#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Identification of Human Proteins That Modify Misfolding and Proteotoxicity of Pathogenic Ataxin-1


Proteins with long, pathogenic polyglutamine (polyQ) sequences have an enhanced propensity to spontaneously misfold and self-assemble into insoluble protein aggregates. Here, we have identified 21 human proteins that influence polyQ-induced ataxin-1 misfolding and proteotoxicity in cell model systems. By analyzing the protein sequences of these modifiers, we discovered a recurrent presence of coiled-coil (CC) domains in ataxin-1 toxicity enhancers, while such domains were not present in suppressors. This suggests that CC domains contribute to the aggregation- and toxicity-promoting effects of modifiers in mammalian cells. We found that the ataxin-1–interacting protein MED15, computationally predicted to possess an N-terminal CC domain, enhances spontaneous ataxin-1 aggregation in cell-based assays, while no such effect was observed with the truncated protein MED15ΔCC, lacking such a domain. Studies with recombinant proteins confirmed these results and demonstrated that the N-terminal CC domain of MED15 (MED15CC) per se is sufficient to promote spontaneous ataxin-1 aggregation in vitro. Moreover, we observed that a hybrid Pum1 protein harboring the MED15CC domain promotes ataxin-1 aggregation in cell model systems. In strong contrast, wild-type Pum1 lacking a CC domain did not stimulate ataxin-1 polymerization. These results suggest that proteins with CC domains are potent enhancers of polyQ-mediated protein misfolding and aggregation in vitro and in vivo.


Vyšlo v časopise: Identification of Human Proteins That Modify Misfolding and Proteotoxicity of Pathogenic Ataxin-1. PLoS Genet 8(8): e32767. doi:10.1371/journal.pgen.1002897
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1002897

Souhrn

Proteins with long, pathogenic polyglutamine (polyQ) sequences have an enhanced propensity to spontaneously misfold and self-assemble into insoluble protein aggregates. Here, we have identified 21 human proteins that influence polyQ-induced ataxin-1 misfolding and proteotoxicity in cell model systems. By analyzing the protein sequences of these modifiers, we discovered a recurrent presence of coiled-coil (CC) domains in ataxin-1 toxicity enhancers, while such domains were not present in suppressors. This suggests that CC domains contribute to the aggregation- and toxicity-promoting effects of modifiers in mammalian cells. We found that the ataxin-1–interacting protein MED15, computationally predicted to possess an N-terminal CC domain, enhances spontaneous ataxin-1 aggregation in cell-based assays, while no such effect was observed with the truncated protein MED15ΔCC, lacking such a domain. Studies with recombinant proteins confirmed these results and demonstrated that the N-terminal CC domain of MED15 (MED15CC) per se is sufficient to promote spontaneous ataxin-1 aggregation in vitro. Moreover, we observed that a hybrid Pum1 protein harboring the MED15CC domain promotes ataxin-1 aggregation in cell model systems. In strong contrast, wild-type Pum1 lacking a CC domain did not stimulate ataxin-1 polymerization. These results suggest that proteins with CC domains are potent enhancers of polyQ-mediated protein misfolding and aggregation in vitro and in vivo.


Zdroje

1. TranPB, MillerRJ (1999) Aggregates in neurodegenerative disease: crowds and power? Trends Neurosci 22: 194–197.

2. ShastryBS (2003) Neurodegenerative disorders of protein aggregation. Neurochem Int 43: 1–7.

3. RossCA, PoirierMA (2005) Opinion: What is the role of protein aggregation in neurodegeneration? Nat Rev Mol Cell Biol 6: 891–898.

4. DobsonCM (2003) Protein folding and misfolding. Nature 426: 884–890.

5. ChaiY, ShaoJ, MillerVM, WilliamsA, PaulsonHL (2002) Live-cell imaging reveals divergent intracellular dynamics of polyglutamine disease proteins and supports a sequestration model of pathogenesis. Proc Natl Acad Sci U S A 99: 9310–9315.

6. StenoienDL, MielkeM, ManciniMA (2002) Intranuclear ataxin1 inclusions contain both fast- and slow-exchanging components. Nat Cell Biol 4: 806–810.

7. ScherzingerE, LurzR, TurmaineM, MangiariniL, HollenbachB, et al. (1997) Huntingtin-encoded polyglutamine expansions form amyloid-like protein aggregates in vitro and in vivo. Cell 90: 549–558.

8. BevivinoAE, LollPJ (2001) An expanded glutamine repeat destabilizes native ataxin-3 structure and mediates formation of parallel beta -fibrils. Proc Natl Acad Sci U S A 98: 11955–11960.

9. PerutzMF, PopeBJ, OwenD, WankerEE, ScherzingerE (2002) Aggregation of proteins with expanded glutamine and alanine repeats of the glutamine-rich and asparagine-rich domains of Sup35 and of the amyloid beta-peptide of amyloid plaques. Proc Natl Acad Sci U S A 99: 5596–5600.

10. LiW, SerpellLC, CarterWJ, RubinszteinDC, HuntingtonJA (2006) Expression and characterization of full-length human huntingtin, an elongated HEAT repeat protein. J Biol Chem 281: 15916–15922.

11. LunkesA, LindenbergKS, Ben-HaiemL, WeberC, DevysD, et al. (2002) Proteases acting on mutant huntingtin generate cleaved products that differentially build up cytoplasmic and nuclear inclusions. Mol Cell 10: 259–269.

12. WellingtonCL, EllerbyLM, GutekunstCA, RogersD, WarbyS, et al. (2002) Caspase cleavage of mutant huntingtin precedes neurodegeneration in Huntington's disease. J Neurosci 22: 7862–7872.

13. ZoghbiHY, OrrHT (2009) Pathogenic mechanisms of a polyglutamine-mediated neurodegenerative disease, spinocerebellar ataxia type 1. J Biol Chem 284: 7425–7429.

14. KochP, BreuerP, PeitzM, JungverdorbenJ, KesavanJ, et al. (2011) Excitation-induced ataxin-3 aggregation in neurons from patients with Machado-Joseph disease. Nature 480: 543–546.

15. BurrightEN, DavidsonJD, DuvickLA, KoshyB, ZoghbiHY, et al. (1997) Identification of a self-association region within the SCA1 gene product, ataxin-1. Hum Mol Genet 6: 513–518.

16. KlementIA, SkinnerPJ, KaytorMD, YiH, HerschSM, et al. (1998) Ataxin-1 nuclear localization and aggregation: role in polyglutamine-induced disease in SCA1 transgenic mice. Cell 95: 41–53.

17. BilenJ, BoniniNM (2007) Genome-wide screen for modifiers of ataxin-3 neurodegeneration in Drosophila. PLoS Genet 3: e177 doi:10.1371/journal.pgen.0030177.

18. DoumanisJ, WadaK, KinoY, MooreAW, NukinaN (2009) RNAi screening in Drosophila cells identifies new modifiers of mutant huntingtin aggregation. PLoS ONE 4: e7275 doi:10.1371/journal.pone.0007275.

19. Fernandez-FunezP, Nino-RosalesML, de GouyonB, SheWC, LuchakJM, et al. (2000) Identification of genes that modify ataxin-1-induced neurodegeneration. Nature 408: 101–106.

20. GhoshS, FeanyMB (2004) Comparison of pathways controlling toxicity in the eye and brain in Drosophila models of human neurodegenerative diseases. Hum Mol Genet 13: 2011–2018.

21. GiorginiF, GuidettiP, NguyenQ, BennettSC, MuchowskiPJ (2005) A genomic screen in yeast implicates kynurenine 3-monooxygenase as a therapeutic target for Huntington disease. Nat Genet 37: 526–531.

22. Kazemi-EsfarjaniP, BenzerS (2000) Genetic suppression of polyglutamine toxicity in Drosophila. Science 287: 1837–1840.

23. MorleyJF, BrignullHR, WeyersJJ, MorimotoRI (2002) The threshold for polyglutamine-expansion protein aggregation and cellular toxicity is dynamic and influenced by aging in Caenorhabditis elegans. Proc Natl Acad Sci U S A 99: 10417–10422.

24. NollenEA, GarciaSM, van HaaftenG, KimS, ChavezA, et al. (2004) Genome-wide RNA interference screen identifies previously undescribed regulators of polyglutamine aggregation. Proc Natl Acad Sci U S A 101: 6403–6408.

25. SteffanJS, BodaiL, PallosJ, PoelmanM, McCampbellA, et al. (2001) Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila. Nature 413: 739–743.

26. WillinghamS, OuteiroTF, DeVitMJ, LindquistSL, MuchowskiPJ (2003) Yeast genes that enhance the toxicity of a mutant huntingtin fragment or alpha-synuclein. Science 302: 1769–1772.

27. CummingsCJ, SunY, OpalP, AntalffyB, MestrilR, et al. (2001) Over-expression of inducible HSP70 chaperone suppresses neuropathology and improves motor function in SCA1 mice. Hum Mol Genet 10: 1511–1518.

28. SayersEW, BarrettT, BensonDA, BoltonE, BryantSH, et al. (2011) Database resources of the National Center for Biotechnology Information. Nucleic Acids Res 39: D38–51.

29. LimJ, HaoT, ShawC, PatelAJ, SzaboG, et al. (2006) A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration. Cell 125: 801–814.

30. KaltenbachLS, RomeroE, BecklinRR, ChettierR, BellR, et al. (2007) Huntingtin interacting proteins are genetic modifiers of neurodegeneration. PLoS Genet 3: e82 doi:10.1371/journal.pgen.0030082.

31. MatsuyamaA, YoshidaM (2009) Systematic cloning of an ORFeome using the Gateway system. Methods Mol Biol 577: 11–24.

32. WankerEE, ScherzingerE, HeiserV, SittlerA, EickhoffH, et al. (1999) Membrane filter assay for detection of amyloid-like polyglutamine-containing protein aggregates. Methods Enzymol 309: 375–386.

33. DietzlG, ChenD, SchnorrerF, SuKC, BarinovaY, et al. (2007) A genome-wide transgenic RNAi library for conditional gene inactivation in Drosophila. Nature 448: 151–156.

34. HosackDA, DennisGJr, ShermanBT, LaneHC, LempickiRA (2003) Identifying biological themes within lists of genes with EASE. Genome Biol 4: R70.

35. KappLD, LorschJR (2004) The molecular mechanics of eukaryotic translation. Annu Rev Biochem 73: 657–704.

36. BertiL, MittlerG, PrzemeckGK, StelzerG, GunzlerB, et al. (2001) Isolation and characterization of a novel gene from the DiGeorge chromosomal region that encodes for a mediator subunit. Genomics 74: 320–332.

37. de MoorCH, MeijerH, LissendenS (2005) Mechanisms of translational control by the 3′ UTR in development and differentiation. Semin Cell Dev Biol 16: 49–58.

38. KrugerM, BegerC, LiQX, WelchPJ, TritzR, et al. (2000) Identification of eIF2Bgamma and eIF2gamma as cofactors of hepatitis C virus internal ribosome entry site-mediated translation using a functional genomics approach. Proc Natl Acad Sci U S A 97: 8566–8571.

39. FiumaraF, FioritiL, KandelER, HendricksonWA (2010) Essential role of coiled coils for aggregation and activity of Q/N-rich prions and PolyQ proteins. Cell 143: 1121–1135.

40. SchaeferMH, WankerEE, Andrade-NavarroMA (2012) Evolution and function of CAG/polyglutamine repeats in protein-protein interaction networks. Nucleic Acids Res

41. LupasA, Van DykeM, StockJ (1991) Predicting coiled coils from protein sequences. Science 252: 1162–1164.

42. FurukawaY, KanekoK, MatsumotoG, KurosawaM, NukinaN (2009) Cross-seeding fibrillation of Q/N-rich proteins offers new pathomechanism of polyglutamine diseases. J Neurosci 29: 5153–5162.

43. PerezMK, PaulsonHL, PendseSJ, SaionzSJ, BoniniNM, et al. (1998) Recruitment and the role of nuclear localization in polyglutamine-mediated aggregation. J Cell Biol 143: 1457–1470.

44. ChaurasiaG, MalhotraS, RussJ, SchnoeglS, HanigC, et al. (2009) UniHI 4: new tools for query, analysis and visualization of the human protein-protein interactome. Nucleic Acids Res 37: D657–660.

45. PalidworGA, ShcherbininS, HuskaMR, RaskoT, StelzlU, et al. (2009) Detection of alpha-rod protein repeats using a neural network and application to huntingtin. PLoS Comput Biol 5: e1000304 doi:10.1371/journal.pcbi.1000304.

46. BoeddrichA, GaumerS, HaackeA, TzvetkovN, AlbrechtM, et al. (2006) An arginine/lysine-rich motif is crucial for VCP/p97-mediated modulation of ataxin-3 fibrillogenesis. EMBO J 25: 1547–1558.

47. MuchowskiPJ, SchaffarG, SittlerA, WankerEE, Hayer-HartlMK, et al. (2000) Hsp70 and hsp40 chaperones can inhibit self-assembly of polyglutamine proteins into amyloid-like fibrils. Proc Natl Acad Sci U S A 97: 7841–7846.

48. JohnsonWC (1999) Analyzing protein circular dichroism spectra for accurate secondary structures. Proteins 35: 307–312.

49. Perez-IratxetaC, Andrade-NavarroMA (2008) K2D2: estimation of protein secondary structure from circular dichroism spectra. BMC Struct Biol 8: 25.

50. McAlindenA, SmithTA, SandellLJ, FicheuxD, ParryDA, et al. (2003) Alpha-helical coiled-coil oligomerization domains are almost ubiquitous in the collagen superfamily. J Biol Chem 278: 42200–42207.

51. AlfadhliA, SteelE, FinlayL, BachingerHP, BarklisE (2002) Hantavirus nucleocapsid protein coiled-coil domains. J Biol Chem 277: 27103–27108.

52. SheehanD, MeadeG, FoleyVM, DowdCA (2001) Structure, function and evolution of glutathione transferases: implications for classification of non-mammalian members of an ancient enzyme superfamily. Biochem J 360: 1–16.

53. TaylorDL (2007) Past, present, and future of high content screening and the field of cellomics. Methods Mol Biol 356: 3–18.

54. VigPJ, HearstS, ShaoQ, LopezME, MurphyHA2nd, et al. (2011) Glial S100B Protein Modulates Mutant Ataxin-1 Aggregation and Toxicity: TRTK12 Peptide, a Potential Candidate for SCA1 Therapy. Cerebellum 10: 254–266.

55. van HamTJ, BreitlingR, SwertzMA, NollenEA (2009) Neurodegenerative diseases: Lessons from genome-wide screens in small model organisms. EMBO Mol Med 1: 360–370.

56. ZoghbiHY, OrrHT (2000) Glutamine repeats and neurodegeneration. Annu Rev Neurosci 23: 217–247.

57. MorimotoRI (2008) Proteotoxic stress and inducible chaperone networks in neurodegenerative disease and aging. Genes Dev 22: 1427–1438.

58. PowersET, MorimotoRI, DillinA, KellyJW, BalchWE (2009) Biological and chemical approaches to diseases of proteostasis deficiency. Annu Rev Biochem 78: 959–991.

59. WarrickJM, ChanHY, Gray-BoardGL, ChaiY, PaulsonHL, et al. (1999) Suppression of polyglutamine-mediated neurodegeneration in Drosophila by the molecular chaperone HSP70. Nat Genet 23: 425–428.

60. HartlFU, BracherA, Hayer-HartlM (2011) Molecular chaperones in protein folding and proteostasis. Nature 475: 324–332.

61. HartlFU (1996) Molecular chaperones in cellular protein folding. Nature 381: 571–579.

62. KoppenM, LangerT (2007) Protein degradation within mitochondria: versatile activities of AAA proteases and other peptidases. Crit Rev Biochem Mol Biol 42: 221–242.

63. Di BellaD, LazzaroF, BruscoA, PlumariM, BattagliaG, et al. (2010) Mutations in the mitochondrial protease gene AFG3L2 cause dominant hereditary ataxia SCA28. Nat Genet 42: 313–321.

64. MalteccaF, MagnoniR, CerriF, CoxGA, QuattriniA, et al. (2009) Haploinsufficiency of AFG3L2, the gene responsible for spinocerebellar ataxia type 28, causes mitochondria-mediated Purkinje cell dark degeneration. J Neurosci 29: 9244–9254.

65. VesseyJP, SchoderboeckL, GinglE, LuziE, RieflerJ, et al. (2010) Mammalian Pumilio 2 regulates dendrite morphogenesis and synaptic function. Proc Natl Acad Sci U S A 107: 3222–3227.

66. LupasAN, GruberM (2005) The structure of alpha-helical coiled coils. Adv Protein Chem 70: 37–78.

67. ParryDA, FraserRD, SquireJM (2008) Fifty years of coiled-coils and alpha-helical bundles: a close relationship between sequence and structure. J Struct Biol 163: 258–269.

68. GasparNJ, KinzyTG, SchererBJ, HumbelinM, HersheyJW, et al. (1994) Translation initiation factor eIF-2. Cloning and expression of the human cDNA encoding the gamma-subunit. J Biol Chem 269: 3415–3422.

69. ScherzingerE, SittlerA, SchweigerK, HeiserV, LurzR, et al. (1999) Self-assembly of polyglutamine-containing huntingtin fragments into amyloid-like fibrils: implications for Huntington's disease pathology. Proc Natl Acad Sci U S A 96: 4604–4609.

70. KimballSR (1999) Eukaryotic initiation factor eIF2. Int J Biochem Cell Biol 31: 25–29.

71. ChangRC, YuMS, LaiCS (2006) Significance of molecular signaling for protein translation control in neurodegenerative diseases. Neurosignals 15: 249–258.

72. JiangHY, WekRC (2005) Phosphorylation of the alpha-subunit of the eukaryotic initiation factor-2 (eIF2alpha) reduces protein synthesis and enhances apoptosis in response to proteasome inhibition. J Biol Chem 280: 14189–14202.

73. ObitaT, SaksenaS, Ghazi-TabatabaiS, GillDJ, PerisicO, et al. (2007) Structural basis for selective recognition of ESCRT-III by the AAA ATPase Vps4. Nature 449: 735–739.

74. LataS, SchoehnG, JainA, PiresR, PiehlerJ, et al. (2008) Helical structures of ESCRT-III are disassembled by VPS4. Science 321: 1354–1357.

75. LeeJA, GaoFB (2008) ESCRT, autophagy, and frontotemporal dementia. BMB Rep 41: 827–832.

76. BellyA, BodonG, BlotB, BouronA, SadoulR, et al. (2010) CHMP2B mutants linked to frontotemporal dementia impair maturation of dendritic spines. J Cell Sci 123: 2943–2954.

77. DiamondMI, RobinsonMR, YamamotoKR (2000) Regulation of expanded polyglutamine protein aggregation and nuclear localization by the glucocorticoid receptor. Proc Natl Acad Sci U S A 97: 657–661.

78. WelchWJ, DiamondMI (2001) Glucocorticoid modulation of androgen receptor nuclear aggregation and cellular toxicity is associated with distinct forms of soluble expanded polyglutamine protein. Hum Mol Genet 10: 3063–3074.

79. PoirierMA, JiangH, RossCA (2005) A structure-based analysis of huntingtin mutant polyglutamine aggregation and toxicity: evidence for a compact beta-sheet structure. Hum Mol Genet 14: 765–774.

80. NagaiY, InuiT, PopielHA, FujikakeN, HasegawaK, et al. (2007) A toxic monomeric conformer of the polyglutamine protein. Nat Struct Mol Biol 14: 332–340.

81. AbediniA, RaleighDP (2009) A role for helical intermediates in amyloid formation by natively unfolded polypeptides? Phys Biol 6: 015005.

82. BalchWE, MorimotoRI, DillinA, KellyJW (2008) Adapting proteostasis for disease intervention. Science 319: 916–919.

83. TaitD, RiccioM, SittlerA, ScherzingerE, SantiS, et al. (1998) Ataxin-3 is transported into the nucleus and associates with the nuclear matrix. Hum Mol Genet 7: 991–997.

84. AbrahamVC, TaylorDL, HaskinsJR (2004) High content screening applied to large-scale cell biology. Trends Biotechnol 22: 15–22.

85. GoujonM, McWilliamH, LiW, ValentinF, SquizzatoS, et al. (2010) A new bioinformatics analysis tools framework at EMBL-EBI. Nucleic Acids Res 38: W695–699.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 8
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#