#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Disruption of Mouse Cenpj, a Regulator of Centriole Biogenesis, Phenocopies Seckel Syndrome


Disruption of the centromere protein J gene, CENPJ (CPAP, MCPH6, SCKL4), which is a highly conserved and ubiquitiously expressed centrosomal protein, has been associated with primary microcephaly and the microcephalic primordial dwarfism disorder Seckel syndrome. The mechanism by which disruption of CENPJ causes the proportionate, primordial growth failure that is characteristic of Seckel syndrome is unknown. By generating a hypomorphic allele of Cenpj, we have developed a mouse (Cenpjtm/tm) that recapitulates many of the clinical features of Seckel syndrome, including intrauterine dwarfism, microcephaly with memory impairment, ossification defects, and ocular and skeletal abnormalities, thus providing clear confirmation that specific mutations of CENPJ can cause Seckel syndrome. Immunohistochemistry revealed increased levels of DNA damage and apoptosis throughout Cenpjtm/tm embryos and adult mice showed an elevated frequency of micronucleus induction, suggesting that Cenpj-deficiency results in genomic instability. Notably, however, genomic instability was not the result of defective ATR-dependent DNA damage signaling, as is the case for the majority of genes associated with Seckel syndrome. Instead, Cenpjtm/tm embryonic fibroblasts exhibited irregular centriole and centrosome numbers and mono- and multipolar spindles, and many were near-tetraploid with numerical and structural chromosomal abnormalities when compared to passage-matched wild-type cells. Increased cell death due to mitotic failure during embryonic development is likely to contribute to the proportionate dwarfism that is associated with CENPJ-Seckel syndrome.


Vyšlo v časopise: Disruption of Mouse Cenpj, a Regulator of Centriole Biogenesis, Phenocopies Seckel Syndrome. PLoS Genet 8(11): e32767. doi:10.1371/journal.pgen.1003022
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003022

Souhrn

Disruption of the centromere protein J gene, CENPJ (CPAP, MCPH6, SCKL4), which is a highly conserved and ubiquitiously expressed centrosomal protein, has been associated with primary microcephaly and the microcephalic primordial dwarfism disorder Seckel syndrome. The mechanism by which disruption of CENPJ causes the proportionate, primordial growth failure that is characteristic of Seckel syndrome is unknown. By generating a hypomorphic allele of Cenpj, we have developed a mouse (Cenpjtm/tm) that recapitulates many of the clinical features of Seckel syndrome, including intrauterine dwarfism, microcephaly with memory impairment, ossification defects, and ocular and skeletal abnormalities, thus providing clear confirmation that specific mutations of CENPJ can cause Seckel syndrome. Immunohistochemistry revealed increased levels of DNA damage and apoptosis throughout Cenpjtm/tm embryos and adult mice showed an elevated frequency of micronucleus induction, suggesting that Cenpj-deficiency results in genomic instability. Notably, however, genomic instability was not the result of defective ATR-dependent DNA damage signaling, as is the case for the majority of genes associated with Seckel syndrome. Instead, Cenpjtm/tm embryonic fibroblasts exhibited irregular centriole and centrosome numbers and mono- and multipolar spindles, and many were near-tetraploid with numerical and structural chromosomal abnormalities when compared to passage-matched wild-type cells. Increased cell death due to mitotic failure during embryonic development is likely to contribute to the proportionate dwarfism that is associated with CENPJ-Seckel syndrome.


Zdroje

1. MajewskiF, GoeckeT (1982) Studies of microcephalic primordial dwarfism I: approach to a delineation of the Seckel syndrome. American journal of medical genetics 12: 7–21.

2. FaivreL, Le MerrerM, LyonnetS, PlauchuH, DagoneauN, et al. (2002) Clinical and genetic heterogeneity of Seckel syndrome. American journal of medical genetics 112: 379–383.

3. Harsha VardhanBG, MuthuMS, SaraswathiK, KoteeswaranD (2007) Bird-headed dwarf of Seckel. Journal of the Indian Society of Pedodontics and Preventive Dentistry 25Suppl: S8–9.

4. Al-DosariMS, ShaheenR, ColakD, AlkurayaFS (2010) Novel CENPJ mutation causes Seckel syndrome. J Med Genet 47: 411–414.

5. KalayE, YigitG, AslanY, BrownKE, PohlE, et al. (2011) CEP152 is a genome maintenance protein disrupted in Seckel syndrome. Nature genetics 43: 23–26.

6. QvistP, HuertasP, JimenoS, NyegaardM, HassanMJ, et al. (2011) CtIP Mutations Cause Seckel and Jawad Syndromes. PLoS genetics 7: e1002310.

7. O'DriscollM, Ruiz-PerezVL, WoodsCG, JeggoPA, GoodshipJA (2003) A splicing mutation affecting expression of ataxia-telangiectasia and Rad3-related protein (ATR) results in Seckel syndrome. Nature genetics 33: 497–501.

8. MajewskiF, RankeM, SchinzelA (1982) Studies of microcephalic primordial dwarfism II: the osteodysplastic type II of primordial dwarfism. American journal of medical genetics 12: 23–35.

9. RauchA, ThielCT, SchindlerD, WickU, CrowYJ, et al. (2008) Mutations in the pericentrin (PCNT) gene cause primordial dwarfism. Science 319: 816–819.

10. WillemsM, GenevieveD, BorckG, BaumannC, BaujatG, et al. (2010) Molecular analysis of pericentrin gene (PCNT) in a series of 24 Seckel/microcephalic osteodysplastic primordial dwarfism type II (MOPD II) families. Journal of medical genetics 47: 797–802.

11. HatchEM, KulukianA, HollandAJ, ClevelandDW, StearnsT (2010) Cep152 interacts with Plk4 and is required for centriole duplication. The Journal of cell biology 191: 721–729.

12. CizmeciogluO, ArnoldM, BahtzR, SetteleF, EhretL, et al. (2010) Cep152 acts as a scaffold for recruitment of Plk4 and CPAP to the centrosome. The Journal of cell biology 191: 731–739.

13. LealGF, RobertsE, SilvaEO, CostaSM, HampshireDJ, et al. (2003) A novel locus for autosomal recessive primary microcephaly (MCPH6) maps to 13q12.2. J Med Genet 40: 540–542.

14. GulA, HassanMJ, HussainS, RazaSI, ChishtiMS, et al. (2006) A novel deletion mutation in CENPJ gene in a Pakistani family with autosomal recessive primary microcephaly. J Hum Genet 51: 760–764.

15. BondJ, RobertsE, SpringellK, LizarragaSB, ScottS, et al. (2005) A centrosomal mechanism involving CDK5RAP2 and CENPJ controls brain size. Nat Genet 37: 353–355.

16. KaindlAM, PassemardS, KumarP, KraemerN, IssaL, et al. (2010) Many roads lead to primary autosomal recessive microcephaly. Progress in neurobiology 90: 363–383.

17. DelavalB, DoxseySJ (2010) Pericentrin in cellular function and disease. The Journal of cell biology 188: 181–190.

18. HungLY, TangCJ, TangTK (2000) Protein 4.1 R-135 interacts with a novel centrosomal protein (CPAP) which is associated with the gamma-tubulin complex. Mol Cell Biol 20: 7813–7825.

19. KohlmaierG, LoncarekJ, MengX, McEwenBF, MogensenMM, et al. (2009) Overly long centrioles and defective cell division upon excess of the SAS-4-related protein CPAP. Current biology: CB 19: 1012–1018.

20. SchmidtTI, Kleylein-SohnJ, WestendorfJ, Le ClechM, LavoieSB, et al. (2009) Control of centriole length by CPAP and CP110. Current biology: CB 19: 1005–1011.

21. TangCJ, FuRH, WuKS, HsuWB, TangTK (2009) CPAP is a cell-cycle regulated protein that controls centriole length. Nat Cell Biol 11: 825–831.

22. NiggEA, RaffJW (2009) Centrioles, centrosomes, and cilia in health and disease. Cell 139: 663–678.

23. ZyssD, GergelyF (2009) Centrosome function in cancer: guilty or innocent? Trends in cell biology 19: 334–346.

24. GanemNJ, GodinhoSA, PellmanD (2009) A mechanism linking extra centrosomes to chromosomal instability. Nature 460: 278–282.

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

26. SkarnesWC, RosenB, WestAP, KoutsourakisM, BushellW, et al. (2011) A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474: 337–342.

27. SirJH, BarrAR, NicholasAK, CarvalhoOP, KhurshidM, et al. (2011) A primary microcephaly protein complex forms a ring around parental centrioles. Nature genetics 43: 1147–1153.

28. FitzgeraldB, O'DriscollM, ChongK, KeatingS, ShannonP (2012) Neuropathology of fetal stage Seckel syndrome: a case report providing a morphological correlate for the emerging molecular mechanisms. Brain & development 34: 238–243.

29. HoriA, TamagawaK, EberSW, WestmeierM, HansmannI (1987) Neuropathology of Seckel syndrome in fetal stage with evidence of intrauterine developmental retardation. Acta neuropathologica 74: 397–401.

30. ArnoldSR, SpicerD, KouseffB, LacsonA, Gilbert-BarnessE (1999) Seckel-like syndrome in three siblings. Pediatric and developmental pathology: the official journal of the Society for Pediatric Pathology and the Paediatric Pathology Society 2: 180–187.

31. Endoh-YamagamiS, KarkarKM, MaySR, CobosI, ThwinMT, et al. (2010) A mutation in the pericentrin gene causes abnormal interneuron migration to the olfactory bulb in mice. Developmental biology 340: 41–53.

32. CarfagniniF, TaniG, AmbrosettoP (1999) MR findings in Seckel's syndrome: report of a case. Pediatric radiology 29: 849–850.

33. AbueloD (2007) Microcephaly syndromes. Seminars in pediatric neurology 14: 118–127.

34. CapovillaG, LorenzettiME, MontagniniA, BorgattiR, PiccinelliP, et al. (2001) Seckel's syndrome and malformations of cortical development: report of three new cases and review of the literature. Journal of child neurology 16: 382–386.

35. ShanskeA, CarideDG, Menasse-PalmerL, BogdanowA, MarionRW (1997) Central nervous system anomalies in Seckel syndrome: report of a new family and review of the literature. American journal of medical genetics 70: 155–158.

36. GruberR, ZhouZ, SukchevM, JoerssT, FrappartPO, et al. (2011) MCPH1 regulates the neuroprogenitor division mode by coupling the centrosomal cycle with mitotic entry through the Chk1-Cdc25 pathway. Nature cell biology 13: 1325–1334.

37. LizarragaSB, MargossianSP, HarrisMH, CampagnaDR, HanAP, et al. (2010) Cdk5rap2 regulates centrosome function and chromosome segregation in neuronal progenitors. Development 137: 1907–1917.

38. Sanchez-AndradeG, KendrickKM (2011) Roles of alpha- and beta-estrogen receptors in mouse social recognition memory: effects of gender and the estrous cycle. Hormones and behavior 59: 114–122.

39. KoganJH, FranklandPW, SilvaAJ (2000) Long-term memory underlying hippocampus-dependent social recognition in mice. Hippocampus 10: 47–56.

40. EngelmannM, HadickeJ, NoackJ (2011) Testing declarative memory in laboratory rats and mice using the nonconditioned social discrimination procedure. Nature protocols 6: 1152–1162.

41. GriffithE, WalkerS, MartinCA, VagnarelliP, StiffT, et al. (2008) Mutations in pericentrin cause Seckel syndrome with defective ATR-dependent DNA damage signaling. Nature genetics 40: 232–236.

42. PoloSE, JacksonSP (2011) Dynamics of DNA damage response proteins at DNA breaks: a focus on protein modifications. Genes & development 25: 409–433.

43. ZhivotovskyB, KroemerG (2004) Apoptosis and genomic instability. Nature reviews Molecular cell biology 5: 752–762.

44. AdiyamanP, BerberogluM, AycanZ, EvliyaogluO, OcalG (2004) Seckel-like syndrome: a patient with precocious puberty associated with nonclassical congenital adrenal hyperplasia. Journal of pediatric endocrinology & metabolism: JPEM 17: 105–110.

45. StoppoloniG, StabileM, RinaldiMM, PriscoF, RabuanoRG, et al. (1992) Seckel syndrome: report of three sibships with the type I primordial dwarfism. Possible linkage with HLA locus. Annales de genetique 35: 213–216.

46. DaughadayW (1941) A comparison of the X-zone of the adrenal cortex in two inbred strains of mice. Cancer Research 1: 883–885.

47. ReddyS, StarrC (2007) Seckel syndrome and spontaneously dislocated lenses. Journal of cataract and refractive surgery 33: 910–912.

48. GuirgisMF, LamBL, HowardCW (2001) Ocular manifestations of Seckel syndrome. American journal of ophthalmology 132: 596–597.

49. CanE, BulbulA, UsluS, DemirinH, ComertS, et al. (2010) A case of Seckel syndrome with Tetralogy of Fallot. Genetic counseling 21: 49–51.

50. UcarB, KilicZ, DinleyiciEC, YakutA, DogruelN (2004) Seckel syndrome associated with atrioventricular canal defect: a case report. Clinical dysmorphology 13: 53–55.

51. RappenU, von BrenndorffAI (1993) [Cardiac symptoms in 2 patients with Seckel syndrome]. Monatsschrift Kinderheilkunde: Organ der Deutschen Gesellschaft fur Kinderheilkunde 141: 584–586.

52. HowanietzH, FrischH, Jedlicka-KohlerI, StegerH (1989) [Seckel dwarfism based on a personal case]. Klinische Padiatrie 201: 139–141.

53. FukudaS, MorishitaY, HashiguchiM, TairaA (1991) [Seckel's syndrome associated with atrial septal defect: a case report and review of the literature in Japan]. Kyobu geka The Japanese journal of thoracic surgery 44: 411–413.

54. Elwell M, Mahler J (1999) Pathology of the Mouse; Maronpot R, editor: Cache River Press.

55. LiuZ, YueS, ChenX, KubinT, BraunT (2010) Regulation of cardiomyocyte polyploidy and multinucleation by CyclinG1. Circulation research 106: 1498–1506.

56. KeenanCM, VidalJD (2006) Standard morphologic evaluation of the heart in the laboratory dog and monkey. Toxicologic pathology 34: 67–74.

57. ChoJH, ChangCJ, ChenCY, TangTK (2006) Depletion of CPAP by RNAi disrupts centrosome integrity and induces multipolar spindles. Biochem Biophys Res Commun 339: 742–747.

58. MayerTU, KapoorTM, HaggartySJ, KingRW, SchreiberSL, et al. (1999) Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science 286: 971–974.

59. GergelyF, BastoR (2008) Multiple centrosomes: together they stand, divided they fall. Genes & development 22: 2291–2296.

60. AldertonGK, JoenjeH, VaronR, BorglumAD, JeggoPA, et al. (2004) Seckel syndrome exhibits cellular features demonstrating defects in the ATR-signalling pathway. Human molecular genetics 13: 3127–3138.

61. CimprichKA, CortezD (2008) ATR: an essential regulator of genome integrity. Nature reviews Molecular cell biology 9: 616–627.

62. PommierY (2006) Topoisomerase I inhibitors: camptothecins and beyond. Nature reviews Cancer 6: 789–802.

63. O'DriscollM, GenneryAR, SeidelJ, ConcannonP, JeggoPA (2004) An overview of three new disorders associated with genetic instability: LIG4 syndrome, RS-SCID and ATR-Seckel syndrome. DNA repair 3: 1227–1235.

64. KjaerI, HansenN, BecktorKB, BirkebaekN, BalslevT (2001) Craniofacial morphology, dentition, and skeletal maturity in four siblings with Seckel syndrome. The Cleft palate-craniofacial journal: official publication of the American Cleft Palate-Craniofacial Association 38: 645–651.

65. GotzM, HuttnerWB (2005) The cell biology of neurogenesis. Nature reviews Molecular cell biology 6: 777–788.

66. LuB, JanL, JanYN (2000) Control of cell divisions in the nervous system: symmetry and asymmetry. Annual review of neuroscience 23: 531–556.

67. EvansPD, VallenderEJ, LahnBT (2006) Molecular evolution of the brain size regulator genes CDK5RAP2 and CENPJ. Gene 375: 75–79.

68. CrastaK, GanemNJ, DagherR, LantermannAB, IvanovaEV, et al. (2012) DNA breaks and chromosome pulverization from errors in mitosis. Nature 482: 53–58.

69. RogakouEP, Nieves-NeiraW, BoonC, PommierY, BonnerWM (2000) Initiation of DNA fragmentation during apoptosis induces phosphorylation of H2AX histone at serine 139. The Journal of biological chemistry 275: 9390–9395.

70. BastoR, LauJ, VinogradovaT, GardiolA, WoodsCG, et al. (2006) Flies without centrioles. Cell 125: 1375–1386.

71. DobbelaereJ, JosueF, SuijkerbuijkS, BaumB, TaponN, et al. (2008) A genome-wide RNAi screen to dissect centriole duplication and centrosome maturation in Drosophila. PLoS biology 6: e224.

72. GraserS, StierhofYD, LavoieSB, GassnerOS, LamlaS, et al. (2007) Cep164, a novel centriole appendage protein required for primary cilium formation. The Journal of cell biology 179: 321–330.

73. UetakeY, SluderG (2010) Prolonged prometaphase blocks daughter cell proliferation despite normal completion of mitosis. Current biology: CB 20: 1666–1671.

74. OrthJD, LoewerA, LahavG, MitchisonTJ (2012) Prolonged mitotic arrest triggers partial activation of apoptosis, resulting in DNA damage and p53 induction. Molecular biology of the cell 23: 567–576.

75. JanssenA, van der BurgM, SzuhaiK, KopsGJ, MedemaRH (2011) Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science 333: 1895–1898.

76. StorchovaZ, PellmanD (2004) From polyploidy to aneuploidy, genome instability and cancer. Nature reviews Molecular cell biology 5: 45–54.

77. BorelF, LohezOD, LacroixFB, MargolisRL (2002) Multiple centrosomes arise from tetraploidy checkpoint failure and mitotic centrosome clusters in p53 and RB pocket protein-compromised cells. Proceedings of the National Academy of Sciences of the United States of America 99: 9819–9824.

78. ThoolenB, MaronpotRR, HaradaT, NyskaA, RousseauxC, et al. (2010) Proliferative and nonproliferative lesions of the rat and mouse hepatobiliary system. Toxicologic pathology 38: 5S–81S.

79. HoeijmakersJH (2009) DNA damage, aging, and cancer. The New England journal of medicine 361: 1475–1485.

80. JefferyAN, MetcalfBS, HoskingJ, StreeterAJ, VossLD, et al. (2012) Age before stage: insulin resistance rises before the onset of puberty: a 9-year longitudinal study (EarlyBird 26). Diabetes care 35: 536–541.

81. Huang-DoranI, BicknellLS, FinucaneFM, RochaN, PorterKM, et al. (2011) Genetic defects in human pericentrin are associated with severe insulin resistance and diabetes. Diabetes 60: 925–935.

82. KlingseisenA, JacksonAP (2011) Mechanisms and pathways of growth failure in primordial dwarfism. Genes & development 25: 2011–2024.

83. JurczykA, PinoSC, O'Sullivan-MurphyB, AddorioM, LidstoneEA, et al. (2010) A novel role for the centrosomal protein, pericentrin, in regulation of insulin secretory vesicle docking in mouse pancreatic beta-cells. PloS one 5: e11812.

84. RichtsmeierJT, BaxterLL, ReevesRH (2000) Parallels of craniofacial maldevelopment in Down syndrome and Ts65Dn mice. Developmental dynamics: an official publication of the American Association of Anatomists 217: 137–145.

85. MahajanVB, SkeieJM, AssefniaAH, MahajanM, TsangSH (2011) Mouse eye enucleation for remote high-throughput phenotyping. Journal of visualized experiments: JoVE 57: e3184.

86. HancockJM, AdamsNC, AidinisV, BlakeA, BogueM, et al. (2007) Mouse Phenotype Database Integration Consortium: integration [corrected] of mouse phenome data resources. Mammalian genome: official journal of the International Mammalian Genome Society 18: 157–163.

87. MasuyaH, InoueM, WadaY, ShimizuA, NaganoJ, et al. (2005) Implementation of the modified-SHIRPA protocol for screening of dominant phenotypes in a large-scale ENU mutagenesis program. Mammalian genome: official journal of the International Mammalian Genome Society 16: 829–837.

88. DertingerSD, TorousDK, TometskoKR (1996) Simple and reliable enumeration of micronucleated reticulocytes with a single-laser flow cytometer. Mutation research 371: 283–292.

89. PierceMJ, MorseRP (2012) The neurologic findings in Taybi-Linder syndrome (MOPD I/III): case report and review of the literature. American journal of medical genetics Part A 158A: 606–610.

90. LoizouJI, SanchoR, KanuN, BollandDJ, YangF, et al. (2011) ATMIN is required for maintenance of genomic stability and suppression of B cell lymphoma. Cancer Cell 19: 587–600.

91. ShannonP, MarkielA, OzierO, BaligaNS, WangJT, et al. (2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome research 13: 2498–2504.

92. MartinA, OchagaviaME, RabasaLC, MirandaJ, Fernandez-de-CossioJ, et al. (2010) BisoGenet: a new tool for gene network building, visualization and analysis. BMC bioinformatics 11: 91.

93. AssenovY, RamirezF, SchelhornSE, LengauerT, AlbrechtM (2008) Computing topological parameters of biological networks. Bioinformatics 24: 282–284.

94. KamburovA, WierlingC, LehrachH, HerwigR (2009) ConsensusPathDB–a database for integrating human functional interaction networks. Nucleic acids research 37: D623–628.

95. Team RDC (2011) R: A language and environment for statistical computing. Vienna, Austria.: R Foundation for Statistical Computing.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 11
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#