Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Aging Cell ; 23(5): e14128, 2024 May.
Article in English | MEDLINE | ID: mdl-38415292

ABSTRACT

Parkinson's disease (PD) is characterized by aggregation of α-synuclein (α-syn) into protein inclusions in degenerating brains. Increasing amounts of aggregated α-syn species indicate significant perturbation of cellular proteostasis. Altered proteostasis depends on α-syn protein levels and the impact of α-syn on other components of the proteostasis network. Budding yeast Saccharomyces cerevisiae was used as eukaryotic reference organism to study the consequences of α-syn expression on protein dynamics. To address this, we investigated the impact of overexpression of α-syn and S129A variant on the abundance and stability of most yeast proteins using a genome-wide yeast library and a tandem fluorescent protein timer (tFT) reporter as a measure for protein stability. This revealed that the stability of in total 377 cellular proteins was altered by α-syn expression, and that the impact on protein stability was significantly enhanced by phosphorylation at Ser129 (pS129). The proteasome assembly chaperone Rpn14 was identified as one of the top candidates for increased protein stability by expression of pS129 α-syn. Elevated levels of Rpn14 enhanced the growth inhibition by α-syn and the accumulation of ubiquitin conjugates in the cell. We found that Rpn14 interacts physically with α-syn and stabilizes pS129 α-syn. The expression of α-syn along with elevated levels of Rpn14 or its human counterpart PAAF1 reduced the proteasome activity in yeast and in human cells, supporting that pS129 α-syn negatively affects the 26S proteasome through Rpn14. This comprehensive study into the alternations of protein homeostasis highlights the critical role of the Rpn14/PAAF1 in α-syn-mediated proteasome dysfunction.


Subject(s)
Proteasome Endopeptidase Complex , Saccharomyces cerevisiae , alpha-Synuclein , alpha-Synuclein/metabolism , Proteasome Endopeptidase Complex/metabolism , Humans , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Molecular Chaperones/metabolism , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics
2.
J Mol Biol ; 435(4): 167935, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36586462

ABSTRACT

Golgi-Associated plant Pathogenesis Related protein 1 (GAPR-1) acts as a negative regulator of autophagy by interacting with Beclin 1 at Golgi membranes in mammalian cells. The molecular mechanism of this interaction is largely unknown. We recently showed that human GAPR-1 (hGAPR-1) has amyloidogenic properties resulting in the formation of protein condensates upon overexpression in Saccharomyces cerevisiae. Here we show that human Beclin 1 (hBeclin 1) has several predicted amyloidogenic regions and that overexpression of hBeclin 1-mCherry in yeast also results in the formation of fluorescent protein condensates. Surprisingly, co-expression of hGAPR-1-GFP and hBeclin 1-mCherry results in a strong reduction of hBeclin 1 condensates. Mutations of the known interaction site on the hGAPR-1 and hBeclin 1 surface abolished the effect on condensate formation during co-expression without affecting the condensate formation properties of the individual proteins. Similarly, a hBeclin 1-derived B18 peptide that is known to bind hGAPR-1 and to interfere with the interaction between hGAPR-1 and hBeclin 1, abolished the reduction of hBeclin 1 condensates by co-expression of hGAPR-1. These results indicate that the same type of protein-protein interactions interfere with condensate formation during co-expression of hGAPR-1 and hBeclin 1 as previously described for their interaction at Golgi membranes. The amyloidogenic properties of the B18 peptide were, however, important for the interaction with hGAPR-1, as mutant peptides with reduced amyloidogenic properties also showed reduced interaction with hGAPR-1 and reduced interference with hGAPR-1/hBeclin 1 condensate formation. We propose that amyloidogenic interactions take place between hGAPR-1 and hBeclin 1 prior to condensate formation.


Subject(s)
Amyloidogenic Proteins , Beclin-1 , Membrane Proteins , Protein Interaction Mapping , Animals , Humans , Beclin-1/genetics , Beclin-1/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Saccharomyces cerevisiae , Mutation , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/metabolism , Protein Multimerization , Protein Interaction Domains and Motifs
3.
Sci Rep ; 12(1): 9625, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35688911

ABSTRACT

Heterologous protein expression is an important method for analysing cellular functions of proteins, in genetic circuit engineering and in overexpressing proteins for biopharmaceutical applications and structural biology research. The degeneracy of the genetic code, which enables a single protein to be encoded by a multitude of synonymous gene sequences, plays an important role in regulating protein expression, but substantial uncertainty exists concerning the details of this phenomenon. Here we analyse the influence of a profiled codon usage adaptation approach on protein expression levels in the eukaryotic model organism Saccharomyces cerevisiae. We selected green fluorescent protein (GFP) and human α-synuclein (αSyn) as representatives for stable and intrinsically disordered proteins and representing a benchmark and a challenging test case. A new approach was implemented to design typical genes resembling the codon usage of any subset of endogenous genes. Using this approach, synthetic genes for GFP and αSyn were generated, heterologously expressed and evaluated in yeast. We demonstrate that GFP is expressed at high levels, and that the toxic αSyn can be adapted to endogenous, low-level expression. The new software is publicly available as a web-application for performing host-specific protein adaptations to a set of the most commonly used model organisms ( https://odysseus.motorprotein.de ).


Subject(s)
Saccharomyces cerevisiae , Software , Codon/genetics , Gene Expression , Green Fluorescent Proteins/genetics , Humans , Saccharomyces cerevisiae/genetics
4.
Cells ; 10(9)2021 08 28.
Article in English | MEDLINE | ID: mdl-34571878

ABSTRACT

Parkinson's disease (PD) is the most prevalent movement disorder characterized with loss of dopaminergic neurons in the brain. One of the pathological hallmarks of the disease is accumulation of aggregated α-synuclein (αSyn) in cytoplasmic Lewy body inclusions that indicates significant dysfunction of protein homeostasis in PD. Accumulation is accompanied with highly elevated S129 phosphorylation, suggesting that this posttranslational modification is linked to pathogenicity and altered αSyn inclusion dynamics. To address the role of S129 phosphorylation on protein dynamics further we investigated the wild type and S129A variants using yeast and a tandem fluorescent timer protein reporter approach to monitor protein turnover and stability. Overexpression of both variants leads to inhibited yeast growth. Soluble S129A is more stable and additional Y133F substitution permits αSyn degradation in a phosphorylation-independent manner. Quantitative cellular proteomics revealed significant αSyn-dependent disturbances of the cellular protein homeostasis, which are increased upon S129 phosphorylation. Disturbances are characterized by decreased abundance of the ubiquitin-dependent protein degradation machinery. Biotin proximity labelling revealed that αSyn interacts with the Rpt2 base subunit. Proteasome subunit depletion by reducing the expression of the corresponding genes enhances αSyn toxicity. Our studies demonstrate that turnover of αSyn and depletion of the proteasome pool correlate in a complex relationship between altered proteasome composition and increased αSyn toxicity.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitin/metabolism , alpha-Synuclein/metabolism , Chromatography, High Pressure Liquid , Down-Regulation , Genotype , Humans , Mass Spectrometry , Mutagenesis, Site-Directed , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation , Proteasome Endopeptidase Complex/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Proteome/analysis , Saccharomyces cerevisiae/genetics , alpha-Synuclein/genetics
5.
J Mol Biol ; 433(19): 167162, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34298062

ABSTRACT

Many proteins that can assemble into higher order structures termed amyloids can also concentrate into cytoplasmic inclusions via liquid-liquid phase separation. Here, we study the assembly of human Golgi-Associated plant Pathogenesis Related protein 1 (GAPR-1), an amyloidogenic protein of the Cysteine-rich secretory proteins, Antigen 5, and Pathogenesis-related 1 proteins (CAP) protein superfamily, into cytosolic inclusions in Saccharomyces cerevisiae. Overexpression of GAPR-1-GFP results in the formation GAPR-1 oligomers and fluorescent inclusions in yeast cytosol. These cytosolic inclusions are dynamic and reversible organelles that gradually increase during time of overexpression and decrease after promoter shut-off. Inclusion formation is, however, a regulated process that is influenced by factors other than protein expression levels. We identified N-myristoylation of GAPR-1 as an important determinant at early stages of inclusion formation. In addition, mutations in the conserved metal-binding site (His54 and His103) enhanced inclusion formation, suggesting that these residues prevent uncontrolled protein sequestration. In agreement with this, we find that addition of Zn2+ metal ions enhances inclusion formation. Furthermore, Zn2+ reduces GAPR-1 protein degradation, which indicates stabilization of GAPR-1 in inclusions. We propose that the properties underlying both the amyloidogenic properties and the reversible sequestration of GAPR-1 into inclusions play a role in the biological function of GAPR-1 and other CAP family members.


Subject(s)
Inclusion Bodies/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Crystallography, X-Ray , Cytosol/chemistry , Cytosol/metabolism , Humans , Membrane Proteins/genetics , Protein Aggregates , Protein Conformation , Protein Domains , Protein Engineering , Proteolysis , Saccharomyces cerevisiae/genetics , Zinc/metabolism
6.
Front Mol Neurosci ; 14: 659926, 2021.
Article in English | MEDLINE | ID: mdl-33912013

ABSTRACT

Aggregation of α-synuclein (αSyn) into proteinaceous deposits is a pathological hallmark of a range of neurodegenerative diseases including Parkinson's disease (PD). Numerous lines of evidence indicate that the accumulation of toxic oligomeric and prefibrillar αSyn species may underpin the cellular toxicity and spread of pathology between cells. Therefore, aggregation of αSyn is considered a priority target for drug development, as aggregation inhibitors are expected to reduce αSyn toxicity and serve as therapeutic agents. Here, we used the budding yeast S. cerevisiae as a platform for the identification of short peptides that inhibit αSyn aggregation and toxicity. A library consisting of approximately one million peptide variants was utilized in two high-throughput screening approaches for isolation of library representatives that reduce αSyn-associated toxicity and aggregation. Seven peptides were isolated that were able to suppress specifically αSyn toxicity and aggregation in living cells. Expression of the peptides in yeast reduced the accumulation of αSyn-induced reactive oxygen species and increased cell viability. Next, the peptides were chemically synthesized and probed for their ability to modulate αSyn aggregation in vitro. Two synthetic peptides, K84s and K102s, of 25 and 19 amino acids, respectively, significantly inhibited αSyn oligomerization and aggregation at sub-stoichiometric molar ratios. Importantly, K84s reduced αSyn aggregation in human cells. These peptides represent promising αSyn aggregation antagonists for the development of future therapeutic interventions.

7.
PLoS Genet ; 17(3): e1009407, 2021 03.
Article in English | MEDLINE | ID: mdl-33657088

ABSTRACT

Parkinson's disease is a neurodegenerative disorder associated with misfolding and aggregation of α-synuclein as a hallmark protein. Two yeast strain collections comprising conditional alleles of essential genes were screened for the ability of each allele to reduce or improve yeast growth upon α-synuclein expression. The resulting 98 novel modulators of α-synuclein toxicity clustered in several major categories including transcription, rRNA processing and ribosome biogenesis, RNA metabolism and protein degradation. Furthermore, expression of α-synuclein caused alterations in pre-rRNA transcript levels in yeast and in human cells. We identified the nucleolar DEAD-box helicase Dbp4 as a prominent modulator of α-synuclein toxicity. Downregulation of DBP4 rescued cells from α-synuclein toxicity, whereas overexpression led to a synthetic lethal phenotype. We discovered that α-synuclein interacts with Dbp4 or its human ortholog DDX10, sequesters the protein outside the nucleolus in yeast and in human cells, and stabilizes a fraction of α-synuclein oligomeric species. These findings provide a novel link between nucleolar processes and α-synuclein mediated toxicity with DDX10 emerging as a promising drug target.


Subject(s)
DEAD-box RNA Helicases/metabolism , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Protein Multimerization , alpha-Synuclein/metabolism , Amyloid/metabolism , Amyloid/ultrastructure , Gene Expression Regulation , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Models, Biological , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Binding , Protein Transport , Yeasts/genetics , Yeasts/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
8.
Methods Mol Biol ; 1948: 145-156, 2019.
Article in English | MEDLINE | ID: mdl-30771176

ABSTRACT

The budding yeast Saccharomyces cerevisiae (S. cerevisiae) has been a remarkable experimental model for the discovery of fundamental biological processes. The high degree of conservation of cellular and molecular processes between the budding yeast and higher eukaryotes has made it a valuable system for the investigation of the molecular mechanisms behind various types of devastating human pathologies. Genetic screens in yeast provided important insight into the toxic mechanisms associated with the accumulation of misfolded proteins. Thus, using yeast genetics and high-throughput screens, novel molecular targets with therapeutic potential have been identified. Here, we describe a yeast screen protocol for the identification of genetic modifiers of alpha-synuclein (aSyn) toxicity, thereby accelerating the identification of novel potential targets for intervention in Parkinson's disease (PD) and other synucleinopathies.


Subject(s)
Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Yeasts/genetics , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/genetics , Humans , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Reproducibility of Results , Yeasts/metabolism , alpha-Synuclein/metabolism
9.
FEMS Yeast Res ; 18(6)2018 09 01.
Article in English | MEDLINE | ID: mdl-30007297

ABSTRACT

Trehalose on both sides of the bilayer is a requirement for full protection of membranes against stress. It was not known yet how trehalose, synthesized in the cytosol when dividing Saccharomyces cerevisiae cells are shifted from 28°C to 40°C, is transported to the outside and degraded when cells return to 28°C. According to our results, the lack of Agt1, a trehalose transporter, although had not affected trehalose synthesis, reduced cell tolerance to 51°C and increased lipid peroxidation. The damage was reversed when external trehalose was added during 40°C adaptation, confirming that the reason for the agt1Δ sensitivity is the absence of trehalose at the outside of the lipid bilayer. The 40-28°C condition caused cytosolic trehalase (Nth1) activation, reducing intracellular trehalose and, consequently, the survival rates after 51°C. Although lower than nth1Δ strain, cells deficient in acid trehalase (ath1Δ)  maintained increased trehalose levels after 40°C-28°C shift, which conferred protection against 51°C. Both Ath1 and Agt1 were found into vesicles near to plasma membrane in response to stress. This suggests that Agt1 containing vesicles would fuse with the membrane under 40°C to transport part of the cytosolic trehalose to the outside. By a similar mechanism, Ath1 would reach the cell surface to hydrolyze the external trehalose but only when the stress would be over. Corroborating this conclusion, Ath1 activity in soluble cell-free extracts increased after 40°C adaptation but decreased when cells returned to 28°C. During 40°C, Ath1 is confined into vesicles, avoiding the cleavage of the outside trehalose.


Subject(s)
Heat-Shock Response , Monosaccharide Transport Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Symporters/metabolism , Trehalase/metabolism , Trehalose/metabolism , Biological Transport , Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Heat-Shock Response/drug effects , Lipid Peroxidation/drug effects , Monosaccharide Transport Proteins/genetics , Mutation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Symporters/genetics , Trehalase/genetics , Trehalose/pharmacology
10.
Front Mol Neurosci ; 11: 94, 2018.
Article in English | MEDLINE | ID: mdl-29636661

ABSTRACT

Aggregation of α-synuclein (αSyn) plays a central role in the pathogenesis of Parkinson's disease (PD). The budding yeast Saccharomyces cerevisiae serves as reference cell to study the interplay between αSyn misfolding, cytotoxicity and post-translational modifications (PTMs). The synuclein family includes α, ß and γ isoforms. ß-synuclein (ßSyn) and αSyn are found at presynaptic terminals and both proteins are presumably involved in disease pathogenesis. Similar to αSyn, expression of ßSyn leads to growth deficiency and formation of intracellular aggregates in yeast. Co-expression of αSyn and ßSyn exacerbates the cytotoxicity. This suggests an important role of ßSyn homeostasis in PD pathology. We show here that the small ubiquitin-like modifier SUMO is an important determinant of protein stability and ßSyn-induced toxicity in eukaryotic cells. Downregulation of sumoylation in a yeast strain, defective for the SUMO-encoding gene resulted in reduced yeast growth, whereas upregulation of sumoylation rescued growth of yeast cell expressing ßSyn. This corroborates a protective role of the cellular sumoylation machinery against ßSyn-induced toxicity. Upregulation of sumoylation significantly reduced ßSyn aggregate formation. This is an indirect molecular process, which is not directly linked to ßSyn sumoylation because amino acid substitutions in the lysine residues required for ßSyn sumoylation decreased aggregation without changing yeast cellular toxicity. αSyn aggregates are more predominantly degraded by the autophagy/vacuole than by the 26S ubiquitin proteasome system. We demonstrate a vice versa situation for ßSyn, which is mainly degraded in the 26S proteasome. Downregulation of sumoylation significantly compromised the clearance of ßSyn by the 26S proteasome and increased protein stability. This effect is specific, because depletion of functional SUMO did neither affect ßSyn aggregate formation nor its degradation by the autophagy/vacuolar pathway. Our data support that cellular ßSyn toxicity and aggregation do not correlate in their cellular impact as for αSyn but rather represent two distinct independent molecular functions and molecular mechanisms. These insights into the relationship between ßSyn-induced toxicity, aggregate formation and degradation demonstrate a significant distinction between the impact of αSyn compared to ßSyn on eukaryotic cells.

11.
PLoS Genet ; 14(2): e1007141, 2018 02.
Article in English | MEDLINE | ID: mdl-29401458

ABSTRACT

The transition from vegetative growth to multicellular development represents an evolutionary hallmark linked to an oxidative stress signal and controlled protein degradation. We identified the Sem1 proteasome subunit, which connects stress response and cellular differentiation. The sem1 gene encodes the fungal counterpart of the human Sem1 proteasome lid subunit and is essential for fungal cell differentiation and development. A sem1 deletion strain of the filamentous fungus Aspergillus nidulans is able to grow vegetatively and expresses an elevated degree of 20S proteasomes with multiplied ATP-independent catalytic activity compared to wildtype. Oxidative stress induces increased transcription of the genes sem1 and rpn11 for the proteasomal deubiquitinating enzyme. Sem1 is required for stabilization of the Rpn11 deubiquitinating enzyme, incorporation of the ubiquitin receptor Rpn10 into the 19S regulatory particle and efficient 26S proteasome assembly. Sem1 maintains high cellular NADH levels, controls mitochondria integrity during stress and developmental transition.


Subject(s)
Aspergillus nidulans/growth & development , Aspergillus nidulans/genetics , Cell Proliferation , Fungal Proteins/physiology , Proteasome Endopeptidase Complex/metabolism , Aspergillus nidulans/metabolism , Cytoplasm/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Organ Specificity , Organisms, Genetically Modified , Proteasome Endopeptidase Complex/genetics , Protein Stability , Ubiquitin/metabolism
12.
PLoS Genet ; 12(6): e1006098, 2016 06.
Article in English | MEDLINE | ID: mdl-27341336

ABSTRACT

Parkinson´s disease (PD) is characterized by the presence of proteinaceous inclusions called Lewy bodies that are mainly composed of α-synuclein (αSyn). Elevated levels of oxidative or nitrative stresses have been implicated in αSyn related toxicity. Phosphorylation of αSyn on serine 129 (S129) modulates autophagic clearance of inclusions and is prominently found in Lewy bodies. The neighboring tyrosine residues Y125, Y133 and Y136 are phosphorylation and nitration sites. Using a yeast model of PD, we found that Y133 is required for protective S129 phosphorylation and for S129-independent proteasome clearance. αSyn can be nitrated and form stable covalent dimers originating from covalent crosslinking of two tyrosine residues. Nitrated tyrosine residues, but not di-tyrosine-crosslinked dimers, contributed to αSyn cytotoxicity and aggregation. Analysis of tyrosine residues involved in nitration and crosslinking revealed that the C-terminus, rather than the N-terminus of αSyn, is modified by nitration and di-tyrosine formation. The nitration level of wild-type αSyn was higher compared to that of A30P mutant that is non-toxic in yeast. A30P formed more dimers than wild-type αSyn, suggesting that dimer formation represents a cellular detoxification pathway in yeast. Deletion of the yeast flavohemoglobin gene YHB1 resulted in an increase of cellular nitrative stress and cytotoxicity leading to enhanced aggregation of A30P αSyn. Yhb1 protected yeast from A30P-induced mitochondrial fragmentation and peroxynitrite-induced nitrative stress. Strikingly, overexpression of neuroglobin, the human homolog of YHB1, protected against αSyn inclusion formation in mammalian cells. In total, our data suggest that C-terminal Y133 plays a major role in αSyn aggregate clearance by supporting the protective S129 phosphorylation for autophagy and by promoting proteasome clearance. C-terminal tyrosine nitration increases pathogenicity and can only be partially detoxified by αSyn di-tyrosine dimers. Our findings uncover a complex interplay between S129 phosphorylation and C-terminal tyrosine modifications of αSyn that likely participates in PD pathology.


Subject(s)
Parkinson Disease/metabolism , Phosphorylation/physiology , Saccharomyces cerevisiae/metabolism , Serine/metabolism , Tyrosine/metabolism , alpha-Synuclein/metabolism , Animals , Autophagy/physiology , Globins/metabolism , Humans , Lewy Bodies/metabolism , Nerve Tissue Proteins/metabolism , Neuroglobin , Oxidation-Reduction , Proteasome Endopeptidase Complex/metabolism
13.
Hum Mol Genet ; 25(2): 275-90, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26586132

ABSTRACT

Synucleins belong to a family of intrinsically unstructured proteins that includes alpha-synuclein (aSyn), beta-synuclein (bSyn) and gamma-synuclein (gSyn). aSyn is the most studied member of the synuclein family due to its central role in genetic and sporadic forms of Parkinson's disease and other neurodegenerative disorders known as synucleionopathies. In contrast, bSyn and gSyn have been less studied, but recent reports also suggest that, unexpectedly, these proteins may also cause neurotoxicity. Here, we explored the yeast toolbox to investigate the cellular effects of bSyn and gSyn. We found that bSyn is toxic and forms cytosolic inclusions that are similar to those formed by aSyn. Moreover, we found that bSyn shares similar toxicity mechanisms with aSyn, including vesicular trafficking impairment and induction of oxidative stress. We demonstrate that co-expression of aSyn and bSyn exacerbates cytotoxicity, due to increased dosage of toxic synuclein forms, and that they are able to form heterodimers in both yeast and in human cells. In contrast, gSyn is not toxic and does not form inclusions in yeast cells. Altogether, our findings shed light into the question of whether bSyn can exert toxic effects and confirms the occurrence of aSyn/bSyn heterodimers, opening novel perspectives for the development of novel strategies for therapeutic intervention in synucleinopathies.


Subject(s)
Oxidative Stress , alpha-Synuclein/metabolism , beta-Synuclein/metabolism , HEK293 Cells , Humans , Mutation , Protein Multimerization , Protein Transport , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transgenes , Transport Vesicles , alpha-Synuclein/genetics , beta-Synuclein/genetics
14.
PLoS One ; 10(9): e0136778, 2015.
Article in English | MEDLINE | ID: mdl-26355961

ABSTRACT

A major challenge in gene library generation is to guarantee a large functional size and diversity that significantly increases the chances of selecting different functional protein variants. The use of trinucleotides mixtures for controlled randomization results in superior library diversity and offers the ability to specify the type and distribution of the amino acids at each position. Here we describe the generation of a high diversity gene library using tHisF of the hyperthermophile Thermotoga maritima as a scaffold. Combining various rational criteria with contingency, we targeted 26 selected codons of the thisF gene sequence for randomization at a controlled level. We have developed a novel method of creating full-length gene libraries by combinatorial assembly of smaller sub-libraries. Full-length libraries of high diversity can easily be assembled on demand from smaller and much less diverse sub-libraries, which circumvent the notoriously troublesome long-term archivation and repeated proliferation of high diversity ensembles of phages or plasmids. We developed a generally applicable software tool for sequence analysis of mutated gene sequences that provides efficient assistance for analysis of library diversity. Finally, practical utility of the library was demonstrated in principle by assessment of the conformational stability of library members and isolating protein variants with HisF activity from it. Our approach integrates a number of features of nucleic acids synthetic chemistry, biochemistry and molecular genetics to a coherent, flexible and robust method of combinatorial gene synthesis.


Subject(s)
Combinatorial Chemistry Techniques/methods , Gene Library , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Genes, Bacterial , Genetic Complementation Test , Molecular Sequence Data , Reference Standards , Software , Thermotoga maritima/genetics , Thermotoga maritima/metabolism
15.
Biomolecules ; 5(2): 617-34, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25915624

ABSTRACT

The budding yeast Saccharomyces cerevisiae represents an established model system to study the molecular mechanisms associated to neurodegenerative disorders. A key-feature of Parkinson's disease is the formation of Lewy bodies, which are cytoplasmic protein inclusions. Misfolded α-synuclein is one of their main constituents. Expression of α-synuclein protein in yeast leads to protein aggregation and cellular toxicity, which is reminiscent to Lewy body containing human cells. The molecular mechanism involved in clearance of α-synuclein aggregates is a central question for elucidating the α-synuclein-related toxicity. Cellular clearance mechanisms include ubiquitin mediated 26S proteasome function as well as lysosome/vacuole associated degradative pathways as autophagy. Various modifications change α-synuclein posttranslationally and alter its inclusion formation, cytotoxicity and the distribution to different clearance pathways. Several of these modification sites are conserved from yeast to human. In this review, we summarize recent findings on the effect of phosphorylation and sumoylation of α-synuclein to the enhanced channeling to either the autophagy or the proteasome degradation pathway in yeast model of Parkinson's disease.


Subject(s)
Parkinson Disease/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae/metabolism , alpha-Synuclein/metabolism , Humans , Protein Aggregates , Saccharomyces cerevisiae/genetics , Sumoylation , alpha-Synuclein/genetics
16.
PLoS Genet ; 10(11): e1004741, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25393002

ABSTRACT

Aggregation of alpha-synuclein (ASYN) in Lewy bodies and Lewy neurites is the typical pathological hallmark of Parkinson's disease (PD) and other synucleinopathies. Furthermore, mutations in the gene encoding for ASYN are associated with familial and sporadic forms of PD, suggesting this protein plays a central role in the disease. However, the precise contribution of ASYN to neuronal dysfunction and death is unclear. There is intense debate about the nature of the toxic species of ASYN and little is known about the molecular determinants of oligomerization and aggregation of ASYN in the cell. In order to clarify the effects of different mutations on the propensity of ASYN to oligomerize and aggregate, we assembled a panel of 19 ASYN variants and compared their behaviour. We found that familial mutants linked to PD (A30P, E46K, H50Q, G51D and A53T) exhibited identical propensities to oligomerize in living cells, but had distinct abilities to form inclusions. While the A30P mutant reduced the percentage of cells with inclusions, the E46K mutant had the opposite effect. Interestingly, artificial proline mutants designed to interfere with the helical structure of the N-terminal domain, showed increased propensity to form oligomeric species rather than inclusions. Moreover, lysine substitution mutants increased oligomerization and altered the pattern of aggregation. Altogether, our data shed light into the molecular effects of ASYN mutations in a cellular context, and established a common ground for the study of genetic and pharmacological modulators of the aggregation process, opening new perspectives for therapeutic intervention in PD and other synucleinopathies.


Subject(s)
Parkinson Disease/genetics , Protein Aggregation, Pathological/genetics , alpha-Synuclein/genetics , Cell Line , Humans , Lewy Bodies/metabolism , Lewy Bodies/pathology , Lysosomes/metabolism , Lysosomes/pathology , Mutagenesis, Site-Directed , Parkinson Disease/pathology , Phosphorylation , Point Mutation , alpha-Synuclein/metabolism
17.
J Biol Chem ; 289(45): 31224-40, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25231978

ABSTRACT

Parkinson disease is associated with the progressive loss of dopaminergic neurons from the substantia nigra. The pathological hallmark of the disease is the accumulation of intracytoplasmic inclusions known as Lewy bodies that consist mainly of post-translationally modified forms of α-synuclein. Whereas phosphorylation is one of the major modifications of α-synuclein in Lewy bodies, sumoylation has recently been described. The interplay between α-synuclein phosphorylation and sumoylation is poorly understood. Here, we examined the interplay between these modifications as well as their impact on cell growth and inclusion formation in yeast. We found that α-synuclein is sumoylated in vivo at the same sites in yeast as in human cells. Impaired sumoylation resulted in reduced yeast growth combined with an increased number of cells with inclusions, suggesting that this modification plays a protective role. In addition, inhibition of sumoylation prevented autophagy-mediated aggregate clearance. A defect in α-synuclein sumoylation could be suppressed by serine 129 phosphorylation by the human G protein-coupled receptor kinase 5 (GRK5) in yeast. Phosphorylation reduced foci formation, alleviated yeast growth inhibition, and partially rescued autophagic α-synuclein degradation along with the promotion of proteasomal degradation, resulting in aggregate clearance in the absence of a small ubiquitin-like modifier. These findings suggest a complex interplay between sumoylation and phosphorylation in α-synuclein aggregate clearance, which may open new horizons for the development of therapeutic strategies for Parkinson disease.


Subject(s)
Lewy Bodies/metabolism , alpha-Synuclein/metabolism , Autophagy , Chromatography, Affinity , G-Protein-Coupled Receptor Kinase 5/metabolism , Humans , Lysine/chemistry , Microscopy, Fluorescence , Phosphorylation , Plasmids/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/metabolism , Sumoylation , Ubiquitin/chemistry
18.
Eukaryot Cell ; 12(8): 1142-54, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23794510

ABSTRACT

Hac1 is the activator of the cellular response to the accumulation of unfolded proteins in the endoplasmic reticulum. Hac1 function requires the activity of Gcn4, which mainly acts as a regulator of the general amino acid control network providing Saccharomyces cerevisiae cells with amino acids. Here, we demonstrate novel functions of Hac1 and describe a mutual connection between Hac1 and Gcn4. Hac1 is required for induction of Gcn4-responsive promoter elements in haploid as well as diploid cells and therefore participates in the cellular amino acid supply. Furthermore, Hac1 and Gcn4 mutually influence their mRNA expression levels. Hac1 is also involved in FLO11 expression and adhesion upon amino acid starvation. Hac1 and Gcn4 act through the same promoter regions of the FLO11 flocculin. The results indicate an indirect effect of both transcription factors on FLO11 expression. Our data suggest a complex mutual cross talk between the Hac1- and Gcn4-controlled networks.


Subject(s)
Amino Acids/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Unfolded Protein Response/genetics , Amino Acids/biosynthesis , Basic-Leucine Zipper Transcription Factors/genetics , Cell Adhesion , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Fungal , Membrane Glycoproteins/metabolism , Promoter Regions, Genetic , Repressor Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics
19.
J Biol Chem ; 287(33): 27567-79, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22722939

ABSTRACT

Parkinson disease is the second most common neurodegenerative disease. The molecular hallmark is the accumulation of proteinaceous inclusions termed Lewy bodies containing misfolded and aggregated α-synuclein. The molecular mechanism of clearance of α-synuclein aggregates was addressed using the bakers' yeast Saccharomyces cerevisiae as the model. Overexpression of wild type α-synuclein or the genetic variant A53T integrated into one genomic locus resulted in a gene copy-dependent manner in cytoplasmic proteinaceous inclusions reminiscent of the pathogenesis of the disease. In contrast, overexpression of the genetic variant A30P resulted only in transient aggregation, whereas the designer mutant A30P/A36P/A76P neither caused aggregation nor impaired yeast growth. The α-synuclein accumulation can be cleared after promoter shut-off by a combination of autophagy and vacuolar protein degradation. Whereas the proteasomal inhibitor MG-132 did not significantly inhibit aggregate clearance, treatment with phenylmethylsulfonyl fluoride, an inhibitor of vacuolar proteases, resulted in significant reduction in clearance. Consistently, a cim3-1 yeast mutant restricted in the 19 S proteasome regulatory subunit was unaffected in clearance, whereas an Δatg1 yeast mutant deficient in autophagy showed a delayed aggregate clearance response. A cim3-1Δatg1 double mutant was still able to clear aggregates, suggesting additional cellular mechanisms for α-synuclein clearance. Our data provide insight into the mechanisms yeast cells use for clearing different species of α-synuclein and demonstrate a higher contribution of the autophagy/vacuole than the proteasome system. This contributes to the understanding of how cells can cope with toxic and/or aggregated proteins and may ultimately enable the development of novel strategies for therapeutic intervention.


Subject(s)
Lewy Bodies/metabolism , Phagosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vacuoles/metabolism , alpha-Synuclein/metabolism , Amino Acid Substitution , Autophagy/physiology , Humans , Lewy Bodies/genetics , Mutation, Missense , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phagosomes/genetics , Proteasome Endopeptidase Complex/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Vacuoles/genetics , alpha-Synuclein/genetics
20.
Methods Mol Biol ; 346: 211-26, 2006.
Article in English | MEDLINE | ID: mdl-16957293

ABSTRACT

Knockouts by homologous recombination are frequently used to investigate the function of genes in Dictyostelium and other organisms. Antisense-mediated gene silencing and RNA interference (RNAi) are convenient alternatives to reduce gene expression to different levels and to silence multigene families. We describe here the methods for efficient RNA interference in Dictyostelium and some useful mutant strains that enhance the success rate or may serve as convenient controls. We believe that it is helpful to also discuss failed attempts to optimize and expand the system because these are rarely discussed in the literature. In addition, a list of in silico and experimentally identified components in the RNAi and antisense pathway is presented.


Subject(s)
Dictyostelium/genetics , Gene Expression Regulation, Fungal/physiology , RNA Helicases/metabolism , RNA, Antisense/genetics , RNA, Small Interfering/pharmacology , Animals , Dictyostelium/enzymology , Dictyostelium/growth & development , RNA Helicases/antagonists & inhibitors , RNA Helicases/genetics , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL
...