Proteins Related to Human Neurodegenerative Diseases inYeast Cells: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: ,

A characteristic feature of neurodegenerative diseases (NDs) is the formation of protein aggregates (inclusion bodies) in cells of the nervous system, formed as a result of the misfolding of proteins. Based on the reviewed results of studies on NDs and related protein aggregation, as well as viral protein aggregation, a new experimental model system for the study of human NDs is proposed. The core of the proposed system is a comparative transcriptomic analysis of changes in signaling pathways during the expression of viral capsid proteins in yeast cells.

  • neurodegenerative diseases
  • Saccharomyces cerevisiae
  • protein aggregation
  • capsid virus proteins

1. Polyglutamine Proteins

There are several known human NDs caused by the expansion of CAG trinucleotide repeats in the coding region of the corresponding genes. These diseases are often called “polyglutamine (polyQ) diseases” because the CAG trinucleotide encodes the amino acid glutamine (Q). To date, glutamine expansions have been described in the following human proteins: huntingtin (Htt); Sca1, Sca2, Sca3, Sca6, Sca7, and Sca17 (different types of spinocerebellar ataxia); androgen receptor protein (spinobulbar muscular atrophy); and atrophin-1 (dentatorubro-pallidoluysian atrophy). The features of the clinical manifestations characteristic of each of these diseases, determined by the type of neurons in which inclusions formed as a result of amyloidization of the described proteins, are observed [1][2][3]. Using yeast as a model system, the aggregation of the huntingtin protein has been studied in the most detail [4].
The human huntingtin locus is large, spanning 180 kb and consisting of 67 exons [5]. This locus encodes a fairly large protein (350 kDa) that performs important, but not fully understood, functions, in particular, those necessary for the normal functioning of synapses [6]. The first exon may contain a variable number of CAG repeats, normally ranging from 6 to 35. Proteins with 36 or more repeats (up to 180 repeats have been described) tend to form amyloid aggregates, which correlates with the development of HD. Importantly, the hallmark of HD is the proteolytic production of an N-terminal fragment of the huntingtin protein (containing a polyQ repeat), which forms aggregates in the nucleus and cytoplasm of affected neurons [7][8].
To study the influence of various factors on the aggregation of the human huntingtin protein in yeast cells, single-copy plasmids containing DNA fragments corresponding to the N-terminal region (amino acids 1-68 of the wild-type protein) of the investigated protein fused to the GFP gene were generated. These vector constructs differed from each other in the number of CAG trinucleotide repeats encoding glutamine (25, 47, 72, or 103 residues). Expression was performed under the control of a strong constitutive yeast promoter (GPD). The results showed that the degree of aggregation varied with polyQ repeat length; in particular, a protein containing 25 glutamine repeats showed no signs of aggregation, whereas when a protein containing 103 glutamine repeats was expressed, one large aggregate was formed [9]. Mutations in yeast cells inhibiting the ubiquitin/proteasome pathway did not affect the aggregation of the studied protein fragments in the series of experiments. At the same time, changes in the expression level of chaperone proteins, in particular, overexpression of Hsp40, Hsp70, and Hsp104, modulated the aggregation of fragments containing 72 and 103 glutamine repeats [10][11].
In the amino acid sequence of the huntingtin protein, a proline-rich region is located next to the polyQ site. Deletion of this proline-rich region effects on the number and shape of formed aggregates [12][13].
Interestingly, when the dependence between the intensity of huntingtin protein aggregate formation in yeast cells and the age of the cell culture was studied, the results showed that proteins containing 25Q did not aggregate during the logarithmic stage of cell growth or in aged cells. Proteins containing 103Q aggregated in both young and aged cells, while proteins containing a moderate amount of glutamine (47Q) aggregated only during cell culture senescence. Mutations in two genes (SIR2 and HSF1), the expression levels of which correlate with aging, affected the dynamics of huntingtin protein aggregation in yeast cells [14].
Currently, when yeast is used as a model organism, transcriptomic, proteomic, and metabolomic analyses are the most promising modern methodological approaches for understanding the molecular mechanisms of the formation of aggregates of proteins containing polyQ tracks, particularly the huntingtin protein [15][16][17][18][19].

2. α-Synuclein

Parkinson’s disease, certain forms of dementia, and multisystem atrophy are characterized by the accumulation of aggregated α-synuclein in protein inclusions called Lewy bodies (LBs) [20]. This class of ND is generally referred to as synucleinopathies [21][22]. It is currently unclear whether the formation of LBs causes cell pathologies or acts as a protective cellular mechanism, leading to the inactivation of soluble prefibrillar oligomeric forms of α-synuclein [23][24][25][26][27].
Under physiological conditions in human cells, α-synuclein is considered a presynaptic protein [28] associated with vesicles and membranes [29]. However, its exact function is not yet well understood [30]. Several studies have suggested that α-synuclein plays a critical role in enabling vesicle fusion; others have reported a chaperone-like activity [31][32][33][34].
Human α-synuclein is encoded by the SNCA gene. The protein consists of 140 amino acids and has a molecular mass of 17 kDa. The protein has N-terminal imperfect repeats based on the amino acid motif KTKEGV. The central region of α-synuclein is strongly hydrophobic, leading to protein dimerization [35][36]. The N-terminus of α-synuclein is critical for oligomerization [37], whereas the C-terminus is highly acidic, facilitating protein–protein interactions [38] and possibly inhibiting aggregation [39]. The α-synuclein protein is intrinsically unfolded; in solution, it lacks a stable tertiary structure and instead forms α-helices, β-sheets, and various complex multimeric structures, such as fibrils, fibers, and aggregates [40].
Multiple wild-type alleles at the SNCA locus, such as duplications or triplications, have been shown to be associated with the familial form of the disease [41][42]. In addition, three missense mutations, A30P, A53T, and E46K, correlate with the autosomal dominant early form of Parkinson’s disease [43][44]. These mutations render α-synuclein more prone to forming amyloid fibrils [45].
It has been shown that α-synuclein in human nerve cells can undergo various posttranslational modifications, namely, phosphorylation [46], ubiquitination [47][48], SUMOylation [49], acetylation [50], nitration [51], and/or glycosylation [52]. Posttranslational modifications of α-synuclein are known to affect its toxicity and ability to form aggregates, but the exact contribution of different posttranslational modifications to the disease mechanism is still unclear.
There is no homolog of the human SNCA gene encoding α-synuclein in the yeast genome. It has been shown that expression of the human α-synuclein gene in yeast affects vesicle trafficking [53][54][55], induces oxidative stress [56], and causes mitochondrial dysfunction [57][58].
It has been demonstrated that when the human α-synuclein gene is heterologously expressed in yeast cells, the resulting protein is transported via the secretory pathway to the plasma membrane, where it forms small aggregates [59][60]. As the level of α-synuclein expression increases, its localization changes significantly, leading to the formation of cytoplasmic inclusions similar to the LBs found in Parkinson’s disease neurons. This is accompanied by an increase in toxicity, defined as reduced cell growth followed by cell death. The increase in toxicity caused by α-synuclein is dependent on dosage. Studies have shown that the expression of three copies of the wild-type α-synuclein gene or two copies of the α-synuclein gene with the A53T mutation under the control of the yeast GAL1 promoter leads to growth inhibition and protein aggregation [61].

3. TDP-4, FUS, and SOD1

Like most NDs, ALS and frontotemporal lobar degeneration (FTLD) are characterized by the misfolding and aggregation of proteins in affected neurons [62]. The aggregates formed in these diseases contain proteins such as TDP-43 (trans-activation response DNA-binding protein 43), FUS (fused in sarcoma), SOD1 (superoxide dismutase), and C9orf72 that are well-known pathologic hallmarks of ALS/FTLD [63][64][65][66][67][68][69][70]. Using yeast as a model system, the aggregation of TDP-43, FUS, and SOD1 has been studied in the most detail. It was shown that the expression of these proteins in yeast reproduces the main features of ALS/FTLD pathology, including aggregation, protein mislocalization, and cellular toxicity [71][72][73][74].
FUS and TDP-43 have similar domain structures and serve many similar functions. TDP-43, which is 414 amino acid residues in length, has been shown to bind to both DNA and RNA and has multiple functions in transcription repression, pre-mRNA splicing, and translation regulation. FUS, which is 526 amino acid residues in length, plays an important role in various cellular processes, such as the regulation of transcription, RNA splicing, RNA transport, and DNA repair. In neuronal cells, it plays a critical role in dendritic outgrowth formation and stability, RNA transport, mRNA stability, and synaptic homeostasis [75].
Both of these proteins have highly conserved RNA recognition motifs (RRMs) and glycine-rich domains [75]. Moreover, using a bioinformatics algorithm originally developed to identify yeast prion domains [76], “prion-like” domains were identified in the N-terminal part of FUS (amino acids 1–239) and in the C-terminal part of TDP-43 (amino acids 277–414) [75][76]. Similar to the prion domains of yeast prion proteins such as Sup35, Ure2, and Rnq1, these domains are rich in uncharged polar amino acids [77][78].
Expression of human FUS and TDP-43 proteins in yeast leads to cytoplasmic aggregation of these proteins and toxicity, thus modeling key aspects of ALS- and FTLD-related proteopathies. The RRM and prion-like domains have been shown to be key to the aggregation of the proteins studied. The patterns of localization of FUS and TDP-43 protein aggregates in yeast cells in terms of size, shape, and number of foci in the cytoplasm are strikingly similar. Indeed, when FUS-YFP and TDP-43-CFP were coexpressed in the same cell, the fluorescence signals colocalized in the same cytoplasmic foci [75].
The aggregates formed by FUS and TDP-43 in yeast cells have been shown not to react with amyloid diagnostic dyes and are SDS-soluble. Thus, FUS and TDP-43 in yeast cells form aggregates that are probably not amyloid in nature, as are aggregates of these proteins observed in patients with ALS and FTLD [75][79][80].
To date, many mutations in the human FUS and TDP-43 genes have been described and have been associated with some familial and sporadic ALS cases [81]. Using a yeast model, the influence of these mutations on the aggregation and toxicity of the proteins under study was tested. The mutations in TDP-43 significantly increased the degree of aggregation and toxicity of this protein in yeast cells. Moreover, almost all ALS-linked mutations in the TDP-43 gene are located in the DNA sequence encoding the prion-like domain of this protein. However, ALS-linked FUS mutations do not promote FUS aggregation in yeast [75].
Superoxide dismutase 1 (Sod1) is an enzyme that, in humans, is encoded by the SOD1 gene located on chromosome 21. Sod1 is 154 amino acid residues long, and alternative splicing results in five forms of the enzyme that differ in their localization in the human body [82]. This enzyme is a 32 kDa homodimer containing an intramolecular disulfide bond and a binuclear Cu/Zn site in each subunit. SOD1 binds copper and zinc ions and is one of three superoxide dismutases responsible for the destruction of free superoxide radicals in human cells. SOD1 is a soluble cytoplasmic and mitochondrial intermembrane protein that converts naturally occurring but harmful superoxide radicals into molecular oxygen and hydrogen peroxide. Recently, it has been shown that under oxidative stress, SOD1 can localize not only in the cytoplasm but also in the nucleus [83][84]. Mutations in this gene (more than 150 identified to date) are associated with familial ALS1 [85]. The protein (both wild-type and ALS1 variants) has a propensity to form fibrillar aggregates in the absence of intramolecular disulfide bonding or bound zinc ions. These aggregates can exert cytotoxic effects. Zinc binding promotes dimerization and stabilizes the wild-type form.
Yeast has been shown to have a homolog of the human SOD1 gene. Expression of the human SOD1 gene in yeast cells lacking the homologous gene showed that human SOD1 can fully complement the biological function of yeast SOD1 [86]. A series of studies have been conducted in which the wild-type human SOD1 gene and SOD1 genes containing various ALS-related mutations were expressed in yeast cells [87][88][89]. Interestingly, neither the wild-type human protein nor any of the mutant variants of this protein corresponding to ALS-related mutations in yeast cells caused impairment of the cell culture growth rate [87][88][89][90][91]. However, it has been shown that the wild-type SOD1 protein, as well as ALS-associated SOD1 mutants, tends to selectively aggregate near mitochondria, where the protein may exert toxic effects that are not yet fully understood [74]. It has been shown that the aggregates formed are SDS-soluble and of a nonamyloid nature [86][92].
Integrated transcriptomic and metabolomic analysis is the most recently developed and promising methodological approach that uses yeast as a model system to understand the biological role of the described proteins in the deregulation of pathways associated with the disease. To date, only preliminary, but encouraging, results have been obtained, and in the researchers' view, further research is warranted [93][94].

4. Tau

Tau, also known as microtubule-associated protein tau (MAPT), is expressed predominantly in cells of the nervous system. The main function of this protein is the stabilization of microtubules, which occurs through a kiss-and-hop mechanism [95][96]. The human MAPT gene contains 16 exons. In cells of the nervous system, alternative splicing yields six tau isoforms, ranging in size from 352 to 441 amino acids. The molecular mass of the tau isoforms ranges from 48 kDa to 68 kDa [97]. Numerous human NDs have been shown to be associated with tau protein aggregation (see Table 1). Although tau is a “naturally unfolded protein”, conformational changes have been detected and could be essential in the formation of aggregates [98]. Various posttranslational modifications of the tau protein (phosphorylation, acetylation, ubiquitination, SUMOylation, methylation, glycosylation, glycation, proteolysis, and numerous others) occur in the brain cells of both healthy people and patients with NDs [99]. Posttranslational modifications play an important role in tau protein aggregation, and phosphorylation is of particular importance.
Table 1. Model substrates used to monitor inclusion formation and localization of misfolded proteins in S. cerevisiae.
Misfolding Protein Origin Experimental Conditions
Luciferase Photinus pyralis Heat shock
Guk 1-7 (guanylate kinase temperature-sensitive)
temperature-sensitive
Saccharomyces cerevisiae Heat shock
Gus 1-3 (glutamyl-tRNA synthetase)
temperature-sensitive
Saccharomyces cerevisiae Heat shock
Pro 3-1 (delta 1-pyrroline-5-carboxylate reductase)
temperature-sensitive
Saccharomyces cerevisiae Heat shock
Ubc9ts (SUMO-conjugating E2 enzyme)
temperature-sensitive
Saccharomyces cerevisiae Heat shock
VHL (von Hippel–Lindau tumor suppressor)
temperature-sensitive
Homo sapiens Constitutively unfolded in yeast cells (absent binding partner)
ΔssCPY* (mutated form of carboxypeptidase Y) Saccharomyces cerevisiae Constitutively unfolded
tGnd1 (truncated phosphogluconate dehydrogenase) Saccharomyces cerevisiae Constitutively unfolded
DegAB (the entire Ndc10 degron) Saccharomyces cerevisiae Constitutively unfolded
Expression of tau in yeast does not result in a negative phenotype [100]. Tau protein expressed in yeast under normal conditions, either under mild proteotoxic stress or in mutants with impaired proteotoxic stress response pathways, did not result in toxicity or the formation of obvious aggregates. In chronologically aged cells, no appreciable Tau aggregates were formed [101].
In S. cerevisiae, tau phosphorylation and hyperphosphorylation are under the control of two protein kinases, Mds1 and Pho85, which are orthologs of two human protein kinases, GSK3β and cdk5, respectively. It has been shown that when tau is expressed in yeasts lacking Pho85, the tau phosphorylation and aggregation levels increase, indicating that Pho85 (and possibly cdk5 in human neuronal cells) is a negative regulator of tau phosphorylation. On the other hand, deletion of the Mds1 gene leads to a decrease in the amount of hyperphosphorylated tau protein and, accordingly, to a decrease in the amount of the aggregated form of this protein. Thus, the tau protein expressed in yeast lacking Pho85 undergoes posttranslational modification by phosphorylation and, as a consequence, changes its conformation and forms aggregates. The hyperphosphorylation and aggregation of the tau protein in yeast cells is similar to processes that are characteristic of neurodegenerative tauopathies, including Alzheimer’s disease [102].

5. Screening of the Compounds Preventing Aggregation and Toxicity of Disease-Specific Proteins

The yeast model systems described above, which allow the study of the aggregation of proteins whose aggregation was originally identified in studies of human NDs, can be used to screen potential drugs that prevent the aggregation and toxicity of these proteins. Indeed, yeast has been shown to be a very useful platform for drug screening to identify potential therapeutic molecules. Importantly, many of these results initially obtained in yeast have subsequently been confirmed in mammalian systems, confirming yeast as reliable. For review, see, e.g., [103][104][105][106].
Although yeast cells are well-suited for high-throughput screening in search of chemical compounds that inhibit aggregation and/or toxicity, one limitation is the inability of many drugs to cross cell membrane and/or cell wall barriers or to accumulate within the cell. This problem can be overcome by using yeast strains containing mutations in the ERG6 or PDR5 genes. Erg6 mutants have increased membrane permeability; pdr5 mutants more easily accumulate drugs intracellularly. For review, see, e.g., [104].

This entry is adapted from the peer-reviewed paper 10.3390/ijms242417213

References

  1. Orr, H.T.; Zoghbi, H.Y. Trinucleotide repeat disorders. Annu. Rev. Neurosci. 2007, 30, 575–621.
  2. Adegbuyiro, A.; Sedighi, F.; Pilkington, A.W.; Groover, S.; Legleiter, J. Proteins Containing Expanded Polyglutamine Tracts and Neurodegenerative Disease. Biochemistry 2017, 56, 1199–1217.
  3. Lieberman, A.P.; Shakkottai, V.G.; Albin, R.L. Polyglutamine Repeats in Neurodegenerative Diseases. Annu. Rev. Pathol. 2019, 14, 1–27.
  4. Hofer, S.; Kainz, K.; Zimmermann, A.; Bauer, M.A.; Pendl, T.; Poglitsch, M.; Madeo, F.; Carmona-Gutierrez, D. Studying Huntington’s Disease in Yeast: From Mechanisms to Pharmacological Approaches. Front. Mol. Neurosci. 2018, 11, 318.
  5. Ambrose, C.M.; Duyao, M.P.; Barnes, G.; Bates, G.P.; Lin, C.S.; Srinidhi, J.; Baxendale, S.; Hummerich, H.; Lehrach, H.; Altherr, M.; et al. Structure and expression of the Huntington’s disease gene: Evidence against simple inactivation due to an expanded CAG repeat. Somat Cell Mol. Genet. 1994, 20, 27–38.
  6. Barron, J.C.; Hurley, E.P.; Parsons, M.P. Huntingtin and the Synapse. Front. Cell Neurosci. 2021, 15, 689332.
  7. DiFiglia, M.; Sapp, E.; Chase, K.O.; Davies, S.W.; Bates, G.P.; Vonsattel, J.P.; Aronin, N. Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain. Science 1997, 277, 1990–1993.
  8. Li, X.J. The early cellular pathology of Huntington’s disease. Mol. Neurobiol. 1999, 20, 111–124.
  9. Krobitsch, S.; Lindquist, S. Aggregation of huntingtin in yeast varies with the length of the polyglutamine expansion and the expression of chaperone proteins. Proc. Natl. Acad. Sci. USA 2000, 97, 1589–1594.
  10. Muchowski, P.J.; Schaffar, G.; Sittler, A.; Wanker, E.E.; Hayer-Hartl, M.K.; Hartl, F.U. Hsp70 and hsp40 chaperones can inhibit self-assembly of polyglutamine proteins into amyloid-like fibrils. Proc. Natl. Acad. Sci. USA 2000, 97, 7841–7846.
  11. Cao, F.; Levine, J.J.; Li, S.H.; Li, X.J. Nuclear aggregation of huntingtin is not prevented by deletion of chaperone Hsp104. Biochim. Biophys. Acta 2001, 1537, 158–166.
  12. Duennwald, M.L.; Jagadish, S.; Muchowski, P.J.; Lindquist, S. Flanking sequences profoundly alter polyglutamine toxicity in yeast. Proc. Natl. Acad. Sci. USA 2006, 103, 11045–11050.
  13. Dehay, B.; Bertolotti, A. Critical role of the proline-rich region in Huntingtin for aggregation and cytotoxicity in yeast. J. Biol. Chem. 2006, 281, 35608–35615.
  14. Cohen, A.; Ross, L.; Nachman, I.; Bar-Nun, S. Aggregation of polyQ proteins is increased upon yeast aging and affected by Sir2 and Hsf1: Novel quantitative biochemical and microscopic assays. PLoS ONE 2012, 7, e44785.
  15. Joyner, P.M.; Matheke, R.M.; Smith, L.M.; Cichewicz, R.H. Probing the metabolic aberrations underlying mutant huntingtin toxicity in yeast and assessing their degree of preservation in humans and mice. J. Proteome Res. 2010, 9, 404–412.
  16. Tauber, E.; Miller-Fleming, L.; Mason, R.P.; Kwan, W.; Clapp, J.; Butler, N.J.; Outeiro, T.F.; Muchowski, P.J.; Giorgini, F. Functional gene expression profiling in yeast implicates translational dysfunction in mutant huntingtin toxicity. J. Biol. Chem. 2011, 286, 410–419.
  17. Papsdorf, K.; Kaiser, C.J.; Drazic, A.; Grotzinger, S.W.; Haessner, C.; Eisenreich, W.; Richter, K. Polyglutamine toxicity in yeast induces metabolic alterations and mitochondrial defects. BMC Genom. 2015, 16, 662.
  18. Pradhan, S.S.; Kanikaram, S.P.; VM, D.D.; Pargaonkar, A.; Dandamudi, R.B.; Sivaramakrishnan, V. Metabolic deregulation associated with aging modulates protein aggregation in the yeast model of Huntington’s disease. J. Biomol. Struct. Dyn. 2023, 1–18.
  19. Pradhan, S.S.; Rao, K.R.; Manjunath, M.; Saiswaroop, R.; Patnana, D.P.; Phalguna, K.S.; Choudhary, B.; Sivaramakrishnan, V. Vitamin B(6,) B(12) and folate modulate deregulated pathways and protein aggregation in yeast model of Huntington disease. 3 Biotech 2023, 13, 96.
  20. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. Alpha-synuclein in Lewy bodies. Nature 1997, 388, 839–840.
  21. Spillantini, M.G.; Crowther, R.A.; Jakes, R.; Hasegawa, M.; Goedert, M. alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies. Proc. Natl. Acad. Sci. USA 1998, 95, 6469–6473.
  22. Goedert, M.; Spillantini, M.G.; Del Tredici, K.; Braak, H. 100 years of Lewy pathology. Nat. Rev. Neurol. 2013, 9, 13–24.
  23. Olanow, C.W.; Perl, D.P.; DeMartino, G.N.; McNaught, K.S. Lewy-body formation is an aggresome-related process: A hypothesis. Lancet Neurol. 2004, 3, 496–503.
  24. Outeiro, T.F.; Putcha, P.; Tetzlaff, J.E.; Spoelgen, R.; Koker, M.; Carvalho, F.; Hyman, B.T.; McLean, P.J. Formation of toxic oligomeric alpha-synuclein species in living cells. PLoS ONE 2008, 3, e1867.
  25. Winner, B.; Jappelli, R.; Maji, S.K.; Desplats, P.A.; Boyer, L.; Aigner, S.; Hetzer, C.; Loher, T.; Vilar, M.; Campioni, S.; et al. In vivo demonstration that alpha-synuclein oligomers are toxic. Proc. Natl. Acad. Sci. USA 2011, 108, 4194–4199.
  26. Tsigelny, I.F.; Sharikov, Y.; Wrasidlo, W.; Gonzalez, T.; Desplats, P.A.; Crews, L.; Spencer, B.; Masliah, E. Role of alpha-synuclein penetration into the membrane in the mechanisms of oligomer pore formation. FEBS J. 2012, 279, 1000–1013.
  27. Lashuel, H.A.; Overk, C.R.; Oueslati, A.; Masliah, E. The many faces of alpha-synuclein: From structure and toxicity to therapeutic target. Nat. Rev. Neurosci. 2013, 14, 38–48.
  28. Maroteaux, L.; Campanelli, J.T.; Scheller, R.H. Synuclein: A neuron-specific protein localized to the nucleus and presynaptic nerve terminal. J. Neurosci. 1988, 8, 2804–2815.
  29. Dikiy, I.; Eliezer, D. N-terminal acetylation stabilizes N-terminal helicity in lipid- and micelle-bound alpha-synuclein and increases its affinity for physiological membranes. J. Biol. Chem. 2014, 289, 3652–3665.
  30. Lazaro, D.F.; Dias, M.C.; Carija, A.; Navarro, S.; Madaleno, C.S.; Tenreiro, S.; Ventura, S.; Outeiro, T.F. The effects of the novel A53E alpha-synuclein mutation on its oligomerization and aggregation. Acta Neuropathol. Commun. 2016, 4, 128.
  31. Kim, T.D.; Paik, S.R.; Yang, C.H.; Kim, J. Structural changes in alpha-synuclein affect its chaperone-like activity in vitro. Protein Sci. 2000, 9, 2489–2496.
  32. Kim, T.D.; Choi, E.; Rhim, H.; Paik, S.R.; Yang, C.H. Alpha-synuclein has structural and functional similarities to small heat shock proteins. Biochem. Biophys. Res. Commun. 2004, 324, 1352–1359.
  33. Chandra, S.; Gallardo, G.; Fernandez-Chacon, R.; Schluter, O.M.; Sudhof, T.C. Alpha-synuclein cooperates with CSPalpha in preventing neurodegeneration. Cell 2005, 123, 383–396.
  34. Ahn, M.; Kim, S.; Kang, M.; Ryu, Y.; Kim, T.D. Chaperone-like activities of alpha-synuclein: Alpha-synuclein assists enzyme activities of esterases. Biochem. Biophys. Res. Commun. 2006, 346, 1142–1149.
  35. Giasson, B.I.; Lee, V.M. Parkin and the molecular pathways of Parkinson’s disease. Neuron 2001, 31, 885–888.
  36. Tong, J.; Wong, H.; Guttman, M.; Ang, L.C.; Forno, L.S.; Shimadzu, M.; Rajput, A.H.; Muenter, M.D.; Kish, S.J.; Hornykiewicz, O.; et al. Brain alpha-synuclein accumulation in multiple system atrophy, Parkinson’s disease and progressive supranuclear palsy: A comparative investigation. Brain 2010, 133 Pt 1, 172–188.
  37. Karube, H.; Sakamoto, M.; Arawaka, S.; Hara, S.; Sato, H.; Ren, C.H.; Goto, S.; Koyama, S.; Wada, M.; Kawanami, T.; et al. N-terminal region of alpha-synuclein is essential for the fatty acid-induced oligomerization of the molecules. FEBS Lett. 2008, 582, 3693–3700.
  38. Cookson, M.R. The biochemistry of Parkinson’s disease. Annu. Rev. Biochem. 2005, 74, 29–52.
  39. Murray, I.V.; Giasson, B.I.; Quinn, S.M.; Koppaka, V.; Axelsen, P.H.; Ischiropoulos, H.; Trojanowski, J.Q.; Lee, V.M. Role of alpha-synuclein carboxy-terminus on fibril formation in vitro. Biochemistry 2003, 42, 8530–8540.
  40. Giasson, B.I.; Uryu, K.; Trojanowski, J.Q.; Lee, V.M. Mutant and wild type human alpha-synucleins assemble into elongated filaments with distinct morphologies in vitro. J. Biol. Chem. 1999, 274, 7619–7622.
  41. Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum, R.; et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science 2003, 302, 841.
  42. Hardy, J.; Cai, H.; Cookson, M.R.; Gwinn-Hardy, K.; Singleton, A. Genetics of Parkinson’s disease and parkinsonism. Ann. Neurol. 2006, 60, 389–398.
  43. Polymeropoulos, M.H.; Lavedan, C.; Leroy, E.; Ide, S.E.; Dehejia, A.; Dutra, A.; Pike, B.; Root, H.; Rubenstein, J.; Boyer, R.; et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997, 276, 2045–2047.
  44. Kruger, R.; Kuhn, W.; Muller, T.; Woitalla, D.; Graeber, M.; Kosel, S.; Przuntek, H.; Epplen, J.T.; Schols, L.; Riess, O. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat. Genet. 1998, 18, 106–108.
  45. Greenbaum, E.A.; Graves, C.L.; Mishizen-Eberz, A.J.; Lupoli, M.A.; Lynch, D.R.; Englander, S.W.; Axelsen, P.H.; Giasson, B.I. The E46K mutation in alpha-synuclein increases amyloid fibril formation. J. Biol. Chem. 2005, 280, 7800–7807.
  46. Fujiwara, H.; Hasegawa, M.; Dohmae, N.; Kawashima, A.; Masliah, E.; Goldberg, M.S.; Shen, J.; Takio, K.; Iwatsubo, T. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat. Cell Biol. 2002, 4, 160–164.
  47. Shimura, H.; Schlossmacher, M.G.; Hattori, N.; Frosch, M.P.; Trockenbacher, A.; Schneider, R.; Mizuno, Y.; Kosik, K.S.; Selkoe, D.J. Ubiquitination of a new form of alpha-synuclein by parkin from human brain: Implications for Parkinson’s disease. Science 2001, 293, 263–269.
  48. Hasegawa, M.; Fujiwara, H.; Nonaka, T.; Wakabayashi, K.; Takahashi, H.; Lee, V.M.; Trojanowski, J.Q.; Mann, D.; Iwatsubo, T. Phosphorylated alpha-synuclein is ubiquitinated in alpha-synucleinopathy lesions. J. Biol. Chem. 2002, 277, 49071–49076.
  49. Dorval, V.; Fraser, P.E. Small ubiquitin-like modifier (SUMO) modification of natively unfolded proteins tau and alpha-synuclein. J. Biol. Chem. 2006, 281, 9919–9924.
  50. Bartels, T.; Choi, J.G.; Selkoe, D.J. alpha-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature 2011, 477, 107–110.
  51. Giasson, B.I.; Duda, J.E.; Murray, I.V.; Chen, Q.; Souza, J.M.; Hurtig, H.I.; Ischiropoulos, H.; Trojanowski, J.Q.; Lee, V.M. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science 2000, 290, 985–989.
  52. Guerrero, E.; Vasudevaraju, P.; Hegde, M.L.; Britton, G.B.; Rao, K.S. Recent advances in alpha-synuclein functions, advanced glycation, and toxicity: Implications for Parkinson’s disease. Mol. Neurobiol. 2013, 47, 525–536.
  53. Cooper, A.A.; Gitler, A.D.; Cashikar, A.; Haynes, C.M.; Hill, K.J.; Bhullar, B.; Liu, K.; Xu, K.; Strathearn, K.E.; Liu, F.; et al. Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson’s models. Science 2006, 313, 324–328.
  54. Gitler, A.D.; Bevis, B.J.; Shorter, J.; Strathearn, K.E.; Hamamichi, S.; Su, L.J.; Caldwell, K.A.; Caldwell, G.A.; Rochet, J.C.; McCaffery, J.M.; et al. The Parkinson’s disease protein alpha-synuclein disrupts cellular Rab homeostasis. Proc. Natl. Acad. Sci. USA 2008, 105, 145–150.
  55. Soper, J.H.; Roy, S.; Stieber, A.; Lee, E.; Wilson, R.B.; Trojanowski, J.Q.; Burd, C.G.; Lee, V.M. Alpha-synuclein-induced aggregation of cytoplasmic vesicles in Saccharomyces cerevisiae. Mol. Biol. Cell 2008, 19, 1093–1103.
  56. Witt, S.N.; Flower, T.R. alpha-Synuclein, oxidative stress and apoptosis from the perspective of a yeast model of Parkinson’s disease. FEMS Yeast Res. 2006, 6, 1107–1116.
  57. Buttner, S.; Bitto, A.; Ring, J.; Augsten, M.; Zabrocki, P.; Eisenberg, T.; Jungwirth, H.; Hutter, S.; Carmona-Gutierrez, D.; Kroemer, G.; et al. Functional mitochondria are required for alpha-synuclein toxicity in aging yeast. J. Biol. Chem. 2008, 283, 7554–7560.
  58. Su, L.J.; Auluck, P.K.; Outeiro, T.F.; Yeger-Lotem, E.; Kritzer, J.A.; Tardiff, D.F.; Strathearn, K.E.; Liu, F.; Cao, S.; Hamamichi, S.; et al. Compounds from an unbiased chemical screen reverse both ER-to-Golgi trafficking defects and mitochondrial dysfunction in Parkinson’s disease models. Dis. Model Mech. 2010, 3, 194–208.
  59. Dixon, C.; Mathias, N.; Zweig, R.M.; Davis, D.A.; Gross, D.S. Alpha-synuclein targets the plasma membrane via the secretory pathway and induces toxicity in yeast. Genetics 2005, 170, 47–59.
  60. Popova, B.; Kleinknecht, A.; Braus, G.H. Posttranslational Modifications and Clearing of alpha-Synuclein Aggregates in Yeast. Biomolecules 2015, 5, 617–634.
  61. Petroi, D.; Popova, B.; Taheri-Talesh, N.; Irniger, S.; Shahpasandzadeh, H.; Zweckstetter, M.; Outeiro, T.F.; Braus, G.H. Aggregate clearance of alpha-synuclein in Saccharomyces cerevisiae depends more on autophagosome and vacuole function than on the proteasome. J. Biol. Chem. 2012, 287, 27567–27579.
  62. Sweeney, P.; Park, H.; Baumann, M.; Dunlop, J.; Frydman, J.; Kopito, R.; McCampbell, A.; Leblanc, G.; Venkateswaran, A.; Nurmi, A.; et al. Protein misfolding in neurodegenerative diseases: Implications and strategies. Transl. Neurodegener. 2017, 6, 6.
  63. Okamoto, K.; Hirai, S.; Yamazaki, T.; Sun, X.Y.; Nakazato, Y. New ubiquitin-positive intraneuronal inclusions in the extra-motor cortices in patients with amyotrophic lateral sclerosis. Neurosci. Lett. 1991, 129, 233–236.
  64. Watanabe, M.; Dykes-Hoberg, M.; Culotta, V.C.; Price, D.L.; Wong, P.C.; Rothstein, J.D. Histological evidence of protein aggregation in mutant SOD1 transgenic mice and in amyotrophic lateral sclerosis neural tissues. Neurobiol. Dis. 2001, 8, 933–941.
  65. Arai, T.; Hasegawa, M.; Akiyama, H.; Ikeda, K.; Nonaka, T.; Mori, H. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem. Biophys. Res. Commun. 2006, 351, 602–611.
  66. Neumann, M.; Sampathu, D.M.; Kwong, L.K.; Truax, A.C.; Micsenyi, M.C.; Chou, T.T.; Bruce, J.; Schuck, T.; Grossman, M.; Clark, C.M.; et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006, 314, 130–133.
  67. Mackenzie, I.R.; Bigio, E.H.; Ince, P.G.; Geser, F.; Neumann, M.; Cairns, N.J.; Kwong, L.K.; Forman, M.S.; Ravits, J.; Stewart, H.; et al. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann. Neurol. 2007, 61, 427–434.
  68. Kwiatkowski, T.J., Jr.; Bosco, D.A.; Leclerc, A.L.; Tamrazian, E.; Vanderburg, C.R.; Russ, C.; Davis, A.; Gilchrist, J.; Kasarskis, E.J.; Munsat, T.; et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 2009, 323, 1205–1208.
  69. Vance, C.; Rogelj, B.; Hortobagyi, T.; De Vos, K.J.; Nishimura, A.L.; Sreedharan, J.; Hu, X.; Smith, B.; Ruddy, D.; Wright, P.; et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 2009, 323, 1208–1211.
  70. Al-Sarraj, S.; King, A.; Troakes, C.; Smith, B.; Maekawa, S.; Bodi, I.; Rogelj, B.; Al-Chalabi, A.; Hortobagyi, T.; Shaw, C.E. p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol. 2011, 122, 691–702.
  71. Johnson, B.S.; McCaffery, J.M.; Lindquist, S.; Gitler, A.D. A yeast TDP-43 proteinopathy model: Exploring the molecular determinants of TDP-43 aggregation and cellular toxicity. Proc. Natl. Acad. Sci. USA 2008, 105, 6439–6444.
  72. Johnson, B.S.; Snead, D.; Lee, J.J.; McCaffery, J.M.; Shorter, J.; Gitler, A.D. TDP-43 is intrinsically aggregation-prone, and amyotrophic lateral sclerosis-linked mutations accelerate aggregation and increase toxicity. J. Biol. Chem. 2009, 284, 20329–20339.
  73. Bastow, E.L.; Gourlay, C.W.; Tuite, M.F. Using yeast models to probe the molecular basis of amyotrophic lateral sclerosis. Biochem. Soc. Trans. 2011, 39, 1482–1487.
  74. Di Gregorio, S.E.; Duennwald, M.L. ALS Yeast Models-Past Success Stories and New Opportunities. Front. Mol. Neurosci. 2018, 11, 394.
  75. Sun, Z.; Diaz, Z.; Fang, X.; Hart, M.P.; Chesi, A.; Shorter, J.; Gitler, A.D. Molecular determinants and genetic modifiers of aggregation and toxicity for the ALS disease protein FUS/TLS. PLoS Biol. 2011, 9, e1000614.
  76. Alberti, S.; Halfmann, R.; King, O.; Kapila, A.; Lindquist, S. A systematic survey identifies prions and illuminates sequence features of prionogenic proteins. Cell 2009, 137, 146–158.
  77. Cushman, M.; Johnson, B.S.; King, O.D.; Gitler, A.D.; Shorter, J. Prion-like disorders: Blurring the divide between transmissibility and infectivity. J. Cell Sci. 2010, 123, 1191–1201.
  78. Shorter, J.; Lindquist, S. Prions as adaptive conduits of memory and inheritance. Nat. Rev. Genet. 2005, 6, 435–450.
  79. Kwong, L.K.; Uryu, K.; Trojanowski, J.Q.; Lee, V.M. TDP-43 proteinopathies: Neurodegenerative protein misfolding diseases without amyloidosis. Neurosignals 2008, 16, 41–51.
  80. Chen-Plotkin, A.S.; Lee, V.M.; Trojanowski, J.Q. TAR DNA-binding protein 43 in neurodegenerative disease. Nat. Rev. Neurol. 2010, 6, 211–220.
  81. Lagier-Tourenne, C.; Polymenidou, M.; Cleveland, D.W. TDP-43 and FUS/TLS: Emerging roles in RNA processing and neurodegeneration. Hum. Mol. Genet. 2010, 19, R46–R64.
  82. Hirano, M.; Hung, W.Y.; Cole, N.; Azim, A.C.; Deng, H.X.; Siddique, T. Multiple transcripts of the human Cu,Zn superoxide dismutase gene. Biochem. Biophys. Res. Commun. 2000, 276, 52–56.
  83. Kawamata, H.; Manfredi, G. Import, maturation, and function of SOD1 and its copper chaperone CCS in the mitochondrial intermembrane space. Antioxid. Redox Signal. 2010, 13, 1375–1384.
  84. Tsang, C.K.; Liu, Y.; Thomas, J.; Zhang, Y.; Zheng, X.F. Superoxide dismutase 1 acts as a nuclear transcription factor to regulate oxidative stress resistance. Nat. Commun. 2014, 5, 3446.
  85. Huai, J.; Zhang, Z. Structural Properties and Interaction Partners of Familial ALS-Associated SOD1 Mutants. Front. Neurol. 2019, 10, 527.
  86. Martins, D.; English, A.M. SOD1 oxidation and formation of soluble aggregates in yeast: Relevance to sporadic ALS development. Redox Biol. 2014, 2, 632–639.
  87. Nishida, C.R.; Gralla, E.B.; Valentine, J.S. Characterization of three yeast copper-zinc superoxide dismutase mutants analogous to those coded for in familial amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 1994, 91, 9906–9910.
  88. Gunther, M.R.; Vangilder, R.; Fang, J.; Beattie, D.S. Expression of a familial amyotrophic lateral sclerosis-associated mutant human superoxide dismutase in yeast leads to decreased mitochondrial electron transport. Arch. Biochem. Biophys. 2004, 431, 207–214.
  89. Bastow, E.L.; Peswani, A.R.; Tarrant, D.S.; Pentland, D.R.; Chen, X.; Morgan, A.; Staniforth, G.L.; Tullet, J.M.; Rowe, M.L.; Howard, M.J.; et al. New links between SOD1 and metabolic dysfunction from a yeast model of amyotrophic lateral sclerosis. J. Cell Sci. 2016, 129, 4118–4129.
  90. Rabizadeh, S.; Gralla, E.B.; Borchelt, D.R.; Gwinn, R.; Valentine, J.S.; Sisodia, S.; Wong, P.; Lee, M.; Hahn, H.; Bredesen, D.E. Mutations associated with amyotrophic lateral sclerosis convert superoxide dismutase from an antiapoptotic gene to a proapoptotic gene: Studies in yeast and neural cells. Proc. Natl. Acad Sci. USA 1995, 92, 3024–3028.
  91. Corson, L.B.; Strain, J.J.; Culotta, V.C.; Cleveland, D.W. Chaperone-facilitated copper binding is a property common to several classes of familial amyotrophic lateral sclerosis-linked superoxide dismutase mutants. Proc. Natl. Acad. Sci. USA 1998, 95, 6361–6366.
  92. Kerman, A.; Liu, H.; Croul, S.; Bilbao, J.; Rogaeva, E.; Zinman, L. Amyotrophic lateral sclerosis is a non-amyloid disease in which extensive misfolding of SOD1 is unique to the familial form. Acta Neuropathol. 2010, 119, 335–344.
  93. Rajaratnam, S.; Soman, A.P.; Phalguna, K.S.; Pradhan, S.S.; Manjunath, M.; Rao, R.K.; Dandamudi, R.B.; Bhagavatham, S.K.S.; Pulukool, S.K.; Rathnakumar, S.; et al. Integrated Omic Analysis Delineates Pathways Modulating Toxic TDP-43 Protein Aggregates in Amyotrophic Lateral Sclerosis. Cells 2023, 12, 1228.
  94. Sai Swaroop, R.; Akhil, P.S.; Sai Sanwid, P.; Bandana, P.; Raksha, R.K.; Meghana, M.; Bibha, C.; Sivaramakrishnan, V. Integrated multi-omic data analysis and validation with yeast model show oxidative phosphorylation modulates protein aggregation in amyotrophic lateral sclerosis. J. Biomol. Struct. Dyn. 2023, 41, 5548–5567.
  95. Janning, D.; Igaev, M.; Sundermann, F.; Bruhmann, J.; Beutel, O.; Heinisch, J.J.; Bakota, L.; Piehler, J.; Junge, W.; Brandt, R. Single-molecule tracking of tau reveals fast kiss-and-hop interaction with microtubules in living neurons. Mol. Biol. Cell 2014, 25, 3541–3551.
  96. Barbier, P.; Zejneli, O.; Martinho, M.; Lasorsa, A.; Belle, V.; Smet-Nocca, C.; Tsvetkov, P.O.; Devred, F.; Landrieu, I. Role of Tau as a Microtubule-Associated Protein: Structural and Functional Aspects. Front. Aging Neurosci. 2019, 11, 204.
  97. Spillantini, M.G.; Goedert, M. Tau protein pathology in neurodegenerative diseases. Trends Neurosci. 1998, 21, 428–433.
  98. Weaver, C.L.; Espinoza, M.; Kress, Y.; Davies, P. Conformational change as one of the earliest alterations of tau in Alzheimer’s disease. Neurobiol. Aging 2000, 21, 719–727.
  99. Alquezar, C.; Arya, S.; Kao, A.W. Tau Post-translational Modifications: Dynamic Transformers of Tau Function, Degradation, and Aggregation. Front. Neurol. 2020, 11, 595532.
  100. Ciaccioli, G.; Martins, A.; Rodrigues, C.; Vieira, H.; Calado, P. A powerful yeast model to investigate the synergistic interaction of alpha-synuclein and tau in neurodegeneration. PLoS ONE 2013, 8, e55848.
  101. Zubcic, K.; Franic, D.; Pravica, M.; Hof, P.R.; Simic, G.; Boban, M. Effects of heterologous human tau protein expression in yeast models of proteotoxic stress response. CNS Neurosci. Ther. 2023.
  102. Vandebroek, T.; Vanhelmont, T.; Terwel, D.; Borghgraef, P.; Lemaire, K.; Snauwaert, J.; Wera, S.; Van Leuven, F.; Winderickx, J. Identification and isolation of a hyperphosphorylated, conformationally changed intermediate of human protein tau expressed in yeast. Biochemistry 2005, 44, 11466–11475.
  103. Rencus-Lazar, S.; DeRowe, Y.; Adsi, H.; Gazit, E.; Laor, D. Yeast Models for the Study of Amyloid-Associated Disorders and Development of Future Therapy. Front. Mol. Biosci. 2019, 6, 15.
  104. Tuite, M.F. Yeast models of neurodegenerative diseases. Prog. Mol. Biol. Transl. Sci. 2019, 168, 351–379.
  105. Zimmermann, A.; Hofer, S.; Pendl, T.; Kainz, K.; Madeo, F.; Carmona-Gutierrez, D. Yeast as a tool to identify anti-aging compounds. FEMS Yeast Res. 2018, 18, foy020.
  106. Chernova, T.A.; Chernoff, Y.O.; Wilkinson, K.D. Yeast Models for Amyloids and Prions: Environmental Modulation and Drug Discovery. Molecules 2019, 24, 3388.
More
This entry is offline, you can click here to edit this entry!
Video Production Service