#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Low Levels of p53 Protein and Chromatin Silencing of p53 Target Genes Repress Apoptosis in Endocycling Cells


In order to maintain genome integrity, eukaryotic cells have evolved multiple ways to respond to DNA damage stress. One of the major cellular responses is apoptosis, during which the cell undergoes programmed cell death in order to prevent the propagation of the damaged genome to daughter cells. Although clinical observations and other studies have shown that tissues can differ in their apoptotic response, the molecular mechanisms underlying these differences are largely unknown. We have shown in our model system, Drosophila, that endocycling cells do not initiate cell death in response to DNA damage. The endocycle is a cell cycle variation that is widely found in nature and conserved from plant to animals. During the endocycle, cells duplicate their genomic DNA but do not enter mitosis to segregate chromosomes, resulting in a polyploid genome content. In this study, we investigate how the apoptotic response to DNA damage is repressed in endocycling cells. We find that the Drosophila ortholog of the human p53 tumor suppressor protein is expressed at very low levels in endocycling cells. Moreover, the downstream pro-apoptotic genes that are regulated by p53 are epigenetically silenced in endocycling cells. Our results provide important insights into tissue-specific apoptotic responses in development, with possible broader impact on understanding radiation therapy response and cancer of different tissues.


Vyšlo v časopise: Low Levels of p53 Protein and Chromatin Silencing of p53 Target Genes Repress Apoptosis in Endocycling Cells. PLoS Genet 10(9): e32767. doi:10.1371/journal.pgen.1004581
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004581

Souhrn

In order to maintain genome integrity, eukaryotic cells have evolved multiple ways to respond to DNA damage stress. One of the major cellular responses is apoptosis, during which the cell undergoes programmed cell death in order to prevent the propagation of the damaged genome to daughter cells. Although clinical observations and other studies have shown that tissues can differ in their apoptotic response, the molecular mechanisms underlying these differences are largely unknown. We have shown in our model system, Drosophila, that endocycling cells do not initiate cell death in response to DNA damage. The endocycle is a cell cycle variation that is widely found in nature and conserved from plant to animals. During the endocycle, cells duplicate their genomic DNA but do not enter mitosis to segregate chromosomes, resulting in a polyploid genome content. In this study, we investigate how the apoptotic response to DNA damage is repressed in endocycling cells. We find that the Drosophila ortholog of the human p53 tumor suppressor protein is expressed at very low levels in endocycling cells. Moreover, the downstream pro-apoptotic genes that are regulated by p53 are epigenetically silenced in endocycling cells. Our results provide important insights into tissue-specific apoptotic responses in development, with possible broader impact on understanding radiation therapy response and cancer of different tissues.


Zdroje

1. WeinertTA, HartwellLH (1993) Cell cycle arrest of cdc mutants and specificity of the RAD9 checkpoint. Genetics 134: 63–80.

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

3. FuchsY, StellerH (2011) Programmed cell death in animal development and disease. Cell 147: 742–758.

4. Di MiccoR, FumagalliM, CicaleseA, PiccininS, GaspariniP, et al. (2006) Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444: 638–642.

5. HanahanD, WeinbergRA (2011) Hallmarks of cancer: the next generation. Cell 144: 646–674.

6. GorgoulisVG, VassiliouLV, KarakaidosP, ZacharatosP, KotsinasA, et al. (2005) Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature 434: 907–913.

7. JacksonJG, PostSM, LozanoG (2011) Regulation of tissue- and stimulus-specific cell fate decisions by p53 in vivo. J Pathol 223: 127–136.

8. BeckermanR, PrivesC (2010) Transcriptional regulation by p53. Cold Spring Harb Perspect Biol 2: a000935.

9. GudkovAV, KomarovaEA (2003) The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer 3: 117–129.

10. LoweSW, SchmittEM, SmithSW, OsborneBA, JacksT (1993) P53 Is Required for Radiation-Induced Apoptosis in Mouse Thymocytes. Nature 362: 847–849.

11. MacCallumDE, HuppTR, MidgleyCA, StuartD, CampbellSJ, et al. (1996) The p53 response to ionising radiation in adult and developing murine tissues. Oncogene 13: 2575–2587.

12. SongS, LambertPF (1999) Different responses of epidermal and hair follicular cells to radiation correlate with distinct patterns of p53 and p21 induction. Am J Pathol 155: 1121–1127.

13. KomarovaEA, ChernovMV, FranksR, WangK, ArminG, et al. (1997) Transgenic mice with p53-responsive lacZ: p53 activity varies dramatically during normal development and determines radiation and drug sensitivity in vivo. EMBO J 16: 1391–1400.

14. RileyT, SontagE, ChenP, LevineA (2008) Transcriptional control of human p53-regulated genes. Nat Rev Mol Cell Biol 9: 402–412.

15. CoatesPJ, LorimoreSA, LindsayKJ, WrightEG (2003) Tissue-specific p53 responses to ionizing radiation and their genetic modification: the key to tissue-specific tumour susceptibility? Journal of Pathology 201: 377–388.

16. BouvardV, ZaitchoukT, VacherM, DuthuA, CanivetM, et al. (2000) Tissue and cell-specific expression of the p53-target genes: bax, fas, mdm2 and waf1/p21, before and following ionising irradiation in mice. Oncogene 19: 649–660.

17. FeiP, BernhardEJ, El-DeiryWS (2002) Tissue-specific induction of p53 targets in vivo. Cancer Res 62: 7316–7327.

18. HamardPJ, BartheleryN, HogstadB, MungamuriSK, TonnessenCA, et al. (2013) The C terminus of p53 regulates gene expression by multiple mechanisms in a target- and tissue-specific manner in vivo. Genes Dev 27: 1868–1885.

19. MehrotraS, MaqboolSB, KolpakasA, MurnenK, CalviBR (2008) Endocycling cells do not apoptose in response to DNA rereplication genotoxic stress. Genes Dev 22: 3158–3171.

20. FoxDT, DuronioRJ (2013) Endoreplication and polyploidy: insights into development and disease. Development 140: 3–12.

21. CalviBR (2013) Making big cells: one size does not fit all. Proc Natl Acad Sci U S A 110: 9621–9622.

22. LeslieM (2014) Strength in numbers? Science 343: 725–727.

23. DavoliT, de LangeT (2011) The causes and consequences of polyploidy in normal development and cancer. Annu Rev Cell Dev Biol 27: 585–610.

24. StorchovaZ, PellmanD (2004) From polyploidy to aneuploidy, genome instability and cancer. Nat Rev Mol Cell Biol 5: 45–54.

25. HasselC, ZhangB, DixonM, CalviBR (2014) Induction of endocycles represses apoptosis independently of differentiation and predisposes cells to genome instability. Development 141: 112–123.

26. PetersM, DeLucaC, HiraoA, StambolicV, PotterJ, et al. (2002) Chk2 regulates irradiation-induced, p53-mediated apoptosis in Drosophila. Proc Natl Acad Sci U S A 99: 11305–11310.

27. SongYH, MireyG, BetsonM, HaberDA, SettlemanJ (2004) The Drosophila ATM ortholog, dATM, mediates the response to ionizing radiation and to spontaneous DNA damage during development. Curr Biol 14: 1354–1359.

28. XuJ, XinS, DuW (2001) Drosophila Chk2 is required for DNA damage-mediated cell cycle arrest and apoptosis. Febs Letters 508: 394–398.

29. BrodskyMH, NordstromW, TsangG, KwanE, RubinGM, et al. (2000) Drosophila p53 binds a damage response element at the reaper locus. Cell 101: 103–113.

30. OllmannM, YoungLM, Di ComoCJ, KarimF, BelvinM, et al. (2000) Drosophila p53 is a structural and functional homolog of the tumor suppressor p53. Cell 101: 91–101.

31. JinS, MartinekS, JooWS, WortmanJR, MirkovicN, et al. (2000) Identification and characterization of a p53 homologue in Drosophila melanogaster. Proc Natl Acad Sci U S A 97: 7301–7306.

32. AbramsJM, WhiteK, FesslerLI, StellerH (1993) Programmed cell death during Drosophila embryogenesis. Development 117: 29–43.

33. ChristichA, KauppilaS, ChenP, SogameN, HoSI, et al. (2002) The damage-responsive Drosophila gene sickle encodes a novel IAP binding protein similar to but distinct from reaper, grim, and hid. Curr Biol 12: 137–140.

34. GretherME, AbramsJM, AgapiteJ, WhiteK, StellerH (1995) The head involution defective gene of Drosophila melanogaster functions in programmed cell death. Genes Dev 9: 1694–1708.

35. SrinivasulaSM, DattaP, KobayashiM, WuJW, FujiokaM, et al. (2002) sickle, a novel Drosophila death gene in the reaper/hid/grim region, encodes an IAP-inhibitory protein. Curr Biol 12: 125–130.

36. WingJP, KarresJS, OgdahlJL, ZhouL, SchwartzLM, et al. (2002) Drosophila sickle is a novel grim-reaper cell death activator. Curr Biol 12: 131–135.

37. WhiteK, GretherME, AbramsJM, YoungL, FarrellK, et al. (1994) Genetic control of programmed cell death in Drosophila. Science 264: 677–683.

38. AkdemirF, ChristichA, SogameN, ChapoJ, AbramsJM (2007) p53 directs focused genomic responses in Drosophila. Oncogene 26: 5184–5193.

39. MaqboolSB, MehrotraS, KolpakasA, DurdenC, ZhangB, et al. (2010) Dampened activity of E2F1-DP and Myb-MuvB transcription factors in Drosophila endocycling cells. J Cell Sci 123: 4095–4106.

40. McNameeLM, BrodskyMH (2009) p53-independent apoptosis limits DNA damage-induced aneuploidy. Genetics 182: 423–435.

41. WichmannA, UyetakeL, SuTT (2010) E2F1 and E2F2 have opposite effects on radiation-induced p53-independent apoptosis in Drosophila. Dev Biol 346: 80–89.

42. MoonNS, Di StefanoL, MorrisEJ, PatelR, WhiteK, et al. (2008) E2F and p53 induce apoptosis independently during Drosophila development but intersect in the context of DNA damage. PLoS Genet 4: e1000153.

43. van BergeijkP, HeimillerJ, UyetakeL, SuTT (2012) Genome-wide expression analysis identifies a modulator of ionizing radiation-induced p53-independent apoptosis in Drosophila melanogaster. PLoS One 7: e36539.

44. Tanaka-MatakatsuM, XuJ, ChengL, DuW (2009) Regulation of apoptosis of rbf mutant cells during Drosophila development. Dev Biol 326: 347–356.

45. SherN, BellGW, LiS, NordmanJ, EngT, et al. (2012) Developmental control of gene copy number by repression of replication initiation and fork progression. Genome Res 22: 64–75.

46. HendersonKD, AndrewDJ (2000) Regulation and function of Scr, exd, and hth in the Drosophila salivary gland. Dev Biol 217: 362–374.

47. TschierschB, HofmannA, KraussV, DornR, KorgeG, et al. (1994) The protein encoded by the Drosophila position-effect variegation suppressor gene Su(var)3-9 combines domains of antagonistic regulators of homeotic gene complexes. EMBO J 13: 3822–3831.

48. JenuweinT (2001) Re-SET-ting heterochromatin by histone methyltransferases. Trends Cell Biol 11: 266–273.

49. StabellM, EskelandR, BjorkmoM, LarssonJ, AalenRB, et al. (2006) The Drosophila G9a gene encodes a multi-catalytic histone methyltransferase required for normal development. Nucleic Acids Res 34: 4609–4621.

50. MisJ, NerSS, GrigliattiTA (2006) Identification of three histone methyltransferases in Drosophila: dG9a is a suppressor of PEV and is required for gene silencing. Mol Genet Genomics 275: 513–526.

51. StankunasK, BergerJ, RuseC, SinclairDA, RandazzoF, et al. (1998) The enhancer of polycomb gene of Drosophila encodes a chromatin protein conserved in yeast and mammals. Development 125: 4055–4066.

52. SotoMC, ChouTB, BenderW (1995) Comparison of germline mosaics of genes in the Polycomb group of Drosophila melanogaster. Genetics 140: 231–243.

53. SinclairDAR, CleggNJ, AntonchukJ, MilneTA, StankunasK, et al. (1998) Enhancer of Polycomb is a suppressor of position-effect variegation in Drosophila melanogaster. Genetics 148: 211–220.

54. PengJC, KarpenGH (2009) Heterochromatic genome stability requires regulators of histone H3 K9 methylation. PLoS Genet 5: e1000435.

55. MarygoldSJ, LeylandPC, SealRL, GoodmanJL, ThurmondJ, et al. (2013) FlyBase: improvements to the bibliography. Nucleic Acids Res 41: D751–757.

56. BourdonJC, FernandesK, Murray-ZmijewskiF, LiuG, DiotA, et al. (2005) p53 isoforms can regulate p53 transcriptional activity. Genes & Development 19: 2122–2137.

57. Dichtel-DanjoyML, MaD, DourlenP, ChatelainG, NapoletanoF, et al. (2013) Drosophila p53 isoforms differentially regulate apoptosis and apoptosis-induced proliferation. Cell Death Differ 20: 108–116.

58. FishMP, GrothAC, CalosMP, NusseR (2007) Creating transgenic Drosophila by microinjecting the site-specific phiC31 integrase mRNA and a transgene-containing donor plasmid. Nat Protoc 2: 2325–2331.

59. BrodskyMH, WeinertBT, TsangG, RongYS, McGinnisNM, et al. (2004) Drosophila melanogaster MNK/Chk2 and p53 regulate multiple DNA repair and apoptotic pathways following DNA damage. Mol Cell Biol 24: 1219–1231.

60. VenkenKJ, CarlsonJW, SchulzeKL, PanH, HeY, et al. (2009) Versatile P[acman] BAC libraries for transgenesis studies in Drosophila melanogaster. Nat Methods 6: 431–434.

61. LiGY, FanB, SuGF (2009) Acute energy reduction induces caspase-dependent apoptosis and activates p53 in retinal ganglion cells (RGC-5). Exp Eye Res 89: 581–589.

62. BeloteJM, FortierE (2002) Targeted expression of dominant negative proteasome mutants in Drosophila melanogaster. Genesis 34: 80–82.

63. KimH, LeeJM, LeeG, BhinJ, OhSK, et al. (2011) DNA damage-induced RORalpha is crucial for p53 stabilization and increased apoptosis. Mol Cell 44: 797–810.

64. NordstromW, AbramsJM (2000) Guardian ancestry: fly p53 and damage-inducible apoptosis. Cell Death Differ 7: 1035–1038.

65. ChenS, WeiHM, LvWW, WangDL, SunFL (2011) E2 ligase dRad6 regulates DMP53 turnover in Drosophila. J Biol Chem 286: 9020–9030.

66. BradyCA, JiangD, MelloSS, JohnsonTM, JarvisLA, et al. (2011) Distinct p53 transcriptional programs dictate acute DNA-damage responses and tumor suppression. Cell 145: 571–583.

67. LaneDP, VermaC (2012) Mdm2 in evolution. Genes Cancer 3: 320–324.

68. PerryME (2010) The regulation of the p53-mediated stress response by MDM2 and MDM4. Cold Spring Harb Perspect Biol 2: a000968.

69. FakharzadehSS, TruskoSP, GeorgeDL (1991) Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J 10: 1565–1569.

70. MomandJ, ZambettiGP, OlsonDC, GeorgeD, LevineAJ (1992) The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 69: 1237–1245.

71. AlltonK, JainAK, HerzHM, TsaiWW, JungSY, et al. (2009) Trim24 targets endogenous p53 for degradation. Proc Natl Acad Sci U S A 106: 11612–11616.

72. RossAJ, LiM, YuB, GaoMX, DerryWB (2011) The EEL-1 ubiquitin ligase promotes DNA damage-induced germ cell apoptosis in C. elegans. Cell Death Differ 18: 1140–1149.

73. WylieA, LuWJ, D'BrotA, BuszczakM, AbramsJM (2014) p53 activity is selectively licensed in the Drosophila stem cell compartment. Elife 3: e01530.

74. AndreyenkovaNG, KokozaEB, SemeshinVF, BelyaevaES, DemakovSA, et al. (2009) Localization and characteristics of DNA underreplication zone in the 75C region of intercalary heterochromatin in Drosophila melanogaster polytene chromosomes. Chromosoma 118: 747–761.

75. PainterTS (1935) The Morphology of the Third Chromosome in the Salivary Gland of Drosophila Melanogaster and a New Cytological Map of This Element. Genetics 20: 301–326.

76. ZhangY, LinN, CarrollPM, ChanG, GuanB, et al. (2008) Epigenetic blocking of an enhancer region controls irradiation-induced proapoptotic gene expression in Drosophila embryos. Dev Cell 14: 481–493.

77. NordmanJ, LiS, EngT, MacalpineD, Orr-WeaverTL (2011) Developmental control of the DNA replication and transcription programs. Genome Res 21: 175–181.

78. NordmanJ, Orr-WeaverTL (2012) Regulation of DNA replication during development. Development 139: 455–464.

79. BelyaevaES, ZhimulevIF, VolkovaEI, AlekseyenkoAA, MoshkinYM, et al. (1998) Su(UR)ES: a gene suppressing DNA underreplication in intercalary and pericentric heterochromatin of Drosophila melanogaster polytene chromosomes. Proc Natl Acad Sci U S A 95: 7532–7537.

80. BaehreckeEH (2005) Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol 6: 505–510.

81. LeeCY, BaehreckeEH (2001) Steroid regulation of autophagic programmed cell death during development. Development 128: 1443–1455.

82. McPheeCK, LoganMA, FreemanMR, BaehreckeEH (2010) Activation of autophagy during cell death requires the engulfment receptor Draper. Nature 465: 1093–1096.

83. HongA, Narbonne-ReveauK, Riesgo-EscovarJ, FuH, AladjemMI, et al. (2007) The cyclin-dependent kinase inhibitor Dacapo promotes replication licensing during Drosophila endocycles. EMBO J 26: 2071–2082.

84. LeachTJ, ChotkowskiHL, WotringMG, DilwithRL, GlaserRL (2000) Replication of heterochromatin and structure of polytene chromosomes. Mol Cell Biol 20: 6308–6316.

85. LaghaM, BothmaJP, LevineM (2012) Mechanisms of transcriptional precision in animal development. Trends Genet 28: 409–416.

86. EspinosaJM, VerdunRE, EmersonBM (2003) p53 functions through stress- and promoter-specific recruitment of transcription initiation components before and after DNA damage. Mol Cell 12: 1015–1027.

87. ChenHZ, OusephMM, LiJ, PecotT, ChokshiV, et al. (2012) Canonical and atypical E2Fs regulate the mammalian endocycle. Nat Cell Biol 14: 1192–1202.

88. PanditSK, WestendorpB, NantasantiS, van LiereE, TootenPC, et al. (2012) E2F8 is essential for polyploidization in mammalian cells. Nat Cell Biol 14: 1181–1191.

89. SherN, Von StetinaJR, BellGW, MatsuuraS, RavidK, et al. (2013) Fundamental differences in endoreplication in mammals and Drosophila revealed by analysis of endocycling and endomitotic cells. Proc Natl Acad Sci U S A 110: 9368–9373.

90. ZielkeN, KimKJ, TranV, ShibutaniST, BravoMJ, et al. (2011) Control of Drosophila endocycles by E2F and CRL4(CDT2). Nature 480: 123–127.

91. MeserveJH, DuronioRJ (2012) Atypical E2Fs drive atypical cell cycles. Nat Cell Biol 14: 1124–1125.

92. UllahZ, KohnMJ, YagiR, VassilevLT, DepamphilisML (2008) Differentiation of trophoblast stem cells into giant cells is triggered by p57/Kip2 inhibition of CDK1 activity. Genes Dev 22: 3024–3036.

93. SolovevaV, LinzerDI (2004) Differentiation of placental trophoblast giant cells requires downregulation of p53 and Rb. Placenta 25: 29–36.

94. ZhengL, DaiH, ZhouM, LiX, LiuC, et al. (2012) Polyploid cells rewire DNA damage response networks to overcome replication stress-induced barriers for tumour progression. Nat Commun 3: 815.

95. VenkenKJ, HeY, HoskinsRA, BellenHJ (2006) P[acman]: a BAC transgenic platform for targeted insertion of large DNA fragments in D. melanogaster. Science 314: 1747–1751.

96. Harlow E, Lane D (1999) Using Antibodies: A Laboratory Manual. Cold Spring Harbor: Cold Spring Harbor Laboratory Press.

97. RothbauerU, ZolghadrK, MuyldermansS, SchepersA, CardosoMC, et al. (2008) A versatile nanotrap for biochemical and functional studies with fluorescent fusion proteins. Mol Cell Proteomics 7: 282–289.

98. SchwedG, MayN, PecherskyY, CalviBR (2002) Drosophila minichromosome maintenance 6 is required for chorion gene amplification and genomic replication. Mol Biol Cell 13: 607–620.

99. KhouryMP, BourdonJC (2010) The isoforms of the p53 protein. Cold Spring Harb Perspect Biol 2: a000927.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 9
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#