The synaptic pathology of alpha-synuclein aggregation in dementia with Lewy bodies, Parkinson's disease and Parkinson's disease dementia

Walter J Schulz-Schaeffer, Walter J Schulz-Schaeffer

Abstract

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are usually associated with loss of dopaminergic neurons. Loss of substantia nigra neurons and presence of Lewy body inclusions in some of the remaining neurons are the hallmark pathology seen in the final stages of the disease. Attempts to correlate Lewy body pathology to either cell death or severity of clinical symptoms, however, have not been successful. While the pathophysiology of the neurodegenerative process can hardly be explained by Lewy bodies, the clinical symptoms do indicate a degenerative process located at the presynapse resulting in a neurotransmitter deficiency. Recently it was shown that 90% or even more of alpha-synuclein aggregates in DLB cases were located at the presynapses in the form of very small deposits. In parallel, dendritic spines are retracted, whereas the presynapses are relatively preserved, suggesting a neurotransmitter deprivation. The same alpha-synuclein pathology can be demonstrated for PD. These findings give rise to the notion that not cell death but rather alpha-synuclein aggregate-related synaptic dysfunction causes the neurodegeneration. This opens new perspectives for understanding PD and DLB. If presynaptic alpha-synuclein aggregation, not neuronal loss, is the key issue of the neurodegenerative process, then PD and DLB may eventually be treatable in the future. The disease may progress via trans-synaptical spread, suggesting that stem cell transplants are of limited use. Future therapies may focus on the regeneration of synapses.

Figures

Fig. 1
Fig. 1
Detection of α-synuclein deposits in DLB with conventional methods. H&E (a) shows 3 Lewy bodies (arrows). Immunohistochemically, Lewy bodies are detectable (b), whereas in the neuropil α-synuclein deposits are not distinguishable from the physiological α-synuclein staining (d) as compared to a control case (mAB 4B12, 1:1,000, abcam). With an antibody against phosphorylated α-synuclein (c), more deposits than just Lewy bodies are detectable (polyAB pSer129, 1:500, LifeSpan BioScience). Bar 100 μm
Fig. 2
Fig. 2
More than 90% of α-synuclein aggregates are located outside of Lewy bodies at synapses in the frontal cortex of DLB. Frontal cortex and cingulate gyrus of a DLB- and control patient as seen using a dissection microscope (a, b). The higher magnification of the PET blot (c) shows the synaptic distribution of aggregates much smaller than Lewy bodies (LB indicated by arrows; mAB 4B12, 1:10,000). Using an antibody against phosphorylated α-synuclein (d), only a fraction of proteinase K-resistant aggregates is detectable (polyAB pSer129, 1:5,000). The detectability of phosphorylated α-synuclein is strongly influenced by the fixation period. Here the tissue was fixated short term using buffered formaldehyde. Bar 100 μm
Fig. 3
Fig. 3
Synaptic α-synuclein aggregates are the main synuclein pathology in Parkinson’s disease as seen in DLB. The substantia nigra shows several proteinase K-resistant α-synuclein aggregates besides Lewy bodies (a). In a Parkinson’s disease patient with dementia (b), the frontal cortex shows a lot of tiny α-synuclein aggregates even though no Lewy bodies are detectable (mAB 4B12, 1:10,000). Bar 100 μm
Fig. 4
Fig. 4
An almost complete loss of dendritic spines accompanies the presynaptic α-synuclein aggregates. Golgy–Cox–Davenport staining of a neuronal dendrite of a frontal cortex neuron in a DLB patient (a) is compared to a control patient of the same age (b). Bar 50 μm

References

    1. Akopian AN, Wood JN. Peripheral nervous system-specific genes identified by subtractive cDNA cloning. J Biol Chem. 1995;270:21264–21270.
    1. Al-Wandi A, Ninkina N, Millership S, Williamson SJ, Jones PA, Buchman VL. Absence of alpha-synuclein affects dopamine metabolism and synaptic markers in the striatum of aging mice. Neurobiol Aging. 2010;31:796–804.
    1. Andreoletti O, Simon S, Lacroux C, et al. PrPSc accumulation in myocytes from sheep incubating natural scrapie. Nat Med. 2004;10:591–593.
    1. Aoki C, Mahadomrongkul V, Fujisawa S, Habersat R, Shirao T. Chemical and morphological alterations of spines within the hippocampus and entorhinal cortex precede the onset of Alzheimer’s disease pathology in double knock-in mice. J Comp Neurol. 2007;505:352–362.
    1. Aoki C, Sekino Y, Hanamura K, et al. Drebrin A is a postsynaptic protein that localizes in vivo to the submembranous surface of dendritic sites forming excitatory synapses. J Comp Neurol. 2005;483:383–402.
    1. Baba M, Nakajo S, Tu PH, et al. Aggregation of alpha-synuclein in Lewy bodies of sporadic Parkinson’s disease and dementia with Lewy bodies. Am J Pathol. 1998;152:879–884.
    1. Baglioni S, Casamenti F, Bucciantini M, et al. Prefibrillar amyloid aggregates could be generic toxins in higher organisms. J Neurosci. 2006;26:8160–8167.
    1. Beach TG, Adler CH, Lue L, et al. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009;117:613–634.
    1. Beach TG, White CL, Hamilton RL, et al. Evaluation of alpha-synuclein immunohistochemical methods used by invited experts. Acta Neuropathol. 2008;116:277–288.
    1. Belichenko PV, Brown D, Jeffrey M, Fraser JR. Dendritic and synaptic alterations of hippocampal pyramidal neurones in scrapie-infected mice. Neuropathol Appl Neurobiol. 2000;26:143–149.
    1. Bergeron C, Petrunka C, Weyer L, Pollanen MS. Altered neurofilament expression does not contribute to Lewy body formation. Am J Pathol. 1996;148:267–272.
    1. Bouzamondo-Bernstein E, Hopkins SD, Spilman P, et al. The neurodegeneration sequence in prion diseases: evidence from functional, morphological and ultrastructural studies of the GABAergic system. J Neuropathol Exp Neurol. 2004;63:882–899.
    1. Braak H, Del TK, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.
    1. Brundin P, Li JY, Holton JL, Lindvall O, Revesz T. Research in motion: the enigma of Parkinson’s disease pathology spread. Nat Rev Neurosci. 2008;9:741–745.
    1. Bucciantini M, Calloni G, Chiti F, et al. Prefibrillar amyloid protein aggregates share common features of cytotoxicity. J Biol Chem. 2004;279:31374–31382.
    1. Bucciantini M, Giannoni E, Chiti F, et al. Inherent toxicity of aggregates implies a common mechanism for protein misfolding diseases. Nature. 2002;416:507–511.
    1. Bucciantini M, Rigacci S, Berti A, et al. Patterns of cell death triggered in two different cell lines by HypF-N prefibrillar aggregates. FASEB J. 2005;19:437–439.
    1. Cabin DE, Shimazu K, Murphy D, et al. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking alpha-synuclein. J Neurosci. 2002;22:8797–8807.
    1. Chandra S, Fornai F, Kwon HB, et al. Double-knockout mice for alpha- and beta-synucleins: effect on synaptic functions. Proc Natl Acad Sci USA. 2004;101:14966–14971.
    1. Chandra S, Gallardo G, Fernandez-Chacon R, Schluter OM, Sudhof TC. Alpha-synuclein cooperates with CSPalpha in preventing neurodegeneration. Cell. 2005;123:383–396.
    1. Clavaguera F, Bolmont T, Crowther RA, et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat Cell Biol. 2009;11:909–913.
    1. Clayton DF, George JM. The synucleins: a family of proteins involved in synaptic function, plasticity, neurodegeneration and disease. Trends Neurosci. 1998;21:249–254.
    1. Conway KA, Harper JD, Lansbury PT., Jr Fibrils formed in vitro from alpha-synuclein and two mutant forms linked to Parkinson’s disease are typical amyloid. Biochemistry. 2000;39:2552–2563.
    1. Davenport HA, Combs CM. Golgi’s dichromate-silver method. 3. Chromating fluids. Stain Technol. 1954;29:165–173.
    1. Davies CA, Mann DM, Sumpter PQ, Yates PO. A quantitative morphometric analysis of the neuronal and synaptic content of the frontal and temporal cortex in patients with Alzheimer’s disease. J Neurol Sci. 1987;78:151–164.
    1. Day M, Wang Z, Ding J, et al. Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models. Nat Neurosci. 2006;9:251–259.
    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;106:13010–13015.
    1. Deutch AY (2006) Striatal plasticity in parkinsonism: dystrophic changes in medium spiny neurons and progression in Parkinson’s disease. J Neural Transm Suppl (70):67–70
    1. Dickson DW, Fujishiro H, DelleDonne A, et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol. 2008;115:437–444.
    1. Ding TT, Lee SJ, Rochet JC, Lansbury PT., Jr Annular alpha-synuclein protofibrils are produced when spherical protofibrils are incubated in solution or bound to brain-derived membranes. Biochemistry. 2002;41:10209–10217.
    1. DiRosa G, Puzzo D, Sant’Angelo A, Trinchese F, Arancio O. Alpha-synuclein: between synaptic function and dysfunction. Histol Histopathol. 2003;18:1257–1266.
    1. Forno LS. Neuropathology of Parkinson’s disease. J Neuropathol Exp Neurol. 1996;55:259–272.
    1. Fortin DL, Nemani VM, Voglmaier SM, Anthony MD, Ryan TA, Edwards RH. Neural activity controls the synaptic accumulation of alpha-synuclein. J Neurosci. 2005;25:10913–10921.
    1. Fortin DL, Troyer MD, Nakamura K, Kubo S, Anthony MD, Edwards RH. Lipid rafts mediate the synaptic localization of alpha-synuclein. J Neurosci. 2004;24:6715–6723.
    1. Fujiwara H, Hasegawa M, Dohmae N, et al. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat Cell Biol. 2002;4:160–164.
    1. Gai WP, Power JH, Blumbergs PC, Blessing WW. Multiple-system atrophy: a new alpha-synuclein disease? Lancet. 1998;352:547–548.
    1. Galvin JE, Pollack J, Morris JC. Clinical phenotype of Parkinson disease dementia. Neurology. 2006;67:1605–1611.
    1. Gelb DJ, Oliver E, Gilman S. Diagnostic criteria for Parkinson disease. Arch Neurol. 1999;56:33–39.
    1. George JM, Jin H, Woods WS, Clayton DF. Characterization of a novel protein regulated during the critical period for song learning in the zebra finch. Neuron. 1995;15:361–372.
    1. Ghetti B, Piccardo P, Spillantini MG, et al. Vascular variant of prion protein cerebral amyloidosis with tau-positive neurofibrillary tangles: the phenotype of the stop codon 145 mutation in PRNP. Proc Natl Acad Sci USA. 1996;93:744–748.
    1. Gibb WR, Lees AJ. The relevance of the Lewy body to the pathogenesis of idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1988;51:745–752.
    1. Goers J, Manning-Bog AB, McCormack AL, et al. Nuclear localization of alpha-synuclein and its interaction with histones. Biochemistry. 2003;42:8465–8471.
    1. Gomez-Isla T, Growdon WB, McNamara M, et al. Clinicopathologic correlates in temporal cortex in dementia with Lewy bodies. Neurology. 1999;53:2003–2009.
    1. Gomez-Tortosa E, Irizarry MC, Gomez-Isla T, Hyman BT. Clinical and neuropathological correlates of dementia with Lewy bodies. Ann NY Acad Sci. 2000;920:9–15.
    1. Gomez-Tortosa E, Newell K, Irizarry MC, Albert M, Growdon JH, Hyman BT. Clinical and quantitative pathologic correlates of dementia with Lewy bodies. Neurology. 1999;53:1284–1291.
    1. Gubellini P, Picconi B, Di FM, Calabresi P. Downstream mechanisms triggered by mitochondrial dysfunction in the basal ganglia: from experimental models to neurodegenerative diseases. Biochim Biophys Acta. 2010;1802:151–161.
    1. Harding AJ, Broe GA, Halliday GM. Visual hallucinations in Lewy body disease relate to Lewy bodies in the temporal lobe. Brain. 2002;125:391–403.
    1. Harding AJ, Halliday GM. Cortical Lewy body pathology in the diagnosis of dementia. Acta Neuropathol. 2001;102:355–363.
    1. Harding AJ, Stimson E, Henderson JM, Halliday GM. Clinical correlates of selective pathology in the amygdala of patients with Parkinson’s disease. Brain. 2002;125:2431–2445.
    1. Hardy JA, Dodd PR, Oakley AE, Kidd AM, Perry RH, Edwardson JA. Use of post-mortem human synaptosomes for studies of metabolism and transmitter amino acid release. Neurosci Lett. 1982;33:317–322.
    1. Haycock JW. Polyvinylpyrrolidone as a blocking agent in immunochemical studies. Anal Biochem. 1993;208:397–399.
    1. Hill WD. Altered neurofilament expression does not contribute to Lewy body formation. Am J Pathol. 1996;149:728–729.
    1. Hornykiewicz O. Basic research on dopamine in Parkinson’s disease and the discovery of the nigrostriatal dopamine pathway: the view of an eyewitness. Neurodegener Dis. 2008;5:114–117.
    1. Huttner WB, Schiebler W, Greengard P, De CP. Synapsin I (protein I), a nerve terminal-specific phosphoprotein. III. Its association with synaptic vesicles studied in a highly purified synaptic vesicle preparation. J Cell Biol. 1983;96:1374–1388.
    1. Hyman BT, Trojanowski JQ. Consensus recommendations for the postmortem diagnosis of Alzheimer disease from the National Institute on Aging and the Reagan Institute Working Group on diagnostic criteria for the neuropathological assessment of Alzheimer disease. J Neuropathol Exp Neurol. 1997;56:1095–1097.
    1. Ingham CA, Hood SH, Arbuthnott GW. Spine density on neostriatal neurones changes with 6-hydroxydopamine lesions and with age. Brain Res. 1989;503:334–338.
    1. Iwai A, Masliah E, Yoshimoto M, et al. The precursor protein of non-A beta component of Alzheimer’s disease amyloid is a presynaptic protein of the central nervous system. Neuron. 1995;14:467–475.
    1. Iwatsubo T, Yamaguchi H, Fujimuro M, et al. Purification and characterization of Lewy bodies from the brains of patients with diffuse Lewy body disease. Am J Pathol. 1996;148:1517–1529.
    1. Jakes R, Spillantini MG, Goedert M. Identification of two distinct synucleins from human brain. FEBS Lett. 1994;345:27–32.
    1. Jankovic J. Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry. 2008;79:368–376.
    1. Javoy-Agid F, Hirsch EC, Dumas S, Duyckaerts C, Mallet J, Agid Y. Decreased tyrosine hydroxylase messenger RNA in the surviving dopamine neurons of the substantia nigra in Parkinson’s disease: an in situ hybridization study. Neuroscience. 1990;38:245–253.
    1. Jeffrey M, Halliday WG, Bell J, et al. Synapse loss associated with abnormal PrP precedes neuronal degeneration in the scrapie-infected murine hippocampus. Neuropathol Appl Neurobiol. 2000;26:41–54.
    1. Jellinger KA. Lewy body-related alpha-synucleinopathy in the aged human brain. J Neural Transm. 2004;111:1219–1235.
    1. Jellinger KA. A critical reappraisal of current staging of Lewy-related pathology in human brain. Acta Neuropathol. 2008;116:1–16.
    1. Ji H, Liu YE, Jia T, et al. Identification of a breast cancer-specific gene, BCSG1, by direct differential cDNA sequencing. Cancer Res. 1997;57:759–764.
    1. Jo E, Darabie AA, Han K, Tandon A, Fraser PE, McLaurin J. alpha-Synuclein–synaptosomal membrane interactions: implications for fibrillogenesis. Eur J Biochem. 2004;271:3180–3189.
    1. Kahle PJ, Neumann M, Ozmen L, et al. Selective insolubility of alpha-synuclein in human Lewy body diseases is recapitulated in a transgenic mouse model. Am J Pathol. 2001;159:2215–2225.
    1. Kitamoto T, Shin RW, Doh-ura K, et al. Abnormal isoform of prion proteins accumulates in the synaptic structures of the central nervous system in patients with Creutzfeldt–Jakob disease. Am J Pathol. 1992;140:1285–1294.
    1. Kobayashi C, Aoki C, Kojima N, Yamazaki H, Shirao T. Drebrin a content correlates with spine head size in the adult mouse cerebral cortex. J Comp Neurol. 2007;503:618–626.
    1. Kontopoulos E, Parvin JD, Feany MB. Alpha-synuclein acts in the nucleus to inhibit histone acetylation and promote neurotoxicity. Hum Mol Genet. 2006;15:3012–3023.
    1. Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease. Nat Med. 2008;14:504–506.
    1. Kordower JH, Chu Y, Hauser RA, Olanow CW, Freeman TB. Transplanted dopaminergic neurons develop PD pathologic changes: a second case report. Mov Disord. 2008;23:2303–2306.
    1. Kordower JH, Freeman TB, Snow BJ, et al. Neuropathological evidence of graft survival and striatal reinnervation after the transplantation of fetal mesencephalic tissue in a patient with Parkinson’s disease. N Engl J Med. 1995;332:1118–1124.
    1. Kovacs GG, Puopolo M, Ladogana A, et al. Genetic prion disease: the EUROCJD experience. Hum Genet. 2005;118:166–174.
    1. Kramer ML, Behrens C, Schulz-Schaeffer WJ. Selective detection, quantification, and subcellular location of alpha-synuclein aggregates with a protein aggregate filtration assay. Biotechniques. 2008;44:403–411.
    1. Kramer ML, Schulz-Schaeffer WJ. Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci. 2007;27:1405–1410.
    1. Kruger R, Kuhn W, Muller T, et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet. 1998;18:106–108.
    1. Lacroux C, Simon S, Benestad SL, et al. Prions in milk from ewes incubating natural scrapie. PLoS Pathog. 2008;4:e1000238.
    1. Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science. 1983;219:979–980.
    1. Langston JW, Sastry S, Chan P, Forno LS, Bolin LM, Di Monte DA. Novel alpha-synuclein-immunoreactive proteins in brain samples from the Contursi kindred, Parkinson’s, and Alzheimer’s disease. Exp Neurol. 1998;154:684–690.
    1. Lansbury PT., Jr Evolution of amyloid: what normal protein folding may tell us about fibrillogenesis and disease. Proc Natl Acad Sci USA. 1999;96:3342–3344.
    1. Lee HJ, Lee SJ. Characterization of cytoplasmic alpha-synuclein aggregates. Fibril formation is tightly linked to the inclusion-forming process in cells. J Biol Chem. 2002;277:48976–48983.
    1. Lee HJ, Patel S, Lee SJ. Intravesicular localization and exocytosis of alpha-synuclein and its aggregates. J Neurosci. 2005;25:6016–6024.
    1. Lee HJ, Suk JE, Bae EJ, Lee JH, Paik SR, Lee SJ. Assembly-dependent endocytosis and clearance of extracellular alpha-synuclein. Int J Biochem Cell Biol. 2008;40:1835–1849.
    1. Lezmi S, Bencsik A, Baron T. PET-blot analysis contributes to BSE strain recognition in C57Bl/6 mice. J Histochem Cytochem. 2006;54:1087–1094.
    1. Li JY, Englund E, Holton JL, et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med. 2008;14:501–503.
    1. Libow LS, Frisina PG, Haroutunian V, Perl DP, Purohit DP. Parkinson’s disease dementia: a diminished role for the Lewy body. Parkinsonism Relat Disord. 2009;15:572–575.
    1. Linazasoro G. Classical Parkinson disease versus Parkinson complex—reflections against staging and in favour of heterogeneity. Eur J Neurol. 2007;14:721–728.
    1. Lippa CF, Fujiwara H, Mann DM, et al. Lewy bodies contain altered alpha-synuclein in brains of many familial Alzheimer’s disease patients with mutations in presenilin and amyloid precursor protein genes. Am J Pathol. 1998;153:1365–1370.
    1. Liu S, Ninan I, Antonova I, et al. alpha-Synuclein produces a long-lasting increase in neurotransmitter release. EMBO J. 2004;23:4506–4516.
    1. Lotharius J, Brundin P. Impaired dopamine storage resulting from alpha-synuclein mutations may contribute to the pathogenesis of Parkinson’s disease. Hum Mol Genet. 2002;11:2395–2407.
    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;106:20051–20056.
    1. Maroteaux L, Campanelli JT, Scheller RH. Synuclein: a neuron-specific protein localized to the nucleus and presynaptic nerve terminal. J Neurosci. 1988;8:2804–2815.
    1. Mattila PM, Rinne JO, Helenius H, Dickson DW, Roytta M. Alpha-synuclein-immunoreactive cortical Lewy bodies are associated with cognitive impairment in Parkinson’s disease. Acta Neuropathol. 2000;100:285–290.
    1. McBride PA, Schulz-Schaeffer WJ, Donaldson M, et al. Early spread of scrapie from the gastrointestinal tract to the central nervous system involves autonomic fibers of the splanchnic and vagus nerves. J Virol. 2001;75:9320–9327.
    1. McKeith I (2007) Dementia with Lewy bodies. In: Koller WC, Melamed E (eds) Parkinson’s disease and related disorders, Part II. Handb Clin Neurol 84:531–548
    1. McKeith IG. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the Consortium on DLB International Workshop. J Alzheimers Dis. 2006;9:417–423.
    1. McKeith IG, Dickson DW, Lowe J, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65:1863–1872.
    1. McKeith IG, Galasko D, Kosaka K, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology. 1996;47:1113–1124.
    1. McNeill TH, Brown SA, Rafols JA, Shoulson I. Atrophy of medium spiny I striatal dendrites in advanced Parkinson’s disease. Brain Res. 1988;455:148–152.
    1. Meissner B, Westner IM, Kallenberg K, et al. Sporadic Creutzfeldt–Jakob disease: clinical and diagnostic characteristics of the rare VV1 type. Neurology. 2005;65:1544–1550.
    1. Meyer-Luehmann M, Coomaraswamy J, Bolmont T, et al. Exogenous induction of cerebral beta-amyloidogenesis is governed by agent and host. Science. 2006;313:1781–1784.
    1. Miake H, Mizusawa H, Iwatsubo T, Hasegawa M. Biochemical characterization of the core structure of alpha-synuclein filaments. J Biol Chem. 2002;277:19213–19219.
    1. Mirra SS, Heyman A, McKeel D, et al. The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology. 1991;41:479–486.
    1. Murphy DD, Rueter SM, Trojanowski JQ, Lee VM. Synucleins are developmentally expressed, and alpha-synuclein regulates the size of the presynaptic vesicular pool in primary hippocampal neurons. J Neurosci. 2000;20:3214–3220.
    1. Nagerl UV, Eberhorn N, Cambridge SB, Bonhoeffer T. Bidirectional activity-dependent morphological plasticity in hippocampal neurons. Neuron. 2004;44:759–767.
    1. Nakajo S, Shioda S, Nakai Y, Nakaya K. Localization of phosphoneuroprotein 14 (PNP 14) and its mRNA expression in rat brain determined by immunocytochemistry and in situ hybridization. Brain Res Mol Brain Res. 1994;27:81–86.
    1. Nakajo S, Tsukada K, Omata K, Nakamura Y, Nakaya K. A new brain-specific 14-kDa protein is a phosphoprotein. Its complete amino acid sequence and evidence for phosphorylation. Eur J Biochem. 1993;217:1057–1063.
    1. Naoi M, Maruyama W, Yi H, Inaba K, Akao Y, Shamoto-Nagai M. Mitochondria in neurodegenerative disorders: regulation of the redox state and death signaling leading to neuronal death and survival. J Neural Transm. 2009;116:1371–1381.
    1. Nikolaus S, Antke C, Muller HW. In vivo imaging of synaptic function in the central nervous system: I. Movement disorders and dementia. Behav Brain Res. 2009;204:1–31.
    1. Pan-Montojo F, Anichtchik O, Dening Y, et al. Progression of Parkinson’s disease pathology is reproduced by intragastric administration of rotenone in mice. PLoS One. 2010;5:e8762.
    1. Parkkinen L, Kauppinen T, Pirttila T, Autere JM, Alafuzoff I. Alpha-synuclein pathology does not predict extrapyramidal symptoms or dementia. Ann Neurol. 2005;57:82–91.
    1. Patt S, Gertz HJ, Gerhard L, Cervos-Navarro J. Pathological changes in dendrites of substantia nigra neurons in Parkinson’s disease: a Golgi study. Histol Histopathol. 1991;6:373–380.
    1. Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276:2045–2047.
    1. Revuelta GJ, Rosso A, Lippa CF. Neuritic pathology as a correlate of synaptic loss in dementia with Lewy bodies. Am J Alzheimers Dis Other Demen. 2008;23:97–102.
    1. Ritchie DL, Head MW, Ironside JW. Advances in the detection of prion protein in peripheral tissues of variant Creutzfeldt–Jakob disease patients using paraffin-embedded tissue blotting. Neuropathol Appl Neurobiol. 2004;30:360–368.
    1. Saito Y, Kawashima A, Ruberu NN, et al. Accumulation of phosphorylated alpha-synuclein in aging human brain. J Neuropathol Exp Neurol. 2003;62:644–654.
    1. Scheibel ME, Scheibel AB. The methods of Golgi. In: Robertson RT, editor. Neuroanatomical research techniques. New York: Academic Press; 1978. pp. 89–114.
    1. Schulz-Schaeffer WJ, Fatzer R, Vandevelde M, Kretzschmar HA (2000) Detection of PrP(Sc) in subclinical BSE with the paraffin-embedded tissue (PET) blot. Arch Virol Suppl 173–180
    1. Schulz-Schaeffer WJ, Tschoke S, Kranefuss N, et al. The paraffin-embedded tissue blot detects PrP(Sc) early in the incubation time in prion diseases. Am J Pathol. 2000;156:51–56.
    1. Selkoe DJ. Alzheimer’s disease is a synaptic failure. Science. 2002;298:789–791.
    1. Sidhu A, Wersinger C, Vernier P. Does alpha-synuclein modulate dopaminergic synaptic content and tone at the synapse? FASEB J. 2004;18:637–647.
    1. Simon-Sanchez J, Schulte C, Bras JM, et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet. 2009;41:1308–1312.
    1. Small DH, Mok SS, Bornstein JC. Alzheimer’s disease and Abeta toxicity: from top to bottom. Nat Rev Neurosci. 2001;2:595–598.
    1. Solis O, Limon DI, Flores-Hernandez J, Flores G. Alterations in dendritic morphology of the prefrontal cortical and striatum neurons in the unilateral 6-OHDA-rat model of Parkinson’s disease. Synapse. 2007;61:450–458.
    1. Specht CG, Tigaret CM, Rast GF, Thalhammer A, Rudhard Y, Schoepfer R. Subcellular localisation of recombinant alpha- and gamma-synuclein. Mol Cell Neurosci. 2005;28:326–334.
    1. Spillantini MG, Crowther RA, Jakes R, Cairns NJ, Lantos PL, Goedert M. Filamentous alpha-synuclein inclusions link multiple system atrophy with Parkinson’s disease and dementia with Lewy bodies. Neurosci Lett. 1998;251:205–208.
    1. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–840.
    1. Stephens B, Mueller AJ, Shering AF, et al. Evidence of a breakdown of corticostriatal connections in Parkinson’s disease. Neuroscience. 2005;132:741–754.
    1. Terry RD. Cell death or synaptic loss in Alzheimer disease. J Neuropathol Exp Neurol. 2000;59:1118–1119.
    1. Terry RD. Do neuronal inclusions kill the cell? J Neural Transm Suppl. 2000;59:91–93.
    1. Terry RD, Masliah E, Salmon DP, et al. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572–580.
    1. Thomzig A, Schulz-Schaeffer W, Kratzel C, Mai J, Beekes M. Preclinical deposition of pathological prion protein PrPSc in muscles of hamsters orally exposed to scrapie. J Clin Invest. 2004;113:1465–1472.
    1. Thomzig A, Schulz-Schaeffer W, Wrede A, et al. Accumulation of pathological prion protein PrPSc in the skin of animals with experimental and natural scrapie. PLoS Pathog. 2007;3:e66.
    1. Thomzig A, Spassov S, Friedrich M, Naumann D, Beekes M. Discriminating scrapie and bovine spongiform encephalopathy isolates by infrared spectroscopy of pathological prion protein. J Biol Chem. 2004;279:33847–33854.
    1. Tompkins MM, Hill WD. Contribution of somal Lewy bodies to neuronal death. Brain Res. 1997;775:24–29.
    1. Totterdell S, Meredith GE. Localization of alpha-synuclein to identified fibers and synapses in the normal mouse brain. Neuroscience. 2005;135:907–913.
    1. Tu PH, Galvin JE, Baba M, et al. Glial cytoplasmic inclusions in white matter oligodendrocytes of multiple system atrophy brains contain insoluble alpha-synuclein. Ann Neurol. 1998;44:415–422.
    1. Ueda K, Fukushima H, Masliah E, et al. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc Natl Acad Sci USA. 1993;90:11282–11286.
    1. Wakabayashi K, Hayashi S, Kakita A, et al. Accumulation of alpha-synuclein/NACP is a cytopathological feature common to Lewy body disease and multiple system atrophy. Acta Neuropathol. 1998;96:445–452.
    1. Wakabayashi K, Tanji K, Mori F, Takahashi H. The Lewy body in Parkinson’s disease: molecules implicated in the formation and degradation of alpha-synuclein aggregates. Neuropathology. 2007;27:494–506.
    1. Wakabayashi K, Yoshimoto M, Tsuji S, Takahashi H. Alpha-synuclein immunoreactivity in glial cytoplasmic inclusions in multiple system atrophy. Neurosci Lett. 1998;249:180–182.
    1. Wanker EE, Scherzinger E, Heiser V, Sittler A, Eickhoff H, Lehrach H. Membrane filter assay for detection of amyloid-like polyglutamine-containing protein aggregates. Methods Enzymol. 1999;309:375–386.
    1. Weisman D, Cho M, Taylor C, Adame A, Thal LJ, Hansen LA. In dementia with Lewy bodies, Braak stage determines phenotype, not Lewy body distribution. Neurology. 2007;69:356–359.
    1. Wemheuer WM, Benestad SL, Wrede A, et al. Similarities between forms of sheep scrapie and Creutzfeldt–Jakob disease are encoded by distinct prion types. Am J Pathol. 2009;175:2566–2573.
    1. Wemheuer WM, Benestad SL, Wrede A, et al. Detection of classical and atypical/Nor98 scrapie by the paraffin-embedded tissue blot method. Vet Rec. 2009;164:677–681.
    1. Whittaker VP. Thirty years of synaptosome research. J Neurocytol. 1993;22:735–742.
    1. Winklhofer KF, Haass C. Mitochondrial dysfunction in Parkinson’s disease. Biochim Biophys Acta. 2010;1802:29–44.
    1. Winklhofer KF, Hartl FU, Tatzelt J. A sensitive filter retention assay for the detection of PrP(Sc) and the screening of anti-prion compounds. FEBS Lett. 2001;503:41–45.
    1. Withers GS, George JM, Banker GA, Clayton DF. Delayed localization of synelfin (synuclein, NACP) to presynaptic terminals in cultured rat hippocampal neurons. Brain Res Dev Brain Res. 1997;99:87–94.
    1. Yavich L, Tanila H, Vepsalainen S, Jakala P. Role of alpha-synuclein in presynaptic dopamine recruitment. J Neurosci. 2004;24:11165–11170.
    1. Yu S, Li X, Liu G, et al. Extensive nuclear localization of alpha-synuclein in normal rat brain neurons revealed by a novel monoclonal antibody. Neuroscience. 2007;145:539–555.
    1. Yuste R, Bonhoeffer T. Morphological changes in dendritic spines associated with long-term synaptic plasticity. Annu Rev Neurosci. 2001;24:1071–1089.
    1. Zaja-Milatovic S, Keene CD, Montine KS, Leverenz JB, Tsuang D, Montine TJ. Selective dendritic degeneration of medium spiny neurons in dementia with Lewy bodies. Neurology. 2006;66:1591–1593.
    1. Zaja-Milatovic S, Milatovic D, Schantz AM, et al. Dendritic degeneration in neostriatal medium spiny neurons in Parkinson disease. Neurology. 2005;64:545–547.
    1. Zarranz JJ, Alegre J, Gomez-Esteban JC, et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol. 2004;55:164–173.
    1. Zhong SC, Luo X, Chen XS, et al. Expression and subcellular location of alpha-synuclein during mouse-embryonic development. Cell Mol Neurobiol. 2010;30:469–482.

Source: PubMed

3
Abonnere