#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Stepwise Activation of the ATR Signaling Pathway upon Increasing Replication Stress Impacts Fragile Site Integrity


Breaks at common fragile sites (CFS) are a recognized source of genome instability in pre-neoplastic lesions, but how such checkpoint-proficient cells escape surveillance and continue cycling is unknown. Here we show, in lymphocytes and fibroblasts, that moderate replication stresses like those inducing breaks at CFSs trigger chromatin loading of sensors and mediators of the ATR pathway but fail to activate Chk1 or p53. Consistently, we found that cells depleted of ATR, but not of Chk1, accumulate single-stranded DNA upon Mre11-dependent resection of collapsed forks. Partial activation of the pathway under moderate stress thus takes steps against fork disassembly but tolerates S-phase progression and mitotic onset. We show that fork protection by ATR is crucial to CFS integrity, specifically in the cell type where a given site displays paucity in backup replication origins. Tolerance to mitotic entry with under-replicated CFSs therefore results in chromosome breaks, providing a pool of cells committed to further instability.


Vyšlo v časopise: Stepwise Activation of the ATR Signaling Pathway upon Increasing Replication Stress Impacts Fragile Site Integrity. PLoS Genet 9(7): e32767. doi:10.1371/journal.pgen.1003643
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003643

Souhrn

Breaks at common fragile sites (CFS) are a recognized source of genome instability in pre-neoplastic lesions, but how such checkpoint-proficient cells escape surveillance and continue cycling is unknown. Here we show, in lymphocytes and fibroblasts, that moderate replication stresses like those inducing breaks at CFSs trigger chromatin loading of sensors and mediators of the ATR pathway but fail to activate Chk1 or p53. Consistently, we found that cells depleted of ATR, but not of Chk1, accumulate single-stranded DNA upon Mre11-dependent resection of collapsed forks. Partial activation of the pathway under moderate stress thus takes steps against fork disassembly but tolerates S-phase progression and mitotic onset. We show that fork protection by ATR is crucial to CFS integrity, specifically in the cell type where a given site displays paucity in backup replication origins. Tolerance to mitotic entry with under-replicated CFSs therefore results in chromosome breaks, providing a pool of cells committed to further instability.


Zdroje

1. DurkinSG, GloverTW (2007) Chromosome fragile sites. Annu Rev Genet 41: 169–192.

2. LetessierA, MillotGA, KoundrioukoffS, LachagesAM, VogtN, et al. (2011) Cell-type-specific replication initiation programs set fragility of the FRA3B fragile site. Nature 470: 120–123.

3. Le TallecB, DutrillauxB, LachagesAM, MillotGA, BrisonO, et al. (2011) Molecular profiling of common fragile sites in human fibroblasts. Nat Struct Mol Biol 18: 1421–1423.

4. DebatisseM, Le TallecB, LetessierA, DutrillauxB, BrisonO (2012) Common fragile sites: mechanisms of instability revisited. Trends Genet 28: 22–32.

5. NegriniS, GorgoulisVG, HalazonetisTD (2010) Genomic instability–an evolving hallmark of cancer. Nat Rev Mol Cell Biol 11: 220–228.

6. JacomeA, Fernandez-CapetilloO (2011) Lac operator repeats generate a traceable fragile site in mammalian cells. EMBO Rep 12: 1032–1038.

7. BesterAC, RonigerM, OrenYS, ImMM, SarniD, et al. (2011) Nucleotide deficiency promotes genomic instability in early stages of cancer development. Cell 145: 435–446.

8. BranzeiD, FoianiM (2010) Maintaining genome stability at the replication fork. Nat Rev Mol Cell Biol 11: 208–219.

9. MatsuokaS, BallifBA, SmogorzewskaA, McDonaldER3rd, HurovKE, et al. (2007) ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science 316: 1160–1166.

10. MenendezD, IngaA, ResnickMA (2009) The expanding universe of p53 targets. Nat Rev Cancer 9: 724–737.

11. JacksonSP, BartekJ (2009) The DNA-damage response in human biology and disease. Nature 461: 1071–1078.

12. CicciaA, ElledgeSJ (2010) The DNA damage response: making it safe to play with knives. Mol Cell 40: 179–204.

13. CasperAM, DurkinSG, ArltMF, GloverTW (2004) Chromosomal instability at common fragile sites in Seckel syndrome. Am J Hum Genet 75: 654–660.

14. MurgaM, BuntingS, MontanaMF, SoriaR, MuleroF, et al. (2009) A mouse model of ATR-Seckel shows embryonic replicative stress and accelerated aging. Nat Genet 41: 891–898.

15. MedhurstAL, WarmerdamDO, AkermanI, VerwayenEH, KanaarR, et al. (2008) ATR and Rad17 collaborate in modulating Rad9 localisation at sites of DNA damage. J Cell Sci 121: 3933–3940.

16. FreireR, van VugtMA, MamelyI, MedemaRH (2006) Claspin: timing the cell cycle arrest when the genome is damaged. Cell Cycle 5: 2831–2834.

17. ShimuraT, MartinMM, TorresMJ, GuC, PluthJM, et al. (2007) DNA-PK is involved in repairing a transient surge of DNA breaks induced by deceleration of DNA replication. J Mol Biol 367: 665–680.

18. SchwartzM, OrenYS, BesterAC, RahatA, SfezR, et al. (2009) Impaired replication stress response in cells from immunodeficiency patients carrying Cernunnos/XLF mutations. PLoS ONE 4: e4516.

19. DurkinSG, ArltMF, HowlettNG, GloverTW (2006) Depletion of CHK1, but not CHK2, induces chromosomal instability and breaks at common fragile sites. Oncogene 25: 4381–4388.

20. PetermannE, Maya-MendozaA, ZachosG, GillespieDA, JacksonDA, et al. (2006) Chk1 requirement for high global rates of replication fork progression during normal vertebrate S phase. Mol Cell Biol 26: 3319–3326.

21. Maya-MendozaA, PetermannE, GillespieDA, CaldecottKW, JacksonDA (2007) Chk1 regulates the density of active replication origins during the vertebrate S phase. Embo J 26: 2719–2731.

22. DupreA, Boyer-ChatenetL, SattlerRM, ModiAP, LeeJH, et al. (2008) A forward chemical genetic screen reveals an inhibitor of the Mre11-Rad50-Nbs1 complex. Nat Chem Biol 4: 119–125.

23. ThompsonR, MontanoR, EastmanA (2012) The mre11 nuclease is critical for the sensitivity of cells to chk1 inhibition. PLoS One 7: e44021.

24. HansenRS, ThomasS, SandstromR, CanfieldTK, ThurmanRE, et al. (2010) Sequencing newly replicated DNA reveals widespread plasticity in human replication timing. Proc Natl Acad Sci U S A 107: 139–144.

25. McMurrayCT (2010) Mechanisms of trinucleotide repeat instability during human development. Nat Rev Genet 11: 786–799.

26. LopesJ, PiazzaA, BermejoR, KriegsmanB, ColosioA, et al. (2011) G-quadruplex-induced instability during leading-strand replication. EMBO J 30: 4033–4046.

27. GilsonE, GeliV (2007) How telomeres are replicated. Nat Rev Mol Cell Biol 8: 825–838.

28. RizzoA, SalvatiE, PorruM, D'AngeloC, StevensMF, et al. (2009) Stabilization of quadruplex DNA perturbs telomere replication leading to the activation of an ATR-dependent ATM signaling pathway. Nucleic Acids Res 37: 5353–5364.

29. SfeirA, KosiyatrakulST, HockemeyerD, MacRaeSL, KarlsederJ, et al. (2009) Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell 138: 90–103.

30. YeJ, LenainC, BauwensS, RizzoA, Saint-LegerA, et al. (2010) TRF2 and apollo cooperate with topoisomerase 2alpha to protect human telomeres from replicative damage. Cell 142: 230–242.

31. BoscoN, de LangeT (2012) A TRF1-controlled common fragile site containing interstitial telomeric sequences. Chromosoma 121: 465–474.

32. AzvolinskyA, GiresiPG, LiebJD, ZakianVA (2009) Highly transcribed RNA polymerase II genes are impediments to replication fork progression in Saccharomyces cerevisiae. Mol Cell 34: 722–734.

33. TuduriS, CrabbeL, ContiC, TourriereH, Holtgreve-GrezH, et al. (2009) Topoisomerase I suppresses genomic instability by preventing interference between replication and transcription. Nat Cell Biol 11: 1315–1324.

34. BermejoR, LaiMS, FoianiM (2012) Preventing replication stress to maintain genome stability: resolving conflicts between replication and transcription. Mol Cell 45: 710–718.

35. SabouriN, McDonaldKR, WebbCJ, CristeaIM, ZakianVA (2012) DNA replication through hard-to-replicate sites, including both highly transcribed RNA Pol II and Pol III genes, requires the S. pombe Pfh1 helicase. Genes Dev 26: 581–593.

36. ErricoA, CostanzoV (2012) Mechanisms of replication fork protection: a safeguard for genome stability. Crit Rev Biochem Mol Biol 47: 222–235.

37. BermejoR, CapraT, JossenR, ColosioA, FrattiniC, et al. (2011) The replication checkpoint protects fork stability by releasing transcribed genes from nuclear pores. Cell 146: 233–246.

38. LabibK, De PiccoliG (2011) Surviving chromosome replication: the many roles of the S-phase checkpoint pathway. Philos Trans R Soc Lond B Biol Sci 366: 3554–3561.

39. El AchkarE, Gerbault-SeureauM, MulerisM, DutrillauxB, DebatisseM (2005) Premature condensation induces breaks at the interface of early and late replicating chromosome bands bearing common fragile sites. Proc Natl Acad Sci U S A 102: 18069–18074.

40. DruscoA, PekarskyY, CostineanS, AntenucciA, ContiL, et al. (2011) Common fragile site tumor suppressor genes and corresponding mouse models of cancer. J Biomed Biotechnol 2011: 984505.

41. OkumuraH, IshiiH, PichiorriF, CroceCM, MoriM, et al. (2009) Fragile gene product, Fhit, in oxidative and replicative stress responses. Cancer Sci 100: 1145–1150.

42. SaldivariJC, MiumaS, BeneJ, HosseiniSA, ShibataH, et al. (2012) Initiation of genome Instability and preneoplastic processes through loss of Fhit expression. PLoS Genetics 8(11): e1003077.

43. CoquelleA, ToledoF, SternS, BiethA, DebatisseM (1998) A new role for hypoxia in tumor progression: induction of fragile site triggering genomic rearrangements and formation of complex DMs and HSRs. Mol Cell 2: 259–265.

44. AnglanaM, ApiouF, BensimonA, DebatisseM (2003) Dynamics of DNA replication in mammalian somatic cells: nucleotide pool modulates origin choice and interorigin spacing. Cell 114: 385–394.

45. TecherH, KoundrioukoffS, AzarD, WilhelmT, CarignonS, et al. (2013) Replication Dynamics: Biases and Robustness of DNA Fiber Analysis. J Mol Biol [epub ahead of print]. doi:10.1016/j.jmb.2013.03.040

46. GreenCM, AlmouzniG (2003) Local action of the chromatin assembly factor CAF-1 at sites of nucleotide excision repair in vivo. Embo J 22: 5163–5174.

47. ChaumeilJ, AuguiS, ChowJC, HeardE (2008) Combined immunofluorescence, RNA fluorescent in situ hybridization, and DNA fluorescent in situ hybridization to study chromatin changes, transcriptional activity, nuclear organization, and X-chromosome inactivation. Methods Mol Biol 463: 297–308.

48. MendezJ, StillmanB (2000) Chromatin association of human origin recognition complex, cdc6, and minichromosome maintenance proteins during the cell cycle: assembly of prereplication complexes in late mitosis. Mol Cell Biol 20: 8602–8612.

49. RaderschallE, GolubEI, HaafT (1999) Nuclear foci of mammalian recombination proteins are located at single-stranded DNA regions formed after DNA damage. Proc Natl Acad Sci U S A 96: 1921–1926.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 7
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#