1,326
Views
11
CrossRef citations to date
0
Altmetric
Addendum

The impact of the genetic background in the Noonan syndrome phenotype induced by K-RasV14I

, , &
Article: e1045169 | Received 14 Jan 2015, Accepted 21 Apr 2015, Published online: 22 May 2015

References

  • Rauen KA. The RASopathies. Annu Rev Genomics Hum Genet 2013; 14:355-69; PMID:23875798; http://dx.doi.org/10.1146/annurev-genom-091212-153523.
  • Roberts AE, Allanson JE, Tartaglia M, Gelb BD. Noonan syndrome. Lancet 2013; 381:333-42; PMID:23312968; http://dx.doi.org/10.1016/S0140-6736(12)61023-X
  • Aoki Y, Niihori T, Banjo T, Okamoto N, Mizuno S, Kurosawa K, Otaga T, Takada F, Yano M, Ando T, et al. Gain-of-function mutations in RIT1 cause Noonan syndrome, a RAS/MAPK pathway syndrome. Am J Hum Genet 2013; 93:173-80; PMID:23791108; http://dx.doi.org/10.1016/j.ajhg.2013.05.021
  • Flex E, Jaiswal M, Pantaleoni F, Martinelli S, Strullu M, Fansa EK, Caye A, De Luca A, Lepri F, Dvorsky R, et al. Activating mutations in RRAS underlie a phenotype within the RASopathy spectrum and contribute to leukaemogenesis. Hum Mol Genet 2014; 23:4315-27; PMID:24705357; http://dx.doi.org/10.1093/hmg/ddu148
  • Tartaglia M, Gelb BD, Zenker M. Noonan syndrome and clinically related disorders. Best Pract Res Clin Endocrinol Metab 2011; 25:161-79; PMID:21396583; http://dx.doi.org/10.1016/j.beem.2010.09.002
  • Schubbert S, Shannon K, Bollag G. Hyperactive Ras in developmental disorders and cancer. Nat Rev Cancer 2007; 7:295-308; PMID:17384584; http://dx.doi.org/10.1038/nrc2109
  • Tartaglia M, Kalidas K, Shaw A, Song X, Musat DL, van der Burgt I, Brunner HG, Bertola DR, Crosby A, Ion A, et al. PTPN11 mutations in Noonan syndrome: molecular spectrum, genotype-phenotype correlation, and phenotypic heterogeneity. Am J Hum Genet 2002; 70:1555-63; PMID:11992261; http://dx.doi.org/10.1086/340847
  • Zenker M, Horn D, Wieczorek D, Allanson J, Pauli S, van der Burgt I, Doerr HG, Gaspar H, Hofbeck M, Gillessen-Kaesbach G, et al. SOS1 is the second most common Noonan gene but plays no major role in cardio-facio-cutaneous syndrome. J Med Genet 2007; 44:651-6; PMID:17586837; http://dx.doi.org/10.1136/jmg.2007.051276
  • Tartaglia M, Gelb BD. Disorders of dysregulated signal traffic through the RAS-MAPK pathway: phenotypic spectrum and molecular mechanisms. Ann N Y Acad Sci 2010; 1214:99-121; PMID:20958325; http://dx.doi.org/10.1111/j.1749-6632.2010.05790.x
  • Sarkozy A, Carta C, Moretti S, Zampino G, Digilio MC, Pantaleoni F, Scioletti AP, Esposito G, Cordeddu V, Lepri F, et al. Germline BRAF mutations in Noonan, LEOPARD, and cardiofaciocutaneous syndromes: molecular diversity and associated phenotypic spectrum. Hum Mutat 2009; 30:695-702; PMID:19206169; http://dx.doi.org/10.1002/humu.20955
  • Lee BH, Kim JM, Jin HY, Kim GH, Choi JH, Yoo HW. Spectrum of mutations in Noonan syndrome and their correlation with phenotypes. J Pediatr 2011; 159:1029-35; PMID:21784453; http://dx.doi.org/10.1016/j.jpeds.2011.05.024
  • Bertola DR, Pereira AC, de Oliveira PS, Kim CA, Krieger JE. Clinical variability in a Noonan syndrome family with a new PTPN11 gene mutation. Am J Med Genet A 2004; 130A:378-83; PMID:15384080; http://dx.doi.org/10.1002/ajmg.a.30270
  • Zenker M, Voss E, Reis A. Mild variable Noonan syndrome in a family with a novel PTPN11 mutation. Eur J Med Genet 2007; 50:43-7; PMID:17052965; http://dx.doi.org/10.1016/j.ejmg.2006.08.003
  • Bentires-Alj M, Kontaridis MI, Neel BG. Stops along the RAS pathway in human genetic disease. Nat Med 2006; 12:283-5; PMID:16520774; http://dx.doi.org/10.1038/nm0306-283
  • Araki T, Chan G, Newbigging S, Morikawa L, Bronson RT, Neel BG. Noonan syndrome cardiac defects are caused by PTPN11 acting in endocardium to enhance endocardial-mesenchymal transformation. Proc Natl Acad Sci U S A 2009; 106:4736-41; PMID:19251646; http://dx.doi.org/10.1073/pnas.0810053106
  • Wu X, Simpson J, Hong JH, Kim KH, Thavarajah NK, Backx PH, Neel BG, Araki T. MEK-ERK pathway modulation ameliorates disease phenotypes in a mouse model of Noonan syndrome associated with the Raf1(L613V) mutation. J Clin Invest 2011; 121:1009-25; PMID:21339642; http://dx.doi.org/10.1172/JCI44929
  • Wu X, Yin J, Simpson J, Kim KH, Gu S, Hong JH, Bayliss P, Backx PH, Neel BG, Araki T. Increased BRAF heterodimerization is the common pathogenic mechanism for noonan syndrome-associated RAF1 mutants. Mol Cell Biol 2012; 32: 3872-90; PMID:22826437; http://dx.doi.org/10.1128/MCB.00751-12
  • Pritchard CA, Bolin L, Slattery R, Murray R, McMahon M. Post-natal lethality and neurological and gastrointestinal defects in mice with targeted disruption of the A-Raf protein kinase gene Curr Biol 1996; 6: 614-7; PMID:8805280; http://dx.doi.org/10.1016/S0960-9822(02)00548-1
  • Hernandez-Porras I, Fabbiano S, Schuhmacher AJ, Aicher A, Canamero M, Camara JA, Cusso L, Desco M, Heeschen C, Mulero F, et al. K-RasV14I recapitulates Noonan syndrome in mice Proc Natl Acad Sci U S A 2014; 111:16395-16400; PMID:25359213; http://dx.doi.org/10.1073/pnas.1418126111
  • Chen PC, Wakimoto H, Conner D, Araki T, Yuan T, Roberts A, Seidman CE, Bronson R, Neel BG, Seidman JG et al. Activation of multiple signaling pathways causes developmental defects in mice with a Noonan syndrome-associated Sos1 mutation. J Clin Invest 2010; 120: 4353-65; PMID:21041952; http://dx.doi.org/10.1172/JCI43910
  • Tidyman WE, Rauen KA. Noonan, Costello and cardio-facio-cutaneous syndromes: dysregulation of the Ras-MAPK pathway. Expert Rev Mol Med 2008; 10:e37; PMID:19063751; http://dx.doi.org/10.1017/S1462399408000902
  • Friedman JM, Gutmann DH, MacCollin M, Riccardi VM. Neurofibromatosis: phenotype, natural history, and pathogenesis. Taylor & Francis, Baltimore 1999.
  • Easton DF, Ponder MA, Huson SM, Ponder BA. An analysis of variation in expression of neurofibromatosis (NF) type 1 (NF1): evidence for modifying genes. Am J Hum Genet 1993; 53:305-13; PMID:8328449
  • Szudek J, Joe H, Friedman JM. Analysis of intrafamilial phenotypic variation in neurofibromatosis 1 (NF1). Genet Epidemiol 2002; 23:150-64; PMID:12214308; http://dx.doi.org/10.1002/gepi.1129
  • Sabbagh A, Pasmant E, Laurendeau I, Parfait B, Barbarot S, Guillot B, Combemale P, Ferkal S, Vidaud M, Aubourg P, et al. Unravelling the genetic basis of variable clinical expression in neurofibromatosis 1. Hum Mol Genet 2009; 18:2768-78; PMID:19417008; http://dx.doi.org/10.1093/hmg/ddp212
  • Pemov A, Sung H, Hyland PL, Sloan JL, Ruppert SL, Baldwin AM, Boland JF, Bass SE, Lee HJ, Jones KM, et al. Genetic Modifiers of Neurofibromatosis Type 1-Associated Cafe-au-Lait Macule Count Identified Using Multi-platform Analysis. PLoS Genet 2014; 10:e1004575; PMID:25329635; http://dx.doi.org/10.1371/journal.pgen.1004575
  • Reilly KM, Broman KW, Bronson RT, Tsang S, Loisel DA, Christy ES, Sun Z, Diehl J, Munroe DJ, Tuskan RG. An imprinted locus epistatically influences Nstr1 and Nstr2 to control resistance to nerve sheath tumors in a neurofibromatosis type 1 mouse model. Cancer Res 2006; 66:62-8; PMID:16397217; http://dx.doi.org/10.1158/0008-5472.CAN-05-1480
  • Walrath JC, Fox K, Truffer E, Gregory Alvord W, Quinones OA, Reilly KM. Chr 19(A/J) modifies tumor resistance in a sex- and parent-of-origin-specific manner. Mamm Genome 2009; 20:214-23; PMID:19347398; http://dx.doi.org/10.1007/s00335-009-9179-4
  • Lin AE, Alexander ME, Colan SD, Kerr B, Rauen KA, Noonan J, Baffa J, Hopkins E, Sol-Church K, Limongelli G, et al. Clinical, pathological, and molecular analyses of cardiovascular abnormalities in Costello syndrome: a Ras/MAPK pathway syndrome. Am J Med Genet A 2011; 155A:486-507; PMID:21344638; http://dx.doi.org/10.1002/ajmg.a.33857
  • Baldassarre G, Mussa A, Banaudi E, Rossi C, Tartaglia M, Silengo M, Ferrero GB. Phenotypic variability associated with the invariant SHOC2 c.4A>G (p.Ser2Gly) missense mutation. Am J Med Genet A 2014; 164A:3120-5; PMID:25331583; http://dx.doi.org/10.1002/ajmg.a.36697
  • Jongmans M, Sistermans EA, Rikken A, Nillesen WM, Tamminga R, Patton M, Maier EM, Tartaglia M, Noordam K, van der Burgt I. Genotypic and phenotypic characterization of Noonan syndrome: new data and review of the literature. Am J Med Genet A 2005; 134A:165-70; PMID:15723289; http://dx.doi.org/10.1002/ajmg.a.30598
  • Musante L, Kehl HG, Majewski F, Meinecke P, Schweiger S, Gillessen-Kaesbach G, Wieczorek D, Hinkel GK, Tinschert S, Hoeltzenbein M, et al. Spectrum of mutations in PTPN11 and genotype-phenotype correlation in 96 patients with Noonan syndrome and five patients with cardio-facio-cutaneous syndrome. Eur J Hum Genet 2003; 11:201-6; PMID:12634870; http://dx.doi.org/10.1038/sj.ejhg.5200935
  • Zenker M, Buheitel G, Rauch R, Koenig R, Bosse K, Kress W, Tietze HU, Doerr HG, Hofbeck M, Singer H, et al. Genotype-phenotype correlations in Noonan syndrome. J Pediatr 2004; 144:368-74; PMID:15001945; http://dx.doi.org/10.1016/j.jpeds.2003.11.032
  • Ezquieta B, Santome JL, Carcavilla A, Guillen-Navarro E, Perez-Aytes A, Sanchez del Pozo J, Garcia-Miñuar S, Castillo E, Alonso M, Vendrell T, et al. Alterations in RAS-MAPK genes in 200 Spanish patients with Noonan and other neuro-cardio-facio-cutaneous syndromes. Genotype and cardiopathy. Rev Esp Cardiol (Engl Ed) 2012; 65:447-55; PMID:22465605; http://dx.doi.org/10.1016/j.recesp.2011.12.016
  • Simpson EM, Linder CC, Sargent EE, Davisson MT, Mobraaten LE, Sharp JJ. Genetic variation among 129 substrains and its importance for targeted mutagenesis in mice. Nat Genet 1997; 16:19-27; PMID:9140391; http://dx.doi.org/10.1038/ng0597-19
  • Chen X, Mitsutake N, LaPerle K, Akeno N, Zanzonico P, Longo VA, Mitsutake S, Kimura ET, Geiger H, Santos E, et al. Endogenous expression of Hras(G12V) induces developmental defects and neoplasms with copy number imbalances of the oncogene. Proc Natl Acad Sci U S A 2009; 106:7979-84; PMID:19416908; http://dx.doi.org/10.1073/pnas.0900343106
  • Schuhmacher AJ, Guerra C, Sauzeau V, Canamero M, Bustelo XR, Barbacid M. A mouse model for Costello syndrome reveals an Ang II-mediated hypertensive condition. J Clin Invest 2008; 118:2169-79; PMID:18483625
  • Urosevic J, Sauzeau V, Soto-Montenegro ML, Reig S, Desco M, Wright EM, Cañamero M, Mulero F, Ortega S, Bustelo XR, et al. Constitutive activation of B-Raf in the mouse germline provides a model for human cardio-facio-cutaneous syndrome. Proc Natl Acad Sci U S A 2011; 108:5015-20; PMID:21383153; http://dx.doi.org/10.1073/pnas.1016933108