#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Inactivation of PNKP by Mutant ATXN3 Triggers Apoptosis by Activating the DNA Damage-Response Pathway in SCA3


Spinocerebellar ataxia type 3 (SCA3) is an untreatable neurodegenerative disease, and the most common dominantly inherited ataxia worldwide. SCA3 is caused by expansion of a CAG tri-nucleotide repeat sequence in the ATXN3 gene’s coding region. The expanded CAG sequences encode a run of the amino acid glutamine; the mutant ATXN3 interacts with multiple proteins in vivo to create insoluble aggregates in SCA3 brains. It is thought that the loss of function of the aggregated proteins contributes to cellular toxicity and neurodegeneration in SCA3. Despite significant progress in understanding SCA3’s etiology, the molecular mechanism by which the mutant protein triggers the death of neurons in SCA3 brains remains unknown. We now report that the mutant ATXN3 protein interacts with and inactivates PNKP (polynucleotide kinase 3’-phosphatase), an essential DNA strand break repair enzyme. This inactivation results in persistent accumulation of DNA damage, and chronic activation of the DNA damage-response ATM signaling pathway in SCA3. Our work suggests that persistent DNA damage/strand breaks and chronic activation of ATM trigger neuronal death in SCA3. Discovery of the mechanism by which mutant ATXN3 induces DNA damage and amplifies the pro-death pathways provides a molecular basis for neurodegeneration in SCA3, and perhaps ultimately for its treatment.


Vyšlo v časopise: Inactivation of PNKP by Mutant ATXN3 Triggers Apoptosis by Activating the DNA Damage-Response Pathway in SCA3. PLoS Genet 11(1): e32767. doi:10.1371/journal.pgen.1004834
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004834

Souhrn

Spinocerebellar ataxia type 3 (SCA3) is an untreatable neurodegenerative disease, and the most common dominantly inherited ataxia worldwide. SCA3 is caused by expansion of a CAG tri-nucleotide repeat sequence in the ATXN3 gene’s coding region. The expanded CAG sequences encode a run of the amino acid glutamine; the mutant ATXN3 interacts with multiple proteins in vivo to create insoluble aggregates in SCA3 brains. It is thought that the loss of function of the aggregated proteins contributes to cellular toxicity and neurodegeneration in SCA3. Despite significant progress in understanding SCA3’s etiology, the molecular mechanism by which the mutant protein triggers the death of neurons in SCA3 brains remains unknown. We now report that the mutant ATXN3 protein interacts with and inactivates PNKP (polynucleotide kinase 3’-phosphatase), an essential DNA strand break repair enzyme. This inactivation results in persistent accumulation of DNA damage, and chronic activation of the DNA damage-response ATM signaling pathway in SCA3. Our work suggests that persistent DNA damage/strand breaks and chronic activation of ATM trigger neuronal death in SCA3. Discovery of the mechanism by which mutant ATXN3 induces DNA damage and amplifies the pro-death pathways provides a molecular basis for neurodegeneration in SCA3, and perhaps ultimately for its treatment.


Zdroje

1. Kawaguchi Y, Okamoto T, Taniwaki M, Aizawa M, Inoue M et al, (1994) CAG expansion in a novel gene for Machado-Joseph disease at chromosome 14q32.1, Nat Genet, 8, 221–228. doi: 10.1038/ng1194-221 7874163

2. Zoghbi HY, Orr HT (2000) Glutamine repeats and neurodegeneration. Ann Rev Neurosci, 23, 213–247. doi: 10.1146/annurev.neuro.23.1.217

3. Kobayashi T, Kakizuka A (2003) Molecular analysis of Machado-Joseph disease, Cytogenetic and Genome Res, 100, 261–275. doi: 10.1159/000072862

4. Rosenberg RN (1992) Machado-Joseph disease: an autosomal dominant system degeneration. Mov Disord, 3, 193–203. doi: 10.1002/mds.870070302

5. Takiyama Y, Oyanagi S, Kawashima S, Sakamoto H, Saito K et al, (1994) A clinical and pathologic study of a large Japanese family with Machado-Joseph disease tightly linked to the DNA markers on chromosome 14q, Neurology, 44, 1302–1308. doi: 10.1212/WNL.44.7.1302 8035935

6. Durr A, Stevanin G, Cancel G, Duyckaerts C, Abbas N et al, (1996) Spinocerebellar ataxia 3 and Machado-Joseph disease: clinical, molecular and neuropathological features, Ann Neurol, 39, 490–499. doi: 10.1002/ana.410390411 8619527

7. Klockgether T, Skalej M, Wedekind D, Luft AR, Welte D et al, (1998) Autosomal dominant cerebellar ataxia type 1: MRI-based volumetry of posterior fossa structures and basal ganglia in spinocerebellar ataxia type 1, 2 and 3, Brain, 121, 1687–1693. doi: 10.1093/brain/121.9.1687 9762957

8. Di Prospero NA, Fishbeck KH (2005) Therapeutic development for triplet repeat expansion diseases, Nat Rev Genet, 6, 756–765. doi: 10.1038/nrg1690 16205715

9. Costa MD and Paulson HL (2012) Toward understanding Machado-Joseph disease, Progress in Neurobiol, 97, 239–257. doi: 10.1016/j.pneurobio.2011.11.006

10. Winborn BJ, Travis SM, Todi SV, Scaglione KM, Xu P et al, (2008) The deubiquitinating enzyme ataxn-3, a polyglutamine disease protein, edits Lys63 linkages in mixed linkage ubiquitin chains, J Biol Chem, 283, 26436–43. doi: 10.1074/jbc.M803692200 18599482

11. Todi SV, Scaglione KM, Blount JR, Basrur V, Conlon KP et al, (2010) Activity and cellular functions of the deubiquitinating enzyme and polyglutamine disease peotein ataxin-3 are regulated by ubiquitination at lysine 117, J Biol Chem, 285, 39303–39313. doi: 10.1074/jbc.M110.181610 20943656

12. Matos CA, de Macedo-Ribeiro S, Carvalho AL (2011) Polyglutamine diseases: the special case of ataxin-3 and Machado Joseph disease, Prog Neurobiol, 95, 26–48. doi: 10.1016/j.pneurobio.2011.06.007 21740957

13. Schmitt I, Linden M, Khazneh H, Evert BO, Breuer P et al, (2007) Inactivation of mouse Atxn3 (ataxin-3) gene increases protein ubiquitination, Biochem Biophy Res Commun, 362, 734–739. doi: 10.1016/j.bbrc.2007.08.062

14. Rodriguez-Lebron E, doCamro Costa M, Luna-Cancalon K, Peron TM, Fischer S et al, (2013): Silencing mutant ATXN3 expression resolves molecular phenotypes in SCA3 transgenic mice, Mol Therapy, 21, 1909–1918. doi: 10.1038/mt.2013.152

15. Chou A-H, Yeh T-H, Kuo Y-L, Kao Y-C, Jou M-J et al, (2006) Polyglutamine-expanded ataxin-3 activates mitochondrial apoptotic pathway by upregulating BAX and downregulating Bcl-XL, Neurobiol Dis, 21, 333–345. doi: 10.1016/j.nbd.2005.07.011 16112867

16. Chou A-H, Lin A-C, Hong K-Y, Hu S-H, Chen Y-L et al, (2011) p53 activation mediates polyglutamine-expanded ataxin-3 upregulation of Bax expression in cerebellar and pontine nuclei neurons, Neurochem Int, 58, 145–152. doi: 10.1016/j.neuint.2010.11.005 21092747

17. Weinfeld M, Mani RS, Abdou I, Aceytuno RD, Glover JN (2011) Tidying up loose ends: the role of polynucleotide kinase/phosphatase in DNA strand break repair, Trends Biochem Sci, 36, 262–71. doi: 10.1016/j.tibs.2011.01.006 21353781

18. Mandal SM, Hegde ML, Chatterjee A, Hedge PM, Szczesny B et al, (2012) Role of human DNA glycosylase Nei-like 2 (NEIL2) and single strand break repair protein polynucleotide kinase 3’-phosphatase in maintenance of mitochondrial genome, J Biol Chem, 287, 2819–2829. doi: 10.1074/jbc.M111.272179 22130663

19. Shyu YJ, Liu H, Deng X, Hu CD (2006) Identification of new fluorescent protein fragments for bimolecular fluorescence complementation analysis under physiological conditions, Biotechniques, 40, 61–66. doi: 10.2144/000112036 16454041

20. Weibrecht I, Leuchowius KJ, Clausson CM, Conze T, Jarvius M et al, (2010) Proximity ligation assay: a recent addition to the proteomics toolbox, Expert Rev Proteomics, 7, 401–409. doi: 10.1586/epr.10.10 20536310

21. Silva-Fernandes A, Duarte-Silva S, Neves-Carvalho A, Amorim M, Soares-Cunha C et al, (2014) Chronic treatment with 17-DMAG improves balance and co-ordination in a new mouse model of Machado-Joseph disease, Neurotherapeutics, 11, 433–49. doi: 10.1007/s13311-013-0255-9 24477711

22. Pacheco LS, da Silveira AF, Trott A, Houenou LJ, Algarve TD et al, (2013) Association between Machado-Joseph disease and oxidative stress biomarkers, Mutat research, 757, 99–103. doi: 10.1016/j.mrgentox.2013.06.023

23. Kurz EU, Lees-Miller SP (2004) DNA damage-induced activation of ATM and ATM-dependent signaling pathways, DNA Repair, 3, 889–900. doi: 10.1016/j.dnarep.2004.03.029 15279774

24. Guo Z, Kozlov S, Lavin MF, Person MD, Paull TT (2010): ATM activation by oxidative stress, Science, 330, 517–521. doi: 10.1126/science.1192912 20966255

25. Miller FD, Pozniak CD, Walsh GS (2000) Neuronal life and death: an essential role for the p53 family, Cell Death Differ, 7, 880–888. doi: 10.1038/sj.cdd.4400736 11279533

26. Culmsee C, Mattson MP (2005) p53 in neuronal apoptosis, Biochem Biophys Res Commun, 331, 761–777. doi: 10.1016/j.bbrc.2005.03.149 15865932

27. Chang JR, Ghafouri M, Mukerjee R, Bagashev A, Chabrashvili T et al, (2011) Role of p53 in neurodegenerative diseases, Neurodegener Dis, 9, 68–80. doi: 10.1159/000329999 22042001

28. Bae BJ, Xu H, Igarashi S, Fujomoto M, Agrawal N et al, (2005) p53 mediates cellular dysfunction and behavioral abnormalities in Huntington disease, Neuron, 47, 29–41. doi: 10.1016/j.neuron.2005.06.005 15996546

29. Wang HL, Chou AH, Lin AC, Chen SY, Weng YH et al, (2010) Polyglutamine-expanded ataxin-7 upregulates Bax expression by activating p53 in cerebellar and inferior olivery neurons. Exp Neurol, 224, 486–494. doi: 10.1016/j.expneurol.2010.05.011 20546728

30. Kharbanda S, Pandey P, Jin S, Inoue S, Bharti A et al, (1997) Functional interaction between DNA-PK and c-Abl in response to DNA damage, Nature, 386, 732–735. doi: 10.1038/386732a0 9109492

31. Baskaran R, Wood LD, Whitaker LL, Canman CE, Morgan SE et al, (1997) Ataxia telangiectasia mutant protein activates c-Abl tyrosine kinase in response to ionizing radiation, Nature, 387, 516–519. doi: 10.1038/387516a0 9168116

32. Shafman T, Khanna KK, Kedar P, Spring K, Kozlov S et al, (1997) Interaction between ATM protein and c-Abl in response to DNA damage, Nature, 387, 520–523. doi: 10.1038/387520a0 9168117

33. Yuan ZM, Utsugisawa T, Ishiko T, Nakada S, Huang Y et al, (1998) Activation of protein kinase c-δ by the c-Abl tyrosine kinase in response to ionizing radiation, Oncogene, 16, 1643–1648. doi: 10.1038/sj.onc.1201698 9582011

34. Adwan TS, Ohm AM, Jones DNM, Humphries MJ, Reyland ME (2011) Regulated binding of Importin-α to protein kinase-Cδ in response to apoptotic signaling facilitates nuclear import, J Biol Chem, 286, 35716–35724. doi: 10.1074/jbc.M111.255950 21865164

35. Basu A, Woolard MD, Johnson CL, (2001) Involvement of protein kinase C-δ in DNA damage-induced apoptosis, Cell Death Differ, 8, 899–908. doi: 10.1038/sj.cdd.4400885 11526445

36. Yoshida K (2008): Nuclear trafficking of pro-apoptotic kinases in response to DNA damage, Trends Mol Med, 14, 305–313. doi: 10.1016/j.molmed.2008.05.003 18539531

37. Fishel ML, Vasko MR, Kelly MR (2007) DNA repair in the neurons: so if they don’t divide what’s to repair, Mutat Res, 614, 24–36. doi: 10.1016/j.mrfmmm.2006.06.007 16879837

38. Weissman L, de souza-Pinto NC, Stevnsner T, Bohr VA (2007) DNA repair, mitochondria, and neurodegeneration, Neuroscience, 145, 1318–1329. doi: 10.1016/j.neuroscience.2006.08.061 17092652

39. LeDoux SP, Druzhyna NM, Hollensworth SB, Harrison JF, Wilson GL (2007) Mitochondrial DNA repair: a critical player in the response of cells of the CNS to genotoxic insults, Neuroscience, 145, 1249–1259. doi: 10.1016/j.neuroscience.2006.10.002 17097236

40. Halliwell B (2006) Oxidative stress and neurodegeneration: where are we now? J Neurochem, 97, 1634–1658. doi: 10.1111/j.1471-4159.2006.03907.x 16805774

41. Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K et al, (2007) Mechanism of cell death in oxidative stress, Antioxid Redox Signal, 9, 49–89. doi: 10.1089/ars.2007.9.49 17115887

42. Wilson DM and McNeil DR 3rd, (2007) Base excision repair and the central nervous system, Neuroscience, 145, 1187–1200. doi: 10.1016/j.neuroscience.2006.07.011 16934943

43. Lee Y, McKinnon PJ (2007) Responding to DNA double strand breaks in the nervous system, Neuroscience, 145, 1365–1374. doi: 10.1016/j.neuroscience.2006.07.026 16934412

44. Date H, Onodera O, Tanaka H, Iwabuchi K, Uekawa K et al, (2001) Early-onset ataxia with ocular motor apraxia and hypoalbuminemia is caused by mutation in a new HIT superfamily gene, Nat Genet, 29, 184–8. doi: 10.1038/ng1001-184 11586299

45. Moreira MC, Barbot C, Tachi N, Kozuka N, Uchida E, et al, (2001) The gene mutated in ataxia-ocular apraxia 1 encodes the new HIT/Zn-finger protein aprataxin, Nat Genet, 29, 189–93. doi: 10.1038/ng1001-189 11586300

46. Takashima H, Boerkoel CF, John J, Saifi GM, Salih MA et al, (2002) Mutation of TDP1, encoding a topoisomerase I-dependent DNA damage repair enzyme, in spinocerebellar ataxia with axonal neuropathy, Nat Genet, 32, 267–72. doi: 10.1038/ng987 12244316

47. Wang W-Y, Pan L, Su SC, Quinn EJ, Sasaki M et al, (2013) Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons, Nat Neurosci, 16, 1383–1391. doi: 10.1038/nn.3514 24036913

48. Shen J, Gilmore EC, Marshall CA, Haddadin M, Reynolds JJ et al, (2010) Mutation in PNKP cause microcepgaly, seizures and defects in DNA repair, Nat Genet, 42, 245–9. doi: 10.1038/ng.526 20118933

49. Poulton C, Oegema R, Heijsman D, Hoogeboom J, Schot R et al, (2013) Progressive cerebellar atrophy and polyneuropathy: expanding the spectrum of PNKP mutations, Neurogenetics, 14, 43–51. doi: 10.1007/s10048-012-0351-8 23224214

50. Reynolds JJ, Walker AK, Gilmore EC, Walsh CA, Caldecott KW (2012) Impact of PNKP mutations associated with microcephaly, seizures and developmental delay on enzyme activity and DNA strand break repair, Nucleic Acids Res, 40, 6608–19. doi: 10.1093/nar/gks318 22508754

51. Liu W, Jiang F, Bi X, Zhang YQ (2012) Drosophila FMRP participates in the DNA damage response by regulating G2/M cell cycle checkpoint and apoptosis, Hum Mol Genet, 21, 4655–4668. doi: 10.1093/hmg/dds307 22843500

52. Alpatov R, Lesch BJ, Nakamoto-Kinoshita M, Blanco A, Chen S, Stutzer A et al, (2014) A chromatin-dependent role of the Fragile X mental retardation protein FMRP in the DNA damage response, Cell, 157, 869–881. doi: 10.1016/j.cell.2014.03.040 24813610

53. Zhang W, Cheng Y, Li Y, Chen Z, Jin P, Chen D (2014) A feed-forward mechanism involving Drosophila fragile X mental retardation protein triggers a replication stress-induced DNA damage response, Hum Mol Genet, 23, 5188–5196. doi: 10.1093/hmg/ddu241 24833720

54. Polidori MC, Mecocci P, Browne SE, Senin U, Beal MF (1999) Oxidative damage to mitochondrial DNA in Huntington disease, Neurosci Lett, 272, 53–6. doi: 10.1016/S0304-3940(99)00578-9 10507541

55. Rolig RL and McKinnon PJ (2000) Linking DNA damage and neurodegeneration, Trends Neurosci, 23, 417–24. doi: 10.1016/S0166-2236(00)01625-8 10941191

56. Martin LJ (2008) DNA damage and repair: Relevance to mechanism of neurodegeneration, J of Neuropathol Exp Neurol, 67, 377–387. doi: 10.1097/NEN.0b013e31816ff780

57. Coppede F, Migliore L (2009) DNA damage and repair in Alzheimer disease, Curr Alzheimer Res, 6, 36–47. doi: 10.2174/156720509787313970 19199873

58. Muller SK, Bender A, Laub C, Hogen T, Schlaudraff F et al, (2013) Lewy body pathology is associated with mitochondrial DNA damage in Parkinson disease, Neurobiol Aging, 34, 2231–3. doi: 10.1016/j.neurobiolaging.2013.03.016 23566333

59. Yoshida K, Miki Y (2010) The cell death machinery governed by the p53 tumor suppressor in response to DNA damage, Cancer Science, 101, 831–5. doi: 10.1111/j.1349-7006.2009.01488.x 20132225

60. Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T et al, (2000) Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis, Science, 288, 1053–8. doi: 10.1126/science.288.5468.1053 10807576

61. Fridman JS, Lowe SW (2003) Control of apoptosis by p53, Oncogene, 22, 9030–9040. doi: 10.1038/sj.onc.1207116 14663481

62. Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Newmeyer DD et al, (2004) Direct activation of BAX by p53 mediates mitochondrial membrane permeabilization and apoptosis, Science, 303, 1010–1014. doi: 10.1126/science.1092734 14963330

63. Shibue T, Takeda K, Oda E, Tanaka H, Murasawa H et al, (2003) Integral role of Noxa in p53-mediated apoptotic response, Genes Dev, 17, 2233–2238. doi: 10.1101/gad.1103603 12952892

64. Nakano K and Vousden KH (2001) PUMA, a novel proapoptotic gene, is induced by p53, Mol Cell, 7, 683–694. doi: 10.1016/S1097-2765(01)00214-3 11463392

65. Du C, Fang M, Li L, Wang X (2000) Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition, Cell, 102, 33–42. doi: 10.1016/S0092-8674(00)00008-8 10929711

66. Cregan SP, Arbour NA, Maclaurin JG, Callaghan SM, Fortin A et al, (2004) p53 activation domain 1 is essential for PUMA upregulation and p53-mediated neuronal cell death, J Neurosci, 24, 10003–10012. doi: 10.1523/JNEUROSCI.2114-04.2004 15525786

67. Chipuk JE, Green DR (2006) Dissecting p53-dependent apoptosis, Cell Death Differ, 13, 94–1002. doi: 10.1038/sj.cdd.4401908

68. Bharti A, Kraeft SK, Gounder M, Pandey P, Jin S et al, (1998) Inactivation of DNA-dependent protein kinase by protein kinase C-δ: implication for apoptosis, Mol Cell Biol, 18, 6719–6728. 9774685

69. Castel AL, Cleary JD, Pearson CE (2010) Repeat instability as the basis for human diseases and as a potential target for therapy, Nat Rev, 11, 165–170. doi: 10.1038/nrm2854

70. Mason AG, Tome S, Simard JP, Libby RT Bammler TK, Beyer RP et al (2014) Expression levels of DNA replication and repair genes predict regional somatic repeat instability in the brain but not altered by polyglutamine disease protein expression or age, Hum Mol Genet, 23, 1606–1618. doi: 10.1093/hmg/ddt551 24191263

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 1
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#