#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Rapid-Throughput Skeletal Phenotyping of 100 Knockout Mice Identifies 9 New Genes That Determine Bone Strength


Osteoporosis is a common polygenic disease and global healthcare priority but its genetic basis remains largely unknown. We report a high-throughput multi-parameter phenotype screen to identify functionally significant skeletal phenotypes in mice generated by the Wellcome Trust Sanger Institute Mouse Genetics Project and discover novel genes that may be involved in the pathogenesis of osteoporosis. The integrated use of primary phenotype data with quantitative x-ray microradiography, micro-computed tomography, statistical approaches and biomechanical testing in 100 unselected knockout mouse strains identified nine new genetic determinants of bone mass and strength. These nine new genes include five whose deletion results in low bone mass and four whose deletion results in high bone mass. None of the nine genes have been implicated previously in skeletal disorders and detailed analysis of the biomechanical consequences of their deletion revealed a novel functional classification of bone structure and strength. The organ-specific and disease-focused strategy described in this study can be applied to any biological system or tractable polygenic disease, thus providing a general basis to define gene function in a system-specific manner. Application of the approach to diseases affecting other physiological systems will help to realize the full potential of the International Mouse Phenotyping Consortium.


Vyšlo v časopise: Rapid-Throughput Skeletal Phenotyping of 100 Knockout Mice Identifies 9 New Genes That Determine Bone Strength. PLoS Genet 8(8): e32767. doi:10.1371/journal.pgen.1002858
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1002858

Souhrn

Osteoporosis is a common polygenic disease and global healthcare priority but its genetic basis remains largely unknown. We report a high-throughput multi-parameter phenotype screen to identify functionally significant skeletal phenotypes in mice generated by the Wellcome Trust Sanger Institute Mouse Genetics Project and discover novel genes that may be involved in the pathogenesis of osteoporosis. The integrated use of primary phenotype data with quantitative x-ray microradiography, micro-computed tomography, statistical approaches and biomechanical testing in 100 unselected knockout mouse strains identified nine new genetic determinants of bone mass and strength. These nine new genes include five whose deletion results in low bone mass and four whose deletion results in high bone mass. None of the nine genes have been implicated previously in skeletal disorders and detailed analysis of the biomechanical consequences of their deletion revealed a novel functional classification of bone structure and strength. The organ-specific and disease-focused strategy described in this study can be applied to any biological system or tractable polygenic disease, thus providing a general basis to define gene function in a system-specific manner. Application of the approach to diseases affecting other physiological systems will help to realize the full potential of the International Mouse Phenotyping Consortium.


Zdroje

1. FarooqiIS, JebbSA, LangmackG, LawrenceE, CheethamCH, et al. (1999) Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med 341: 879–884.

2. MontagueCT, FarooqiIS, WhiteheadJP, SoosMA, RauH, et al. (1997) Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387: 903–908.

3. PearsonER, FlechtnerI, NjolstadPR, MaleckiMT, FlanaganSE, et al. (2006) Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med 355: 467–477.

4. YamagataK, OdaN, KaisakiPJ, MenzelS, FurutaH, et al. (1996) Mutations in the hepatocyte nuclear factor-1alpha gene in maturity-onset diabetes of the young (MODY3). Nature 384: 455–458.

5. BrunkowME, GardnerJC, Van NessJ, PaeperBW, KovacevichBR, et al. (2001) Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. Am J Hum Genet 68: 577–589.

6. ChandrasekharappaSC, GuruSC, ManickamP, OlufemiSE, CollinsFS, et al. (1997) Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science 276: 404–407.

7. GongY, SleeRB, FukaiN, RawadiG, Roman-RomanS, et al. (2001) LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 107: 513–523.

8. CollinsFS, RossantJ, WurstW (2007) A mouse for all reasons. Cell 128: 9–13.

9. 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.

10. JohnellO, KanisJA (2006) An estimate of the worldwide prevalence and disability associated with osteoporotic fractures. Osteoporos Int 17: 1726–1733.

11. RalstonSH, UitterlindenAG (2010) Genetics of osteoporosis. Endocr Rev 31: 629–662.

12. RivadeneiraF, StyrkarsdottirU, EstradaK, HalldorssonBV, HsuYH, et al. (2009) Twenty bone-mineral-density loci identified by large-scale meta-analysis of genome-wide association studies. Nat Genet 41: 1199–1206.

13. CirulliET, GoldsteinDB (2010) Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nat Rev Genet 11: 415–425.

14. BlackDM, ThompsonDE, BauerDC, EnsrudK, MuslinerT, et al. (2000) Fracture risk reduction with alendronate in women with osteoporosis: the Fracture Intervention Trial. FIT Research Group. J Clin Endocrinol Metab 85: 4118–4124.

15. MackeyDC, BlackDM, BauerDC, McCloskeyEV, EastellR, et al. (2011) Effects of antiresorptive treatment on non-vertebral fracture outcomes. J Bone Miner Res 26: 2411–2418.

16. BoydenLM, MaoJ, BelskyJ, MitznerL, FarhiA, et al. (2002) High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med 346: 1513–1521.

17. RachnerTD, KhoslaS, HofbauerLC (2011) Osteoporosis: now and the future. Lancet 377: 1276–1287.

18. BrownSD, ChambonP, de AngelisMH (2005) EMPReSS: standardized phenotype screens for functional annotation of the mouse genome. Nat Genet 37: 1155.

19. JusticeMJ (2008) Removing the cloak of invisibility: phenotyping the mouse. Dis Model Mech 1: 109–112.

20. KarpNA, BakerLA, GerdinAK, AdamsNC, Ramirez-SolisR, et al. (2010) Optimising experimental design for high-throughput phenotyping in mice: a case study. Mamm Genome 21: 467–476.

21. Hardisty-HughesRE, ParkerA, BrownSD (2010) A hearing and vestibular phenotyping pipeline to identify mouse mutants with hearing impairment. Nat Protoc 5: 177–190.

22. BoskeyAL, MooreDJ, AmlingM, CanalisE, DelanyAM (2003) Infrared analysis of the mineral and matrix in bones of osteonectin-null mice and their wildtype controls. J Bone Miner Res 18: 1005–1011.

23. DelanyAM, AmlingM, PriemelM, HoweC, BaronR, et al. (2000) Osteopenia and decreased bone formation in osteonectin-deficient mice. J Clin Invest 105: 915–923.

24. Everitt B, editor. An R and S-PLUS Companion to Multivariate Analysis. London: Springer.

25. MitchellAFS, KrzanowskiWJ (1985) The Mahalanobis distance and elliptic distributions. Biometrika 72: 464–467.

26. RousseeuwPJ (1985) Multivariate estimation with high breakdown point;. W. Grossmann GP, I. Vinceze, and W. Wertz, Eds., editor The Netherlands: Reidel. 283–297.

27. RousseeuwPJ, Van ZomerenBC (1990) Unmasking Multivariate Outliers and Leverage Points. Journal of the American Statistical Association 85: 633–639.

28. AmmannP, RizzoliR (2003) Bone strength and its determinants. Osteoporos Int 14 Suppl 3: S13–18.

29. BoyleWJ, SimonetWS, LaceyDL (2003) Osteoclast differentiation and activation. Nature 423: 337–342.

30. HaradaS, RodanGA (2003) Control of osteoblast function and regulation of bone mass. Nature 423: 349–355.

31. DickensonRP, HuttonWC, StottJR (1981) The mechanical properties of bone in osteoporosis. J Bone Joint Surg Br 63-B: 233–238.

32. Marchler-BauerA, AndersonJB, ChitsazF, DerbyshireMK, DeWeese-ScottC, et al. (2009) CDD: specific functional annotation with the Conserved Domain Database. Nucleic Acids Res 37: D205–210.

33. DaileyL, BasilicoC (2001) Coevolution of HMG domains and homeodomains and the generation of transcriptional regulation by Sox/POU complexes. J Cell Physiol 186: 315–328.

34. LaudetV, StehelinD, CleversH (1993) Ancestry and diversity of the HMG box superfamily. Nucleic Acids Res 21: 2493–2501.

35. LoveJJ, LiX, CaseDA, GieseK, GrosschedlR, et al. (1995) Structural basis for DNA bending by the architectural transcription factor LEF-1. Nature 376: 791–795.

36. PohlerJR, NormanDG, BramhamJ, BianchiME, LilleyDM (1998) HMG box proteins bind to four-way DNA junctions in their open conformation. EMBO J 17: 817–826.

37. BernardP, TangP, LiuS, DewingP, HarleyVR, et al. (2003) Dimerization of SOX9 is required for chondrogenesis, but not for sex determination. Hum Mol Genet 12: 1755–1765.

38. MundlosS, OlsenBR (1997) Heritable diseases of the skeleton. Part I: Molecular insights into skeletal development-transcription factors and signaling pathways. FASEB J 11: 125–132.

39. MacDonaldBT, TamaiK, HeX (2009) Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell 17: 9–26.

40. BalemansW, Van HulW (2007) The genetics of low-density lipoprotein receptor-related protein 5 in bone: a story of extremes. Endocrinology 148: 2622–2629.

41. BaronR, RawadiG (2007) Wnt signaling and the regulation of bone mass. Curr Osteoporos Rep 5: 73–80.

42. ChanA, van BezooijenRL, LowikCW (2007) A new paradigm in the treatment of osteoporosis: Wnt pathway proteins and their antagonists. Curr Opin Investig Drugs 8: 293–298.

43. LuytenFP, TylzanowskiP, LoriesRJ (2009) Wnt signaling and osteoarthritis. Bone 44: 522–527.

44. GiangrecoA, JensenKB, TakaiY, MiyoshiJ, WattFM (2009) Necl2 regulates epidermal adhesion and wound repair. Development 136: 3505–3514.

45. KuramochiM, FukuharaH, NobukuniT, KanbeT, MaruyamaT, et al. (2001) TSLC1 is a tumor-suppressor gene in human non-small-cell lung cancer. Nat Genet 27: 427–430.

46. BiedererT, SaraY, MozhayevaM, AtasoyD, LiuX, et al. (2002) SynCAM, a synaptic adhesion molecule that drives synapse assembly. Science 297: 1525–1531.

47. TakayanagiY, FujitaE, YuZ, YamagataT, MomoiMY, et al. (2010) Impairment of social and emotional behaviors in Cadm1-knockout mice. Biochem Biophys Res Commun 396: 703–708.

48. GiangrecoA, HosteE, TakaiY, RosewellI, WattFM (2012) Epidermal Cadm1 expression promotes autoimmune alopecia via enhanced T cell adhesion and cytotoxicity. J Immunol 188: 1514–1522.

49. ItoA, HagiyamaM, OonumaJ (2008) Nerve-mast cell and smooth muscle-mast cell interaction mediated by cell adhesion molecule-1, CADM1. J Smooth Muscle Res 44: 83–93.

50. FujitaE, KourokuY, OzekiS, TanabeY, ToyamaY, et al. (2006) Oligo-astheno-teratozoospermia in mice lacking RA175/TSLC1/SynCAM/IGSF4A, a cell adhesion molecule in the immunoglobulin superfamily. Mol Cell Biol 26: 718–726.

51. RauchF, GlorieuxFH (2004) Osteogenesis imperfecta. Lancet 363: 1377–1385.

52. TatibanaM, KitaK, TairaM, IshijimaS, SonodaT, et al. (1995) Mammalian phosphoribosyl-pyrophosphate synthetase. Adv Enzyme Regul 35: 229–249.

53. KatashimaR, IwahanaH, FujimuraM, YamaokaT, ItakuraM (1998) Assignment of the human phosphoribosylpyrophosphate synthetase-associated protein 41 gene (PRPSAP2) to 17p11.2–p12. Genomics 54: 180–181.

54. BothJ, WuT, BrasJ, SchaapGR, BaasF, et al. (2012) Identification of novel candidate oncogenes in chromosome region 17p11.2–p12 in human osteosarcoma. PLoS One 7: e30907.

55. SundbergBE, WaagE, JacobssonJA, StephanssonO, RumaksJ, et al. (2008) The evolutionary history and tissue mapping of amino acid transporters belonging to solute carrier families SLC32, SLC36, and SLC38. J Mol Neurosci 35: 179–193.

56. Franchi-GazzolaR, Dall'AstaV, SalaR, VisigalliR, BevilacquaE, et al. (2006) The role of the neutral amino acid transporter SNAT2 in cell volume regulation. Acta Physiol (Oxf) 187: 273–283.

57. KronenbergHM (2003) Developmental regulation of the growth plate. Nature 423: 332–336.

58. HiraokaS, FuruichiT, NishimuraG, ShibataS, YanagishitaM, et al. (2007) Nucleotide-sugar transporter SLC35D1 is critical to chondroitin sulfate synthesis in cartilage and skeletal development in mouse and human. Nat Med 13: 1363–1367.

59. Superti-FurgaA, HastbackaJ, RossiA, van der HartenJJ, WilcoxWR, et al. (1996) A family of chondrodysplasias caused by mutations in the diastrophic dysplasia sulfate transporter gene and associated with impaired sulfation of proteoglycans. Ann N Y Acad Sci 785: 195–201.

60. LiX, OminskyMS, NiuQT, SunN, DaughertyB, et al. (2008) Targeted deletion of the sclerostin gene in mice results in increased bone formation and bone strength. J Bone Miner Res 23: 860–869.

61. AkhterMP, WellsDJ, ShortSJ, CullenDM, JohnsonML, et al. (2004) Bone biomechanical properties in LRP5 mutant mice. Bone 35: 162–169.

62. LeeSW, ChoYS, NaJM, ParkUH, KangM, et al. (2010) ASXL1 represses retinoic acid receptor-mediated transcription through associating with HP1 and LSD1. J Biol Chem 285: 18–29.

63. FisherCL, LeeI, BloyerS, BozzaS, ChevalierJ, et al. (2010) Additional sex combs-like 1 belongs to the enhancer of trithorax and polycomb group and genetically interacts with Cbx2 in mice. Dev Biol 337: 9–15.

64. WellikDM (2007) Hox patterning of the vertebrate axial skeleton. Dev Dyn 236: 2454–2463.

65. WilliamsJA, KondoN, OkabeT, TakeshitaN, PilchakDM, et al. (2009) Retinoic acid receptors are required for skeletal growth, matrix homeostasis and growth plate function in postnatal mouse. Dev Biol 328: 315–327.

66. HoischenA, van BonBW, Rodriguez-SantiagoB, GilissenC, VissersLE, et al. (2011) De novo nonsense mutations in ASXL1 cause Bohring-Opitz syndrome. Nat Genet 43: 729–731.

67. SchultzDC, AyyanathanK, NegorevD, MaulGG, RauscherFJ3rd (2002) SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev 16: 919–932.

68. YangL, XiaL, WuDY, WangH, ChanskyHA, et al. (2002) Molecular cloning of ESET, a novel histone H3-specific methyltransferase that interacts with ERG transcription factor. Oncogene 21: 148–152.

69. NifujiA, IdenoH, OhyamaY, TakanabeR, ArakiR, et al. (2010) Nemo-like kinase (NLK) expression in osteoblastic cells and suppression of osteoblastic differentiation. Exp Cell Res 316: 1127–1136.

70. TakadaI, MiharaM, SuzawaM, OhtakeF, KobayashiS, et al. (2007) A histone lysine methyltransferase activated by non-canonical Wnt signalling suppresses PPAR-gamma transactivation. Nat Cell Biol 9: 1273–1285.

71. HisanoY, KobayashiN, KawaharaA, YamaguchiA, NishiT (2011) The sphingosine 1-phosphate transporter, SPNS2, functions as a transporter of the phosphorylated form of the immunomodulating agent FTY720. J Biol Chem 286: 1758–1766.

72. IshiiM, EgenJG, KlauschenF, Meier-SchellersheimM, SaekiY, et al. (2009) Sphingosine-1-phosphate mobilizes osteoclast precursors and regulates bone homeostasis. Nature 458: 524–528.

73. IshiiT, ShimazuY, NishiyamaI, KikutaJ, IshiiM (2011) The role of sphingosine 1-phosphate in migration of osteoclast precursors; an application of intravital two-photon microscopy. Mol Cells 31: 399–403.

74. PedersonL, RuanM, WestendorfJJ, KhoslaS, OurslerMJ (2008) Regulation of bone formation by osteoclasts involves Wnt/BMP signaling and the chemokine sphingosine-1-phosphate. Proc Natl Acad Sci U S A 105: 20764–20769.

75. RyuJ, KimHJ, ChangEJ, HuangH, BannoY, et al. (2006) Sphingosine 1-phosphate as a regulator of osteoclast differentiation and osteoclast-osteoblast coupling. EMBO J 25: 5840–5851.

76. ChuY, YangX (2011) SUMO E3 ligase activity of TRIM proteins. Oncogene 30: 1108–1116.

77. NapolitanoLM, JaffrayEG, HayRT, MeroniG (2011) Functional interactions between ubiquitin E2 enzymes and TRIM proteins. Biochem J 434: 309–319.

78. OzatoK, ShinDM, ChangTH, MorseHC3rd (2008) TRIM family proteins and their emerging roles in innate immunity. Nat Rev Immunol 8: 849–860.

79. WangY, LiY, QiX, YuanW, AiJ, et al. (2004) TRIM45, a novel human RBCC/TRIM protein, inhibits transcriptional activities of ElK-1 and AP-1. Biochem Biophys Res Commun 323: 9–16.

80. KomoriT (2006) Regulation of osteoblast differentiation by transcription factors. J Cell Biochem 99: 1233–1239.

81. WagnerEF (2010) Bone development and inflammatory disease is regulated by AP-1 (Fos/Jun). Ann Rheum Dis 69 Suppl 1: i86–88.

82. RichardsJB, RivadeneiraF, InouyeM, PastinenTM, SoranzoN, et al. (2008) Bone mineral density, osteoporosis, and osteoporotic fractures: a genome-wide association study. Lancet 371: 1505–1512.

83. StyrkarsdottirU, HalldorssonBV, GretarsdottirS, GudbjartssonDF, WaltersGB, et al. (2008) Multiple genetic loci for bone mineral density and fractures. N Engl J Med 358: 2355–2365.

84. EstradaK, StyrkarsdottirU, EvangelouE, HsuYH, DuncanEL, et al. (2012) Genome-wide meta-analysis identifies 56 bone mineral density loci and reveals 14 loci associated with risk of fracture. Nat Genet 44: 491–501.

85. BassettJH, BoydeA, HowellPG, BassettRH, GallifordTM, et al. (2010) Optimal bone strength and mineralization requires the type 2 iodothyronine deiodinase in osteoblasts. Proc Natl Acad Sci U S A 107: 7604–7609.

86. ParfittAM, DreznerMK, GlorieuxFH, KanisJA, MallucheH, et al. (1987) Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Miner Res 2: 595–610.

87. Venables WN, Ripley BD (2002) Modern Applied Statistics with S New York, NY, USA: Springer.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 8
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#