55
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Extracellular Dopamine Potentiates Mn-Induced Oxidative Stress, Lifespan Reduction, and Dopaminergic Neurodegeneration in a BLI-3–Dependent Manner in Caenorhabditis elegans

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Parkinson's disease (PD)-mimicking drugs and pesticides, and more recently PD-associated gene mutations, have been studied in cell cultures and mammalian models to decipher the molecular basis of PD. Thus far, a dozen of genes have been identified that are responsible for inherited PD. However they only account for about 8% of PD cases, most of the cases likely involving environmental contributions. Environmental manganese (Mn) exposure represents an established risk factor for PD occurrence, and both PD and Mn-intoxicated patients display a characteristic extrapyramidal syndrome primarily involving dopaminergic (DAergic) neurodegeneration with shared common molecular mechanisms. To better understand the specificity of DAergic neurodegeneration, we studied Mn toxicity in vivo in Caenorhabditis elegans. Combining genetics and biochemical assays, we established that extracellular, and not intracellular, dopamine (DA) is responsible for Mn-induced DAergic neurodegeneration and that this process (1) requires functional DA-reuptake transporter (DAT-1) and (2) is associated with oxidative stress and lifespan reduction. Overexpression of the anti-oxidant transcription factor, SKN-1, affords protection against Mn toxicity, while the DA-dependency of Mn toxicity requires the NADPH dual-oxidase BLI-3. These results suggest that in vivo BLI-3 activity promotes the conversion of extracellular DA into toxic reactive species, which, in turn, can be taken up by DAT-1 in DAergic neurons, thus leading to oxidative stress and cell degeneration.

          Author Summary

          In Parkinson's disease (PD), motor neurons that produce dopamine degenerate, leading to a characteristic syndrome including tremor, rigidity, and bradykinesia. The mechanisms leading to PD have been under intense investigation, identifying hereditary mutations responsible for about 8% of the cases. However, multiple environmental factors contribute to PD; and, amongst those, manganese (Mn) exposure from pesticides, industrial fumes, and gasoline additives has been robustly associated with PD. To gain insights into processes leading to the specific degeneration of dopaminergic neurons, we used a simple animal model, the nematode Caenorhabditis elegans, which, upon Mn exposure, recapitulates key molecular processes known to be involved in PD. Combining biochemistry and genetics, we demonstrate that dopamine secreted by the neurons and not intracellular dopamine is directly involved in the generation of toxic reactive oxygen species. We identify two essential mediators of this dopamine-dependent effect which are an extracellularly active enzyme called dual-oxidase and the dopamine re-uptake transporter. We also reveal that a transcription factor which is strongly expressed in two neurons involved in the regulation of aging is a powerful modulator of the dopamine-dependent toxicity. Our study establishes novel evidence of the link among PD, aging, and oxidative stress within the context of exposure to Mn.

          Related collections

          Most cited references154

          • Record: found
          • Abstract: found
          • Article: not found

          Mechanism of toxicity in rotenone models of Parkinson's disease.

          Exposure of rats to the pesticide and complex I inhibitor rotenone reproduces features of Parkinson's disease, including selective nigrostriatal dopaminergic degeneration and alpha-synuclein-positive cytoplasmic inclusions (Betarbet et al., 2000; Sherer et al., 2003). Here, we examined mechanisms of rotenone toxicity using three model systems. In SK-N-MC human neuroblastoma cells, rotenone (10 nm to 1 microm) caused dose-dependent ATP depletion, oxidative damage, and death. To determine the molecular site of action of rotenone, cells were transfected with the rotenone-insensitive single-subunit NADH dehydrogenase of Saccharomyces cerevisiae (NDI1), which incorporates into the mammalian ETC and acts as a "replacement" for endogenous complex I. In response to rotenone, NDI1-transfected cells did not show mitochondrial impairment, oxidative damage, or death, demonstrating that these effects of rotenone were caused by specific interactions at complex I. Although rotenone caused modest ATP depletion, equivalent ATP loss induced by 2-deoxyglucose was without toxicity, arguing that bioenergetic defects were not responsible for cell death. In contrast, reducing oxidative damage with antioxidants, or by NDI1 transfection, blocked cell death. To determine the relevance of rotenone-induced oxidative damage to dopaminergic neuronal death, we used a chronic midbrain slice culture model. In this system, rotenone caused oxidative damage and dopaminergic neuronal loss, effects blocked by alpha-tocopherol. Finally, brains from rotenone-treated animals demonstrated oxidative damage, most notably in midbrain and olfactory bulb, dopaminergic regions affected by Parkinson's disease. These results, using three models of increasing complexity, demonstrate the involvement of oxidative damage in rotenone toxicity and support the evaluation of antioxidant therapies for Parkinson's disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Alpha-synuclein is part of a diverse and highly conserved interaction network that includes PARK9 and manganese toxicity.

            Parkinson's disease (PD), dementia with Lewy bodies and multiple system atrophy, collectively referred to as synucleinopathies, are associated with a diverse group of genetic and environmental susceptibilities. The best studied of these is PD. alpha-Synuclein (alpha-syn) has a key role in the pathogenesis of both familial and sporadic PD, but evidence linking it to other predisposition factors is limited. Here we report a strong genetic interaction between alpha-syn and the yeast ortholog of the PD-linked gene ATP13A2 (also known as PARK9). Dopaminergic neuron loss caused by alpha-syn overexpression in animal and neuronal PD models is rescued by coexpression of PARK9. Further, knockdown of the ATP13A2 ortholog in Caenorhabditis elegans enhances alpha-syn misfolding. These data provide a direct functional connection between alpha-syn and another PD susceptibility locus. Manganese exposure is an environmental risk factor linked to PD and PD-like syndromes. We discovered that yeast PARK9 helps to protect cells from manganese toxicity, revealing a connection between PD genetics (alpha-syn and PARK9) and an environmental risk factor (PARK9 and manganese). Finally, we show that additional genes from our yeast screen, with diverse functions, are potent modifiers of alpha-syn-induced neuron loss in animals, establishing a diverse, highly conserved interaction network for alpha-syn.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              C. elegans Model Identifies Genetic Modifiers of α-Synuclein Inclusion Formation During Aging

              Introduction Sporadic as well as familial Parkinson's disease are characterized by protein inclusions in the brain containing α-synuclein [1]. Similar inclusions are also present in other neurodegenerative diseases, including dementia with Lewy bodies [2]. The α-synuclein gene is causatively related to Parkinson's disease, since mutations in the gene, and duplication or triplication of the α-synuclein locus cause familial forms of Parkinson's disease in humans [3]–[5]. Sporadic Parkinson's disease, seen in 1–4% of the population over 65 years of age, appears to be unrelated to mutations or multiplications of the α-synuclein locus. How α-synuclein inclusions are produced is unknown, but identifying cellular factors and processes involved in the formation of these inclusions may provide some understanding of the molecular cause of Parkinson's disease and of the link between aging and the sporadic form of the disease. To study pathological α-synuclein accumulation, we used a genetic model organism, the nematode Caenorhabditis elegans. We chose C. elegans for its thoroughly characterized aging properties, its amenability to genome-wide RNAi screening, and its transparency throughout its lifetime, which allows visualization of inclusions in living animals during aging. We expressed human α-synuclein fused to yellow fluorescent protein in the body wall muscle of C. elegans, where it, age-dependently, accumulated into inclusions. In old age these inclusions contained aggregated material, similar to human pathological inclusions. We used a genome-wide RNAi screen to identify genes and cellular processes involved in age-related α-synuclein accumulation in inclusions. Results/Discussion To visually trace expression of α-synuclein, we expressed human α-synuclein fused to yellow fluorescent protein (YFP) in C. elegans under control of the unc-54 promoter, which drives expression to the body wall muscle cells. Muscle expression rather than neuronal expression was chosen for several reasons. The unc-54 promoter is strong and muscle cells are large, allowing for visual detection of α-synuclein expression and its subcellular localization. Furthermore, RNAi by feeding seems to work more efficiently in muscles than in neurons, which better allows for genome-wide RNAi screening. Finally, muscle expression has been used successfully to model protein-misfolding diseases and to identify modifier genes in previous studies [6]–[8]. The α-synuclein-YFP chimaeric protein is recognized by an antibody specific for human α-synuclein and an antibody for YFP (Figure 1B). YFP fused to human α-synuclein relocates to inclusions (Figure 1A), which are visible as early as day 2 after hatching and increase in number and size during the animals' aging up to late adulthood. As YFP alone remains diffusely localized throughout aging, this indicates that relocation of α-synuclein-YFP into foci is caused by intrinsic properties of the α-synuclein protein. 10.1371/journal.pgen.1000027.g001 Figure 1 Α-synuclein-YFP in Transgenic Animals Relocalizes to Discrete Inclusions during Aging. (A) Confocal laser scanning images showing α-synuclein-YFP expression in the head region of transgenic C. elegans during aging. (B) Immunoblotting analysis of SDS/PAGE separated protein extracts from α-synuclein-YFP, N2 (wt) and YFP animals using α-synuclein (LB509) and YFP (anti-GFP) antibodies. Loading control is α-actin. (C) Immunoblotting analysis of protein extracts from 3-, 5-, 11- and 17-day old α-synuclein-YFP synchronized animals using anti-α- synuclein antibody. One of the characteristics of late inclusions in the brains of Parkinson's patients is the presence of electron-dense filamentous and granular protein material, which is typical for aggregated protein [9]. To address whether α-synuclein was aggregated within the inclusions in our C. elegans model, we measured the mobility of the α-synuclein-YFP chimaera by fluorescence recovery after photo bleaching (FRAP) [10]. We observed two types of inclusions. One type contained mostly mobile material (Figure 2P–2T, 2W; ∼80% fluorescence recovery), whereas the other type contained immobilized material (Figure 2K-2O, 2X; ∼40% fluorescence recovery), similar to Q40- YFP aggregates (Figure 2F-2J, V; ∼30% fluorescence recovery), indicating aggregated protein, a characteristic of α-synuclein deposits in Parkinson's disease. 10.1371/journal.pgen.1000027.g002 Figure 2 Fluorescent Recovery after Photo Bleaching Reveals α-Synuclein Inclusions Contain Mobile as well as Immobilized Protein Material. (A,F,K,P) Images of YFP, Q40-YFP and α-synuclein-YFP transgenic animals. (B-E,G-J, L-O,Q-T) High magnification images of the area indicated (red box) before photo bleaching and during recovery. (U-X) Graphical representation of fluorescence recovery after photo bleaching in (B-E, G-J, L-O, Q-T). Relative fluorescence intensity (RFI) value on y-axis represents percentage fluorescence corrected for background bleaching. (Y) Average number of inclusions larger than ∼2 µm2 per animal between tip of the nose and pharyngeal bulb during aging (n = 9 for day 11, n = 10 for days 9, 13, 15 and 17). (Z) Percentage of foci containing immobile material during aging. Bar in d-g represents 50 µm (overview) and 5 µm in higher magnification images. Error bars in (Y) indicate standard deviation. Notably, there was an increase in the number of “immobile” inclusions relative to “mobile” inclusions during aging (Figure 2Y and 2Z), which appeared unrelated to the expression level of α- synuclein-YFP (Figure 1C). Interestingly, immobile inclusions were not observed before late adulthood, which is consistent with age-related aggregation in Parkinson's disease patients [11]. We noted that the motility of the mobile material contained in inclusions was similar at all ages, suggesting that aggregation was not a gradual process but caused by a sharp transition from mobile to immobile material (data not shown). Taken together, the time-dependent accumulation and immobilization of α-synuclein in inclusions verify the suitability of our α-synuclein-YFP C. elegans system as a model for human Parkinson's disease. To identify processes involved in inclusion formation we searched for genes that, when inactivated, increased the amount of inclusions using a genome-wide RNAi screen. Worms were screened by visual inspection using fluorescence microscopy at days 4 and 5 after transfer of synchronized, first stage (L1) larvae to RNAi clones. Clones were scored positive when at least five out of the first ten worms screened showed an increase in the amount of inclusions (Figure 3). Genes for which RNAi results were confirmed in duplicate, in liquid culture and on agar plates, were considered suppressors of inclusion formation. 10.1371/journal.pgen.1000027.g003 Figure 3 Suppressors of Inclusion Formation Identified by RNAi. (A) Confocal images showing head region of α-synuclein-YFP transgenic animals fed on OP50 bacteria, bacteria containing L4440 (empty vector) and expressing double stranded RNA targeting two representative genes (F26H11.4 and Y48G1A.6) found to increase inclusion formation. Phenotypes of increased inclusion formation were analyzed in liquid culture by observing at least five out of the first ten animals screened to show an increased presence of inclusions compared to wild type. Scale bar represents 50 µm. (B) Quantification of the number of inclusions present in worms as shown in (A) (n = 2). In this screen we found 80 suppressors of inclusion formation (Table 1). Forty-nine of these genes have an established human ortholog (Blast E-value ≤2.5e-5), indicating involvement of these genes in molecular pathways conserved between humans and nematodes (Table S1 online). The effect of RNAi was confirmed in genetic deletion strains for three genes: sir-2.1, an NAD+-dependent protein deacetylase, lagr-1, a sphingolipid synthase, and ymel-1, a mitochondrial protease, which is an ortholog of the human presenilin associated metalloprotease (PAMP) (Figure 4). 10.1371/journal.pgen.1000027.g004 Figure 4 Sir-2.1, ymel-1 and lagr-1 Are Suppressors of α-Synuclein Inclusion Formation. (A,B) Confocal images showing α-synuclein-YFP transgenic animals and the transgenic strains containing a deletion in the sir-2.1gene (sir-2.1(ok434)) on day 9. (C) Average number of inclusions between tip of the head and pharyngeal bulb of the worm (n = 8). *P≤0.025 (Student's t test). (D, E, F) Confocal images showing α-synuclein-YFP transgenic worms and the transgenic strains containing a deletion in the ymel-1and lagr-1 gene (ymel-1(tm1920) and lagr-1(gk331)), on day 9. (G) Average number of inclusions between tip and pharyngeal bulb of the worm (n = 8 (wt and ymel-1), n = 7 (lagr-1)). *P≤0.05, **P≤0.05 (Student's t test). 10.1371/journal.pgen.1000027.t001 Table 1 Suppressors of α-Synuclein Inclusion Formation. Function Cosmid no. Gene Description Strength CS R05H10.6 cdh-7 FAT tumor suppressor homolog 3 + CS Y54E2A.2 Similar to Ca2+-binding actin-bundling protein (spectrin) + DR E03A3.2 rcq-5 DEAD/DEAH box helicase ++ DR F32D1.10 mcm-7 DNA-replication licensing factor + DR W02D9.1 pri-2 Eukaryotic-type DNA primase, large subunit ++ ECM F41F3.4 col-139 Structural component of the cuticle, phosphate transport + EM C07A9.8 Bestrophin, anion channel ++ EM C28H8.11 Tryptophan 2,3-dioxygenase ++ EM R03E1.2 Renin-precursor, lysosomal ATPase H+ transporter ++ EM T14F9.1 vha-15 (phi-52) ATPase coupled proton transport, Vacuolar ATP synthase sub + EM Y37H9A.6 ndx-4 NUDIX hydrolase + ET B0213.12 cyp-34A7 Cytochrome P450 2B6 + ET W01B11.2 sulp-6 STAS domain, Sulfate transporter family ++ ET W01B11.6 Thioredoxin, Yeast: required for ER-Golgi transport, stress protection ++ GM F29F11.2 UDP-glucuronosyltransferase 1–8 precursor ++ GM K07A3.1 fbp-1(K07A3.b) Fructose-1-6-bisphosphatase + LM C28C12.7 spp-10 sphingolipid metabolism/lysosomal ++ LM F28H1.4 Membrane-associating domain, Plasmolipin ++ LM F54F11.1 Lipolytic enzyme, putative phospholipase ++ LM K09H9.6 lpd-6 RNA-binding protein required for 60S ribosomal subunit biogenesis + LM T08H10.1 Aldo-keto reductase family 1 member B10, oxido-reductase + LM W03G9.6 paf-1 acetylhydrolase/phospholipase A2 ++ LM Y48G9A.10 Carnitine O-palmitoyltransferase I ++ LM Y6B3B.10 lagr-1 LAG1, (dihydro)ceramide synthase, spingholipid synthesis, ER ++ PD C46F9.3 math-24 MATH domain, Ubiquitin carboxyl-terminal hydrolase 7 ++ PD C47B2.1 F-box domain + PD F32H2.7 E3 ubiquitin-protein ligase HECTD1 + PD F49B2.6 Peptidase family M1 + PD M03C11.5 ymel-1 Peptidase M41, FtsH, metallo protease: unfolded mito-proteins ++ PD R09B3.4 ubc-12 Ub-conj. enzyme (E2), NEDD8-conj. enzyme NCE2 + PD R151.6 Human Derlin-2, protein degradation in ER + PD T06A4.1 Peptidase M14, carboxypeptidase A, Zinc-metalloprotease + PD Y63D3A.9 fbxb-93 F-box + PF F52C12.2 Uncharacterized conserved protein + PF R151.7 Hsp90 protein ++ PS F42A10.4 efk-1 Ca/calmodulin-dep. kinase, Elongation factor 2 kinase + PS F56H6.9 Protein of unknown function, DUF288 + PS Y67D8A_370.a puf-4 Translational repression, Splice Isoform 2 of Pumilio homolog 2 ++ RSP C04F5.5 srab-2 7TM chemoreceptor, GPCR activity, DNA biding/Zn-finger + RSP C28H8.5 ShTK domain ++ RSP D2089.1 rsp-7 Splicing factor, arginine/serine-rich ++ RSP F26H11.4 Neurofilament triplet H protein ++ RSP T12A2.3 Protein AF-9 ++ RSP Y113G7B.18 mdt-17 Transcriptional cofactor + RSP Y116A8C.35 uaf-2 Splicing factor U2AF 35 kDa subunit + RSP Y48G1A.6 Y48G1A_53.a Polycomb group protein SCM/L(3)MBT ++ SG C24B9.8 str-13 7TM chemoreceptor, GPCR activity ++ SG F10E9.3 Splice Isoform 1 of Death domain-associated protein 6 ++ SG F12A10.6 Serine/threonine kinase (haspin family) ++ SG F19H8.2 ∼to Rho kinase + SG F21F3.3 Isoprenylcysteine carboxyl methyltransferase family, yeast: ER ++ SG R09E12.1 srbc-59 7TM chemoreceptor, srbc family ++ SG R52.4 Chemoreceptor + SG T25B9.2 protein phosphatase 1, catalytic subunit, alpha isoform 3 ++ SG T28F2.2 Splice Isoform 1 of COMM domain-containing protein 4 ++ SG W05B5.2 GPCR, membrane ++ SG Y71G12B.20 mab-20 Semaphorin-4G precursor ++ SG ZK355.1 ZK355.h Tyr-kinase Receptor ++ U B0238.11 HMG box-containing protein, transcriptional + U C02E7.6 Splice Isoform 1 of AMME syndrome candidate gene 1 protein ++ U C29F9.1 Unknown ++ U D1022.5 Dienelactone hydrolase family + U F01G12.5a let-2 Unknown ++ U F47F6.5 Similarity to C-type lectin ++ U F54E7.6 Unknown ++ U F56C4.1 Unknown + U H23L24.e Unknown ++ U K01G12.3 HIV TAT specific factor 1 + U R09H3.3 Unknown ++ U R10H10.2 spe-26 kelch-like 20 ++ U R11A8.4 sir-2.1 NAD-dependent deacetylase sirtuin-1 ++ U T05A10.4 Similarity to cysteine-rich secretory protein 2 precursor + U T06G6.8 Unknown ++ U XE249 Chondroitin 6-sulfotransferase ++ U Y19D10A.j C-type lectin + U ZC334.9 ins-28 Insulin-like peptide ++ U ZK1290.11 Unknown ++ VT C07G1.5 hgrs-1 (pqn-9) HGF-reg tyr-kinase substrate, membrane trafficking/protein sorting + VT C34C12.2 Role in preribosome assembly or transport/t-snare domain ++ VT M151.3 Girdin, Yeast: ER-Golgi transport, SNARE assembly ++ VT T05G5.9 Ran-binding protein 2-like 4/yeast: ER-Golgi/SNARE assembly ++ VT W02A11.2 ESCRT-II complex subunit, Yeast: vacuolar protein sorting protein 25 + U: Unknown, RSP: RNA synthesis and processing, PT: Protein Transport, PS: Protein synthesis, PF: Protein Folding, DR: DNA replication, ECM: Extracellular matrix, CS: Cytoskeleton, EM: Energy metabolism, GM: Glucose Metabolism, SG: Signaling, VT: vesicle transport, ET: electron transport. +: up to a two-fold increase, ++: more than a two-fold increase in the amount of inclusions. The modifier genes function in a variety of biological processes, some of which have previously been suggested to be involved in Parkinson's disease, such as vesicular transport and lipid metabolism. Lipid metabolism, lipid membranes, and vesicle-mediated transport have previously been linked to α-synuclein pathology in a yeast model [12],[13]. In Parkinson's patients, lipids and membrane material have been found to be directly associated with α-synuclein in lipid droplets and Lewy bodies [11], [14]–[16]. Although we did identify a C. elegans ortholog of the recently discovered modifier of neuronal alpha-synuclein toxicity SIRT2, we did not pick up two other modifiers of neuronal alpha-synuclein pathology: the G-protein coupled receptor kinase 2 and the molecular chaperone Hsp70 [17]–[19]. In Drosophila, overexpression of G-protein coupled receptor kinase 2 (Gprk2) increases neuronal toxicity of α-synuclein [18]. In addition, expression of an S129A mutant of α-synuclein, which cannot be phosphorylated by Grpk2, is less toxic, while forming more aggregates [18]. Based on these observations, one might expect that knockdown of Gprk2 would result in an increase in the amount of inclusions as well. Two orthologs of the Gprk2 gene are present in C. elegans, grk-1 and grk-2. We tested the effect of knockdown of these two genes by RNAi on the formation of inclusions. Unexpectedly, RNAi knockdown of grk-1 or grk-2, and not RNAi of random tyrosine or serine kinases, resulted in a decrease in the amount of inclusions (Figure 5A and data not shown), indicating that we could not have recovered these genes in our screen for more inclusions. There was no obvious difference in the level of α-synuclein-YFP expression at day 5 after synchronization, indicating that formation of the inclusions themselves is affected (Figure 5D). The effect of knockdown of grk-1 and grk-2 by RNAi was confirmed by crossing in deletion alleles of both genes, grk-1(ok1239) and grk-2(gk268), into the α-synuclein-YFP strain (Figure 5B). Note that an RNAi screen for a reduction in the amount of inclusions yielded only one other kinase, which supports the idea that GRKs act specifically (data not shown). In all, C. elegans orthologs of Gprk2 act as modifiers of α-synuclein inclusion formation. Knockdown of Gprk2 in Drosophila or overexpression of grk-1 or grk-2 in C. elegans will be required to establish whether their specific role in alpha-synuclein pathology is comparable between the two species. 10.1371/journal.pgen.1000027.g005 Figure 5 RNAi and Deletion of grk-1 and grk-2 Decreases Inclusion Formation. (A) Confocal images showing head region of α-synuclein-YFP transgenic animals fed on OP50 bacteria, bacteria containing empty RNAi vector (L4440), and expressing double stranded RNA targeting grk-1 and grk-2. (B) Confocal images of α-synuclein-YFP C. elegans, in wild type background, grk-1(ok1239) and grk-2(gk268) background. Scale bar represents 25 μm. (C) Quantification of the number of worms within the population (n = 20) with the same amount (wt), fewer (
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Genet
                plos
                plosgen
                PLoS Genetics
                Public Library of Science (San Francisco, USA )
                1553-7390
                1553-7404
                August 2010
                August 2010
                26 August 2010
                : 6
                : 8
                : e1001084
                Affiliations
                [1 ]Department of Pediatrics, Vanderbilt University, Nashville, Tennessee, United States of America
                [2 ]Children's Hospital, Vanderbilt University, Nashville, Tennessee, United States of America
                [3 ]London Centre for Nanotechnology, University College London, London, United Kingdom
                [4 ]Center for Molecular Toxicology, Vanderbilt University, Nashville, Tennessee, United States of America
                University of California San Francisco, United States of America
                Author notes

                Conceived and designed the experiments: AB CA DSdA MA. Performed the experiments: AB CA DSdA. Analyzed the data: AB CA DSdA DM. Contributed reagents/materials/analysis tools: DM MA. Wrote the paper: AB MA.

                Article
                10-PLGE-RA-2613R2
                10.1371/journal.pgen.1001084
                2928785
                20865164
                b2558149-994b-4d3e-8b51-dbba659c385d
                Benedetto et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
                History
                : 16 February 2010
                : 22 July 2010
                Page count
                Pages: 18
                Categories
                Research Article
                Cell Biology/Cellular Death and Stress Responses
                Genetics and Genomics/Disease Models
                Genetics and Genomics/Genetics of Disease
                Neurological Disorders

                Genetics
                Genetics

                Comments

                Comment on this article