Activation of multiple signaling pathways causes developmental defects in mice with a Noonan syndrome–associated Sos1 mutation.

Noonan syndrome (NS) is an autosomal dominant genetic disorder characterized by short stature, unique facial features, and congenital heart disease. About 10%-15% of individuals with NS have mutations in son of sevenless 1 (SOS1), which encodes a RAS and RAC guanine nucleotide exchange factor (GEF). To understand the role of SOS1 in the pathogenesis of NS, we generated mice with the NS-associated Sos1E846K gain-of-function mutation. Both heterozygous and homozygous mutant mice showed many NS-associated pheno-types, including growth delay, distinctive facial dysmorphia, hematologic abnormalities, and cardiac defects. We found that the Ras/MAPK pathway as well as Rac and Stat3 were activated in the mutant hearts. These data provide in vivo molecular and cellular evidence that Sos1 is a GEF for Rac under physiological conditions and suggest that Rac and Stat3 activation might contribute to NS phenotypes. Furthermore, prenatal administration of a MEK inhibitor ameliorated the embryonic lethality, cardiac defects, and NS features of the homozygous mutant mice, demonstrating that this signaling pathway might represent a promising therapeutic target for NS.

[1]  Randhir Singh,et al.  Molecular targets and regulators of cardiac hypertrophy. , 2010, Pharmacological research.

[2]  M. Geletu,et al.  Activated Rac1 requires gp130 for Stat3 activation, cell proliferation and migration. , 2010, Experimental cell research.

[3]  J. Molkentin,et al.  Extracellular signal‐regulated kinase 1/2 (ERK1/2) signaling in cardiac hypertrophy , 2010, Annals of the New York Academy of Sciences.

[4]  Rony Seger,et al.  The subcellular localization of MEK and ERK—A novel nuclear translocation signal (NTS) paves a way to the nucleus , 2010, Molecular and Cellular Endocrinology.

[5]  H. Hasle Malignant Diseases in Noonan Syndrome and Related Disorders , 2009, Hormone Research in Paediatrics.

[6]  J. Schmitt,et al.  Cardiac hypertrophy: targeting Raf/MEK/ERK1/2-signaling. , 2009, The international journal of biochemistry & cell biology.

[7]  Tomoki Nakamura,et al.  Noonan syndrome is associated with enhanced pERK activity, the repression of which can prevent craniofacial malformations , 2009, Proceedings of the National Academy of Sciences.

[8]  Ravi Iyengar,et al.  Mutation in SHOC2 promotes aberrant protein N-myristoylation and underlies Noonan-like syndrome with loose anagen hair , 2009, Nature Genetics.

[9]  Tomoki Nakamura,et al.  Protein tyrosine phosphatase activity in the neural crest is essential for normal heart and skull development , 2009, Proceedings of the National Academy of Sciences.

[10]  Xin-Yuan Fu,et al.  Negative Regulation of Stat3 by Activating PTPN11 Mutants Contributes to the Pathogenesis of Noonan Syndrome and Juvenile Myelomonocytic Leukemia* , 2009, The Journal of Biological Chemistry.

[11]  K. Rauen,et al.  The RASopathies: developmental syndromes of Ras/MAPK pathway dysregulation. , 2009, Current opinion in genetics & development.

[12]  Susan A. Elmore,et al.  Histology Atlas of the Developing Mouse Heart with Emphasis on E11.5 to E18.5 , 2009, Toxicologic pathology.

[13]  B. Neel,et al.  Noonan syndrome cardiac defects are caused by PTPN11 acting in endocardium to enhance endocardial-mesenchymal transformation , 2009, Proceedings of the National Academy of Sciences.

[14]  J. Molkentin,et al.  Role of ERK1/2 signaling in congenital valve malformations in Noonan syndrome , 2008, Proceedings of the National Academy of Sciences.

[15]  Anthony J. Muslin,et al.  MAPK signalling in cardiovascular health and disease: molecular mechanisms and therapeutic targets. , 2008, Clinical science.

[16]  T. Zimmers,et al.  Body surface area prediction in normal, hypermuscular, and obese mice. , 2008, The Journal of surgical research.

[17]  K. Kuo,et al.  Angiotensin II Activates Signal Transducer and Activators of Transcription 3 via Rac1 in Atrial Myocytes and Fibroblasts: Implication for the Therapeutic Effect of Statin in Atrial Structural Remodeling , 2008, Circulation.

[18]  P. Cohen,et al.  The selectivity of protein kinase inhibitors: a further update. , 2007, The Biochemical journal.

[19]  G. Dorn,et al.  Mediating ERK 1/2 signaling rescues congenital heart defects in a mouse model of Noonan syndrome. , 2007, The Journal of clinical investigation.

[20]  Michael J Ackerman,et al.  Gain-of-function RAF1 mutations cause Noonan and LEOPARD syndromes with hypertrophic cardiomyopathy , 2007, Nature Genetics.

[21]  Suzanne Schubbert,et al.  Hyperactive Ras in developmental disorders and cancer , 2007, Nature Reviews Cancer.

[22]  Kenichi Nomoto,et al.  Recent advances of MEK inhibitors and their clinical progress. , 2007, Current topics in medicinal chemistry.

[23]  D. Hilfiker-Kleiner,et al.  Survival pathways in hypertrophy and heart failure: The gp130-STAT3 axis , 2007, Basic Research in Cardiology.

[24]  J. Settleman,et al.  GEFs in growth factor signaling , 2007, Growth factors.

[25]  Wendy Schackwitz,et al.  Gain-of-function SOS1 mutations cause a distinctive form of Noonan syndrome , 2006, Nature Genetics.

[26]  D. Kwiatkowski,et al.  Requirement of Rac1 in the development of cardiac hypertrophy. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[27]  Kam Y. J. Zhang,et al.  Germline KRAS mutations cause Noonan syndrome , 2006, Nature Genetics.

[28]  J. Lyons,et al.  Recent progress in targeting the Raf/MEK/ERK pathway with inhibitors in cancer drug discovery. , 2005, Current opinion in pharmacology.

[29]  F. McCormick,et al.  Killing time for cancer cells , 2005, Nature Reviews Cancer.

[30]  C. Der,et al.  GEF means go: turning on RHO GTPases with guanine nucleotide-exchange factors , 2005, Nature Reviews Molecular Cell Biology.

[31]  M. Kotlikoff,et al.  Gene trap and gene inversion methods for conditional gene inactivation in the mouse , 2005, Nucleic acids research.

[32]  J. Sebolt-Leopold,et al.  Targeting the mitogen-activated protein kinase cascade to treat cancer , 2004, Nature Reviews Cancer.

[33]  D. Gilliland,et al.  Mouse model of Noonan syndrome reveals cell type– and gene dosage–dependent effects of Ptpn11 mutation , 2004, Nature Medicine.

[34]  Hua Yu,et al.  The STATs of cancer — new molecular targets come of age , 2004, Nature Reviews Cancer.

[35]  P. Goldschmidt-Clermont,et al.  Rac regulates cardiovascular superoxide through diverse molecular interactions: more than a binary GTP switch. , 2003, American journal of physiology. Cell physiology.

[36]  M. Hori,et al.  The Small GTP-binding Protein Rac1 Induces Cardiac Myocyte Hypertrophy through the Activation of Apoptosis Signal-regulating Kinase 1 and Nuclear Factor-κB* , 2003, Journal of Biological Chemistry.

[37]  S. Meloche,et al.  Rho Family GTPases Are Required for Activation of Jak/STAT Signaling by G Protein-Coupled Receptors , 2003, Molecular and Cellular Biology.

[38]  Jian-Mei Li,et al.  Fluvastatin Enhances the Inhibitory Effects of a Selective Angiotensin II Type 1 Receptor Blocker, Valsartan, on Vascular Neointimal Formation , 2003, Circulation.

[39]  G. Booz,et al.  Interplay between the cardiac renin angiotensin system and JAK-STAT signaling: role in cardiac hypertrophy, ischemia/reperfusion dysfunction, and heart failure. , 2002, Journal of molecular and cellular cardiology.

[40]  D. Bar-Sagi,et al.  The Two Hats of SOS , 2002, Science's STKE.

[41]  Giorgio Scita,et al.  Mechanisms through which Sos-1 coordinates the activation of Ras and Rac , 2002, The Journal of cell biology.

[42]  Michael A. Patton,et al.  Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome , 2001, Nature Genetics.

[43]  D. Bar-Sagi,et al.  Rac1 mediates STAT3 activation by autocrine IL-6 , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[44]  D. Bar-Sagi,et al.  Ras and Rho GTPases A Family Reunion , 2000, Cell.

[45]  H. Vikis,et al.  Regulation of STAT3 by direct binding to the Rac1 GTPase. , 2000, Science.

[46]  M. Siddiqui,et al.  The role of Jak/STAT signaling in heart tissue renin-angiotensin system , 2000, Molecular and Cellular Biochemistry.

[47]  G. Scita,et al.  Signaling from Ras to Rac and beyond: not just a matter of GEFs , 2000, The EMBO journal.

[48]  A. Clerk,et al.  Small guanine nucleotide-binding proteins and myocardial hypertrophy. , 2000, Circulation research.

[49]  T. Hewett,et al.  Altered focal adhesion regulation correlates with cardiomyopathy in mice expressing constitutively active rac1. , 2000, The Journal of clinical investigation.

[50]  H. Rossbach,et al.  Duchenne muscular dystrophy and concomitant metastatic alveolar rhabdomyosarcoma. , 1999, Journal of pediatric hematology/oncology.

[51]  E. Mantadakis,et al.  Prevalence of priapism in children and adolescents with sickle cell anemia. , 1999, Journal of pediatric hematology/oncology.

[52]  K. Tanaka,et al.  A requirement for the rac1 GTPase in the signal transduction pathway leading to cardiac myocyte hypertrophy. , 1998, The Journal of clinical investigation.

[53]  S. Ogawa,et al.  Biphasic activation of the JAK/STAT pathway by angiotensin II in rat cardiomyocytes. , 1998, Circulation research.

[54]  D. Bar-Sagi,et al.  Coupling of Ras and Rac guanosine triphosphatases through the Ras exchanger Sos. , 1998, Science.

[55]  S. Ogawa,et al.  Role of angiotensin II in activation of the JAK/STAT pathway induced by acute pressure overload in the rat heart. , 1997, Circulation research.

[56]  F. Alt,et al.  Efficient in vivo manipulation of mouse genomic sequences at the zygote stage. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[57]  B. Berk,et al.  Direct stimulation of Jak/STAT pathway by the angiotensin II AT1 receptor , 1995, Nature.

[58]  D. Horn,et al.  A restricted spectrum of NRAS mutations causes Noonan syndrome , 2010, Nature Genetics.

[59]  Ravi Iyengar,et al.  Mutation of SHOC2 promotes aberrant protein N-myristoylation and causes Noonan-like syndrome with loose anagen hair , 2009 .

[60]  J. Sebolt-Leopold,et al.  Targeting MEK is effective chemoprevention of hepatocellular carcinoma in TGF-alpha-transgenic mice. , 2008, Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract.

[61]  Li Li,et al.  Germline gain-of-function mutations in SOS1 cause Noonan syndrome , 2007, Nature Genetics.

[62]  J. Sebolt-Leopold,et al.  Targeting MEK is Effective Chemoprevention of Hepatocellular Carcinoma in TGF-α-Transgenic Mice , 2007, Journal of Gastrointestinal Surgery.

[63]  Michiko Furutani,et al.  Germline gain-of-function mutations in RAF1 cause Noonan syndrome , 2007, Nature Genetics.

[64]  D. Hilfiker-Kleiner,et al.  Survival pathways in hypertrophy and heart failure: The gp130-STAT3 axis , 2007, Basic Research in Cardiology.

[65]  A. Brice,et al.  Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome , 2002, Nature Genetics.

[66]  M. Arıcı,et al.  A unifying hypothesis for the renin-angiotensin system and hematopoiesis: sticking the pieces together with the JAK-STAT pathway. , 2000, Medical hypotheses.

[67]  J B Gibbs,et al.  Anticancer drug targets: growth factors and growth factor signaling. , 2000, The Journal of clinical investigation.

[68]  D. Chitayat,et al.  Juvenile myelomonocytic leukemia and Noonan syndrome. , 1999, Journal of pediatric hematology/oncology.

[69]  G. J. Bhat,et al.  Cross-talk between angiotensin II and interleukin-6-induced signaling through Stat3 transcription factor , 1998, Basic Research in Cardiology.