#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Reduced Life- and Healthspan in Mice Carrying a Mono-Allelic MVA Mutation


Mosaic Variegated Aneuploidy (MVA) syndrome is a rare autosomal recessive disorder characterized by inaccurate chromosome segregation and high rates of near-diploid aneuploidy. Children with MVA syndrome die at an early age, are cancer prone, and have progeroid features like facial dysmorphisms, short stature, and cataracts. The majority of MVA cases are linked to mutations in BUBR1, a mitotic checkpoint gene required for proper chromosome segregation. Affected patients either have bi-allelic BUBR1 mutations, with one allele harboring a missense mutation and the other a nonsense mutation, or mono-allelic BUBR1 mutations combined with allelic variants that yield low amounts of wild-type BubR1 protein. Parents of MVA patients that carry single allele mutations have mild mitotic defects, but whether they are at risk for any of the pathologies associated with MVA syndrome is unknown. To address this, we engineered a mouse model for the nonsense mutation 2211insGTTA (referred to as GTTA) found in MVA patients with bi-allelic BUBR1 mutations. Here we report that both the median and maximum lifespans of the resulting BubR1+/GTTA mice are significantly reduced. Furthermore, BubR1+/GTTA mice develop several aging-related phenotypes at an accelerated rate, including cataract formation, lordokyphosis, skeletal muscle wasting, impaired exercise ability, and fat loss. BubR1+/GTTA mice develop mild aneuploidies and show enhanced growth of carcinogen-induced tumors. Collectively, these data demonstrate that the BUBR1 GTTA mutation compromises longevity and healthspan, raising the interesting possibility that mono-allelic changes in BUBR1 might contribute to differences in aging rates in the general population.


Vyšlo v časopise: Reduced Life- and Healthspan in Mice Carrying a Mono-Allelic MVA Mutation. PLoS Genet 8(12): e32767. doi:10.1371/journal.pgen.1003138
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003138

Souhrn

Mosaic Variegated Aneuploidy (MVA) syndrome is a rare autosomal recessive disorder characterized by inaccurate chromosome segregation and high rates of near-diploid aneuploidy. Children with MVA syndrome die at an early age, are cancer prone, and have progeroid features like facial dysmorphisms, short stature, and cataracts. The majority of MVA cases are linked to mutations in BUBR1, a mitotic checkpoint gene required for proper chromosome segregation. Affected patients either have bi-allelic BUBR1 mutations, with one allele harboring a missense mutation and the other a nonsense mutation, or mono-allelic BUBR1 mutations combined with allelic variants that yield low amounts of wild-type BubR1 protein. Parents of MVA patients that carry single allele mutations have mild mitotic defects, but whether they are at risk for any of the pathologies associated with MVA syndrome is unknown. To address this, we engineered a mouse model for the nonsense mutation 2211insGTTA (referred to as GTTA) found in MVA patients with bi-allelic BUBR1 mutations. Here we report that both the median and maximum lifespans of the resulting BubR1+/GTTA mice are significantly reduced. Furthermore, BubR1+/GTTA mice develop several aging-related phenotypes at an accelerated rate, including cataract formation, lordokyphosis, skeletal muscle wasting, impaired exercise ability, and fat loss. BubR1+/GTTA mice develop mild aneuploidies and show enhanced growth of carcinogen-induced tumors. Collectively, these data demonstrate that the BUBR1 GTTA mutation compromises longevity and healthspan, raising the interesting possibility that mono-allelic changes in BUBR1 might contribute to differences in aging rates in the general population.


Zdroje

1. RickeRM, van ReeJH, van DeursenJM (2008) Whole chromosome instability and cancer: a complex relationship. Trends Genet 24: 457–466.

2. SiegelJJ, AmonA (2012) New Insights into the Troubles of Aneuploidy. Annu Rev Cell Dev Biol 28: 189–214.

3. NagaokaSI, HassoldTJ, HuntPA (2012) Human aneuploidy: mechanisms and new insights into an age-old problem. Nat Rev Genet 13: 493–504.

4. HollandAJ, ClevelandDW (2012) Losing balance: the origin and impact of aneuploidy in cancer. EMBO Rep 13: 501–514.

5. BakhoumSF, ComptonDA (2012) Chromosomal instability and cancer: a complex relationship with therapeutic potential. J Clin Invest 122: 1138–1143.

6. PfauSJ, AmonA (2012) Chromosomal instability and aneuploidy in cancer: from yeast to man. EMBO Rep 13: 515–527.

7. BakerDJ, DawlatyMM, GalardyP, van DeursenJM (2007) Mitotic regulation of the anaphase-promoting complex. Cellular and molecular life sciences: CMLS 64: 589–600.

8. MusacchioA, SalmonED (2007) The spindle-assembly checkpoint in space and time. Nature Reviews Molecular Cell Biology 8: 379–393.

9. PetersJM (2006) The anaphase promoting complex/cyclosome: a machine designed to destroy. Nat Rev Mol Cell Biol 7: 644–656.

10. LampsonMA, KapoorTM (2005) The human mitotic checkpoint protein BubR1 regulates chromosome-spindle attachments. Nature Cell Biology 7: 93–+.

11. CahillDP, LengauerC, YuJ, RigginsGJ, WillsonJK, et al. (1998) Mutations of mitotic checkpoint genes in human cancers. Nature 392: 300–303.

12. LengauerC, KinzlerKW, VogelsteinB (1997) Genetic instability in colorectal cancers. Nature 386: 623–627.

13. Rio FrioT, LavoieJ, HamelN, GeyerFC, KushnerYB, et al. (2010) Homozygous BUB1B mutation and susceptibility to gastrointestinal neoplasia. N Engl J Med 363: 2628–2637.

14. WeaverBA, ClevelandDW (2006) Does aneuploidy cause cancer? Curr Opin Cell Biol 18: 658–667.

15. HanksS, ColemanK, ReidS, PlajaA, FirthH, et al. (2004) Constitutional aneuploidy and cancer predisposition caused by biallelic mutations in BUB1B. Nat Genet 36: 1159–1161.

16. MatsuuraS, MatsumotoY, MorishimaK, IzumiH, MatsumotoH, et al. (2006) Monoallelic BUB1B mutations and defective mitotic-spindle checkpoint in seven families with premature chromatid separation (PCS) syndrome. Am J Med Genet A 140: 358–367.

17. Garcia-CastilloH, Vasquez-VelasquezAI, RiveraH, Barros-NunezP (2008) Clinical and genetic heterogeneity in patients with mosaic variegated aneuploidy: Delineation of clinical subtypes. Am J Med Genet A 146A: 1687–1695.

18. SuijkerbuijkSJ, van OschMH, BosFL, HanksS, RahmanN, et al. (2010) Molecular causes for BUBR1 dysfunction in the human cancer predisposition syndrome mosaic variegated aneuploidy. Cancer Res 70: 4891–4900.

19. DaiW, WangQ, LiuT, SwamyM, FangY, et al. (2004) Slippage of mitotic arrest and enhanced tumor development in mice with BubR1 haploinsufficiency. Cancer Res 64: 440–445.

20. WangQ, LiuT, FangY, XieS, HuangX, et al. (2004) BUBR1 deficiency results in abnormal megakaryopoiesis. Blood 103: 1278–1285.

21. BakerDJ, JeganathanKB, CameronJD, ThompsonM, JunejaS, et al. (2004) BubR1 insufficiency causes early onset of aging-associated phenotypes and infertility in mice. Nature genetics 36: 744–749.

22. BakerDJ, JeganathanKB, MalureanuL, Perez-TerzicC, TerzicA, et al. (2006) Early aging-associated phenotypes in Bub3/Rae1 haploinsufficient mice. J Cell Biol 172: 529–540.

23. BakerDJ, Perez-TerzicC, JinF, PitelK, NiederlanderNJ, et al. (2008) Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by BubR1 insufficiency. Nature cell biology 10: 825–836.

24. MatsumotoT, BakerDJ, d'UscioLV, MozammelG, KatusicZS, et al. (2007) Aging-associated vascular phenotype in mutant mice with low levels of BubR1. Stroke 38: 1050–1056.

25. HanksS, ColemanK, SummersgillB, MessahelB, WilliamsonD, et al. (2006) Comparative genomic hybridization and BUB1B mutation analyses in childhood cancers associated with mosaic variegated aneuploidy syndrome. Cancer letters 239: 234–238.

26. LimwongseC, SchwartzS, BocianM, RobinNH (1999) Child with mosaic variegated aneuploidy and embryonal rhabdomyosarcoma. Am J Med Genet 82: 20–24.

27. HanksS, RahmanN (2005) Aneuploidy-cancer predisposition syndromes: a new link between the mitotic spindle checkpoint and cancer. Cell Cycle 4: 225–227.

28. KajiiT, IkeuchiT, YangZQ, NakamuraY, TsujiY, et al. (2001) Cancer-prone syndrome of mosaic variegated aneuploidy and total premature chromatid separation: Report of five infants. American Journal of Medical Genetics 104: 57–64.

29. Garcia-CastilloH, Vasquez-VelasquezAI, RiveraH, Barros-NunezP (2008) Clinical and genetic heterogeneity in patients with mosaic variegated aneuploidy: Delineation of clinical subtypes. American Journal of Medical Genetics Part A 146A: 1687–1695.

30. KajiiT, KawaiT, TakumiT, MisuH, MabuchiO, et al. (1998) Mosaic variegated aneuploidy with multiple congenital abnormalities: Homozygosity for total premature chromatid separation trait. American Journal of Medical Genetics 78: 245–249.

31. PlajaA, VendrellT, SmeetsD, SarretE, GiliT, et al. (2001) Variegated aneuploidy related to premature centromere division (PCD) is expressed in vivo and is a cancer-prone disease. Am J Med Genet 98: 216–223.

32. GanmoreI, SmoohaG, IzraeliS (2009) Constitutional aneuploidy and cancer predisposition. Hum Mol Genet 18: R84–93.

33. MatsuuraS, ItoE, TauchiH, KomatsuK, IkeuchiT, et al. (2000) Chromosomal instability syndrome of total premature chromatid separation with mosaic variegated aneuploidy is defective in mitotic-spindle checkpoint. Am J Hum Genet 67: 483–486.

34. KawameH, SugioY, FuyamaY, HayashiY, SuzukiH, et al. (1999) Syndrome of microcephaly, Dandy-Walker malformation, and Wilms tumor caused by mosaic variegated aneuploidy with premature centromere division (PCD): report of a new case and review of the literature. J Hum Genet 44: 219–224.

35. RickeRM, JeganathanKB, van DeursenJM (2011) Bub1 overexpression induces aneuploidy and tumor formation through Aurora B kinase hyperactivation. J Cell Biol 193: 1049–1064.

36. van ReeJH, JeganathanKB, MalureanuL, van DeursenJM (2010) Overexpression of the E2 ubiquitin-conjugating enzyme UbcH10 causes chromosome missegregation and tumor formation. J Cell Biol 188: 83–100.

37. MalureanuL, JeganathanKB, JinF, BakerDJ, van ReeJH, et al. (2010) Cdc20 hypomorphic mice fail to counteract de novo synthesis of cyclin B1 in mitosis. J Cell Biol 191: 313–329.

38. BakerDJ, WijshakeT, TchkoniaT, LeBrasseurNK, ChildsBG, et al. (2011) Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479: 232–236.

39. KnollmannBC, CasimiroMC, KatchmanAN, SirenkoSG, SchoberT, et al. (2004) Isoproterenol exacerbates a long QT phenotype in Kcnq1-deficient neonatal mice: Possible roles for human-like Kcnq1 isoform 1 and slow delayed rectifier K+ current. Journal of Pharmacology and Experimental Therapeutics 310: 311–318.

40. WangL, YangL, DebiddaM, WitteD, ZhengY (2007) Cdc42 GTPase-activating protein deficiency promotes genomic instability and premature aging-like phenotypes. Proc Natl Acad Sci U S A 104: 1248–1253.

41. TynerSD, VenkatachalamS, ChoiJ, JonesS, GhebraniousN, et al. (2002) p53 mutant mice that display early ageing-associated phenotypes. Nature 415: 45–53.

42. BakerDJ, JeganathanKB, MalureanuL, Perez-TerzicC, TerzicA, et al. (2006) Early aging-associated phenotypes in Bub3/Rae1 haploinsufficient mice. The Journal of cell biology 172: 529–540.

43. LeBrasseurNK, SchelhornTM, BernardoBL, CosgrovePG, LoriaPM, et al. (2009) Myostatin Inhibition Enhances the Effects of Exercise on Performance and Metabolic Outcomes in Aged Mice. Journals of Gerontology Series a-Biological Sciences and Medical Sciences 64: 940–948.

44. RoubenoffR (2000) Sarcopenia and its implications for the elderly. Eur J Clin Nutr 54 Suppl 3: S40–47.

45. TchkoniaT, MorbeckDE, Von ZglinickiT, Van DeursenJ, LustgartenJ, et al. (2010) Fat tissue, aging, and cellular senescence. Aging cell 9: 667–684.

46. BakerDJ, JeganathanKB, CameronJD, ThompsonM, JunejaS, et al. (2004) BubR1 insufficiency causes early onset of aging-associated phenotypes and infertility in mice. Nat Genet 36: 744–749.

47. BakerDJ, Perez-TerzicC, JinF, PitelK, NiederlanderNJ, et al. (2008) Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by BubR1 insufficiency. Nat Cell Biol 10: 825–836.

48. BurtnerCR, KennedyBK (2010) Progeria syndromes and ageing: what is the connection? Nat Rev Mol Cell Biol 11: 567–578.

49. HastyP, CampisiJ, HoeijmakersJ, van SteegH, VijgJ (2003) Aging and genome maintenance: lessons from the mouse? Science 299: 1355–1359.

50. GarinisGA, van der HorstGT, VijgJ, HoeijmakersJH (2008) DNA damage and ageing: new-age ideas for an age-old problem. Nat Cell Biol 10: 1241–1247.

51. MoserMJ, BigbeeWL, GrantSG, EmondMJ, LangloisRG, et al. (2000) Genetic instability and hematologic disease risk in Werner syndrome patients and heterozygotes. Cancer Res 60: 2492–2496.

52. LebelM, LederP (1998) A deletion within the murine Werner syndrome helicase induces sensitivity to inhibitors of topoisomerase and loss of cellular proliferative capacity. Proc Natl Acad Sci U S A 95: 13097–13102.

53. van der HorstGT, MeiraL, GorgelsTG, de WitJ, Velasco-MiguelS, et al. (2002) UVB radiation-induced cancer predisposition in Cockayne syndrome group A (Csa) mutant mice. DNA Repair (Amst) 1: 143–157.

54. AndressooJO, WeedaG, de WitJ, MitchellJR, BeemsRB, et al. (2009) An Xpb mouse model for combined xeroderma pigmentosum and cockayne syndrome reveals progeroid features upon further attenuation of DNA repair. Mol Cell Biol 29: 1276–1290.

55. de BoerJ, DonkerI, de WitJ, HoeijmakersJH, WeedaG (1998) Disruption of the mouse xeroderma pigmentosum group D DNA repair/basal transcription gene results in preimplantation lethality. Cancer Res 58: 89–94.

56. de BoerJ, AndressooJO, de WitJ, HuijmansJ, BeemsRB, et al. (2002) Premature aging in mice deficient in DNA repair and transcription. Science 296: 1276–1279.

57. BakerDJ, JinF, van DeursenJM (2008) The yin and yang of the Cdkn2a locus in senescence and aging. Cell Cycle 7: 2795–2802.

58. SchvartzmanJM, SotilloR, BenezraR (2010) Mitotic chromosomal instability and cancer: mouse modelling of the human disease. Nat Rev Cancer 10: 102–115.

59. KimAH, PuramSV, BilimoriaPM, IkeuchiY, KeoughS, et al. (2009) A centrosomal Cdc20-APC pathway controls dendrite morphogenesis in postmitotic neurons. Cell 136: 322–336.

60. YangY, KimAH, YamadaT, WuB, BilimoriaPM, et al. (2009) A Cdc20-APC ubiquitin signaling pathway regulates presynaptic differentiation. Science 326: 575–578.

61. BabuJR, JeganathanKB, BakerDJ, WuX, Kang-DeckerN, et al. (2003) Rae1 is an essential mitotic checkpoint regulator that cooperates with Bub3 to prevent chromosome missegregation. The Journal of cell biology 160: 341–353.

62. MalureanuLA (2011) Targeting vector construction through recombineering. Methods Mol Biol 693: 181–203.

63. O'GormanS, DagenaisNA, QianM, MarchukY (1997) Protamine-Cre recombinase transgenes efficiently recombine target sequences in the male germ line of mice, but not in embryonic stem cells. Proc Natl Acad Sci U S A 94: 14602–14607.

64. KasperLH, BrindlePK, SchnabelCA, PritchardCEJ, ClearyML, et al. (1999) CREB binding protein interacts with nucleoporin-specific FG repeats that activate transcription and mediate NUP98-HOXA9 oncogenicity. Molecular and Cellular Biology 19: 764–776.

65. HamadaM, MalureanuLA, WijshakeT, ZhouW, van DeursenJM (2012) Reprogramming to pluripotency can conceal somatic cell chromosomal instability. PLoS Genet 8: e1002913 doi:10.1371/journal.pgen.1002913.

66. BabuJR, JeganathanKB, BakerDJ, WuX, Kang-DeckerN, et al. (2003) Rae1 is an essential mitotic checkpoint regulator that cooperates with Bub3 to prevent chromosome missegregation. J Cell Biol 160: 341–353.

67. PajvaniUB, TrujilloME, CombsTP, IyengarP, JelicksL, et al. (2005) Fat apoptosis through targeted activation of caspase 8: a new mouse model of inducible and reversible lipoatrophy. Nature Medicine 11: 797–803.

68. Martinez-FernandezA, NelsonTJ, YamadaS, ReyesS, AlekseevAE, et al. (2009) iPS programmed without c-MYC yield proficient cardiogenesis for functional heart chimerism. Circulation Research 105: 648–656.

69. SerranoM, LeeHW, ChinL, CordonCardoC, BeachD, et al. (1996) Role of the INK4a locus in tumor suppression and cell mortality. Cell 85: 27–37.

70. WangC, LiQ, ReddenDT, WeindruchR, AllisonDB (2004) Statistical methods for testing effects on “maximum lifespan”. Mech Ageing Dev 125: 629–632.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 12
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#