#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

ATXN2-CAG42 Sequesters PABPC1 into Insolubility and Induces FBXW8 in Cerebellum of Old Ataxic Knock-In Mice


Spinocerebellar Ataxia Type 2 (SCA2) is caused by expansion of a polyglutamine encoding triplet repeat in the human ATXN2 gene beyond (CAG)31. This is thought to mediate toxic gain-of-function by protein aggregation and to affect RNA processing, resulting in degenerative processes affecting preferentially cerebellar neurons. As a faithful animal model, we generated a knock-in mouse replacing the single CAG of murine Atxn2 with CAG42, a frequent patient genotype. This expansion size was inherited stably. The mice showed phenotypes with reduced weight and later motor incoordination. Although brain Atxn2 mRNA became elevated, soluble ATXN2 protein levels diminished over time, which might explain partial loss-of-function effects. Deficits in soluble ATXN2 protein correlated with the appearance of insoluble ATXN2, a progressive feature in cerebellum possibly reflecting toxic gains-of-function. Since in vitro ATXN2 overexpression was known to reduce levels of its protein interactor PABPC1, we studied expansion effects on PABPC1. In cortex, PABPC1 transcript and soluble and insoluble protein levels were increased. In the more vulnerable cerebellum, the progressive insolubility of PABPC1 was accompanied by decreased soluble protein levels, with PABPC1 mRNA showing no compensatory increase. The sequestration of PABPC1 into insolubility by ATXN2 function gains was validated in human cell culture. To understand consequences on mRNA processing, transcriptome profiles at medium and old age in three different tissues were studied and demonstrated a selective induction of Fbxw8 in the old cerebellum. Fbxw8 is encoded next to the Atxn2 locus and was shown in vitro to decrease the level of expanded insoluble ATXN2 protein. In conclusion, our data support the concept that expanded ATXN2 undergoes progressive insolubility and affects PABPC1 by a toxic gain-of-function mechanism with tissue-specific effects, which may be partially alleviated by the induction of FBXW8.


Vyšlo v časopise: ATXN2-CAG42 Sequesters PABPC1 into Insolubility and Induces FBXW8 in Cerebellum of Old Ataxic Knock-In Mice. PLoS Genet 8(8): e32767. doi:10.1371/journal.pgen.1002920
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1002920

Souhrn

Spinocerebellar Ataxia Type 2 (SCA2) is caused by expansion of a polyglutamine encoding triplet repeat in the human ATXN2 gene beyond (CAG)31. This is thought to mediate toxic gain-of-function by protein aggregation and to affect RNA processing, resulting in degenerative processes affecting preferentially cerebellar neurons. As a faithful animal model, we generated a knock-in mouse replacing the single CAG of murine Atxn2 with CAG42, a frequent patient genotype. This expansion size was inherited stably. The mice showed phenotypes with reduced weight and later motor incoordination. Although brain Atxn2 mRNA became elevated, soluble ATXN2 protein levels diminished over time, which might explain partial loss-of-function effects. Deficits in soluble ATXN2 protein correlated with the appearance of insoluble ATXN2, a progressive feature in cerebellum possibly reflecting toxic gains-of-function. Since in vitro ATXN2 overexpression was known to reduce levels of its protein interactor PABPC1, we studied expansion effects on PABPC1. In cortex, PABPC1 transcript and soluble and insoluble protein levels were increased. In the more vulnerable cerebellum, the progressive insolubility of PABPC1 was accompanied by decreased soluble protein levels, with PABPC1 mRNA showing no compensatory increase. The sequestration of PABPC1 into insolubility by ATXN2 function gains was validated in human cell culture. To understand consequences on mRNA processing, transcriptome profiles at medium and old age in three different tissues were studied and demonstrated a selective induction of Fbxw8 in the old cerebellum. Fbxw8 is encoded next to the Atxn2 locus and was shown in vitro to decrease the level of expanded insoluble ATXN2 protein. In conclusion, our data support the concept that expanded ATXN2 undergoes progressive insolubility and affects PABPC1 by a toxic gain-of-function mechanism with tissue-specific effects, which may be partially alleviated by the induction of FBXW8.


Zdroje

1. BauerPO, NukinaN (2009) The pathogenic mechanisms of polyglutamine diseases and current therapeutic strategies. J Neurochem 110: 1737–1765.

2. ImbertG, SaudouF, YvertG, DevysD, TrottierY, et al. (1996) Cloning of the gene for spinocerebellar ataxia 2 reveals a locus with high sensitivity to expanded CAG/glutamine repeats. Nat Genet 14: 285–291.

3. PulstSM, NechiporukA, NechiporukT, GispertS, ChenXN, et al. (1996) Moderate expansion of a normally biallelic trinucleotide repeat in spinocerebellar ataxia type 2. Nat Genet 14: 269–276.

4. SanpeiK, TakanoH, IgarashiS, SatoT, OyakeM, et al. (1996) Identification of the spinocerebellar ataxia type 2 gene using a direct identification of repeat expansion and cloning technique, DIRECT. Nat Genet 14: 277–284.

5. EldenAC, KimHJ, HartMP, Chen-PlotkinAS, JohnsonBS, et al. (2010) Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 466: 1069–1075.

6. GispertS, KurzA, WaibelS, BauerP, LiepeltI, et al. (2012) The modulation of Amyotrophic Lateral Sclerosis risk by Ataxin-2 intermediate polyglutamine expansions is a specific effect. Neurobiol Dis 45: 356–361.

7. RossOA, RutherfordNJ, BakerM, Soto-OrtolazaAI, CarrasquilloMM, et al. (2011) Ataxin-2 repeat-length variation and neurodegeneration. Hum Mol Genet 20: 3207–3212.

8. AuburgerGW (2012) Spinocerebellar ataxia type 2. Handb Clin Neurol 103: 423–436.

9. GiuntiP, SabbadiniG, SweeneyMG, DavisMB, VenezianoL, et al. (1998) The role of the SCA2 trinucleotide repeat expansion in 89 autosomal dominant cerebellar ataxia families. Frequency, clinical and genetic correlates. Brain 121(Pt 3):459–467.

10. JacobiH, BauerP, GiuntiP, LabrumR, SweeneyMG, et al. (2011) The natural history of spinocerebellar ataxia type 1, 2, 3, and 6: a 2-year follow-up study. Neurology 77: 1035–1041.

11. ScholsL, GispertS, VorgerdM, Menezes Vieira-SaeckerAM, BlankeP, et al. (1997) Spinocerebellar ataxia type 2. Genotype and phenotype in German kindreds. Arch Neurol 54: 1073–1080.

12. TrejoA, TarratsRM, AlonsoME, BollMC, OchoaA, et al. (2004) Assessment of the nutrition status of patients with Huntington's disease. Nutrition 20: 192–196.

13. AzizNA, van der MarckMA, PijlH, Olde RikkertMG, BloemBR, et al. (2008) Weight loss in neurodegenerative disorders. J Neurol 255: 1872–1880.

14. Abdel-AleemA, ZakiMS (2008) Spinocerebellar ataxia type 2 (SCA2) in an Egyptian family presenting with polyphagia and marked CAG expansion in infancy. J Neurol 255: 413–419.

15. EstradaR, GalarragaJ, OrozcoG, NodarseA, AuburgerG (1999) Spinocerebellar ataxia 2 (SCA2): morphometric analyses in 11 autopsies. Acta Neuropathol 97: 306–310.

16. GiergaK, BurkK, BauerM, Orozco DiazG, AuburgerG, et al. (2005) Involvement of the cranial nerves and their nuclei in spinocerebellar ataxia type 2 (SCA2). Acta Neuropathol 109: 617–631.

17. HocheF, SeidelK, BruntER, AuburgerG, ScholsL, et al. (2008) Involvement of the auditory brainstem system in spinocerebellar ataxia type 2 (SCA2), type 3 (SCA3) and type 7 (SCA7). Neuropathol Appl Neurobiol 34: 479–491.

18. Orozco DiazG, Nodarse FleitesA, Cordoves SagazR, AuburgerG (1990) Autosomal dominant cerebellar ataxia: clinical analysis of 263 patients from a homogeneous population in Holguin, Cuba. Neurology 40: 1369–1375.

19. RubU, SchultzC, Del TrediciK, GiergaK, ReifenbergerG, et al. (2003) Anatomically based guidelines for systematic investigation of the central somatosensory system and their application to a spinocerebellar ataxia type 2 (SCA2) patient. Neuropathol Appl Neurobiol 29: 418–433.

20. RubU, SeidelK, OzerdenI, GiergaK, BruntER, et al. (2007) Consistent affection of the central somatosensory system in spinocerebellar ataxia type 2 and type 3 and its significance for clinical symptoms and rehabilitative therapy. Brain Res Rev 53: 235–249.

21. ScherzedW, BruntER, HeinsenH, de VosRA, SeidelK, et al. (2011) Pathoanatomy of Cerebellar Degeneration in Spinocerebellar Ataxia Type 2 (SCA2) and Type 3 (SCA3). Cerebellum

22. TuinI, VossU, KangJS, KesslerK, RubU, et al. (2006) Stages of sleep pathology in spinocerebellar ataxia type 2 (SCA2). Neurology 67: 1966–1972.

23. Velazquez-PerezL, SeifriedC, Santos-FalconN, AbeleM, ZiemannU, et al. (2004) Saccade velocity is controlled by polyglutamine size in spinocerebellar ataxia 2. Ann Neurol 56: 444–447.

24. CharlesP, CamuzatA, BenammarN, SellalF, DesteeA, et al. (2007) Are interrupted SCA2 CAG repeat expansions responsible for parkinsonism? Neurology 69: 1970–1975.

25. Gwinn-HardyK, ChenJY, LiuHC, LiuTY, BossM, et al. (2000) Spinocerebellar ataxia type 2 with parkinsonism in ethnic Chinese. Neurology 55: 800–805.

26. RubU, GiergaK, BruntER, de VosRA, BauerM, et al. (2005) Spinocerebellar ataxias types 2 and 3: degeneration of the pre-cerebellar nuclei isolates the three phylogenetically defined regions of the cerebellum. J Neural Transm 112: 1523–1545.

27. HocheF, BalikoL, den DunnenW, SteineckerK, BartosL, et al. (2011) Spinocerebellar ataxia type 2 (SCA2): identification of early brain degeneration in one monozygous twin in the initial disease stage. Cerebellum 10: 245–253.

28. RagothamanM, SarangmathN, ChaudharyS, KhareV, MittalU, et al. (2004) Complex phenotypes in an Indian family with homozygous SCA2 mutations. Ann Neurol 55: 130–133.

29. SasakiH, WakisakaA, SanpeiK, TakanoH, IgarashiS, et al. (1998) Phenotype variation correlates with CAG repeat length in SCA2–a study of 28 Japanese patients. J Neurol Sci 159: 202–208.

30. FreundHJ, BarnikolUB, NolteD, TreuerH, AuburgerG, et al. (2007) Subthalamic-thalamic DBS in a case with spinocerebellar ataxia type 2 and severe tremor-A unusual clinical benefit. Mov Disord 22: 732–735.

31. HuynhDP, FigueroaK, HoangN, PulstSM (2000) Nuclear localization or inclusion body formation of ataxin-2 are not necessary for SCA2 pathogenesis in mouse or human. Nat Genet 26: 44–50.

32. KoyanoS, IwabuchiK, YagishitaS, KuroiwaY, UchiharaT (2002) Paradoxical absence of nuclear inclusion in cerebellar Purkinje cells of hereditary ataxias linked to CAG expansion. J Neurol Neurosurg Psychiatry 73: 450–452.

33. PangJT, GiuntiP, ChamberlainS, AnSF, VitalianiR, et al. (2002) Neuronal intranuclear inclusions in SCA2: a genetic, morphological and immunohistochemical study of two cases. Brain 125: 656–663.

34. OrrHT, ZoghbiHY (2007) Trinucleotide repeat disorders. Annu Rev Neurosci 30: 575–621.

35. FryerJD, YuP, KangH, Mandel-BrehmC, CarterAN, et al. (2011) Exercise and genetic rescue of SCA1 via the transcriptional repressor Capicua. Science 334: 690–693.

36. LiebermanAP, HarmisonG, StrandAD, OlsonJM, FischbeckKH (2002) Altered transcriptional regulation in cells expressing the expanded polyglutamine androgen receptor. Hum Mol Genet 11: 1967–1976.

37. ZuccatoC, ValenzaM, CattaneoE (2010) Molecular mechanisms and potential therapeutical targets in Huntington's disease. Physiol Rev 90: 905–981.

38. NonisD, SchmidtMH, van de LooS, EichF, DikicI, et al. (2008) Ataxin-2 associates with the endocytosis complex and affects EGF receptor trafficking. Cell Signal 20: 1725–1739.

39. RalserM, AlbrechtM, NonhoffU, LengauerT, LehrachH, et al. (2005) An integrative approach to gain insights into the cellular function of human ataxin-2. J Mol Biol 346: 203–214.

40. SatterfieldTF, PallanckLJ (2006) Ataxin-2 and its Drosophila homolog, ATX2, physically assemble with polyribosomes. Hum Mol Genet 15: 2523–2532.

41. NonhoffU, RalserM, WelzelF, PicciniI, BalzereitD, et al. (2007) Ataxin-2 interacts with the DEAD/H-box RNA helicase DDX6 and interferes with P-bodies and stress granules. Mol Biol Cell 18: 1385–1396.

42. van de LooS, EichF, NonisD, AuburgerG, NowockJ (2009) Ataxin-2 associates with rough endoplasmic reticulum. Exp Neurol 215: 110–118.

43. HuynhDP, Del BigioMR, HoDH, PulstSM (1999) Expression of ataxin-2 in brains from normal individuals and patients with Alzheimer's disease and spinocerebellar ataxia 2. Ann Neurol 45: 232–241.

44. WataseK, ZoghbiHY (2003) Modelling brain diseases in mice: the challenges of design and analysis. Nat Rev Genet 4: 296–307.

45. LescureA, LutzY, EberhardD, JacqX, KrolA, et al. (1994) The N-terminal domain of the human TATA-binding protein plays a role in transcription from TATA-containing RNA polymerase II and III promoters. Embo J 13: 1166–1175.

46. FisherCD, JacksonJP, LickteigAJ, AugustineLM, CherringtonNJ (2008) Drug metabolizing enzyme induction pathways in experimental non-alcoholic steatohepatitis. Arch Toxicol 82: 959–964.

47. Lastres-BeckerI, BrodesserS, LutjohannD, AzizovM, BuchmannJ, et al. (2008) Insulin receptor and lipid metabolism pathology in ataxin-2 knock-out mice. Hum Mol Genet 17: 1465–1481.

48. NishimuraH, KimE, NakanishiT, BabaT (2004) Possible function of the ADAM1a/ADAM2 Fertilin complex in the appearance of ADAM3 on the sperm surface. J Biol Chem 279: 34957–34962.

49. HoMS, OuC, ChanYR, ChienCT, PiH (2008) The utility F-box for protein destruction. Cell Mol Life Sci 65: 1977–2000.

50. Thangima ZannatM, BhattacharjeeRB, BagJ (2011) Depletion of cellular poly (A) binding protein prevents protein synthesis and leads to apoptosis in HeLa cells. Biochem Biophys Res Commun 408: 375–381.

51. SvitkinYV, EvdokimovaVM, BraseyA, PestovaTV, FantusD, et al. (2009) General RNA-binding proteins have a function in poly(A)-binding protein-dependent translation. Embo J 28: 58–68.

52. KedershaN, AndersonP (2007) Mammalian stress granules and processing bodies. Methods Enzymol 431: 61–81.

53. MuhlemannO (2008) Recognition of nonsense mRNA: towards a unified model. Biochem Soc Trans 36: 497–501.

54. GehmanLT, StoilovP, MaguireJ, DamianovA, LinCH, et al. (2011) The splicing regulator Rbfox1 (A2BP1) controls neuronal excitation in the mammalian brain. Nat Genet 43: 706–711.

55. UnderwoodJG, BoutzPL, DoughertyJD, StoilovP, BlackDL (2005) Homologues of the Caenorhabditis elegans Fox-1 protein are neuronal splicing regulators in mammals. Mol Cell Biol 25: 10005–10016.

56. HallenL, KleinH, StoschekC, WehrmeyerS, NonhoffU, et al. (2011) The KRAB-containing zinc-finger transcriptional regulator ZBRK1 activates SCA2 gene transcription through direct interaction with its gene product, ataxin-2. Hum Mol Genet 20: 104–114.

57. LeeT, LiYR, IngreC, WeberM, GrehlT, et al. (2011) Ataxin-2 intermediate-length polyglutamine expansions in European ALS patients. Hum Mol Genet 20: 1697–1700.

58. Liu-YesucevitzL, BassellGJ, GitlerAD, HartAC, KlannE, et al. (2011) Local RNA translation at the synapse and in disease. J Neurosci 31: 16086–16093.

59. ThomasPSJr, FraleyGS, DamianV, WoodkeLB, ZapataF, et al. (2006) Loss of endogenous androgen receptor protein accelerates motor neuron degeneration and accentuates androgen insensitivity in a mouse model of X-linked spinal and bulbar muscular atrophy. Hum Mol Genet 15: 2225–2238.

60. EverettCM, WoodNW (2004) Trinucleotide repeats and neurodegenerative disease. Brain 127: 2385–2405.

61. McCulloughSD, GrantPA (2010) Histone acetylation, acetyltransferases, and ataxia–alteration of histone acetylation and chromatin dynamics is implicated in the pathogenesis of polyglutamine-expansion disorders. Adv Protein Chem Struct Biol 79: 165–203.

62. TsutsumiT, KuwabaraH, AraiT, XiaoY, DecaprioJA (2008) Disruption of the Fbxw8 gene results in pre- and postnatal growth retardation in mice. Mol Cell Biol 28: 743–751.

63. LittermanN, IkeuchiY, GallardoG, O'ConnellBC, SowaME, et al. (2011) An OBSL1-Cul7Fbxw8 ubiquitin ligase signaling mechanism regulates Golgi morphology and dendrite patterning. PLoS Biol 9: e1001060 doi:10.1371/journal.pbio.1001060.

64. HuynhDP, NguyenDT, Pulst-KorenbergJB, BriceA, PulstSM (2007) Parkin is an E3 ubiquitin-ligase for normal and mutant ataxin-2 and prevents ataxin-2-induced cell death. Exp Neurol 203: 531–541.

65. Lagier-TourenneC, ClevelandDW (2010) Neurodegeneration: An expansion in ALS genetics. Nature 466: 1052–1053.

66. Al-RamahiI, PerezAM, LimJ, ZhangM, SorensenR, et al. (2007) dAtaxin-2 mediates expanded Ataxin-1-induced neurodegeneration in a Drosophila model of SCA1. PLoS Genet 3: e234 doi:10.1371/journal.pgen.0030234.

67. ClarkHB, BurrightEN, YunisWS, LarsonS, WilcoxC, et al. (1997) Purkinje cell expression of a mutant allele of SCA1 in transgenic mice leads to disparate effects on motor behaviors, followed by a progressive cerebellar dysfunction and histological alterations. J Neurosci 17: 7385–7395.

68. LivakKJ, SchmittgenTD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408.

69. EdgarR, DomrachevM, LashAE (2002) Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res 30: 207–210.

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#