Skip to main content
Erschienen in: Orphanet Journal of Rare Diseases 1/2013

Open Access 01.12.2013 | Research

Unfolded protein response in Gaucher disease: from human to Drosophila

verfasst von: Gali Maor, Sigal Rencus-Lazar, Mirella Filocamo, Hermann Steller, Daniel Segal, Mia Horowitz

Erschienen in: Orphanet Journal of Rare Diseases | Ausgabe 1/2013

Abstract

Background

In Gaucher disease (GD), resulting from mutations in the GBA gene, mutant β-glucocerebrosidase (GCase) molecules are recognized as misfolded in the endoplasmic reticulum (ER). They are retrotranslocated to the cytoplasm, where they are ubiquitinated and undergo proteasomal degradation in a process known as the ER Associated Degradation (ERAD). We have shown in the past that the degree of ERAD of mutant GCase correlates with GD severity.
Persistent presence of mutant, misfolded protein molecules in the ER leads to ER stress and evokes the unfolded protein response (UPR).

Methods

We investigated the presence of UPR in several GD models, using molecular and behavioral assays.

Results

Our results show the existence of UPR in skin fibroblasts from GD patients and carriers of GD mutations. We could recapitulate UPR in two different Drosophila models for carriers of GD mutations: flies heterozygous for the endogenous mutant GBA orthologs and flies expressing the human N370S or L444P mutant GCase variants. We encountered early death in both fly models, indicating the deleterious effect of mutant GCase during development. The double heterozygous flies, and the transgenic flies, expressing mutant GCase in dopaminergic/serotonergic cells developed locomotion deficit.

Conclusion

Our results strongly suggest that mutant GCase induces the UPR in GD patients as well as in carriers of GD mutations and leads to development of locomotion deficit in flies heterozygous for GD mutations.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1750-1172-8-140) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare no competing interests.

Authors’ contributions

GM designed the experiments, performed them and wrote the manuscript; SRL constructed the Drosophila expressing plasmids: MF supplied most human cell lines; HS supplied Drosophila materials and participated in manuscript preparation; DS participated in designing of experiments and in preparation of the manuscript; MH designed the experiments and participated in the manuscript preparation. All authors read and approved the final manuscript.
Abkürzungen
GD
Gaucher disease
GCase
Glucocerebrosidase
ER
Endoplasmic reticulum
ERQC
ER quality control
ERAD
ER associated degradation
UPR
Unfolded protein response
ATF6
Activating transcription factor 6
GlcCer
Glucosylceramide
eIF2α
Eukaryotic Initiation Factor 2α
CBE
Conduritol-B-epoxide
PD
Parkinson disease
LB
Lewy body.

Background

Gaucher disease (GD) is a lysosomal storage disease, caused by mutations in the gene encoding lysosomal acid β-glucocerebrosidase (GCase), designated GBA. As a result, glucosylceramide (GlcCer) is not properly degraded and accumulates primarily in cells of mononuclear phagocyte origin [1, 2].
More than 300 mutations were identified in the GBA gene. A large fraction of them are missense mutations, though premature termination, splice site mutations, deletions and recombinant alleles have been recognized as well [3]. There are several abundant mutations. Thus, the N370S mutation [4] is the most prevalent among type 1 GD patients, while the L444P mutation [5] is most common among the neuronopathic types of GD. The majority of patients homozygous for this mutation develop type 3 GD. The 84GG mutation is an insertion of a guanine 84 nucleotides downstream from the first initiator methionine of the GBA mRNA, resulting in premature protein termination [6].
As a lysosomal enzyme, GCase is synthesized on endoplasmic reticulum (ER) bound polyribosomes [7]. Upon its entry into the ER, it undergoes N-linked glycosylation on four asparagines, after which it is subject to ER quality control (ERQC). When correctly folded it shuttles to the Golgi compartment for further modifications on the N-glycans and finally it traffics to the lysosomes. Mutant GCase variants are recognized as misfolded proteins and undergo various degrees of ER associated degradation (ERAD). The accumulation of misfolded molecules in the ER, activate signaling events known as the unfolded protein response (UPR) [8]. UPR monitors the conditions in the ER, by sensing insufficiency in protein folding capacity and translates this information into gene expression [9]. The ER membrane harbors three ER stress sensors: The type 1 transmembrane protein kinase endoribonuclease IRE1, the type 1 protein kinase PERK, and the activating transcription factor 6 (ATF6). These three branches operate simultaneously and use unique mechanisms of signal transductions. The three UPR transducers are constitutively expressed in metazoan cells [10], and are maintained in an inactive state through interaction with the ER protein chaperone BiP. Accumulated unfolded protein(s) binds and sequesters BiP, thus promoting BiP dissociation from PERK, IRE1 and ATF6. Dissociation of BiP from the three stress sensors allows their modification and signal transduction, which results in a response to the accumulation of misfolded proteins [11]. Thus, IRE1 undergoes dimerization and phosphorylation and participates in a cytoplasmic complex, which splices the transcription factor X-box binding protein 1 (Xbp1). Upon its splicing the Xbp1 mRNA is translated into a protein that translocates into the nucleus and turns on UPR related genes [9, 12, 13]. PERK is a kinase that undergoes dimerization and autophosphorylation and mediates phosphorylation of the eukaryotic translation initiation factor 2α (eIF2α). Phosphorylated eIF2α attenuates general protein translation in the cells [9, 11, 14]. ATF6, the third component among the UPR sensors, shuttles to the Golgi, where it is sequentially cleaved by proteases. Its cleaved N-terminal cytosolic fragment enters the nucleus where it serves as a transcription factor of UPR upregulated genes, including the induction of the proapoptotic bZIP transcription factor CCAAT/enhancer-binding protein homologous protein (CHOP) [15], which is essential for cell cycle arrest and the apoptotic response to chronic ER stress [9, 11, 13, 14, 16]. Manifestation of UPR in GD derived cells has already been noted in cell lines that originated from GD patients, homozygous for the N370S or the L444P mutations [17, 18]. Yet, accumulation of glucosylceramide (GlcCer) per se, induced by conduritol-β-epoxide (CBE), did not result in UPR [19]. Likewise, in the absence of mutant GCase there was no UPR [19], underscoring the importance of mutant GCase in the activation of ERAD and UPR.
In this study we tested whether UPR is activated in GD. Our results show the occurrence of UPR in GD derived skin fibroblasts and in carriers of GD mutations, both in humans and Drosophila. In heterozygous flies, and flies expressing the human N370S or L444P mutant GCase variants there were significant developmental defects. Locomotion deficit was evident in aging flies, reminiscent of Parkinson disease (PD).

Materials and methods

Materials

The following primary antibodies were used in this study: rabbit polyclonal anti-GRP78 antibodies (Cell Signaling Technology, Beverly, MA, USA), mouse monoclonal anti-CHOP antibody (Cell Signaling Technology, Beverly, MA, USA), rabbit polyclonal anti-phospho-eIF2α (Ser51) antibodies, rabbit polyclonal anti-eIF2α antibodies (from cell signaling Technology, Beverly, MA, USA), mouse monoclonal anti-actin antibody (Sigma-Aldrich, Israel), rabbit polyclonal anti human GCase antibodies (Sigma-Aldrich, Israel) and mouse monoclonal anti-tubulin antibody (Sigma-Aldrich, Israel).
Secondary antibodies used were: horseradish peroxidase-conjugated goat anti-mouse antibodies and Horseradish peroxidase-conjugated goat anti-rabbit antibodies (both from Jackson Immuno Research Laboratories, West Grove, PA, USA). Leupeptin, phenylmethylsulfonyl fluoride (PMSF) and aprotinin were from Sigma–Aldrich (Rehovot, Israel). Absolute Blue qPCR SYBR Green ROX Mix was from Thermo Scientific (Logan, UT, USA).

Cell lines

Human primary skin fibroblasts were provided by two publically available, sources: by “Cell Line and DNA Biobank from Patients Affected by Genetic Diseases” (G. Gaslini Institute), Telethon Genetic Biobank for GD skin fibroblasts or by Prof. R. O. Brady, NIH. The patients signed an informed consent. Work with the cell lines was in accordance with the institutional guidelines of Tel Aviv University. Identifiable clinical and personal data from the patients were not available for this study. Cells were grown in DMEM supplemented with 20% FBS (Biological Industries, Beit Haemek, Israel). All cells were grown at 37°C in the presence of 5% CO2.

Fly strains

Canton-S flies (WT) served as a wild-type control. Strains were maintained on cornmeal-molasses medium at 25°C. Strains harboring a minos transposable element in CG31414 [Mi{ET1}CG31414] or in CG31148 [Mi{ET1}CG31148] were from the Bloomington Stock Center (#23602 and #23435, respectively). Da-Gal4 and Ddc-Gal4 driver lines were obtained from Bloomington Stock Center. Transgenic flies, harboring pUASTmycHisGCase, pUASTmycHisN370SGCase or pUASTmycHisL444PGCase on the second chromosome, were established by BestGene (Chino Hills, CA, USA).

Methods

Construction of plasmids

An Xba I-Sap I fragment, isolated from pcDNA4 (Invitrogen Life Technologies Co., Carlsbad, CA, USA) was subcloned between Xba I and Sap I restriction sites of pUAST, to create pUASTmycHis. Eco RI-Xho I fragments, containing either the normal or the N370S or the L444P mutant human GCase cDNAs, isolated from the plasmids MycHis WT GCase, MycHis N370S GCase or MycHis L444P GCase [20], respectively, were subcloned in pUASTmycHis, cleaved with the same restriction enzymes, to create pUASTmycHisGCase, pUASTmycHisN370SGCase or pUASTmycHisL444PGCase, respectively.

RNA preparation

Total RNA was isolated using EZ-RNA kit (Biological Industries, Beit Haemek, Israel), according to the manufacturer’s instructions. For RNA extraction from flies, adult flies were frozen in liquid nitrogen and then homogenized in TRI Reagent solution (MRC, Cincinnati, Ohio, USA). The extraction was performed according to the manufacturer’s recommendations.

RT PCR

Two μg of RNA were reverse transcribed with M-MLV reverse transcriptase (Promega Corporation, CA, USA), using oligo dT primer in a total volume of 20 μl, at 42°C for 60 minutes. Reactions were stopped by incubation at 70°C for 15 minutes. One-two microliters of the resulting cDNA were amplified by PCR or by quantitative real time PCR.

PCR

PCR was executed in 25 μl containing 0.4 mM dNTPs, 10 ρM of each primer, 1 unit of Taq polymerase (Takara, Shiga, Japan) and 10× Taq buffer (10 mM Tris HCL pH 8.3, 50 mM KCl and 1.5 mM MgCl2). Thirty cycles of 94°C (1 minute), 58°C (1 minute) and 72°C (1 minute) were performed, following by 10 minutes at 72°C for final extension. PCR reactions were carried out in an Eppendorff Master-cycler EP Gradient S (Eppendorf, Hamburg, Germany). PCR products were separated by agarose gel electrophoresis (1–1.5%) and visualized with 0.1% ethidium bromide. Sequence of the primers used appears in Table 1.
Table 1
Primers used in this study
Human-GAPDH-RT-F
5′-CTCCTCCTGTTCGACAGTCA-3′
Human-GAPDH-RT-R
5′-GTTGACTCCGACCTTCACCT-3′
Human-CHOP-RT-F
5′-AGCGACAGAGCCAAAATCAG-3′
Human-CHOP-RT-R
5′-TCTGCTTTCAGGTGTGGTGA-3′
Human-GRP78-RT-F
5′-CATCAAGTTCTTGCCGTTCA-3′
Human-GRP78-RT-R
5′-ATGTCTTTGTTTGCCCACCT-3′
Human-GAPDH-F
5′-CCATCAATGACCCCTTCATTGACC-3′
Human-GAPDH-R
5′-CTCAYGGYYCACACCCATGAC-3′
Human-s-XBP1-F
5′-TCTGCTGAGTCCGCAGCAG-3′
Human-s-XBP1-R
5′-GAAAAGGGAGGCTGGTAAGGAAC-3′
Drosophila-Hsc-70-3-RT-F
5′-GCTGGTGTTATTGCCGGTCTGC-3′
Drosophila-Hsc-70-3-RT-R
5′-GATGCCTCGGGATGGTTCCTTGC-3′
Drosophila-s-Xbp1-RT-F
5′-CCGAACTGAAGCAGCAACAGC-3′
Drosophila-s-Xbp1-RT-R
5′-GTATACCCTGCGGCAGATCC-3′
Drosophila-RP49-RT-F
5′-TAAGAAGCGCACAAAGCACT-3′
Drosophila-RP49-RT-R
5′-GGGCATCAGATATTGTCCCT-3′
The table contains the sequence of all the primers used in this work. RT real time, R reverse, F forward.

Quantitative real time PCR

One μl of cDNA was used for quantitative real time PCR. PCR was performed using “power SYBR green QPCR mix reagent kit” (Applied Biosystems, Foster City, CA, USA) by Rotor-Gene 6000 (Qiagen, Valencia, CA, USA). The reaction mixture contained 50% QPCR mix, 300 nM of forward primer and 300 nM of reverse primer, in a final volume of 10 μl. Thermal cycling conditions were 95°C (10 minutes), and 40 cycles of 95°C (10 seconds) 60°C (20 seconds) and 72°C (20 seconds). Relative gene expression was determined by Ct value. Human cDNA was amplified with primers specific for human BiP (Human-GRP78-RT-F and Human-GRP78-RT-R, Table 1) or human CHOP (Human-CHOP-RT-F and Human-CHOP-RT-R, Table 1). GAPDH was used as a normalizing control for human genes (amplified with primers: Human-GAPDH-RT-F and Human-GAPDH-RT-R, Table 1). Amplification of Drosophila genes was conducted with primers specific for Drosophila Hsc-70-3 (Drosophila-Hsc-70-3-RT-F and Drosophila-Hsc-70-3-RT-R, Table 1), or for the spliced form of Drosophila Xbp1 (Drosophila-s-Xbp1-RT-F and Drosophila-s-Xbp1-RT-R, Table 1). RP49 was used as a normalizing control (amplified with primers: Drosophila RP49-RT-F and Drosophila RP49-RT-R, Table 1).

Detection of spliced Xbp1 mRNA processing

Human spliced Xbp1 was amplified from cDNA using the primers: Human s-Xbp1 F and Human s-Xbp1-R (Table 1). GAPDH was used as a normalizing control (amplified with primers: Human-GAPDH-F and Human-GAPDH-R, Table 1). To amplify Drosophila spliced Xbp1 the primers: Drosophila s-Xbp1-RT-F and Drosophila s-Xbp1-RT-R (see Table 1) were used, with Drosophila RP49 as a normalizing control. The forward primer could anneal only to the spliced form of Xbp1 mRNA.

SDS-PAGE and western blotting

Cultured cells

Cell monolayers were washed three times with ice-cold phosphate-buffered saline (PBS) and lysed at 4°C in lysis buffer (10 mM HEPES pH 8.0, 100 mM NaCl, 1 mM MgCl2 and 1% Triton X-100) containing 10 μg/ml aprotinin, 0.1 mM phenylmethylsulfonyl fluoride (PMSF) and 10 μg/ml leupeptin. Lysates were incubated on ice for 30 minutes and centrifuged at 10,000 g for 15 minutes at 4°C.

Flies

For each preparation, 10 flies were homogenized in RIPA lysis buffer (50 mM Tris/HCL, 150 mM NaCl, 1 mM EDTA, 1% TritonX-100, 1% sodiumdeoxycholate, 0.1% SDS) containing protease inhibitors (10 μg/ml leupeptin, 10 μg/ml aprotinin and 0.1 mM PMSF- all from Sigma-Aldrich, Israel). Samples containing the same amount of protein were electrophoresed through 10% SDS–PAGE and electroblotted onto a nitrocellulose membrane (Schleicher and Schuell BioScience, Keene, NH, USA). Further treatment of membranes and ECL detection was as described elsewhere [21].

Enzymatic activity

Confluent primary skin fibroblasts were washed twice with ice-cold PBS and collected with a rubber policeman in 150 μl sterile water. Cell lysates, containing 40 μg of protein, were assayed for GCase activity in 0.2 ml of 100 mM potassium phosphate buffer, pH 4.5, containing 0.15% Triton X-100 (Sigma-Aldrich, Israel) and 0.125% taurocholate (Calbiochem, La Jolla, CA, USA) in the presence of 1.5 mM 4-methyl-umbeliferyl-glucopyranoside (MUG) (Genzyme Corporation. Boston, MA, USA) for 1 h at 37°C. The reaction was stopped by the addition of 0.5 ml of stop solution (0.1 M glycine, 0.1 M NaOH, pH 10) and the amount of 4-methyl-umbeliferone (4-MU) was quantified using Perkin Elmer Luminescence Spectrometer LS 50 (excitation wavelength: 340 nm; emission: 448 nm) [22].

Endonuclease-H (endo-H) treatment

Samples of cell lysates, containing 100 μg of total protein, were subjected to an overnight incubation with endo-H (New England Biolabs, Beverly, MA, USA), according to the manufacturer’s instructions. They were electrophoresed through 10% SDS-PAGE and the corresponding blot was interacted with anti GCase and anti actin antibodies. Total GCase amount was divided by that of actin at the same lane and normalized to WT GCase, which was considered 100. To determine the endo-H resistant fraction, the intensity of GCase resistant fraction was divided by the intensity of the entire amount of GCase in the same lane. GDPV (GD Predictive Value) was determined as described elsewhere [23] (GCase amount X GCase resistant fraction: 100).

Climbing assay of flies

Vials, each containing 10 male flies, were tapped gently on the table and left standing for 15 seconds. The number of flies that climbed at least five cm was recorded. The experiment was repeated 10 times.

Blot quantitation

The blots were scanned using Image Scan scanner (Amersham Pharmacia Biotech, Buckinghamshire, England), and the intensity of each band was measured by the Image Master 1DPrime densitometer (Amersham Pharmacia Biotech, Buckinghamshire, England) and GelQuant (BiochemLabSolutions).

Statistics

All the results were statistically analyzed using the student t-test.

Results

Activation of UPR in GD derived fibroblasts

Mutant GCase is recognized as misfolded in the ER. After several unsuccessful attempts to refold it, it undergoes ERAD [21]. The level of ERAD correlates with GD severity, since it determines the amount of mutant enzyme that reaches the lysosomes and degrades the substrate there, depending on its residual activity. Moreover, skin fibroblasts that derived from GD patients homozygous for the N370S or the L444P mutations exhibited UPR [17, 24]. We, therefore, decided to extend the study and tested whether UPR is activated in additional GD derived cells. The UPR induces increased transcription of the molecular chaperone BiP and the transcription factor CHOP [25]. In addition, the UPR induces splicing of the Xbp1 transcript and phosphorylation of eIF2α [25].
Based on the above, we first examined the activation of UPR in GD by testing mRNA levels of BiP and CHOP in skin fibroblasts obtained from GD patients. To do so, we used the quantitative RT-PCR approach, using normal fibroblasts as control. Our results, (Figure 1A), showed a significant increase in BiP and CHOP mRNA levels in GD derived fibroblasts, compared to normal cells. A concomitant increase was detected in the protein levels of BiP and CHOP (Figure 1B-D).
In GD, there is accumulation of the GCase substrate GlcCer, along with the presence of mutant GCase, which undergoes ERAD. In order to test possible contribution of substrate accumulation to UPR, we induced substrate accumulation using the non-competitive inhibitor of GCase, CBE, for 10 days, as has done by Farfel et al. [19]. It has already been shown in the past that CBE treatment leads to substrate accumulation in skin fibroblasts [26]. Treatment of normal skin fibroblasts with 200 μM CBE, which completely abolished GCase activity, (Additional file 1: Figure S1A) did not induce elevation in mRNA levels of either BiP or CHOP (Additional file 1: Figure S1B). Thus, substrate accumulation in GD derived fibroblasts does not activate the UPR.

Splicing of Xbp1 as a UPR marker in GD derived fibroblasts

Splicing of Xbp1 is a central hinge of the IRE1 pathway [25, 27], which is another branch activated in the UPR. The Xbp1 mRNA contains two overlapping reading frames, A and B. Under normal conditions, only A frame is transcribed, producing an unspliced version of Xbp1 with no protein product (see Figure 2A) [28]. Upon UPR activation, IRE1 dimerizes and participates in cytoplasmic splicing of Xbp1, thus, removing a 26 bp intron from the Xbp1 mRNA. The spliced Xbp1 mRNA (frame B) encodes a transcription factor that binds to the UPRE or ERSE consensus sequences of promoters of UPR target genes, thus leading to their transcription [28]. Expression of spliced Xbp1 in GD derived skin fibroblasts was tested (see Figure 2B). Our results, presented in Figure 2B, showed that in GD derived fibroblasts the spliced Xbp1 product was significantly elevated, in comparison to normal fibroblasts.

Elevation in phosphorylation of eIF2α in GD derived fibroblasts

Dissociation of PERK from BiP leads to its dimerization and autophosphorylation, and mediates phosphorylation of the eukaryotic translation initiation factor 2α (eIF2α). eIF2α is a subunit of eIF2, a heterotrimeric GTPase, required to bring the initiator methionyl-tRNA to the 40S ribosomal subunit for AUG initiation codon selection. Phosphorylation of eIF2α inhibits the GDP/GTP exchange reaction on eIF2, thus preventing eIF2 recycling and its initial step of protein synthesis [9]. Phosphorylated eIF2α attenuates general protein translation in cells [9, 11, 14]. We therefore tested possible changes in the levels of phosphorylation of eIF2α, using western blotting and interaction with anti-phosphorylated eIF2α antibodies. Our results, presented in Figure 2D, E, showed that eIF2α phosphorylation was increased in GD derived fibroblasts in comparison to normal fibroblasts.

Activation of UPR in carriers of GD mutations

Our results strongly suggested that UPR results from the presence of mutant GCase in the cells, since substrate accumulation by itself did not lead to activation of the UPR machinery. To confirm our observations we decided to investigate the occurrence of UPR in cells derived from carriers of different GD mutations. To this end we tested elevation in BiP and CHOP mRNA, in Xbp1 splicing and in phosphorylation of eIF2α in cells that derived from carriers of several GD mutations. Our results, presented in Figure 3, showed a significant elevation in the amount of BiP and CHOP mRNAs as well as in Xbp1 splicing in carriers of GD mutations. Likewise, phosphorylation of eIF2α increased significantly in cells that originated from carriers in comparison to normal cells. Interestingly, we observed activation of UPR also in cells carrying the 84GG mutation, strongly suggesting that existence of a mutant GBA mRNA evokes UPR.

Activation of UPR in Drosophila GD-like carriers

There are two GBA homologs in Drosophila, designated CG31414 and CG31148, both encoding proteins showing ~31% identity and ~49% similarity to the human GCase. Two Drosophila strains are available, each carrying a minos transposable element insertion in one of the fly GBA orthologs. Insertion of minos in the CG31414 gene leads to translation of truncated GCase, lacking 129 C-terminal amino acids (out of the 448 amino acids of the predicted normal fly GCase protein). The minos insertion in CG31148 leads to a truncated protein lacking 34 C-terminal amino acids. Due to their close proximity on chromosome 3, simple genetic manipulations cannot be employed to create a chromosome with both mutated genes. However, double heterozygous fly is an authentic model for the GD carrier state in human. We generated double heterozygous flies, which exhibited ~30% decrease in GCase activity, as expected (Figure 4A). We then tested possible activation of UPR in these flies. The fly does not contain a CHOP homolog in its genome and UPR activation is measured by elevation in mRNA of the BiP ortholog heat-shock cognate 70–3 (Hsc-70-3 gene), in level of Xbp1 splicing and in the level of phosphorylated eIF2α. We observed a significant elevation in the level of Hsc-70-3 mRNA and Xbp1 splicing in the double heterozygotes, as well as in phosphorylation of eIF2α (Figure 4), compared to WT flies. These results clearly point to existence of UPR in carriers of mutations in the GBA orthologs of Drosophila. A significant defect in development from larva to pupa and from pupa to adult was found in the double heterozygous animals, in comparison to WT Drosophila (Figure 4E). This highlights the significance of normal GCase during development and the deleterious effect of mutant GCase.
We also ectopically expressed the N370S or the L444P human mutant GCase variants in the fly, using the Gal4/UAS system. In this system the yeast Gal4 transcription factor, expressed from a tissue specific Drosophila promoter, is used to express a specific transgene (in our case, the human normal or mutant mycHis tagged GCase cDNAs) coupled to the yeast UAS. Thus, expression of the transgene depends on the specificity of the promoter used for the expression of the Gal4 transcription factor. We used daughterless-Gal4, which drives ubiquitous expression of the transgene. We verified expression of the transgene using western blotting and interaction with anti myc antibody (Figure 5A). The results showed expression of the normal human as well as the mutant proteins in the transgenic flies. In comparison to the normal human protein expressed in the fly, the N370S or the L444P mutant variants presented higher endo-H sensitivity (Figure 5A) [21, 29, 30], illustrating the existence of ERAD of mutant human GCase in the flies. The fraction of lysosomal N370S human GCase, (endo-H resistant fraction, labeled by black circles in Figure 5A) was higher than that of L444P human GCase, as expected for these two mutations and shown for endogenous human N370S and L444P mutant GCase variants [21, 29]. Furthermore, the results (Figure 5B-D) demonstrated higher activation of the UPR in flies expressing the mutant variants than those expressing the WT human GCase. UPR was measured by increase in the mRNA levels of Hsc-70-3, in the mRNA levels of spliced Xbp1 and in the levels of phosphorylated eIF2α.
A significant defect in development from larva to pupa and from pupa to adult was found in animals expressing the human mutant GCase variants, in comparison to animals expressing the WT human GCase (Figure 5E), similar to the double heterozygous flies. This highlights the fact that overexpression of human mutant GCase in the fly leads to deleterious effects, regardless of the presence of endogenous WT GCase.

Locomotor dysfunction in flies expressing human mutant GCase variant

An association has been found between GD patients and carriers of GD mutations and PD. Thus, GD patients and carriers of GD mutations are prone to develop PD [3155]. We used the Ddc-Gal4 driver in order to drive specific expression of the human N370S and the L444P GCase mutants in the dopaminergic/serotonergic neurons of the fly. The locomotion (climbing) assay, commonly used for assaying behavior of flies expressing familial Parkinson related proteins in their brain [56], was used to assess the neural dysfunction caused to the flies due to expression of mutant GCase in the nervous system. We monitored the climbing ability of the flies at the age of 7, 12 and 22 days. Measurements, conducted at the age of 22 days, revealed significant locomotion dysfunction in both fly models for carriers of GD mutations, namely, flies double heterozygotes in their two GBA orthologs and flies transgenic for the N370S or the L444P mutant human GCase variants (Figure 6).

Discussion

In this work we show that persistent presence of mutant GCase activates the UPR, in humans and in Drosophila.
Several previous publications have already noticed the existence of UPR in GD derived skin fibroblasts. Wei et al. showed in one N370S/N370S Type 1 GD derived skin fibroblast line, a significant increase in mRNA level of the UPR related genes ATF6, BiP and Xbp1. This was accompanied by a concomitant increase in the protein level of active ATF6, BiP and phosphorylated eIF2α [17]. Mu et al. reported on UPR activation in cell lines that derived from GD patients homozygous for the N370S and the L444P mutations [24]. In another publication the efficacy of catechin in alleviating UPR in one GD derived line (N370S/N370S) has been documented [18].
In the present work we show UPR activation in skin fibroblasts derived from GD patients manifested by an increase in mRNA and protein levels of BiP and CHOP, splicing of Xbp1 and phosphorylation of eIF2α. We also show that in fibroblast lines derived from carriers of GD mutations there is UPR as well. Interestingly, the 84GG mutation is not expected to culminate in a mature protein [6]. Since there is UPR in carriers of this mutation, one has to assume that either the 84GG mutant RNA participates in translation of a very short peptide (26 amino acids long), shorter than the full size leader of GCase (38 amino acids long), thereby blocking ER entrance of newly synthesized proteins, or the 84GG mRNA-bound polyribosomes block ER entrance, thereby leading to development of ER stress and, as a result, to UPR. These hypotheses will have to be further tested.
ERAD and UPR are well conserved across species and Drosophila has become an important tool in studying these phenomena. The short life span of the fly with the sophisticated molecular and genetic tools for fast establishment of transgenic lines, and availability of deletions and mutations in any chosen gene have made it an attractive model, which allows fast screening and analyses of large populations. We could recapitulate UPR in two fly models: one corresponding to carriers of GD mutations and the other involving transgenic flies expressing the human N370S or the L444P mutant proteins.
All lysosomal enzymes are synthesized on ER bound polyribosomes and upon their entry into the ER undergo N-linked glycosylation and quality control, after which they shuttle to the Golgi apparatus. Following further modifications there, they are trafficked to the lysosomes. Therefore, all mutant lysosomal enzymes are expected to undergo ERAD and induce the UPR machinery. UPR has already been documented in other lysosomal diseases. Thus, in Fabry disease which results from mutation in the α-galactosidase-A encoding gene (α-Gal-A) and accumulation of the globotrioside Gb3, mutant variants undergo ERAD, which induces the UPR [57, 58]. UPR has also been documented in skin fibroblasts from patients suffering from ceroid lupofusinosis (CLN) 1, 2, 3, 6 and 8, as well as in cells of patients with GM1 gangliosidosis (suffering from reduced activity of β-galactosidase), Tay Sachs disease (reduced activity of β-hexosaminidase A) and Niemann Pick type C2 (mutations in the NPC2 gene) [17]. Interestingly, in some model systems for lysosomal diseases UPR was not recapitulated. Farfel et al. [19] were unable to demonstrate UPR in neuronal cells that derived from knockout mice, lacking the GBA gene, or in animals or cells, treated with the GCase non-competitive inhibitor CBE. Lack of UPR in both of these cases is likely due to the absence of mutant GCase in the ER. Likewise, in NPC1-deficient mice and in an NPC1 cell-based model, created by knocking down the expression of NPC1 using RNA interference, there was no UPR [59]. Again, in both cases, no mutant protein was present in the ER to induce the UPR machinery.
In recent years association has been elegantly demonstrated between GD and PD, a neurodegenerative disease affecting 1% of individuals over 60 years old. Thus, there is a higher propensity among Type 1 GD patients and among carriers of GD mutations to develop PD in comparison to the non-GD population [3155]. Brains of carriers of GD mutations who develop PD display Lewy bodies (LB) and loss of substantia nigra neurons [60]. Carriers of GBA mutations tend to have more cortical LBs than those of non-carriers (82% versus 43%, respectively) [61], suggesting that mutant GCase variants promote α-synuclein aggregation directly. Cullen et al. showed that expression of mutant human GCase, but not that of the normal counterpart, led to increase in α-synuclein accumulation in MES23.5, PC12, and HEK293 cell lines, arguing that mutant GCase has a direct role in α-synuclein accumulation, and most probably, aggregation [62].
Since carriers of GD mutations do not accumulate GlcCer in their brain and its build-up has not been demonstrated in brains of Type 1 GD patients, we raised the possibility that ERAD of mutant GCase contributes to the development of PD among GD patients and carriers of GD mutations. We have shown that parkin interacts with mutant GCase and mediates its Lysine 48 ubiquitination and proteasomal degradation [20]. We proposed that this interaction between parkin and mutant GCase leads to deleterious effect in dopaminergic cells caused by the accumulation of parkin substrates, which are potentially toxic. Our recent results (Bendikov-Bar and Horowitz, unpublished) show that mutant GCase competes with two known substrates of parkin, PARIS [63] and ARTS [64], whose accumulation in cells leads to apoptosis. PARIS is a transcription repressor of peroxisome proliferator-activated receptor gamma (PPARγ) coactivator-1α (PGC-1α) expression. PGC-1α is a master regulator of mitochondrial biogenesis [65]. Thus, PARIS accumulation impedes mitochondria biogenesis. Interestingly, PARIS accumulates in mouse models of parkin inactivation and in PD patients’ brains [63]. ARTS is a mitochondrial protein that initiates caspase activation, upstream of cytochrome c release in the mitochondrial apoptotic pathway [66].
We now extend our hypothesis for the role of mutant GCase in the development of PD. We propose that ERAD of mutant GCase leads to accumulation of parkin substrates, some of which are deleterious, like PARIS and ARTS. This accumulation leads to cell death. We also assume that UPR, as part of the cellular ER stress, induced by the persistent presence of mutant GCase in the ER, leads to cellular death. Therefore, both, ERAD of mutant GCase and UPR contribute to dopaminergic cell death and development of PD. It still remains to be tested whether and how mutant GCase leads to aggregation of α-synuclein.
In the present work we show that expression of the mutant fly orthologs of GBA or expression of human mutant N370S or L444P proteins leads to death at early stages of the fly development. The flies expressing mutant GCase in the dopaminergic/serotonergic cells develop locomotion dysfunction, reminiscent of PD. This is the first animal model in which carriers of GD mutations develop parkisonian signs.
In a recent publication, parkin insolubility was associated with lack of degradation of ubiquitinated proteins and accumulation of α-synuclein and parkin in autophagosomes, suggesting autophagic defects in PD [67]. To test parkin’s role in mediating autophagic clearance, the authors used lentiviral gene transfer to express human wild type or mutant parkin (T240R) with α-synuclein in the rat striatum. Lentiviral expression of α-synuclein led to accumulation of autophagic vacuoles, while co-expression of parkin with α-synuclein facilitated autophagic clearance. Expression of parkin loss-of-function mutation did not affect autophagic clearance. Taken together, the data suggested that functional parkin regulates autophagosome clearance. It is possible, and remains to be proven, that the interaction between parkin and mutant GCase variants in dopaminergic cells attenuates normal autophagy, which leads to α-synuclein aggregation.
Our results strongly indicate a direct association between mutant GCase and development of Parkinsonian signs in the fly. However, there is another paradigm, arguing that insufficient lysosomal mutant GCase activity leads to substrate accumulation (GlcCer or glucosylsphingosine), α-synuclein aggregation, block in trafficking of GCase to lysosomes and development of PD [60, 62, 6870]. Thus, Sardi et al. showed that in brain sections derived from 12 months old D409V homozygous mice (but not from D409V heterozygous animals) there are α-synuclein and ubiquitin aggregates in the hippocampus, cerebral cortex and cerebellum. Memory deficits were detected in these mice at 6 months of age [68]. Administration of normal enzyme to the brain using gene therapy with an AAV derived vector, expressing a normal human GBA cDNA, significantly reduced the aggregation of ubiquitin and α-synuclein and ameliorated the memory deficit [71].
Development of PD in carriers of GD mutations implies that the presence of a mutant GBA allele is a dominant predisposing factor. This is a unique case of an autosomal recessive metabolic disease with a dominant element, namely the tendency of carriers of GD mutations to develop PD.
Dominance results either from haploinsufficiency or from gain of function. If haploinsufficiency accounts for the development of PD in carriers of GD mutations, it implies insufficient GCase activity in the dopaminergic neurons. Why is it not manifested in macrophages, in which case the disease would have been dominant? If, alternatively, the dominance results from gain of function, then its development depends on accumulation of enough deleterious product (mutant GCase, in our case), as in the case of Alzheimer disease, which displays age dependent accumulation of β-amyloid and tau, or Huntington disease, which exhibits accumulation of huntingtin [7274]. Our results suggest the gain of function alternative.

Conclusions

In this study we show that UPR is activated in GD patients as well as in carriers of GD mutations. In Drosophila models for carriers of different GD mutations, UPR is activated as well, and locomotion deficits are observed in the aging flies as a result of the presence of mutant GCase.
Written informed consent was obtained from the patients for publication of this paper.

Acknowledgements

We thank the “Cell Line and DNA Biobank from Patients Affected by Genetic Diseases” (G. Gaslini Institute), Telethon Genetic Biobank and Dr. R. O. Brady from NIH, MD, USA for GD skin fibroblasts or fibroblasts form carriers of GD mutations. This work was supported by a grant from the Israeli Ministry of Health (to MH) and the Telethon Genetic Biobank Network Project No. GTB07001A (to MF).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare no competing interests.

Authors’ contributions

GM designed the experiments, performed them and wrote the manuscript; SRL constructed the Drosophila expressing plasmids: MF supplied most human cell lines; HS supplied Drosophila materials and participated in manuscript preparation; DS participated in designing of experiments and in preparation of the manuscript; MH designed the experiments and participated in the manuscript preparation. All authors read and approved the final manuscript.
Anhänge

Electronic supplementary material

Additional file 1: Figure S1: CBE treatment does not induce UPR. A. To test the effect of substrate accumulation on induction of the UPR machinery, normal skin fibroblasts were treated with 200 mM of the GCase non-competitive inhibitor, CBE (Sigma-Aldrich, Israel) for 10 days. Enzymatic activity was tested after a day and 10 days of treatment, using 4-MUG as a substrate. B. Following 10 days of CBE treatment RNA was prepared from the cells and activation of UPR was tested by following CHOP and BiP mRNA, using quantitative RT-PCR with primers specific for human BiP (Human-GRP78-RT-F and Human-GRP78-RT-R, Table 1) or CHOP (Human-CHOP-RT-F and Human-CHOP-RT-R, Table 1). GAPDH was used as a normalizing control (amplified with primers: Human-GAPDH-RT-F and Human-GAPDH-RT-R, Table 1). As control for UPR activation, cells were treated for 3 hours with 150 nM of Thapsigargin (Sigma Aldrich, Israel), after which RNA was prepared and used for quantitative RT-PCR. Significance: * < 0.05; ** < 0.01. Dark box: CHOP; Light box: BiP. (PDF 23 KB)
Literatur
1.
2.
Zurück zum Zitat Jmoudiak M, Futerman AH: Gaucher disease: pathological mechanisms and modern management. Br J Haematol. 2005, 129: 178-188.PubMedCrossRef Jmoudiak M, Futerman AH: Gaucher disease: pathological mechanisms and modern management. Br J Haematol. 2005, 129: 178-188.PubMedCrossRef
3.
Zurück zum Zitat Hruska KS, LaMarca ME, Scott CR, Sidransky E: Gaucher disease: mutation and polymorphism spectrum in the glucocerebrosidase gene (GBA). Hum Mutat. 2008, 29: 567-583.PubMedCrossRef Hruska KS, LaMarca ME, Scott CR, Sidransky E: Gaucher disease: mutation and polymorphism spectrum in the glucocerebrosidase gene (GBA). Hum Mutat. 2008, 29: 567-583.PubMedCrossRef
4.
Zurück zum Zitat Tsuji S, Martin BM, Barranger JA, Stubblefield BK, LaMarca ME, Ginns EI: Genetic heterogeneity in type 1 Gaucher disease: multiple genotypes in Ashkenazic and non-Ashkenazic individuals. Proc Natl Acad Sci USA. 1988, 85: 2349-2352.PubMedCentralPubMedCrossRef Tsuji S, Martin BM, Barranger JA, Stubblefield BK, LaMarca ME, Ginns EI: Genetic heterogeneity in type 1 Gaucher disease: multiple genotypes in Ashkenazic and non-Ashkenazic individuals. Proc Natl Acad Sci USA. 1988, 85: 2349-2352.PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Tsuji S, Choudary PV, Martin BM, Stubblefield BK, Mayor JA, Barranger JA, Ginns EI: A mutation in the human glucocerebrosidase gene in neuronopathic Gaucher’s disease. N Engl J Med. 1987, 316: 570-575.PubMedCrossRef Tsuji S, Choudary PV, Martin BM, Stubblefield BK, Mayor JA, Barranger JA, Ginns EI: A mutation in the human glucocerebrosidase gene in neuronopathic Gaucher’s disease. N Engl J Med. 1987, 316: 570-575.PubMedCrossRef
6.
Zurück zum Zitat Beutler E, Gelbart T, Kuhl W, Sorge J, West C: Identification of the second common Jewish Gaucher disease mutation makes possible population-based screening for the heterozygous state. Proc Natl Acad Sci USA. 1991, 88: 10544-10547.PubMedCentralPubMedCrossRef Beutler E, Gelbart T, Kuhl W, Sorge J, West C: Identification of the second common Jewish Gaucher disease mutation makes possible population-based screening for the heterozygous state. Proc Natl Acad Sci USA. 1991, 88: 10544-10547.PubMedCentralPubMedCrossRef
7.
Zurück zum Zitat Erickson AH, Ginns EI, Barranger JA: Biosynthesis of the lysosomal enzyme glucocerebrosidase. J Biol Chem. 1985, 260: 14319-14324.PubMed Erickson AH, Ginns EI, Barranger JA: Biosynthesis of the lysosomal enzyme glucocerebrosidase. J Biol Chem. 1985, 260: 14319-14324.PubMed
9.
Zurück zum Zitat Walter P, Ron D: The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011, 334: 1081-1086.PubMedCrossRef Walter P, Ron D: The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011, 334: 1081-1086.PubMedCrossRef
11.
Zurück zum Zitat Kaufman RJ, Back SH, Song B, Han J, Hassler J: The unfolded protein response is required to maintain the integrity of the endoplasmic reticulum, prevent oxidative stress and preserve differentiation in beta-cells. Diabetes Obes Metab. 2010, 12 (Suppl 2): 99-107.PubMedCentralPubMedCrossRef Kaufman RJ, Back SH, Song B, Han J, Hassler J: The unfolded protein response is required to maintain the integrity of the endoplasmic reticulum, prevent oxidative stress and preserve differentiation in beta-cells. Diabetes Obes Metab. 2010, 12 (Suppl 2): 99-107.PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Badiola N, Penas C, Minano-Molina A, Barneda-Zahonero B, Fado R, Sanchez-Opazo G, Comella JX, Sabria J, Zhu C, Blomgren K, et al: Induction of ER stress in response to oxygen-glucose deprivation of cortical cultures involves the activation of the PERK and IRE-1 pathways and of caspase-12. Cell Death Dis. 2011, 2: e149.PubMedCentralPubMedCrossRef Badiola N, Penas C, Minano-Molina A, Barneda-Zahonero B, Fado R, Sanchez-Opazo G, Comella JX, Sabria J, Zhu C, Blomgren K, et al: Induction of ER stress in response to oxygen-glucose deprivation of cortical cultures involves the activation of the PERK and IRE-1 pathways and of caspase-12. Cell Death Dis. 2011, 2: e149.PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Foufelle F, Ferre P: [Unfolded protein response: its role in physiology and physiopathology]. Med Sci (Paris). 2007, 23: 291-296.CrossRef Foufelle F, Ferre P: [Unfolded protein response: its role in physiology and physiopathology]. Med Sci (Paris). 2007, 23: 291-296.CrossRef
14.
Zurück zum Zitat Benyair R, Ron E, Lederkremer GZ: Protein quality control, retention, and degradation at the endoplasmic reticulum. Int Rev Cell Mol Biol. 2011, 292: 197-280.PubMedCrossRef Benyair R, Ron E, Lederkremer GZ: Protein quality control, retention, and degradation at the endoplasmic reticulum. Int Rev Cell Mol Biol. 2011, 292: 197-280.PubMedCrossRef
15.
Zurück zum Zitat Janssens S, Tinel A, Lippens S, Tschopp J: PIDD mediates NF-kappaB activation in response to DNA damage. Cell. 2005, 123: 1079-1092.PubMedCrossRef Janssens S, Tinel A, Lippens S, Tschopp J: PIDD mediates NF-kappaB activation in response to DNA damage. Cell. 2005, 123: 1079-1092.PubMedCrossRef
17.
Zurück zum Zitat Wei H, Kim SJ, Zhang Z, Tsai PC, Wisniewski KE, Mukherjee AB: ER and oxidative stresses are common mediators of apoptosis in both neurodegenerative and non-neurodegenerative lysosomal storage disorders and are alleviated by chemical chaperones. Hum Mol Genet. 2008, 17: 469-477.PubMedCrossRef Wei H, Kim SJ, Zhang Z, Tsai PC, Wisniewski KE, Mukherjee AB: ER and oxidative stresses are common mediators of apoptosis in both neurodegenerative and non-neurodegenerative lysosomal storage disorders and are alleviated by chemical chaperones. Hum Mol Genet. 2008, 17: 469-477.PubMedCrossRef
18.
Zurück zum Zitat Lee YJ, Kim SJ, Heo TH: Protective effect of catechin in type I Gaucher disease cells by reducing endoplasmic reticulum stress. Biochem Biophys Res Commun. 2011, 413: 254-258.PubMedCrossRef Lee YJ, Kim SJ, Heo TH: Protective effect of catechin in type I Gaucher disease cells by reducing endoplasmic reticulum stress. Biochem Biophys Res Commun. 2011, 413: 254-258.PubMedCrossRef
19.
Zurück zum Zitat Farfel-Becker T, Vitner E, Dekel H, Leshem N, Enquist IB, Karlsson S, Futerman AH: No evidence for activation of the unfolded protein response in neuronopathic models of Gaucher disease. Hum Mol Genet. 2009, 18: 1482-1488.PubMedCrossRef Farfel-Becker T, Vitner E, Dekel H, Leshem N, Enquist IB, Karlsson S, Futerman AH: No evidence for activation of the unfolded protein response in neuronopathic models of Gaucher disease. Hum Mol Genet. 2009, 18: 1482-1488.PubMedCrossRef
20.
Zurück zum Zitat Ron I, Rapaport D, Horowitz M: Interaction between parkin and mutant glucocerebrosidase variants: a possible link between Parkinson disease and Gaucher disease. Hum Mol Genet. 2010, 19: 3771-3781.PubMedCrossRef Ron I, Rapaport D, Horowitz M: Interaction between parkin and mutant glucocerebrosidase variants: a possible link between Parkinson disease and Gaucher disease. Hum Mol Genet. 2010, 19: 3771-3781.PubMedCrossRef
21.
Zurück zum Zitat Ron I, Horowitz M: ER retention and degradation as the molecular basis underlying Gaucher disease heterogeneity. Hum Mol Genet. 2005, 14: 2387-2398.PubMedCrossRef Ron I, Horowitz M: ER retention and degradation as the molecular basis underlying Gaucher disease heterogeneity. Hum Mol Genet. 2005, 14: 2387-2398.PubMedCrossRef
22.
Zurück zum Zitat Ron I, Dagan A, Gatt S, Pasmanik-Chor M, Horowitz M: Use of fluorescent substrates for characterization of Gaucher disease mutations. Blood Cells Mol Dis. 2005, 35: 57-65.PubMedCrossRef Ron I, Dagan A, Gatt S, Pasmanik-Chor M, Horowitz M: Use of fluorescent substrates for characterization of Gaucher disease mutations. Blood Cells Mol Dis. 2005, 35: 57-65.PubMedCrossRef
23.
Zurück zum Zitat Bendikov-Bar I, Horowitz M: Gaucher disease paradigm: from ERAD to co-morbidity. Hum Mutat. 2012, 33: 1398-1407.PubMedCrossRef Bendikov-Bar I, Horowitz M: Gaucher disease paradigm: from ERAD to co-morbidity. Hum Mutat. 2012, 33: 1398-1407.PubMedCrossRef
24.
Zurück zum Zitat Mu TW, Ong DS, Wang YJ, Balch WE, Yates JR, Segatori L, Kelly JW: Chemical and biological approaches synergize to ameliorate protein-folding diseases. Cell. 2008, 134: 769-781.PubMedCentralPubMedCrossRef Mu TW, Ong DS, Wang YJ, Balch WE, Yates JR, Segatori L, Kelly JW: Chemical and biological approaches synergize to ameliorate protein-folding diseases. Cell. 2008, 134: 769-781.PubMedCentralPubMedCrossRef
25.
Zurück zum Zitat Davenport EL, Morgan GJ, Davies FE: Untangling the unfolded protein response. Cell Cycle. 2008, 7: 865-869.PubMedCrossRef Davenport EL, Morgan GJ, Davies FE: Untangling the unfolded protein response. Cell Cycle. 2008, 7: 865-869.PubMedCrossRef
26.
Zurück zum Zitat Sasagasako N, Kobayashi T, Yamaguchi Y, Shinnoh N, Goto I: Glucosylceramide and glucosylsphingosine metabolism in cultured fibroblasts deficient in acid beta-glucosidase activity. J Biochem. 1994, 115: 113-119.PubMed Sasagasako N, Kobayashi T, Yamaguchi Y, Shinnoh N, Goto I: Glucosylceramide and glucosylsphingosine metabolism in cultured fibroblasts deficient in acid beta-glucosidase activity. J Biochem. 1994, 115: 113-119.PubMed
28.
Zurück zum Zitat van Schadewijk A, Van’t Wout EF, Stolk J, Hiemstra PS: A quantitative method for detection of spliced X-box binding protein-1 (XBP1) mRNA as a measure of endoplasmic reticulum (ER) stress. Cell Stress Chaperones. 2012, 17: 275-279.PubMedCentralPubMedCrossRef van Schadewijk A, Van’t Wout EF, Stolk J, Hiemstra PS: A quantitative method for detection of spliced X-box binding protein-1 (XBP1) mRNA as a measure of endoplasmic reticulum (ER) stress. Cell Stress Chaperones. 2012, 17: 275-279.PubMedCentralPubMedCrossRef
29.
Zurück zum Zitat Bendikov-Bar I, Ron I, Filocamo M, Horowitz M: Characterization of the ERAD process of the L444P mutant glucocerebrosidase variant. Blood Cells Mol Dis. 2011, 46: 4-10.PubMedCrossRef Bendikov-Bar I, Ron I, Filocamo M, Horowitz M: Characterization of the ERAD process of the L444P mutant glucocerebrosidase variant. Blood Cells Mol Dis. 2011, 46: 4-10.PubMedCrossRef
30.
Zurück zum Zitat Ron I, Horowitz M: Intracellular cholesterol modifies the ERAD of glucocerebrosidase in Gaucher disease patients. Mol Genet Metab. 2008, 93: 426-436.PubMedCrossRef Ron I, Horowitz M: Intracellular cholesterol modifies the ERAD of glucocerebrosidase in Gaucher disease patients. Mol Genet Metab. 2008, 93: 426-436.PubMedCrossRef
31.
Zurück zum Zitat Tayebi N, Callahan M, Madike V, Stubblefield BK, Orvisky E, Krasnewich D, Fillano JJ, Sidransky E: Gaucher disease and parkinsonism: a phenotypic and genotypic characterization. Mol Genet Metab. 2001, 73: 313-321.PubMedCrossRef Tayebi N, Callahan M, Madike V, Stubblefield BK, Orvisky E, Krasnewich D, Fillano JJ, Sidransky E: Gaucher disease and parkinsonism: a phenotypic and genotypic characterization. Mol Genet Metab. 2001, 73: 313-321.PubMedCrossRef
32.
Zurück zum Zitat Tayebi N, Walker J, Stubblefield B, Orvisky E, LaMarca ME, Wong K, Rosenbaum H, Schiffmann R, Bembi B, Sidransky E: Gaucher disease with parkinsonian manifestations: does glucocerebrosidase deficiency contribute to a vulnerability to parkinsonism?. Mol Genet Metab. 2003, 79: 104-109.PubMedCrossRef Tayebi N, Walker J, Stubblefield B, Orvisky E, LaMarca ME, Wong K, Rosenbaum H, Schiffmann R, Bembi B, Sidransky E: Gaucher disease with parkinsonian manifestations: does glucocerebrosidase deficiency contribute to a vulnerability to parkinsonism?. Mol Genet Metab. 2003, 79: 104-109.PubMedCrossRef
33.
Zurück zum Zitat Varkonyi J, Rosenbaum H, Baumann N, MacKenzie JJ, Simon Z, Aharon-Peretz J, Walker JM, Tayebi N, Sidransky E: Gaucher disease associated with parkinsonism: four further case reports. Am J Med Genet A. 2003, 116A: 348-351.PubMedCrossRef Varkonyi J, Rosenbaum H, Baumann N, MacKenzie JJ, Simon Z, Aharon-Peretz J, Walker JM, Tayebi N, Sidransky E: Gaucher disease associated with parkinsonism: four further case reports. Am J Med Genet A. 2003, 116A: 348-351.PubMedCrossRef
34.
Zurück zum Zitat Bembi B, Zambito Marsala S, Sidransky E, Ciana G, Carrozzi M, Zorzon M, Martini C, Gioulis M, Pittis MG, Capus L: Gaucher’s disease with Parkinson’s disease: clinical and pathological aspects. Neurology. 2003, 61: 99-101.PubMedCrossRef Bembi B, Zambito Marsala S, Sidransky E, Ciana G, Carrozzi M, Zorzon M, Martini C, Gioulis M, Pittis MG, Capus L: Gaucher’s disease with Parkinson’s disease: clinical and pathological aspects. Neurology. 2003, 61: 99-101.PubMedCrossRef
35.
Zurück zum Zitat Aharon-Peretz J, Rosenbaum H, Gershoni-Baruch R: Mutations in the glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2004, 351: 1972-1977.PubMedCrossRef Aharon-Peretz J, Rosenbaum H, Gershoni-Baruch R: Mutations in the glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2004, 351: 1972-1977.PubMedCrossRef
36.
Zurück zum Zitat Goker-Alpan O, Schiffmann R, LaMarca ME, Nussbaum RL, McInerney-Leo A, Sidransky E: Parkinsonism among Gaucher disease carriers. J Med Genet. 2004, 41: 937-940.PubMedCentralPubMedCrossRef Goker-Alpan O, Schiffmann R, LaMarca ME, Nussbaum RL, McInerney-Leo A, Sidransky E: Parkinsonism among Gaucher disease carriers. J Med Genet. 2004, 41: 937-940.PubMedCentralPubMedCrossRef
37.
Zurück zum Zitat Lwin A, Orvisky E, Goker-Alpan O, LaMarca ME, Sidransky E: Glucocerebrosidase mutations in subjects with parkinsonism. Mol Genet Metab. 2004, 81: 70-73.PubMedCrossRef Lwin A, Orvisky E, Goker-Alpan O, LaMarca ME, Sidransky E: Glucocerebrosidase mutations in subjects with parkinsonism. Mol Genet Metab. 2004, 81: 70-73.PubMedCrossRef
38.
Zurück zum Zitat Clark LN, Nicolai A, Afridi S, Harris J, Mejia-Santana H, Strug L, Cote LJ, Louis ED, Andrews H, Waters C, et al: Pilot association study of the beta-glucocerebrosidase N370S allele and Parkinson’s disease in subjects of Jewish ethnicity. Mov Disord. 2005, 20: 100-103.PubMedCrossRef Clark LN, Nicolai A, Afridi S, Harris J, Mejia-Santana H, Strug L, Cote LJ, Louis ED, Andrews H, Waters C, et al: Pilot association study of the beta-glucocerebrosidase N370S allele and Parkinson’s disease in subjects of Jewish ethnicity. Mov Disord. 2005, 20: 100-103.PubMedCrossRef
39.
Zurück zum Zitat Eblan MJ, Walker JM, Sidransky E: The glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2005, 352: 728-731. author reply 728–731.PubMedCrossRef Eblan MJ, Walker JM, Sidransky E: The glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2005, 352: 728-731. author reply 728–731.PubMedCrossRef
40.
Zurück zum Zitat Sato C, Morgan A, Lang AE, Salehi-Rad S, Kawarai T, Meng Y, Ray PN, Farrer LA, St George-Hyslop P, Rogaeva E: Analysis of the glucocerebrosidase gene in Parkinson’s disease. Mov Disord. 2005, 20: 367-370.PubMedCrossRef Sato C, Morgan A, Lang AE, Salehi-Rad S, Kawarai T, Meng Y, Ray PN, Farrer LA, St George-Hyslop P, Rogaeva E: Analysis of the glucocerebrosidase gene in Parkinson’s disease. Mov Disord. 2005, 20: 367-370.PubMedCrossRef
41.
Zurück zum Zitat Schlossmacher MG, Cullen V, Muthing J: The glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2005, 352: 728-731. author reply 728–731.PubMedCrossRef Schlossmacher MG, Cullen V, Muthing J: The glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2005, 352: 728-731. author reply 728–731.PubMedCrossRef
42.
43.
Zurück zum Zitat Zimran A, Neudorfer O, Elstein D: The glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2005, 352: 728-731. author reply 728–731.PubMedCrossRef Zimran A, Neudorfer O, Elstein D: The glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med. 2005, 352: 728-731. author reply 728–731.PubMedCrossRef
44.
Zurück zum Zitat Eblan MJ, Scholz S, Stubblefield B, Gutti U, Goker-Alpan O, Hruska KS, Singleton AB, Sidransky E: Glucocerebrosidase mutations are not found in association with LRRK2 G2019S in subjects with parkinsonism. Neurosci Lett. 2006, 404: 163-165.PubMedCrossRef Eblan MJ, Scholz S, Stubblefield B, Gutti U, Goker-Alpan O, Hruska KS, Singleton AB, Sidransky E: Glucocerebrosidase mutations are not found in association with LRRK2 G2019S in subjects with parkinsonism. Neurosci Lett. 2006, 404: 163-165.PubMedCrossRef
45.
Zurück zum Zitat Goker-Alpan O, Giasson BI, Eblan MJ, Nguyen J, Hurtig HI, Lee VM, Trojanowski JQ, Sidransky E: Glucocerebrosidase mutations are an important risk factor for Lewy body disorders. Neurology. 2006, 67: 908-910.PubMedCrossRef Goker-Alpan O, Giasson BI, Eblan MJ, Nguyen J, Hurtig HI, Lee VM, Trojanowski JQ, Sidransky E: Glucocerebrosidase mutations are an important risk factor for Lewy body disorders. Neurology. 2006, 67: 908-910.PubMedCrossRef
46.
Zurück zum Zitat Itokawa K, Tamura N, Kawai N, Shimazu K, Ishii K: Parkinsonism in type I Gaucher’s disease. Intern Med. 2006, 45: 1165-1167.PubMedCrossRef Itokawa K, Tamura N, Kawai N, Shimazu K, Ishii K: Parkinsonism in type I Gaucher’s disease. Intern Med. 2006, 45: 1165-1167.PubMedCrossRef
47.
48.
Zurück zum Zitat Clark LN, Ross BM, Wang Y, Mejia-Santana H, Harris J, Louis ED, Cote LJ, Andrews H, Fahn S, Waters C, et al: Mutations in the glucocerebrosidase gene are associated with early-onset Parkinson disease. Neurology. 2007, 69: 1270-1277.PubMedCentralPubMedCrossRef Clark LN, Ross BM, Wang Y, Mejia-Santana H, Harris J, Louis ED, Cote LJ, Andrews H, Fahn S, Waters C, et al: Mutations in the glucocerebrosidase gene are associated with early-onset Parkinson disease. Neurology. 2007, 69: 1270-1277.PubMedCentralPubMedCrossRef
49.
Zurück zum Zitat Tan EK, Tong J, Fook-Chong S, Yih Y, Wong MC, Pavanni R, Zhao Y: Glucocerebrosidase mutations and risk of Parkinson disease in Chinese patients. Arch Neurol. 2007, 64: 1056-1058.PubMedCrossRef Tan EK, Tong J, Fook-Chong S, Yih Y, Wong MC, Pavanni R, Zhao Y: Glucocerebrosidase mutations and risk of Parkinson disease in Chinese patients. Arch Neurol. 2007, 64: 1056-1058.PubMedCrossRef
50.
Zurück zum Zitat Goker-Alpan O, Lopez G, Vithayathil J, Davis J, Hallett M, Sidransky E: The spectrum of parkinsonian manifestations associated with glucocerebrosidase mutations. Arch Neurol. 2008, 65: 1353-1357.PubMedCentralPubMedCrossRef Goker-Alpan O, Lopez G, Vithayathil J, Davis J, Hallett M, Sidransky E: The spectrum of parkinsonian manifestations associated with glucocerebrosidase mutations. Arch Neurol. 2008, 65: 1353-1357.PubMedCentralPubMedCrossRef
51.
Zurück zum Zitat Mitsui J, Mizuta I, Toyoda A, Ashida R, Takahashi Y, Goto J, Fukuda Y, Date H, Iwata A, Yamamoto M, et al: Mutations for Gaucher disease confer high susceptibility to Parkinson disease. Arch Neurol. 2009, 66: 571-576.PubMedCrossRef Mitsui J, Mizuta I, Toyoda A, Ashida R, Takahashi Y, Goto J, Fukuda Y, Date H, Iwata A, Yamamoto M, et al: Mutations for Gaucher disease confer high susceptibility to Parkinson disease. Arch Neurol. 2009, 66: 571-576.PubMedCrossRef
52.
Zurück zum Zitat Sidransky E, Samaddar T, Tayebi N: Mutations in GBA are associated with familial Parkinson disease susceptibility and age at onset. Neurology. 2009, 73: 1424-1425. author reply 1425–1426.PubMedCrossRef Sidransky E, Samaddar T, Tayebi N: Mutations in GBA are associated with familial Parkinson disease susceptibility and age at onset. Neurology. 2009, 73: 1424-1425. author reply 1425–1426.PubMedCrossRef
53.
Zurück zum Zitat Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER, Bar-Shira A, Berg D, Bras J, Brice A, et al: Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. N Engl J Med. 2009, 361: 1651-1661.PubMedCentralPubMedCrossRef Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER, Bar-Shira A, Berg D, Bras J, Brice A, et al: Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. N Engl J Med. 2009, 361: 1651-1661.PubMedCentralPubMedCrossRef
54.
Zurück zum Zitat Lesage S, Condroyer C, Hecham N, Anheim M, Belarbi S, Lohman E, Viallet F, Pollak P, Abada M, Durr A, et al: Mutations in the glucocerebrosidase gene confer a risk for Parkinson disease in North Africa. Neurology. 2011, 76: 301-303.PubMedCrossRef Lesage S, Condroyer C, Hecham N, Anheim M, Belarbi S, Lohman E, Viallet F, Pollak P, Abada M, Durr A, et al: Mutations in the glucocerebrosidase gene confer a risk for Parkinson disease in North Africa. Neurology. 2011, 76: 301-303.PubMedCrossRef
55.
Zurück zum Zitat Lesage S, Anheim M, Condroyer C, Pollak P, Durif F, Dupuits C, Viallet F, Lohmann E, Corvol JC, Honore A, et al: Large-scale screening of the Gaucher’s disease-related glucocerebrosidase gene in Europeans with Parkinson’s disease. Hum Mol Genet. 2011, 20: 202-210.PubMedCrossRef Lesage S, Anheim M, Condroyer C, Pollak P, Durif F, Dupuits C, Viallet F, Lohmann E, Corvol JC, Honore A, et al: Large-scale screening of the Gaucher’s disease-related glucocerebrosidase gene in Europeans with Parkinson’s disease. Hum Mol Genet. 2011, 20: 202-210.PubMedCrossRef
56.
Zurück zum Zitat Feany MB, Bender WW: A Drosophila model of Parkinson’s disease. Nature. 2000, 404: 394-398.PubMedCrossRef Feany MB, Bender WW: A Drosophila model of Parkinson’s disease. Nature. 2000, 404: 394-398.PubMedCrossRef
58.
Zurück zum Zitat Yam GH, Bosshard N, Zuber C, Steinmann B, Roth J: Pharmacological chaperone corrects lysosomal storage in Fabry disease caused by trafficking-incompetent variants. Am J Physiol Cell Physiol. 2006, 290: C1076-C1082.PubMedCrossRef Yam GH, Bosshard N, Zuber C, Steinmann B, Roth J: Pharmacological chaperone corrects lysosomal storage in Fabry disease caused by trafficking-incompetent variants. Am J Physiol Cell Physiol. 2006, 290: C1076-C1082.PubMedCrossRef
59.
Zurück zum Zitat Klein A, Mosqueira M, Martinez G, Robledo F, Gonzalez M, Caballero B, Cancino GI, Alvarez AR, Hetz C, Zanlungo S: Lack of activation of the unfolded protein response in mouse and cellular models of Niemann-Pick type C disease. Neurodegener Dis. 2011, 8: 124-128.PubMedCrossRef Klein A, Mosqueira M, Martinez G, Robledo F, Gonzalez M, Caballero B, Cancino GI, Alvarez AR, Hetz C, Zanlungo S: Lack of activation of the unfolded protein response in mouse and cellular models of Niemann-Pick type C disease. Neurodegener Dis. 2011, 8: 124-128.PubMedCrossRef
60.
Zurück zum Zitat Mazzulli JR, Xu YH, Sun Y, Knight AL, McLean PJ, Caldwell GA, Sidransky E, Grabowski GA, Krainc D: Gaucher disease glucocerebrosidase and alpha-synuclein form a bidirectional pathogenic loop in synucleinopathies. Cell. 2011, 146: 37-52.PubMedCentralPubMedCrossRef Mazzulli JR, Xu YH, Sun Y, Knight AL, McLean PJ, Caldwell GA, Sidransky E, Grabowski GA, Krainc D: Gaucher disease glucocerebrosidase and alpha-synuclein form a bidirectional pathogenic loop in synucleinopathies. Cell. 2011, 146: 37-52.PubMedCentralPubMedCrossRef
61.
Zurück zum Zitat Clark LN, Kartsaklis LA, Wolf Gilbert R, Dorado B, Ross BM, Kisselev S, Verbitsky M, Mejia-Santana H, Cote LJ, Andrews H, et al: Association of glucocerebrosidase mutations with dementia with lewy bodies. Arch Neurol. 2009, 66: 578-583.PubMedCentralPubMedCrossRef Clark LN, Kartsaklis LA, Wolf Gilbert R, Dorado B, Ross BM, Kisselev S, Verbitsky M, Mejia-Santana H, Cote LJ, Andrews H, et al: Association of glucocerebrosidase mutations with dementia with lewy bodies. Arch Neurol. 2009, 66: 578-583.PubMedCentralPubMedCrossRef
62.
Zurück zum Zitat Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, Kolodziej P, Kahn I, Saftig P, Woulfe J, et al: Acid beta-glucosidase mutants linked to Gaucher disease, Parkinson disease, and Lewy body dementia alter alpha-synuclein processing. Ann Neurol. 2011, 69: 940-953.PubMedCrossRef Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, Kolodziej P, Kahn I, Saftig P, Woulfe J, et al: Acid beta-glucosidase mutants linked to Gaucher disease, Parkinson disease, and Lewy body dementia alter alpha-synuclein processing. Ann Neurol. 2011, 69: 940-953.PubMedCrossRef
63.
Zurück zum Zitat Shin JH, Ko HS, Kang H, Lee Y, Lee YI, Pletinkova O, Troconso JC, Dawson VL, Dawson TM: PARIS (ZNF746) repression of PGC-1alpha contributes to neurodegeneration in Parkinson’s disease. Cell. 2011, 144: 689-702.PubMedCentralPubMedCrossRef Shin JH, Ko HS, Kang H, Lee Y, Lee YI, Pletinkova O, Troconso JC, Dawson VL, Dawson TM: PARIS (ZNF746) repression of PGC-1alpha contributes to neurodegeneration in Parkinson’s disease. Cell. 2011, 144: 689-702.PubMedCentralPubMedCrossRef
64.
Zurück zum Zitat Kemeny S, Dery D, Loboda Y, Rovner M, Lev T, Zuri D, Finberg JP, Larisch S: Parkin promotes degradation of the mitochondrial pro-apoptotic ARTS protein. PLoS One. 2012, 7: e38837.PubMedCentralPubMedCrossRef Kemeny S, Dery D, Loboda Y, Rovner M, Lev T, Zuri D, Finberg JP, Larisch S: Parkin promotes degradation of the mitochondrial pro-apoptotic ARTS protein. PLoS One. 2012, 7: e38837.PubMedCentralPubMedCrossRef
65.
Zurück zum Zitat Lindholm D, Eriksson O, Makela J, Belluardo N, Korhonen L: PGC-1alpha: a master gene that is hard to master. Cell Mol Life Sci. 2012, 69: 2465-2468.PubMedCrossRef Lindholm D, Eriksson O, Makela J, Belluardo N, Korhonen L: PGC-1alpha: a master gene that is hard to master. Cell Mol Life Sci. 2012, 69: 2465-2468.PubMedCrossRef
66.
Zurück zum Zitat Larisch S, Yi Y, Lotan R, Kerner H, Eimerl S, Tony Parks W, Gottfried Y, Birkey Reffey S, de Caestecker MP, Danielpour D, et al: A novel mitochondrial septin-like protein, ARTS, mediates apoptosis dependent on its P-loop motif. Nat Cell Biol. 2000, 2: 915-921.PubMedCrossRef Larisch S, Yi Y, Lotan R, Kerner H, Eimerl S, Tony Parks W, Gottfried Y, Birkey Reffey S, de Caestecker MP, Danielpour D, et al: A novel mitochondrial septin-like protein, ARTS, mediates apoptosis dependent on its P-loop motif. Nat Cell Biol. 2000, 2: 915-921.PubMedCrossRef
67.
Zurück zum Zitat Lonskaya I, Hebron ML, Algarzae NK, Desforges N, Moussa CE: Decreased parkin solubility is associated with impairment of autophagy in the nigrostriatum of sporadic Parkinson’s disease. Neuroscience. 2012, 232C: 90. Lonskaya I, Hebron ML, Algarzae NK, Desforges N, Moussa CE: Decreased parkin solubility is associated with impairment of autophagy in the nigrostriatum of sporadic Parkinson’s disease. Neuroscience. 2012, 232C: 90.
68.
Zurück zum Zitat Sardi SP, Clarke J, Kinnecom C, Tamsett TJ, Li L, Stanek LM, Passini MA, Grabowski GA, Schlossmacher MG, Sidman RL, et al: CNS expression of glucocerebrosidase corrects {alpha}-synuclein pathology and memory in a mouse model of Gaucher-related synucleinopathy. Proc Natl Acad Sci USA. 2011, 108: 12101-12106.PubMedCentralPubMedCrossRef Sardi SP, Clarke J, Kinnecom C, Tamsett TJ, Li L, Stanek LM, Passini MA, Grabowski GA, Schlossmacher MG, Sidman RL, et al: CNS expression of glucocerebrosidase corrects {alpha}-synuclein pathology and memory in a mouse model of Gaucher-related synucleinopathy. Proc Natl Acad Sci USA. 2011, 108: 12101-12106.PubMedCentralPubMedCrossRef
69.
Zurück zum Zitat Xu YH, Sun Y, Ran H, Quinn B, Witte D, Grabowski GA: Accumulation and distribution of alpha-synuclein and ubiquitin in the CNS of Gaucher disease mouse models. Mol Genet Metab. 2011, 102: 436-447.PubMedCentralPubMedCrossRef Xu YH, Sun Y, Ran H, Quinn B, Witte D, Grabowski GA: Accumulation and distribution of alpha-synuclein and ubiquitin in the CNS of Gaucher disease mouse models. Mol Genet Metab. 2011, 102: 436-447.PubMedCentralPubMedCrossRef
70.
Zurück zum Zitat Sardi SP, Singh P, Cheng SH, Shihabuddin LS, Schlossmacher MG: Mutant GBA1 expression and synucleinopathy risk: first insights from cellular and mouse models. Neurodegener Dis. 2012, 10: 195-202.PubMedCrossRef Sardi SP, Singh P, Cheng SH, Shihabuddin LS, Schlossmacher MG: Mutant GBA1 expression and synucleinopathy risk: first insights from cellular and mouse models. Neurodegener Dis. 2012, 10: 195-202.PubMedCrossRef
71.
Zurück zum Zitat Sardi SP, Clarke J, Viel C, Chan M, Tamsett TJ, Treleaven CM, Bu J, Sweet L, Passini MA, Dodge JC, et al: Augmenting CNS glucocerebrosidase activity as a therapeutic strategy for parkinsonism and other Gaucher-related synucleinopathies. Proc Natl Acad Sci USA. 2013, 110: 3537-3542.PubMedCentralPubMedCrossRef Sardi SP, Clarke J, Viel C, Chan M, Tamsett TJ, Treleaven CM, Bu J, Sweet L, Passini MA, Dodge JC, et al: Augmenting CNS glucocerebrosidase activity as a therapeutic strategy for parkinsonism and other Gaucher-related synucleinopathies. Proc Natl Acad Sci USA. 2013, 110: 3537-3542.PubMedCentralPubMedCrossRef
72.
Zurück zum Zitat Zheng Z, Diamond MI: Huntington disease and the huntingtin protein. Prog Mol Biol Transl Sci. 2012, 107: 189-214.PubMedCrossRef Zheng Z, Diamond MI: Huntington disease and the huntingtin protein. Prog Mol Biol Transl Sci. 2012, 107: 189-214.PubMedCrossRef
73.
Zurück zum Zitat Halliday GM, Holton JL, Revesz T, Dickson DW: Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 2011, 122: 187-204.PubMedCrossRef Halliday GM, Holton JL, Revesz T, Dickson DW: Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 2011, 122: 187-204.PubMedCrossRef
74.
Zurück zum Zitat Seeman P, Seeman N: Alzheimer’s disease: beta-amyloid plaque formation in human brain. Synapse. 2011, 65: 1289-1297.PubMedCrossRef Seeman P, Seeman N: Alzheimer’s disease: beta-amyloid plaque formation in human brain. Synapse. 2011, 65: 1289-1297.PubMedCrossRef
Metadaten
Titel
Unfolded protein response in Gaucher disease: from human to Drosophila
verfasst von
Gali Maor
Sigal Rencus-Lazar
Mirella Filocamo
Hermann Steller
Daniel Segal
Mia Horowitz
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Orphanet Journal of Rare Diseases / Ausgabe 1/2013
Elektronische ISSN: 1750-1172
DOI
https://doi.org/10.1186/1750-1172-8-140

Weitere Artikel der Ausgabe 1/2013

Orphanet Journal of Rare Diseases 1/2013 Zur Ausgabe