#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

: A Mouse Strain with an Ift140 Mutation That Results in a Skeletal Ciliopathy Modelling Jeune Syndrome


Cilia are architecturally complex organelles that protrude from the cell membrane and have signalling, sensory and motility functions that are central to normal tissue development and homeostasis. There are two broad categories of cilia; motile and non-motile, or primary, cilia. The central role of primary cilia in health and disease has become prominent in the past decade with the recognition of a number of human syndromes that result from defects in the formation or function of primary cilia. This rapidly growing class of conditions, now known as ciliopathies, impact the development of a diverse range of tissues including the neural axis, craniofacial structures, skeleton, kidneys, eyes and lungs. The broad impact of cilia dysfunction on development reflects the pivotal position of the primary cilia within a signalling nexus involving a growing number of growth factor systems including Hedgehog, Pdgf, Fgf, Hippo, Notch and both canonical Wnt and planar cell polarity. We have identified a novel ENU mutant allele of Ift140, which causes a mid-gestation embryonic lethal phenotype in homozygous mutant mice. Mutant embryos exhibit a range of phenotypes including exencephaly and spina bifida, craniofacial dysmorphism, digit anomalies, cardiac anomalies and somite patterning defects. A number of these phenotypes can be attributed to alterations in Hedgehog signalling, although additional signalling systems are also likely to be involved. We also report the identification of a homozygous recessive mutation in IFT140 in a Jeune syndrome patient. This ENU-induced Jeune syndrome model will be useful in delineating the origins of dysmorphology in human ciliopathies.


Vyšlo v časopise: : A Mouse Strain with an Ift140 Mutation That Results in a Skeletal Ciliopathy Modelling Jeune Syndrome. PLoS Genet 9(8): e32767. doi:10.1371/journal.pgen.1003746
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003746

Souhrn

Cilia are architecturally complex organelles that protrude from the cell membrane and have signalling, sensory and motility functions that are central to normal tissue development and homeostasis. There are two broad categories of cilia; motile and non-motile, or primary, cilia. The central role of primary cilia in health and disease has become prominent in the past decade with the recognition of a number of human syndromes that result from defects in the formation or function of primary cilia. This rapidly growing class of conditions, now known as ciliopathies, impact the development of a diverse range of tissues including the neural axis, craniofacial structures, skeleton, kidneys, eyes and lungs. The broad impact of cilia dysfunction on development reflects the pivotal position of the primary cilia within a signalling nexus involving a growing number of growth factor systems including Hedgehog, Pdgf, Fgf, Hippo, Notch and both canonical Wnt and planar cell polarity. We have identified a novel ENU mutant allele of Ift140, which causes a mid-gestation embryonic lethal phenotype in homozygous mutant mice. Mutant embryos exhibit a range of phenotypes including exencephaly and spina bifida, craniofacial dysmorphism, digit anomalies, cardiac anomalies and somite patterning defects. A number of these phenotypes can be attributed to alterations in Hedgehog signalling, although additional signalling systems are also likely to be involved. We also report the identification of a homozygous recessive mutation in IFT140 in a Jeune syndrome patient. This ENU-induced Jeune syndrome model will be useful in delineating the origins of dysmorphology in human ciliopathies.


Zdroje

1. BerbariNF, O'ConnorAK, HaycraftCJ, YoderBK (2009) The Primary Cilium as a Complex Signaling Center. Current Biology 19: R526–R535.

2. RosenbaumJL, WitmanGB (2002) Intraflagellar transport. Nat Rev Mol Cell Biol 3: 813–825.

3. KozminskiKG, BeechPL, RosenbaumJL (1995) The Chlamydomonas kinesin-like protein FLA10 is involved in motility associated with the flagellar membrane. The Journal of Cell Biology 131: 1517–1527.

4. ColeDG, DienerDR, HimelblauAL, BeechPL, FusterJC, et al. (1998) Chlamydomonas Kinesin-II–dependent Intraflagellar Transport (IFT): IFT Particles Contain Proteins Required for Ciliary Assembly in Caenorhabditis elegans Sensory Neurons. The Journal of Cell Biology 141: 993–1008.

5. PipernoG, SiudaE, HendersonS, SegilM, VaananenH, et al. (1998) Distinct Mutants of Retrograde Intraflagellar Transport (IFT) Share Similar Morphological and Molecular Defects. The Journal of Cell Biology 143: 1591–1601.

6. PipernoG, MeadK (1997) Transport of a novel complex in the cytoplasmic matrix of Chlamydomonas flagella. Proceedings of the National Academy of Sciences 94: 4457–4462.

7. KozminskiKG, JohnsonKA, ForscherP, RosenbaumJL (1993) A motility in the eukaryotic flagellum unrelated to flagellar beating. Proceedings of the National Academy of Sciences 90: 5519–5523.

8. HirokawaN (1998) Kinesin and Dynein Superfamily Proteins and the Mechanism of Organelle Transport. Science 279: 519–526.

9. TaschnerM, BhogarajuS, LorentzenE (2012) Architecture and function of IFT complex proteins in ciliogenesis. Differentiation 83: S12–S22.

10. PazourGJ, DickertBL, WitmanGB (1999) The DHC1b (DHC2) Isoform of Cytoplasmic Dynein Is Required for Flagellar Assembly. The Journal of Cell Biology 144: 473–481.

11. PorterME, BowerR, KnottJA, ByrdP, DentlerW (1999) Cytoplasmic Dynein Heavy Chain 1b Is Required for Flagellar Assembly in Chlamydomonas. Molecular Biology of the Cell 10: 693–712.

12. PiginoG, GeimerS, LanzavecchiaS, PaccagniniE, CanteleF, et al. (2009) Electron-tomographic analysis of intraflagellar transport particle trains in situ. The Journal of Cell Biology 187: 135–148.

13. NovarinoG, AkizuN, Gleeson JosephG (2011) Modeling Human Disease in Humans: The Ciliopathies. Cell 147: 70–79.

14. HuberC, Cormier-DaireV (2012) Ciliary disorder of the skeleton. American Journal of Medical Genetics Part C: Seminars in Medical Genetics 160C: 165–174.

15. WatersA, BealesP (2011) Ciliopathies: an expanding disease spectrum. Pediatric Nephrology 26: 1039–1056.

16. ChristensenST, ClementCA, SatirP, PedersenLB (2012) Primary cilia and coordination of receptor tyrosine kinase (RTK) signalling. The Journal of Pathology 226: 172–184.

17. CaruanaG, FarliePG, HartAH, Bagheri-FamS, WallaceMJ, et al. (2013) Genome-wide ENU mutagenesis in combination with high density SNP analysis and exome sequencing provides a rapid route to the identification of novel mouse models of developmental disease. PLoS ONE 8: e55429.

18. SchmidtsM, FrankV, EisenbergerT, al TurkiS, BizetAA, et al. (2013) Combined NGS Approaches Identify Mutations in the Intraflagellar Transport Gene IFT140 in Skeletal Ciliopathies with Early Progressive Kidney Disease. Human Mutation 34: 714–724.

19. PerraultI, SaunierS, HaneinS, FilholE, Bizet AlbaneA, et al. (2012) Mainzer-Saldino Syndrome Is a Ciliopathy Caused by IFT140 Mutations. The American Journal of Human Genetics 90: 864–870.

20. JonassenJA, SanAgustinJ, BakerSP, PazourGJ (2012) Disruption of IFT complex A causes cystic kidneys without mitotic spindle misorientation. J Am Soc Nephrol 23: 641–651.

21. CameronDA, PennimpedeT, PetkovichM (2009) Tulp3 is a critical repressor of mouse hedgehog signaling. Developmental Dynamics 238: 1140–1149.

22. PattersonVL, DamrauC, PaudyalA, ReeveB, GrimesDT, et al. (2009) Mouse hitchhiker mutants have spina bifida, dorso-ventral patterning defects and polydactyly: identification of Tulp3 as a novel negative regulator of the Sonic hedgehog pathway. Hum Mol Genet 18: 1719–1739.

23. AsheA, ButterfieldNC, TownL, CourtneyAD, CooperAN, et al. (2012) Mutations in mouse Ift144 model the craniofacial, limb and rib defects in skeletal ciliopathies. Human Molecular Genetics 21: 1808–1823.

24. LupasA, Van DykeM, StockJ (1991) Predicting coiled coils from protein sequences. Science 252: 1162–1164.

25. BuchanDWA, WardSM, LobleyAE, NugentTCO, BrysonK, et al. (2010) Protein annotation and modelling servers at University College London. Nucleic Acids Research 38: W563–W568.

26. BenazetJ-D, BischofbergerM, TieckeE, GoncalvesA, MartinJF, et al. (2009) A Self-Regulatory System of Interlinked Signaling Feedback Loops Controls Mouse Limb Patterning. Science 323: 1050–1053.

27. KnustE, BossingerO (2002) Composition and formation of intercellular junctions in epithelial cells. Science 298: 1955–1959.

28. TsukitaS, FuruseM, ItohM (2001) Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol 2: 285–293.

29. BealesPL, BlandE, TobinJL, BacchelliC, TuysuzB, et al. (2007) IFT80, which encodes a conserved intraflagellar transport protein, is mutated in Jeune asphyxiating thoracic dystrophy. Nat Genet 39: 727–729.

30. DavisEE, ZhangQ, LiuQ, DiplasBH, DaveyLM, et al. (2011) TTC21B contributes both causal and modifying alleles across the ciliopathy spectrum. Nat Genet 43: 189–196.

31. DagoneauN, GouletM, GenevièveD, SznajerY, MartinovicJ, et al. (2009) DYNC2H1 Mutations Cause Asphyxiating Thoracic Dystrophy and Short Rib-Polydactyly Syndrome, Type III. The American Journal of Human Genetics 84: 706–711.

32. BredrupC, SaunierS, Oud MachteldM, FiskerstrandT, HoischenA, et al. (2011) Ciliopathies with Skeletal Anomalies and Renal Insufficiency due to Mutations in the IFT-A Gene WDR19. The American Journal of Human Genetics 89: 634–643.

33. SchmidtsM, ArtsHH, BongersEMHF, YapZ, OudMM, et al. (2013) Exome sequencing identifies DYNC2H1 mutations as a common cause of asphyxiating thoracic dystrophy (Jeune syndrome) without major polydactyly, renal or retinal involvement. Journal of Medical Genetics 50: 309–323.

34. LiemKF, AsheA, HeM, SatirP, MoranJ, et al. (2012) The IFT-A complex regulates Shh signaling through cilia structure and membrane protein trafficking. The Journal of Cell Biology 197: 789–800.

35. QinJ, LinY, NormanRX, KoHW, EggenschwilerJT (2011) Intraflagellar transport protein 122 antagonizes Sonic Hedgehog signaling and controls ciliary localization of pathway components. Proceedings of the National Academy of Sciences 108: 1456–1461.

36. ChangY-F, ImamJS, WilkinsonMF (2007) The Nonsense-Mediated Decay RNA Surveillance Pathway. Annual Review of Biochemistry 76: 51–74.

37. WangJ, SmithPJ, KrainerAR, ZhangMQ (2005) Distribution of SR protein exonic splicing enhancer motifs in human protein-coding genes. Nucleic Acids Research 33: 5053–5062.

38. LiuH-X, ZhangM, KrainerAR (1998) Identification of functional exonic splicing enhancer motifs recognized by individual SR proteins. Genes & Development 12: 1998–2012.

39. DietzHC, ValleD, FrancomanoCA, KendziorRJ, PyeritzRE, et al. (1993) The skipping of constitutive exons in vivo induced by nonsense mutations. Science 259: 680–683.

40. WoolfeA, MullikinJ, ElnitskiL (2010) Genomic features defining exonic variants that modulate splicing. Genome Biology 11: R20.

41. CaputiM, KendziorRJ, BeemonKL (2002) A nonsense mutation in the fibrillin-1 gene of a Marfan syndrome patient induces NMD and disrupts an exonic splicing enhancer. Genes & Development 16: 1754–1759.

42. ErikssonM, BrownWT, GordonLB, GlynnMW, SingerJ, et al. (2003) Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature 423: 293–298.

43. MoseleyCT, MullisPE, PrinceMA, PhillipsJA (2002) An Exon Splice Enhancer Mutation Causes Autosomal Dominant GH Deficiency. Journal of Clinical Endocrinology & Metabolism 87: 847–852.

44. BoichardA, VenetL, NaasT, BoutronA, ChevretL, et al. (2008) Two silent substitutions in the PDHA1 gene cause exon 5 skipping by disruption of a putative exonic splicing enhancer. Molecular Genetics and Metabolism 93: 323–330.

45. CartegniL, KrainerAR (2002) Disruption of an SF2/ASF-dependent exonic splicing enhancer in SMN2 causes spinal muscular atrophy in the absence of SMN1. Nat Genet 30: 377–384.

46. LiuH-X, CartegniL, ZhangMQ, KrainerAR (2001) A mechanism for exon skipping caused by nonsense or missense mutations in BRCA1 and other genes. Nat Genet 27: 55–58.

47. KatoN, AoyamaH (1998) Dermomyotomal origin of the ribs as revealed by extirpation and transplantation experiments in chick and quail embryos. Development 125: 3437–3443.

48. FerranteMI, RomioL, CastroS, CollinsJE, GouldingDA, et al. (2009) Convergent extension movements and ciliary function are mediated by ofd1, a zebrafish orthologue of the human oral-facial-digital type 1 syndrome gene. Human Molecular Genetics 18: 289–303.

49. ZengH, HooverAN, LiuA (2010) PCP effector gene Inturned is an important regulator of cilia formation and embryonic development in mammals. Developmental Biology 339: 418–428.

50. WallingfordJB, HarlandRM (2002) Neural tube closure requires Dishevelled-dependent convergent extension of the midline. Development 129: 5815–5825.

51. HuiC-c, JoynerAL (1993) A mouse model of Greig cephalo-polysyndactyly syndrome: the extra-toesJ mutation contains an intragenic deletion of the Gli3 gene. Nat Genet 3: 241–246.

52. HuangfuD, AndersonKV (2005) Cilia and Hedgehog responsiveness in the mouse. Proceedings of the National Academy of Sciences of the United States of America 102: 11325–11330.

53. WangB, FallonJF, BeachyPA (2000) Hedgehog-Regulated Processing of Gli3 Produces an Anterior/Posterior Repressor Gradient in the Developing Vertebrate Limb. Cell 100: 423–434.

54. MaySR, AshiqueAM, KarlenM, WangB, ShenY, et al. (2005) Loss of the retrograde motor for IFT disrupts localization of Smo to cilia and prevents the expression of both activator and repressor functions of Gli. Developmental Biology 287: 378–389.

55. HaycraftCJ, BanizsB, Aydin-SonY, ZhangQ, MichaudEJ, et al. (2005) Gli2 and Gli3 Localize to Cilia and Require the Intraflagellar Transport Protein Polaris for Processing and Function. PLoS Genet 1: e53.

56. LiuA, WangB, NiswanderLA (2005) Mouse intraflagellar transport proteins regulate both the activator and repressor functions of Gli transcription factors. Development 132: 3103–3111.

57. ShethR, BastidaMF, RosM (2007) Hoxd and Gli3 interactions modulate digit number in the amniote limb. Developmental Biology 310: 430–441.

58. HillP, GötzK, RütherU (2009) A SHH-independent regulation of Gli3 is a significant determinant of anteroposterior patterning of the limb bud. Developmental Biology 328: 506–516.

59. ChariteJ, McFaddenDG, OlsonEN (2000) The bHLH transcription factor dHAND controls Sonic hedgehog expression and establishment of the zone of polarizing activity during limb development. Development 127: 2461–2470.

60. O'RourkeMP, SooK, BehringerRR, HuiCC, TamPP (2002) Twist plays an essential role in FGF and SHH signal transduction during mouse limb development. Dev Biol 248: 143–156.

61. KrawchukD, WeinerSJ, ChenY-T, LuBC, CostantiniF, et al. (2010) Twist1 activity thresholds define multiple functions in limb development. Developmental Biology 347: 133–146.

62. SloughJ, CooneyL, BruecknerM (2008) Monocilia in the embryonic mouse heart suggest a direct role for cilia in cardiac morphogenesis. Developmental Dynamics 237: 2304–2314.

63. TanSY, RosenthalJ, ZhaoX-Q, FrancisRJ, ChatterjeeB, et al. (2007) Heterotaxy and complex structural heart defects in a mutant mouse model of primary ciliary dyskinesia. The Journal of Clinical Investigation 117: 3742–3752.

64. Ibañez-TallonI, HeintzN, OmranH (2003) To beat or not to beat: roles of cilia in development and disease. Human Molecular Genetics 12: R27–R35.

65. HoffmannAD, PetersonMA, Friedland-LittleJM, AndersonSA, MoskowitzIP (2009) sonic hedgehog is required in pulmonary endoderm for atrial septation. Development 136: 1761–1770.

66. KentWJ, SugnetCW, FureyTS, RoskinKM, PringleTH, et al. (2002) The human genome browser at UCSC. Genome Res 12: 996–1006.

67. FrankDU, FotheringhamLK, BrewerJA, MugliaLJ, Tristani-FirouziM, et al. (2002) An Fgf8 mouse mutant phenocopies human 22q11 deletion syndrome. Development 129: 4591–4603.

68. RamenskyV, BorkP, SunyaevS (2002) Human non-synonymous SNPs: server and survey. Nucleic Acids Res 30: 3894–3900.

69. BorodovskyM, McIninchJ (1993) Recognition of genes in DNA sequence with ambiguities. Biosystems 30: 161–171.

70. AdzhubeiIA, SchmidtS, PeshkinL, RamenskyVE, GerasimovaA, et al. (2010) A method and server for predicting damaging missense mutations. Nat Methods 7: 248–249.

71. WallinJ, WiltingJ, KosekiH, FritschR, ChristB, et al. (1994) The role of Pax-1 in axial skeleton development. Development 120: 1109–1121.

72. ManjiSS, MillerKA, WilliamsLH, AndreasenL, SiboeM, et al. (2011) An ENU-induced mutation of Cdh23 causes congenital hearing loss, but no vestibular dysfunction, in mice. Am J Pathol 179: 903–914.

73. FowlesLF, BennettsJS, BerkmanJL, WilliamsE, KoopmanP, et al. (2003) Genomic screen for genes involved in mammalian craniofacial development. Genesis 35: 73–87.

74. SherryST, WardMH, KholodovM, BakerJ, PhanL, et al. (2001) dbSNP: the NCBI database of genetic variation. Nucleic Acids Res 29: 308–311.

75. Genomes Project C (2010) A map of human genome variation from population-scale sequencing. Nature 467: 1061–1073.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 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#