1,454
Views
17
CrossRef citations to date
0
Altmetric
Extra View

Proteolytic clearance of extracellular α-synuclein as a new therapeutic approach against Parkinson disease

&
Pages 121-126 | Received 16 Sep 2012, Accepted 11 Nov 2012, Published online: 15 Nov 2012

Abstract

Many neurodegenerative diseases such as Alzheimer disease and Parkinson disease show similar characteristics. They typically show deposits of protein aggregates, the formation of which is considered important in their pathogenesis. Recently, aggregation-prone proteins have been shown to spread between cells and so may contribute to the pathogenesis of diseases like prion disease. Such a pathogenesis pathway is possibly common to many neurodegenerative diseases. If confirmed, it could allow the development of therapeutic interventions against many such diseases. In Parkinson disease, α-synuclein, a major component of cytosolic protein inclusions named Lewy body, has been shown to be released and taken up by cells, which may facilitate its progressive pathological spreading between cells. Accordingly, inhibition of spreading by targeting extracellular α-synuclein may represent a new therapy against Parkinson disease. Research into the intercellular spreading of extracellular protein aggregations of α-synuclein and its clearance pathway are reviewed here with a focus on the proteolytic clearance pathway as a therapeutic target for the treatment of Parkinson disease. Considering the similar characteristics of aggregation-prone proteins, these clearance systems might allow treatment of other neurodegenerative diseases beyond Parkinson disease.

This article refers to:

Introduction

Age-related progressive neurodegenerative diseases such as Alzheimer (AD) and Parkinson (PD) diseases are increasingly affecting the world’s aging population. Despite much research, their pathogenesis still remains insufficiently understood to allow the rational design of therapeutic interventions that reduce their progression.

Interestingly, many neurodegenerative diseases involve protein aggregate inclusions, despite displaying different symptoms. PD shows cytosolic Lewy bodies or Lewy neurites composed of mainly α-synuclein (α-syn). AD shows extracellular senile plaques of mainly Aβ and cytosolic neurofibrillary tangles comprising mainly hyperphosphorylated tau. Moreover, polyglutamine diseases, Amyotropic Lateral Sclerosis (ALS) and prion disease show typical protein inclusions composed of mainly mutated polyglutamine expanded proteins, mutated superoxide dismutase (SOD)-1 and PrPsc, respectively.Citation1 Furthermore, the process of protein aggregate formation is considered to be significant in the pathogenesis of neurodegenerative diseases and aggregation-prone proteins are promising therapeutic targets for the treatment of such neurodegenerative diseases.

Recently, these aggregation-prone proteins have been observed to show unexpected yet interesting similar characteristics. Their regional and intercellular spreading has been observed irrespective of cytosolic or extracellular proteins and these processes are suspected to be significant in the pathogenesis of neurodegenerative diseases. Therefore, the inhibition of these proteins’ spreading may represent a new possible treatment of such diseases.

This review discusses the intercellular spreading of extracellular α-syn and the proteolytic clearance of extracellular α-syn as a new therapeutic target for PD. The characteristics of α-syn are compared with those of other aggregation-prone proteins implicated in neurodegenerative diseases.

The Relationship Between PD and α-Syn

PD is the second most common neurodegenerative disease after AD. It is characterized by a progressive dopaminergic neuronal loss in the substantia nigra pars compacta. Also characteristic is the presence of intracytoplasmic protein aggregates, Lewy bodies or Lewy neurites, as observed during other similar neurodegenerative diseases associated with aggregation-prone protein.Citation2

The intracytoplasmic protein aggregates observed in PD patients are mainly composed of α-syn, a small protein that is expressed abundantly in neuronal cells; it is localized mainly in the presynaptic nerve terminals.Citation3 Mutations of α-syn gene (A30P, E46K and A53T) and multiplications of the wild-type gene have been found to be associated with familial cases of early onset PD. Moreover, genetic variations in both the promoter and the region of the SNCA gene encoding the α-syn protein have been found to increase the susceptibility to PD.Citation4 Recent genome-wide association studies (GWASs) have identified variants of the SNCA gene that are coupled to increase PD susceptibility.Citation5,Citation6 Overall, a clear link has been found between this protein and idiopathic and familial PD. In support of these genetic studies, animal models with transgenic overexpression of α-syn have been shown to mimic several aspects of PD.Citation7 These observations in addition to several in vitro studies have firmly established the involvement of α-syn in the pathogenesis of PD.Citation7

Extracellular α-Syn and the Spreading of α-Syn Pathology Between Cells

α-Syn is normally considered a cytoplasmic protein and its function in cells’ cytoplasm has been received most attention. However, this view is beginning to change.

Monomeric and oligomeric α-syn have been continuously reported to be present in both healthy and diseased human cerebrospinal fluid (CSF) and blood plasma.Citation8-Citation10 In addition, α-syn and its aggregates have been shown to be secreted from neuronal cells by a non-classical exocytic pathwayCitation11 and its release is increased under various protein misfolding stress conditions.Citation12 α-Syn has also been reported to be secreted in a calcium-dependent manner by exosomes,Citation13 and lysosomal dysfunction increases exosome-mediated α-syn release.Citation14 These studies imply that extracellular α-syn is not only due to cellular leakage by cell death, but also its release from cells. This phenomenon is proposed to be probably part of a cellular quality control mechanism for the removal of damaged and harmful proteins by exocytosis.

Uptake of α-syn into cells has also been observed.Citation15-Citation18 Although the exact mechanism of α-syn uptake has not been established, 11-amino acid imperfect repeats found in the α-syn sequence have been shown to be critical.Citation16 α-Syn has been shown to be internalized via GM1 and hitherto-unknown protein receptors via a lipid raft-dependent endocytosis mechanism,Citation18 suggesting that cytosolic α-syn can be released from cells and taken up by other cells. In addition to studies of its release and uptake, extracellular α-syn has been reported to have effects on neurotoxicityCitation15,Citation19,Citation20 and inflammation.Citation14,Citation18,Citation21-Citation24

Recent observations that the transplants grafted into the brain of PD patients displayed Lewy bodiesCitation25,Citation26 were considered to be connected with Braak et al.’s proposal that Lewy body pathology spreads from one brain area to another according to a stereotypic pattern in specific stages.Citation27 Consequently, more recent in vitro and in vivo experimentsCitation28-Citation33 have shown that α-syn aggregates released from neuronal cells can be transferred to neighboring neurons and form Lewy body-like inclusions, providing a mechanistic basis for the spread of α-syn pathology in PD patients and a hypothesis that a prion-like mechanism may underlie the progression of PD.

Interestingly, other cytosolic proteins, such as tau, SOD-1 and polyglutamine expanding proteins, implicated in the pathogenesis of other neurodegenerative diseases have also been observed to have similar characteristics to α-syn.

Tau, a major component of cytosolic neurofibrillary tangles observed in AD, has been found in both healthy and AD CSF.Citation34,Citation35 It is secreted from cells by unconventional exocytosis or exosomeCitation36-Citation38 and its aggregates are taken up by cells via a hitherto-unknown mechanism.Citation39 In addition, extracellular tau has been shown to be toxic to cultured neuronal cells,Citation40 and prion-like intercellular spreading of tau aggregates has been reported both in vivo and in vitro.Citation41,Citation42

Native and misfolded SOD-1 have also been observed in the CSF of control and ALS patients.Citation43 Once secreted,Citation44,Citation45 it causes neurotoxicity and glial cell activation.Citation45,Citation46 Prion-like propagation of SOD-1 aggregates have also been reported both in vivo and in vitro.Citation47,Citation48

Polyglutamine peptide aggregates observed in polyglutamine diseases have also been reported to be internalized by cells and become co-sequestered in aggreasomes with cytosolic proteins,Citation49 implying the possibility of the intercellular spreading of polyglutamine aggregates.

Accordingly, the prion-like characteristics of aggregation-prone proteins responsible for many neurodegenerative diseases may be a common pathogenic mechanism and thus the reason for the recent growing interest (see reviews in refs. Citation1 and Citation50Citation54 for more details).

Proteolytic Clearance as a Therapeutic Approach Against PD and Other Diseases

Although this hypothesis needs further proof, it has led to the investigation of therapies that slow the progression of neurodegeneration by preventing the intercellular spreading of these proteins.

Preventing the spread of extracellular α-syn may be a suitable means of halting the progression of PD. The level of extracellular α-syn depends both on the rate of α-syn release from neuronal cells and the rate of its removal through various clearance pathways such as cell-mediated clearance, proteolytic degradation, chaperone-mediated clearance and active/passive transport out of the brain. Therefore, the treatment of PD could be achieved through targeting the regulation of α-syn release and uptake, or the removal of extracellular α-syn by a variety of clearance systems.

Proteolytic clearance is possible using any of several proteases that have been identified to be able to cleave and degrade α-syn: these include neurosin,Citation55 matrix metalloproteinases (MMPs),Citation56 calpain,Citation57 cathepsin DCitation58 and plasmin.Citation59 Among them, neurosin, MMPs and plasmin have been reported to cleave and degrade extracellular α-syn. Neurosin, a serine protease, is preferentially expressed in neurons and oligodendrocytes in the brain.Citation55 It was first observed to be colocalized in some senile plaques in AD patients as well as Lewy bodies in PD patients.Citation60 It has also been reported to degrade intracellular α-syn, but less efficiently A53T α-syn and also to inhibit α-syn polymerization.Citation55 Tatebe et al. later demonstrated in vitro that secreted neurosin degrades extracellular α-syn.Citation61 Recently, the viral mediated delivery of neurosin has been shown to promote the clearance of α-syn and reduces pathology in an α-syn model,Citation62 implying that neurosin may be a new therapeutic target for PD.

MMPs, particularly MMP-3, have also been reported to cleave extracellular α-syn.Citation56 Sung et al. demonstrated that oxidative injury induces the cleavage of extracellular α-syn released from neuronal cells and this is eventually cleaved by MMPs. However, the cleavage of α-syn by MMP-3 further induces its aggregation,Citation56 lessens any likely effects on PD of cleaving extracellular α-syn by MMPs.

The plasmin system may be a therapeutic target for preventing the intercellular spreading of extracellular α-syn. It is one of the proteases that can cleave extracellular α-syn.Citation59 Plasmin is an extracellular serine protease that is important in fibrinolysis. It is derived from its inactive form, plasminogen, by tissue type plasminogen activator (tPA) or urokinase plasminogen activator (uPA).Citation63 Although plasmin is synthesized mainly in the liver, it has also been detected in the CNS and is mainly expressed in neurons and astrocytes. Additionally, plasmin in the CNS is physiologically and pathologically important in such as neuronal development, synaptic plasticity and excitotoxicity through the cleaving of extracellular matrix components such as fibronectin, laminin and MMPs in addition to fibrin.Citation64-Citation66 Plasmin cleaves monomeric and further oligomeric and fibrillar forms of α-syn irrespective of the familial type of point mutation, unlike neurosin. However, tPA, uPA and thrombin do not cleave α-syn. Plasmin also inhibits the intercellular spreading of α-syn released from neuronal cells and glial activation by extracellular α-syn by cleaving the N-terminal region of α-syn into small fragments. This suggests that the plasmin system in the CNS may prevent the progression of PD through inhibiting extracellular α-syn’s detrimental intercellular spreading and glial activation.Citation59

Interestingly, plasmin can also degrade several forms of Aβ and block Aβ-induced toxicity, which contribute to the progression of AD,Citation67,Citation68 suggesting that it could also act against other neurodegenerative diseases besides PD. Overall, proteolytic enzymes such as neurosin and plasmin which cleave extracellular α-syn appear to be potential therapies against PD.

Proteolytic systems against Aβ are actively being studied for the treatment of AD. Several proteases including neprilysin, insulin degrading enzyme and MMPs as well as plasmin have been identified to cleave Aβ.Citation69-Citation72 Ex vivo gene delivery of neprilysin has been reported to reduce amyloid plaque burden in AD models.Citation73 Also, the inhibitor of PAI-1, which inhibits tPA activity and further plasmin activity, augments the activity of the plasmin system, thereby reducing the Aβ level and restoring memory deficit in AD models.Citation74 Consequently, several in vitro and in vivo studies have implicated amyloid degrading enzymes as new therapeutic targets against AD (see reviews in refs. Citation75Citation77 for more details). With regard to prion disease, cysteine proteases such as cathepsin B and L have been reported to degrade prions in CD11c+ dendritic cells and in GT1–1 neuronal cells.Citation78 Efforts to find proteases that can degrade PrPsc and further inhibit the amplification of pathologic effects of PrPsc are still ongoing.

In addition, dysregulation of these protease systems has also been observed to be associated with neurodegenerative diseases. In PD, reduced expression of neurosin has been observed in the brain of an animal model of PD and in patients with dementia with Lewy bodies.Citation62 Alterations of MMPs have also been observed in neurodegenerative diseases other than PD,Citation79 and extracellular α-syn has been reported to regulate the activity of MMPs.Citation22,Citation80,Citation81 Although the association between the plasmin system in the CNS and PD has yet to be established, the reduction of tPA activity by extracellular α-syn in primary astrocytes and microglia has been reported.Citation80 Extracellular α-syn has also been shown to increase PAI-1 expression in neurons, astrocytes and microglia and thus may inhibit plasmin activity,Citation59 suggesting that the plasmin system may be dysregulated in PD. Furthermore, the association between the plasmin system and other neurodegenerative diseases has been well reported. In AD, decreased tPA activity has been observed in AD models and its activity has been proposed to be controlled by a substantial increase of PAI-1.Citation82 Increased PAI-1 has been observed in APP transgenic miceCitation83 and in the CSF of AD patients.Citation84 Brain plasmin activity is also reduced in AD brains.Citation70 In prion disease, tPA accelerates the cleavage of prion protein by plasmin, implying that the plasmin system may be involved in the pathogenesis.Citation85 Dysregulation of neprilysin and insulin degrading enzyme, major Aβ degrading enzymes, has also been observed in AD.Citation86-Citation88 Therefore, dysregulation of the proteolytic clearance systems may be a common pathologic mechanism of neurodegenerative diseases beyond PD.

Other Clearance Systems as Therapeutic Approaches Against PD and Other Diseases

In addition to proteolytic degradation, other clearance pathways could represent potential therapeutic targets. Cell-mediated clearance pathways including endocytosis or phagocytosis have been reported to clear extracellular α-syn.Citation17 Immunization against α-syn can improve α-syn pathology, possibly due to increased cell-mediated clearance.Citation89,Citation90 As a chaperone-mediated clearance pathway, HSP70 was reported to reduce extracellular α-syn oligomer formation and related toxicity.Citation91

A variety of clearance pathways of Aβ in AD have been actively studied.Citation92 Microglia and astrocytes have been reported to be able to phagocytose Aβ and immunization with Aβ was shown to promote clearance,Citation93 which are currently considered the most effective therapeutic targets against AD.Citation94 Clusterin has also been reported to bind to Aβ and enhance Aβ clearance as a chaperone.Citation93 Passive immunization against prion has shown to decrease CNS pathology.Citation95 Immunization strategies against tau and mutant SOD-1 could also be used to treat AD and ALS, respectively.Citation96-Citation98 However, it remains uncertain whether their primary targets are cytosolic or extracellular proteins.

Concluding Remarks

Several common characteristic of neurodegenerative diseases are coming to be known. Precise understanding of these diseases’ pathogenesis could aid the development of common therapeutic interventions to stop their progression. In this sense, the spreading of aggregation-prone proteins, particularly cytosolic proteins such as α-syn, tau, SOD-1 and polyglutamine expanding proteins, into neighboring cells is potentially important in pathogenesis. Furthermore, interventions against their spreading could form the bases of new treatments. Accordingly, the proteolytic clearance system and other clearance systems which block the proteins’ spreading and hence their detrimental effects could serve as good targets for treatment. There may be more proteolytic enzymes responsible for the degradation of aggregation-prone proteins and efforts to find them and so regulate them directly or indirectly should aid the elucidation of the pathogenesis of many neurodegenerative diseases and hence help the development of therapeutic strategies of them.

Acknowledgments

This work was supported by the Basic Science Research Program (2010–0003623), the Mid-carrier Researcher Program (2011–0016603) and the MRC program (2012051428) through the National Research Foundation of Korea (NRF) funded by the Korean Ministry of Education, Science and Technology.

References

  • Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci 2010; 11:155 - 9; PMID: 20029438
  • Saiki S, Sato S, Hattori N. Molecular pathogenesis of Parkinson’s disease: update. J Neurol Neurosurg Psychiatry 2012; 83:430 - 6; http://dx.doi.org/10.1136/jnnp-2011-301205; PMID: 22138181
  • Surguchov A. Chapter 6 Molecular and Cellular Biology of Synucleins. 2008; 270:225-317.
  • Martin I, Dawson VL, Dawson TM. Recent advances in the genetics of Parkinson’s disease. Annu Rev Genomics Hum Genet 2011; 12:301 - 25; http://dx.doi.org/10.1146/annurev-genom-082410-101440; PMID: 21639795
  • Satake W, Nakabayashi Y, Mizuta I, Hirota Y, Ito C, Kubo M, et al. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson’s disease. Nat Genet 2009; 41:1303 - 7; http://dx.doi.org/10.1038/ng.485; PMID: 19915576
  • Simón-Sánchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet 2009; 41:1308 - 12; http://dx.doi.org/10.1038/ng.487; PMID: 19915575
  • Vekrellis K, Xilouri M, Emmanouilidou E, Rideout HJ, Stefanis L. Pathological roles of α-synuclein in neurological disorders. Lancet Neurol 2011; 10:1015 - 25; http://dx.doi.org/10.1016/S1474-4422(11)70213-7; PMID: 22014436
  • El-Agnaf OM, Salem SA, Paleologou KE, Cooper LJ, Fullwood NJ, Gibson MJ, et al. Alpha-synuclein implicated in Parkinson’s disease is present in extracellular biological fluids, including human plasma. FASEB J 2003; 17:1945 - 7; PMID: 14519670
  • Mollenhauer B, Locascio JJ, Schulz-Schaeffer W, Sixel-Döring F, Trenkwalder C, Schlossmacher MG. α-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: a cohort study. Lancet Neurol 2011; 10:230 - 40; http://dx.doi.org/10.1016/S1474-4422(11)70014-X; PMID: 21317042
  • Tokuda T, Qureshi MM, Ardah MT, Varghese S, Shehab SA, Kasai T, et al. Detection of elevated levels of α-synuclein oligomers in CSF from patients with Parkinson disease. Neurology 2010; 75:1766 - 72; http://dx.doi.org/10.1212/WNL.0b013e3181fd613b; PMID: 20962290
  • Lee HJ, Patel S, Lee SJ. Intravesicular localization and exocytosis of alpha-synuclein and its aggregates. J Neurosci 2005; 25:6016 - 24; http://dx.doi.org/10.1523/JNEUROSCI.0692-05.2005; PMID: 15976091
  • Jang A, Lee HJ, Suk JE, Jung JW, Kim KP, Lee SJ. Non-classical exocytosis of alpha-synuclein is sensitive to folding states and promoted under stress conditions. J Neurochem 2010; 113:1263 - 74; PMID: 20345754
  • Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, et al. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci 2010; 30:6838 - 51; http://dx.doi.org/10.1523/JNEUROSCI.5699-09.2010; PMID: 20484626
  • Alvarez-Erviti L, Couch Y, Richardson J, Cooper JM, Wood MJ. Alpha-synuclein release by neurons activates the inflammatory response in a microglial cell line. Neurosci Res 2011; 69:337 - 42; http://dx.doi.org/10.1016/j.neures.2010.12.020; PMID: 21255620
  • Sung JY, Kim J, Paik SR, Park JH, Ahn YS, Chung KC. Induction of neuronal cell death by Rab5A-dependent endocytosis of alpha-synuclein. J Biol Chem 2001; 276:27441 - 8; http://dx.doi.org/10.1074/jbc.M101318200; PMID: 11316809
  • Ahn KJ, Paik SR, Chung KC, Kim J. Amino acid sequence motifs and mechanistic features of the membrane translocation of alpha-synuclein. J Neurochem 2006; 97:265 - 79; http://dx.doi.org/10.1111/j.1471-4159.2006.03731.x; PMID: 16524375
  • 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 - 49; http://dx.doi.org/10.1016/j.biocel.2008.01.017; PMID: 18291704
  • Park JY, Kim KS, Lee SB, Ryu JS, Chung KC, Choo YK, et al. On the mechanism of internalization of alpha-synuclein into microglia: roles of ganglioside GM1 and lipid raft. J Neurochem 2009; 110:400 - 11; http://dx.doi.org/10.1111/j.1471-4159.2009.06150.x; PMID: 19457104
  • El-Agnaf OM, Jakes R, Curran MD, Middleton D, Ingenito R, Bianchi E, et al. Aggregates from mutant and wild-type alpha-synuclein proteins and NAC peptide induce apoptotic cell death in human neuroblastoma cells by formation of beta-sheet and amyloid-like filaments. FEBS Lett 1998; 440:71 - 5; http://dx.doi.org/10.1016/S0014-5793(98)01418-5; PMID: 9862428
  • Seo JH, Rah JC, Choi SH, Shin JK, Min K, Kim HS, et al. Alpha-synuclein regulates neuronal survival via Bcl-2 family expression and PI3/Akt kinase pathway. FASEB J 2002; 16:1826 - 8; PMID: 12223445
  • Klegeris A, Giasson BI, Zhang H, Maguire J, Pelech S, McGeer PL. Alpha-synuclein and its disease-causing mutants induce ICAM-1 and IL-6 in human astrocytes and astrocytoma cells. FASEB J 2006; 20:2000 - 8; http://dx.doi.org/10.1096/fj.06-6183com; PMID: 17012252
  • Lee EJ, Woo MS, Moon PG, Baek MC, Choi IY, Kim WK, et al. Alpha-synuclein activates microglia by inducing the expressions of matrix metalloproteinases and the subsequent activation of protease-activated receptor-1. J Immunol 2010; 185:615 - 23; http://dx.doi.org/10.4049/jimmunol.0903480; PMID: 20511551
  • Lee SB, Park SM, Ahn KJ, Chung KC, Paik SR, Kim J. Identification of the amino acid sequence motif of alpha-synuclein responsible for macrophage activation. Biochem Biophys Res Commun 2009; 381:39 - 43; http://dx.doi.org/10.1016/j.bbrc.2009.02.002; PMID: 19351591
  • Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, et al. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson’s disease. FASEB J 2005; 19:533 - 42; http://dx.doi.org/10.1096/fj.04-2751com; PMID: 15791003
  • 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 - 6; http://dx.doi.org/10.1038/nm1747; PMID: 18391962
  • Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med 2008; 14:501 - 3; http://dx.doi.org/10.1038/nm1746; PMID: 18391963
  • Braak H, Del Tredici K, Rüb 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; http://dx.doi.org/10.1016/S0197-4580(02)00065-9; PMID: 12498954
  • Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci U S A 2009; 106:13010 - 5; http://dx.doi.org/10.1073/pnas.0903691106; PMID: 19651612
  • Luk KC, Song C, O’Brien P, Stieber A, Branch JR, Brunden KR, et al. Exogenous alpha-synuclein fibrils seed the formation of Lewy body-like intracellular inclusions in cultured cells. Proc Natl Acad Sci U S A 2009; 106:20051 - 6; PMID: 19892735
  • Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, et al. Exogenous α-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 2011; 72:57 - 71; http://dx.doi.org/10.1016/j.neuron.2011.08.033; PMID: 21982369
  • Hansen C, Angot E, Bergström AL, Steiner JA, Pieri L, Paul G, et al. α-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J Clin Invest 2011; 121:715 - 25; http://dx.doi.org/10.1172/JCI43366; PMID: 21245577
  • Kordower JH, Dodiya HB, Kordower AM, Terpstra B, Paumier K, Madhavan L, et al. Transfer of host-derived α synuclein to grafted dopaminergic neurons in rat. Neurobiol Dis 2011; 43:552 - 7; http://dx.doi.org/10.1016/j.nbd.2011.05.001; PMID: 21600984
  • Mougenot AL, Nicot S, Bencsik A, Morignat E, Verchère J, Lakhdar L, et al. Prion-like acceleration of a synucleinopathy in a transgenic mouse model. Neurobiol Aging 2012; 33:2225 - 8; http://dx.doi.org/10.1016/j.neurobiolaging.2011.06.022; PMID: 21813214
  • Buerger K, Ewers M, Pirttilä T, Zinkowski R, Alafuzoff I, Teipel SJ, et al. CSF phosphorylated tau protein correlates with neocortical neurofibrillary pathology in Alzheimer’s disease. Brain 2006; 129:3035 - 41; http://dx.doi.org/10.1093/brain/awl269; PMID: 17012293
  • Vandermeeren M, Mercken M, Vanmechelen E, Six J, van de Voorde A, Martin JJ, et al. Detection of tau proteins in normal and Alzheimer’s disease cerebrospinal fluid with a sensitive sandwich enzyme-linked immunosorbent assay. J Neurochem 1993; 61:1828 - 34; http://dx.doi.org/10.1111/j.1471-4159.1993.tb09823.x; PMID: 8228996
  • Chai X, Dage JL, Citron M. Constitutive secretion of tau protein by an unconventional mechanism. Neurobiol Dis 2012; 48:356 - 66; http://dx.doi.org/10.1016/j.nbd.2012.05.021; PMID: 22668776
  • Saman S, Kim W, Raya M, Visnick Y, Miro S, Saman S, et al. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J Biol Chem 2012; 287:3842 - 9; http://dx.doi.org/10.1074/jbc.M111.277061; PMID: 22057275
  • Simón D, García-García E, Royo F, Falcón-Pérez JM, Avila J. Proteostasis of tau. Tau overexpression results in its secretion via membrane vesicles. FEBS Lett 2012; 586:47 - 54; http://dx.doi.org/10.1016/j.febslet.2011.11.022; PMID: 22138183
  • Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem 2009; 284:12845 - 52; http://dx.doi.org/10.1074/jbc.M808759200; PMID: 19282288
  • Gómez-Ramos A, Díaz-Hernández M, Cuadros R, Hernández F, Avila J. Extracellular tau is toxic to neuronal cells. FEBS Lett 2006; 580:4842 - 50; http://dx.doi.org/10.1016/j.febslet.2006.07.078; PMID: 16914144
  • Clavaguera F, Bolmont T, Crowther RA, Abramowski D, Frank S, Probst A, et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat Cell Biol 2009; 11:909 - 13; http://dx.doi.org/10.1038/ncb1901; PMID: 19503072
  • Guo JL, Lee VM. Seeding of normal Tau by pathological Tau conformers drives pathogenesis of Alzheimer-like tangles. J Biol Chem 2011; 286:15317 - 31; http://dx.doi.org/10.1074/jbc.M110.209296; PMID: 21372138
  • Zetterström P,, Andersen PM, Brännström T, Marklund SL. Misfolded superoxide dismutase-1 in CSF from amyotrophic lateral sclerosis patients. J Neurochem 2011; 117:91 - 9; http://dx.doi.org/10.1111/j.1471-4159.2011.07177.x; PMID: 21226712
  • Mondola P, Annella T, Santillo M, Santangelo F. Evidence for secretion of cytosolic CuZn superoxide dismutase by Hep G2 cells and human fibroblasts. Int J Biochem Cell Biol 1996; 28:677 - 81; http://dx.doi.org/10.1016/1357-2725(96)00004-0; PMID: 8673732
  • Urushitani M, Sik A, Sakurai T, Nukina N, Takahashi R, Julien JP. Chromogranin-mediated secretion of mutant superoxide dismutase proteins linked to amyotrophic lateral sclerosis. Nat Neurosci 2006; 9:108 - 18; http://dx.doi.org/10.1038/nn1603; PMID: 16369483
  • Zhao W, Beers DR, Henkel JS, Zhang W, Urushitani M, Julien JP, et al. Extracellular mutant SOD1 induces microglial-mediated motoneuron injury. Glia 2010; 58:231 - 43; http://dx.doi.org/10.1002/glia.20919; PMID: 19672969
  • Chia R, Tattum MH, Jones S, Collinge J, Fisher EM, Jackson GS. Superoxide dismutase 1 and tgSOD1 mouse spinal cord seed fibrils, suggesting a propagative cell death mechanism in amyotrophic lateral sclerosis. PLoS One 2010; 5:e10627; http://dx.doi.org/10.1371/journal.pone.0010627; PMID: 20498711
  • Münch C, O’Brien J, Bertolotti A. Prion-like propagation of mutant superoxide dismutase-1 misfolding in neuronal cells. Proc Natl Acad Sci U S A 2011; 108:3548 - 53; http://dx.doi.org/10.1073/pnas.1017275108; PMID: 21321227
  • Ren PH, Lauckner JE, Kachirskaia I, Heuser JE, Melki R, Kopito RR. Cytoplasmic penetration and persistent infection of mammalian cells by polyglutamine aggregates. Nat Cell Biol 2009; 11:219 - 25; http://dx.doi.org/10.1038/ncb1830; PMID: 19151706
  • Miller G. Neurodegeneration. Could they all be prion diseases?. Science 2009; 326:1337 - 9; http://dx.doi.org/10.1126/science.326.5958.1337; PMID: 19965731
  • Goedert M, Clavaguera F, Tolnay M. The propagation of prion-like protein inclusions in neurodegenerative diseases. Trends Neurosci 2010; 33:317 - 25; http://dx.doi.org/10.1016/j.tins.2010.04.003; PMID: 20493564
  • Lee SJ, Desplats P, Sigurdson C, Tsigelny I, Masliah E. Cell-to-cell transmission of non-prion protein aggregates. Nat Rev Neurol 2010; 6:702 - 6; http://dx.doi.org/10.1038/nrneurol.2010.145; PMID: 21045796
  • Krammer C, Schätzl HM, Vorberg I. Prion-like propagation of cytosolic protein aggregates: insights from cell culture models. Prion 2009; 3:206 - 12; http://dx.doi.org/10.4161/pri.3.4.10013; PMID: 19901539
  • Aguzzi A, Rajendran L. The transcellular spread of cytosolic amyloids, prions and prionoids. Neuron 2009; 64:783 - 90; http://dx.doi.org/10.1016/j.neuron.2009.12.016; PMID: 20064386
  • Iwata A, Maruyama M, Akagi T, Hashikawa T, Kanazawa I, Tsuji S, et al. Alpha-synuclein degradation by serine protease neurosin: implication for pathogenesis of synucleinopathies. Hum Mol Genet 2003; 12:2625 - 35; http://dx.doi.org/10.1093/hmg/ddg283; PMID: 12928483
  • Sung JY, Park SM, Lee CH, Um JW, Lee HJ, Kim J, et al. Proteolytic cleavage of extracellular secreted alpha-synuclein via matrix metalloproteinases. J Biol Chem 2005; 280:25216 - 24; http://dx.doi.org/10.1074/jbc.M503341200; PMID: 15863497
  • Mishizen-Eberz AJ, Norris EH, Giasson BI, Hodara R, Ischiropoulos H, Lee VM, et al. Cleavage of alpha-synuclein by calpain: potential role in degradation of fibrillized and nitrated species of alpha-synuclein. Biochemistry 2005; 44:7818 - 29; http://dx.doi.org/10.1021/bi047846q; PMID: 15909996
  • Sevlever D, Jiang P, Yen SH. Cathepsin D is the main lysosomal enzyme involved in the degradation of alpha-synuclein and generation of its carboxy-terminally truncated species. Biochemistry 2008; 47:9678 - 87; http://dx.doi.org/10.1021/bi800699v; PMID: 18702517
  • Kim KS, Choi YR, Park JY, Lee JH, Kim DK, Lee SJ, et al. Proteolytic cleavage of extracellular α-synuclein by plasmin: implications for Parkinson disease. J Biol Chem 2012; 287:24862 - 72; http://dx.doi.org/10.1074/jbc.M112.348128; PMID: 22619171
  • Ogawa K, Yamada T, Tsujioka Y, Taguchi J, Takahashi M, Tsuboi Y, et al. Localization of a novel type trypsin-like serine protease, neurosin, in brain tissues of Alzheimer’s disease and Parkinson’s disease. Psychiatry Clin Neurosci 2000; 54:419 - 26; http://dx.doi.org/10.1046/j.1440-1819.2000.00731.x; PMID: 10997858
  • Tatebe H, Watanabe Y, Kasai T, Mizuno T, Nakagawa M, Tanaka M, et al. Extracellular neurosin degrades α-synuclein in cultured cells. Neurosci Res 2010; 67:341 - 6; http://dx.doi.org/10.1016/j.neures.2010.04.008; PMID: 20403393
  • Spencer B, Michael S, Shen J, Kosberg K, Rockenstein E, Patrick C, et al. Lentivirus Mediated Delivery of Neurosin Promotes Clearance of Wild-type α-Synuclein and Reduces the Pathology in an α-Synuclein Model of LBD. Mol Ther 2012; http://dx.doi.org/10.1038/mt.2012.66; PMID: 22508489
  • Schaller J, Gerber SS. The plasmin-antiplasmin system: structural and functional aspects. Cell Mol Life Sci 2011; 68:785 - 801; http://dx.doi.org/10.1007/s00018-010-0566-5; PMID: 21136135
  • Dotti CG, Galvan C, Ledesma MD. Plasmin deficiency in Alzheimer’s disease brains: causal or casual?. Neurodegener Dis 2004; 1:205 - 12; http://dx.doi.org/10.1159/000080987; PMID: 16908991
  • Syrovets T, Simmet T. Novel aspects and new roles for the serine protease plasmin. Cell Mol Life Sci 2004; 61:873 - 85; http://dx.doi.org/10.1007/s00018-003-3348-5; PMID: 15095009
  • Sheehan JJ, Tsirka SE. Fibrin-modifying serine proteases thrombin, tPA and plasmin in ischemic stroke: a review. Glia 2005; 50:340 - 50; http://dx.doi.org/10.1002/glia.20150; PMID: 15846799
  • Van Nostrand WE, Porter M. Plasmin cleavage of the amyloid beta-protein: alteration of secondary structure and stimulation of tissue plasminogen activator activity. Biochemistry 1999; 38:11570 - 6; http://dx.doi.org/10.1021/bi990610f; PMID: 10471309
  • Tucker HM, Kihiko M, Caldwell JN, Wright S, Kawarabayashi T, Price D, et al. The plasmin system is induced by and degrades amyloid-beta aggregates. J Neurosci 2000; 20:3937 - 46; PMID: 10818128
  • Iwata N, Tsubuki S, Takaki Y, Watanabe K, Sekiguchi M, Hosoki E, et al. Identification of the major Abeta1-42-degrading catabolic pathway in brain parenchyma: suppression leads to biochemical and pathological deposition. Nat Med 2000; 6:143 - 50; http://dx.doi.org/10.1038/77399; PMID: 10655101
  • Ledesma MD, Da Silva JS, Crassaerts K, Delacourte A, De Strooper B, Dotti CG. Brain plasmin enhances APP alpha-cleavage and Abeta degradation and is reduced in Alzheimer’s disease brains. EMBO Rep 2000; 1:530 - 5; PMID: 11263499
  • Backstrom JR, Lim GP, Cullen MJ, Tökés ZA. Matrix metalloproteinase-9 (MMP-9) is synthesized in neurons of the human hippocampus and is capable of degrading the amyloid-beta peptide (1-40). J Neurosci 1996; 16:7910 - 9; PMID: 8987819
  • Qiu WQ, Walsh DM, Ye Z, Vekrellis K, Zhang J, Podlisny MB, et al. Insulin-degrading enzyme regulates extracellular levels of amyloid beta-protein by degradation. J Biol Chem 1998; 273:32730 - 8; http://dx.doi.org/10.1074/jbc.273.49.32730; PMID: 9830016
  • Hemming ML, Patterson M, Reske-Nielsen C, Lin L, Isacson O, Selkoe DJ. Reducing amyloid plaque burden via ex vivo gene delivery of an Abeta-degrading protease: a novel therapeutic approach to Alzheimer disease. PLoS Med 2007; 4:e262; http://dx.doi.org/10.1371/journal.pmed.0040262; PMID: 17760499
  • Jacobsen JS, Comery TA, Martone RL, Elokdah H, Crandall DL, Oganesian A, et al. Enhanced clearance of Abeta in brain by sustaining the plasmin proteolysis cascade. Proc Natl Acad Sci U S A 2008; 105:8754 - 9; http://dx.doi.org/10.1073/pnas.0710823105; PMID: 18559859
  • Leissring MA. The AbetaCs of Abeta-cleaving proteases. J Biol Chem 2008; 283:29645 - 9; http://dx.doi.org/10.1074/jbc.R800022200; PMID: 18723506
  • Miners JS, Barua N, Kehoe PG, Gill S, Love S. Aβ-degrading enzymes: potential for treatment of Alzheimer disease. J Neuropathol Exp Neurol 2011; 70:944 - 59; http://dx.doi.org/10.1097/NEN.0b013e3182345e46; PMID: 22002425
  • Nalivaeva NN, Beckett C, Belyaev ND, Turner AJ. Are amyloid-degrading enzymes viable therapeutic targets in Alzheimer’s disease?. J Neurochem 2012; 120:Suppl 1 167 - 85; http://dx.doi.org/10.1111/j.1471-4159.2011.07510.x; PMID: 22122230
  • Luhr KM, Nordström EK, Löw P, Ljunggren HG, Taraboulos A, Kristensson K. Scrapie protein degradation by cysteine proteases in CD11c+ dendritic cells and GT1-1 neuronal cells. J Virol 2004; 78:4776 - 82; http://dx.doi.org/10.1128/JVI.78.9.4776-4782.2004; PMID: 15078959
  • Kim EM, Hwang O. Role of matrix metalloproteinase-3 in neurodegeneration. J Neurochem 2011; 116:22 - 32; http://dx.doi.org/10.1111/j.1471-4159.2010.07082.x; PMID: 21044079
  • Joo SH, Kwon KJ, Kim JW, Kim JW, Hasan MR, Lee HJ, et al. Regulation of matrix metalloproteinase-9 and tissue plasminogen activator activity by alpha-synuclein in rat primary glial cells. Neurosci Lett 2010; 469:352 - 6; http://dx.doi.org/10.1016/j.neulet.2009.12.026; PMID: 20026244
  • Kim S, Cho SH, Kim KY, Shin KY, Kim HS, Park CH, et al. Alpha-synuclein induces migration of BV-2 microglial cells by up-regulation of CD44 and MT1-MMP. J Neurochem 2009; 109:1483 - 96; http://dx.doi.org/10.1111/j.1471-4159.2009.06075.x; PMID: 19457162
  • Melchor JP, Pawlak R, Strickland S. The tissue plasminogen activator-plasminogen proteolytic cascade accelerates amyloid-beta (Abeta) degradation and inhibits Abeta-induced neurodegeneration. J Neurosci 2003; 23:8867 - 71; PMID: 14523088
  • Cacquevel M, Launay S, Castel H, Benchenane K, Chéenne S, Buée L, et al. Ageing and amyloid-beta peptide deposition contribute to an impaired brain tissue plasminogen activator activity by different mechanisms. Neurobiol Dis 2007; 27:164 - 73; http://dx.doi.org/10.1016/j.nbd.2007.04.004; PMID: 17566751
  • Sutton R, Keohane ME, VanderBerg SR, Gonias SL. Plasminogen activator inhibitor-1 in the cerebrospinal fluid as an index of neurological disease. Blood Coagul Fibrinolysis 1994; 5:167 - 71; http://dx.doi.org/10.1097/00001721-199404000-00002; PMID: 8054448
  • Kornblatt JA, Marchal S, Rezaei H, Kornblatt MJ, Balny C, Lange R, et al. The fate of the prion protein in the prion/plasminogen complex. Biochem Biophys Res Commun 2003; 305:518 - 22; http://dx.doi.org/10.1016/S0006-291X(03)00804-0; PMID: 12763023
  • Apelt J, Ach K, Schliebs R. Aging-related down-regulation of neprilysin, a putative beta-amyloid-degrading enzyme, in transgenic Tg2576 Alzheimer-like mouse brain is accompanied by an astroglial upregulation in the vicinity of beta-amyloid plaques. Neurosci Lett 2003; 339:183 - 6; http://dx.doi.org/10.1016/S0304-3940(03)00030-2; PMID: 12633883
  • Ferretti MT, Partridge V, Leon WC, Canneva F, Allard S, Arvanitis DN, et al. Transgenic mice as a model of pre-clinical Alzheimer’s disease. Curr Alzheimer Res 2011; 8:4 - 23; http://dx.doi.org/10.2174/156720511794604561; PMID: 21143159
  • Nalivaevaa NN, Fisk L, Kochkina EG, Plesneva SA, Zhuravin IA, Babusikova E, et al. Effect of hypoxia/ischemia and hypoxic preconditioning/reperfusion on expression of some amyloid-degrading enzymes. Ann N Y Acad Sci 2004; 1035:21 - 33; http://dx.doi.org/10.1196/annals.1332.002; PMID: 15681798
  • Masliah E, Rockenstein E, Adame A, Alford M, Crews L, Hashimoto M, et al. Effects of alpha-synuclein immunization in a mouse model of Parkinson’s disease. Neuron 2005; 46:857 - 68; http://dx.doi.org/10.1016/j.neuron.2005.05.010; PMID: 15953415
  • Masliah E, Rockenstein E, Mante M, Crews L, Spencer B, Adame A, et al. Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease. PLoS One 2011; 6:e19338; http://dx.doi.org/10.1371/journal.pone.0019338; PMID: 21559417
  • Danzer KM, Ruf WP, Putcha P, Joyner D, Hashimoto T, Glabe C, et al. Heat-shock protein 70 modulates toxic extracellular α-synuclein oligomers and rescues trans-synaptic toxicity. FASEB J 2011; 25:326 - 36; http://dx.doi.org/10.1096/fj.10-164624; PMID: 20876215
  • Kurz A, Perneczky R. Amyloid clearance as a treatment target against Alzheimer’s disease. J Alzheimers Dis 2011; 24:Suppl 2 61 - 73; PMID: 21422524
  • Sokolowski JD, Mandell JW. Phagocytic clearance in neurodegeneration. Am J Pathol 2011; 178:1416 - 28; http://dx.doi.org/10.1016/j.ajpath.2010.12.051; PMID: 21435432
  • Lemere CA, Masliah E. Can Alzheimer disease be prevented by amyloid-beta immunotherapy?. Nat Rev Neurol 2010; 6:108 - 19; http://dx.doi.org/10.1038/nrneurol.2009.219; PMID: 20140000
  • Sadowski MJ, Pankiewicz J, Prelli F, Scholtzova H, Spinner DS, Kascsak RB, et al. Anti-PrP Mab 6D11 suppresses PrP(Sc) replication in prion infected myeloid precursor line FDC-P1/22L and in the lymphoreticular system in vivo. Neurobiol Dis 2009; 34:267 - 78; http://dx.doi.org/10.1016/j.nbd.2009.01.013; PMID: 19385058
  • Götz J, Ittner A, Ittner LM. Tau-targeted treatment strategies in Alzheimer’s disease. Br J Pharmacol 2012; 165:1246 - 59; http://dx.doi.org/10.1111/j.1476-5381.2011.01713.x; PMID: 22044248
  • Gros-Louis F, Soucy G, Larivière R, Julien JP. Intracerebroventricular infusion of monoclonal antibody or its derived Fab fragment against misfolded forms of SOD1 mutant delays mortality in a mouse model of ALS. J Neurochem 2010; 113:1188 - 99; PMID: 20345765
  • Urushitani M, Ezzi SA, Julien JP. Therapeutic effects of immunization with mutant superoxide dismutase in mice models of amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A 2007; 104:2495 - 500; http://dx.doi.org/10.1073/pnas.0606201104; PMID: 17277077

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.