    • Iron chelation therapy in aceruloplasminaemia: study of a patient with a novel missense mutation
    • Mariani R, Arosio C, Pelucchi S, et al. Iron chelation therapy in aceruloplasminaemia: study of a patient with a novel missense mutation. Gut 2004, 53:756-758.
    • (2004) Gut , vol.53 , pp. 756-758
    • Mariani, R.1    Arosio, C.2    Pelucchi, S.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.