Rapamycin reverses hypertrophic cardiomyopathy in a mouse model of LEOPARD syndrome-associated PTPN11 mutation.

LEOPARD syndrome (LS) is an autosomal dominant "RASopathy" that manifests with congenital heart disease. Nearly all cases of LS are caused by catalytically inactivating mutations in the protein tyrosine phosphatase (PTP), non-receptor type 11 (PTPN11) gene that encodes the SH2 domain-containing PTP-2 (SHP2). RASopathies typically affect components of the RAS/MAPK pathway, yet it remains unclear how PTPN11 mutations alter cellular signaling to produce LS phenotypes. We therefore generated knockin mice harboring the Ptpn11 mutation Y279C, one of the most common LS alleles. Ptpn11(Y279C/+) (LS/+) mice recapitulated the human disorder, with short stature, craniofacial dysmorphia, and morphologic, histologic, echocardiographic, and molecular evidence of hypertrophic cardiomyopathy (HCM). Heart and/or cardiomyocyte lysates from LS/+ mice showed enhanced binding of Shp2 to Irs1, decreased Shp2 catalytic activity, and abrogated agonist-evoked Erk/Mapk signaling. LS/+ mice also exhibited increased basal and agonist-induced Akt and mTor activity. The cardiac defects in LS/+ mice were completely reversed by treatment with rapamycin, an inhibitor of mTOR. Our results demonstrate that LS mutations have dominant-negative effects in vivo, identify enhanced mTOR activity as critical for causing LS-associated HCM, and suggest that TOR inhibitors be considered for treatment of HCM in LS patients.

[1]  B. Neel,et al.  MEK-ERK pathway modulation ameliorates disease phenotypes in a mouse model of Noonan syndrome associated with the Raf1(L613V) mutation. , 2011, The Journal of clinical investigation.

[2]  J. Seidman,et al.  Activation of multiple signaling pathways causes developmental defects in mice with a Noonan syndrome–associated Sos1 mutation. , 2010, The Journal of clinical investigation.

[3]  S. Colan Hypertrophic cardiomyopathy in childhood. , 2010, Heart failure clinics.

[4]  Hongbing Shen,et al.  A nonsynonymous polymorphism in IL23R gene is associated with risk of gastric cancer in a Chinese population , 2010, Molecular carcinogenesis.

[5]  F. Haj,et al.  Altered Glucose Homeostasis in Mice with Liver-specific Deletion of Src Homology Phosphatase 2* , 2010, The Journal of Biological Chemistry.

[6]  D. Horn,et al.  The face of Noonan syndrome: Does phenotype predict genotype , 2010, American journal of medical genetics. Part A.

[7]  E. Peng,et al.  Rapamycin regulates Akt and ERK phosphorylation through mTORC1 and mTORC2 signaling pathways , 2010, Molecular carcinogenesis.

[8]  A. Look,et al.  Phosphatase-dependent and -independent functions of Shp2 in neural crest cells underlie LEOPARD syndrome pathogenesis. , 2010, Developmental cell.

[9]  S. Lyonnet,et al.  Functional Effects of PTPN11 (SHP2) Mutations Causing LEOPARD Syndrome on Epidermal Growth Factor-Induced Phosphoinositide 3-Kinase/AKT/Glycogen Synthase Kinase 3β Signaling , 2010, Molecular and Cellular Biology.

[10]  Lei Yang,et al.  Patient-specific induced pluripotent stem cell derived models of LEOPARD syndrome , 2010, Nature.

[11]  G. Germino,et al.  Rapamycin ameliorates PKD resulting from conditional inactivation of Pkd1. , 2010, Journal of the American Society of Nephrology : JASN.

[12]  B. Gelb,et al.  Noonan Syndrome: Clinical Aspects and Molecular Pathogenesis , 2010, Molecular Syndromology.

[13]  K. Otsu,et al.  Cardiac fibroblasts are essential for the adaptive response of the murine heart to pressure overload. , 2010, The Journal of clinical investigation.

[14]  M. Zenker Genetic and Pathogenetic Aspects of Noonan Syndrome and Related Disorders , 2009, Hormone Research in Paediatrics.

[15]  E. Abel,et al.  Mammalian Target of Rapamycin Is a Critical Regulator of Cardiac Hypertrophy in Spontaneously Hypertensive Rats , 2009, Hypertension.

[16]  W. Lieberthal,et al.  The role of the mammalian target of rapamycin (mTOR) in renal disease. , 2009, Journal of the American Society of Nephrology : JASN.

[17]  J. Dancey,et al.  Evaluating temsirolimus activity in multiple tumors: a review of clinical trials. , 2009, Seminars in oncology.

[18]  R. Wüthrich,et al.  Mammalian target of rapamycin and autosomal dominant polycystic kidney disease. , 2009, Transplantation proceedings.

[19]  Adriaan P IJzerman,et al.  Rapamycin weekly maintenance dosing and the potential efficacy of combination sorafenib plus rapamycin but not atorvastatin or doxycycline in tuberous sclerosis preclinical models , 2009, BMC pharmacology.

[20]  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.

[21]  D. Srivastava,et al.  Cardiac fibroblasts regulate myocardial proliferation through beta1 integrin signaling. , 2009, Developmental cell.

[22]  W. Rottbauer,et al.  MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts , 2008, Nature.

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

[24]  K. Rauen,et al.  Noonan, Costello and cardio–facio–cutaneous syndromes: dysregulation of the Ras–MAPK pathway , 2008, Expert Reviews in Molecular Medicine.

[25]  A. Ralph-Edwards,et al.  Hypertrophic cardiomyopathy: current understanding and treatment objectives , 2008, Journal of Clinical Pathology.

[26]  Í. Lopes-Cendes,et al.  Shp2 Negatively Regulates Growth in Cardiomyocytes by Controlling Focal Adhesion Kinase/Src and mTOR Pathways , 2008, Circulation research.

[27]  Margit Rosner,et al.  The mTOR pathway and its role in human genetic diseases. , 2008, Mutation research.

[28]  M. Rubart,et al.  Cardiac Restricted Overexpression of Kinase-dead Mammalian Target of Rapamycin (mTOR) Mutant Impairs the mTOR-mediated Signaling and Cardiac Function* , 2008, Journal of Biological Chemistry.

[29]  N. Bodyak,et al.  Deletion of Ptpn11 (Shp2) in Cardiomyocytes Causes Dilated Cardiomyopathy via Effects on the Extracellular Signal–Regulated Kinase/Mitogen-Activated Protein Kinase and RhoA Signaling Pathways , 2008, Circulation.

[30]  B. Neel,et al.  The tyrosine phosphatase Shp2 (PTPN11) in cancer , 2008, Cancer and Metastasis Reviews.

[31]  J. den Hertog,et al.  Shp2 Knockdown and Noonan/LEOPARD Mutant Shp2–Induced Gastrulation Defects , 2007, PLoS genetics.

[32]  D. Weishaupt,et al.  Clinical proof-of-concept trial to assess the therapeutic effect of sirolimus in patients with autosomal dominant polycystic kidney disease: SUISSE ADPKD study , 2007, BMC nephrology.

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

[34]  S. Pikkarainen,et al.  Signaling pathways mediating cardiac myocyte gene expression in physiological and stress responses , 2007, Journal of cellular physiology.

[35]  B. Gelb,et al.  The genetics of congenital heart disease: a review of recent developments , 2007, Current opinion in cardiology.

[36]  K. Rauen HRAS and the Costello syndrome , 2007, Clinical genetics.

[37]  I. Shiojima,et al.  Microarray analysis of Akt1 activation in transgenic mouse hearts reveals transcript expression profiles associated with compensatory hypertrophy and failure. , 2006, Physiological genomics.

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

[39]  P. Syrris,et al.  Mutation screening of the PTPN11 gene in hypertrophic cardiomyopathy. , 2006, European journal of medical genetics.

[40]  A. Dart,et al.  Inhibition of mTOR reduces chronic pressure-overload cardiac hypertrophy and fibrosis , 2006, Journal of hypertension.

[41]  M. Vidaud,et al.  Reduced phosphatase activity of SHP‐2 in LEOPARD syndrome: Consequences for PI3K binding on Gab1 , 2006, FEBS letters.

[42]  B. Dallapiccola,et al.  LEOPARD syndrome: Clinical diagnosis in the first year of life , 2006, American journal of medical genetics. Part A.

[43]  Stephan Rohr,et al.  Electrotonic Modulation of Cardiac Impulse Conduction by Myofibroblasts , 2006, Circulation research.

[44]  D. Barford,et al.  PTPN11 (Shp2) Mutations in LEOPARD Syndrome Have Dominant Negative, Not Activating, Effects* , 2006, Journal of Biological Chemistry.

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

[46]  R. Foà,et al.  Diversity and functional consequences of germline and somatic PTPN11 mutations in human disease. , 2006, American journal of human genetics.

[47]  Malcolm McGregor,et al.  Diverse Biochemical Properties of Shp2 Mutants , 2005, Journal of Biological Chemistry.

[48]  Bruce D Gelb,et al.  Noonan syndrome and related disorders: genetics and pathogenesis. , 2005, Annual review of genomics and human genetics.

[49]  T. Ogata,et al.  PTPN11 mutations and genotype-phenotype correlations in Noonan and LEOPARD syndromes. , 2005, Pediatric endocrinology reviews : PER.

[50]  M. Digilio,et al.  Congenital intrahepatic portosystemic venous shunt: An unusual feature in LEOPARD syndrome and in neurofibromatosis type 1 , 2005, American journal of medical genetics. Part A.

[51]  H. Ohashi,et al.  Functional analysis of PTPN11/SHP-2 mutants identified in Noonan syndrome and childhood leukemia , 2005, Journal of Human Genetics.

[52]  G. Dorn,et al.  Protein kinase cascades in the regulation of cardiac hypertrophy. , 2005, The Journal of clinical investigation.

[53]  I. Lax,et al.  The docking protein Gab1 is the primary mediator of EGF-stimulated activation of the PI-3K/Akt cell survival pathway , 2004, BMC Biology.

[54]  M. Loh,et al.  Functional analysis of leukemia-associated PTPN11 mutations in primary hematopoietic cells. , 2004, Blood.

[55]  T. Ogata,et al.  Two novel and one recurrent PTPN11 mutations in LEOPARD syndrome , 2004, American journal of medical genetics. Part A.

[56]  C. Parent Faculty Opinions recommendation of PI3Kgamma modulates the cardiac response to chronic pressure overload by distinct kinase-dependent and -independent effects. , 2004 .

[57]  L. Silengo,et al.  PI3Kγ Modulates the Cardiac Response to Chronic Pressure Overload by Distinct Kinase-Dependent and -Independent Effects , 2004, Cell.

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

[59]  B. Gelb,et al.  Genetic evidence for lineage-related and differentiation stage-related contribution of somatic PTPN11 mutations to leukemogenesis in childhood acute leukemia. , 2004, Blood.

[60]  M. Loh,et al.  Mutations in PTPN11 implicate the SHP-2 phosphatase in leukemogenesis. , 2004, Blood.

[61]  Jie Wu,et al.  Noonan syndrome–associated SHP2/PTPN11 mutants cause EGF‐dependent prolonged GAB1 binding and sustained ERK2/MAPK1 activation , 2004, Human mutation.

[62]  P. Kang,et al.  Phosphoinositide 3-kinase(p110α) plays a critical role for the induction of physiological, but not pathological, cardiac hypertrophy , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[63]  D. Seripa,et al.  Correlation between PTPN11 gene mutations and congenital heart defects in Noonan and LEOPARD syndromes , 2003, Journal of medical genetics.

[64]  B. Neel,et al.  The 'Shp'ing news: SH2 domain-containing tyrosine phosphatases in cell signaling. , 2003, Trends in biochemical sciences.

[65]  W. Manning,et al.  Rapamycin Attenuates Load-Induced Cardiac Hypertrophy in Mice , 2003, Circulation.

[66]  N. Copeland,et al.  A highly efficient recombineering-based method for generating conditional knockout mutations. , 2003, Genome research.

[67]  C. D. dos Remedios,et al.  Activation of signal transducer and activator of transcription (STAT) pathways in failing human hearts. , 2003, Cardiovascular research.

[68]  C. Proud,et al.  Ras/Erk Signaling Is Essential for Activation of Protein Synthesis by Gq Protein-Coupled Receptor Agonists in Adult Cardiomyocytes , 2002, Circulation research.

[69]  Mohit M. Jain,et al.  Cardiac-Specific Overexpression of GLUT1 Prevents the Development of Heart Failure Attributable to Pressure Overload in Mice , 2002, Circulation.

[70]  C. Kahn,et al.  Regulation of Myocardial Contractility and Cell Size by Distinct PI3K-PTEN Signaling Pathways , 2002, Cell.

[71]  J. Fryns,et al.  PTPN11 mutations in LEOPARD syndrome , 2002, Journal of medical genetics.

[72]  M. Digilio,et al.  Grouping of multiple-lentigines/LEOPARD and Noonan syndromes on the PTPN11 gene. , 2002, American journal of human genetics.

[73]  B. Neel,et al.  Receptor-Specific Regulation of Phosphatidylinositol 3′-Kinase Activation by the Protein Tyrosine Phosphatase Shp2 , 2002, Molecular and Cellular Biology.

[74]  Bruce D Gelb,et al.  PTPN11 mutations in Noonan syndrome: molecular spectrum, genotype-phenotype correlation, and phenotypic heterogeneity. , 2002, American journal of human genetics.

[75]  P. Kang,et al.  Akt/Protein Kinase B Promotes Organ Growth in Transgenic Mice , 2002, Molecular and Cellular Biology.

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

[77]  Qiang Wang,et al.  The tyrosine phosphatase SHP-2 is required for mediating phosphatidylinositol 3-kinase/Akt activation by growth factors , 2001, Oncogene.

[78]  B. Neel,et al.  Combinatorial control of the specificity of protein tyrosine phosphatases. , 2001, Current opinion in cell biology.

[79]  Y. Hakak,et al.  Shp-2 mediates v-Src-induced morphological changes and activation of the anti-apoptotic protein kinase Akt , 2000, Oncogene.

[80]  P. Kang,et al.  The conserved phosphoinositide 3‐kinase pathway determines heart size in mice , 2000, The EMBO journal.

[81]  C. Proud,et al.  Activation of mRNA translation in rat cardiac myocytes by insulin involves multiple rapamycin-sensitive steps. , 2000, American journal of physiology. Heart and circulatory physiology.

[82]  R. Kitsis,et al.  Akt promotes survival of cardiomyocytes in vitro and protects against ischemia-reperfusion injury in mouse heart. , 2000, Circulation.

[83]  B. Marino,et al.  Congenital heart diseases in children with Noonan syndrome: An expanded cardiac spectrum with high prevalence of atrioventricular canal. , 1999, The Journal of pediatrics.

[84]  G. Feng Shp-2 tyrosine phosphatase: signaling one cell or many. , 1999, Experimental cell research.

[85]  J. Alexandre,et al.  La rapamycine et le CCI-779 , 1999 .

[86]  S. Shamsadini,et al.  Leopard syndrome , 1999, The Lancet.

[87]  M. Digilio,et al.  Noonan syndrome and aortic coarctation. , 1998, American journal of medical genetics.

[88]  J. Ross,et al.  Distinct molecular phenotypes in murine cardiac muscle development, growth, and hypertrophy. , 1998, Journal of molecular and cellular cardiology.

[89]  H Niimura,et al.  Mutations in the gene for cardiac myosin-binding protein C and late-onset familial hypertrophic cardiomyopathy. , 1998, The New England journal of medicine.

[90]  D. Barford,et al.  Revealing mechanisms for SH2 domain mediated regulation of the protein tyrosine phosphatase SHP-2. , 1998, Structure.

[91]  Jiahuai Han,et al.  Cardiac Hypertrophy Induced by Mitogen-activated Protein Kinase Kinase 7, a Specific Activator for c-Jun NH2-terminal Kinase in Ventricular Muscle Cells* , 1998, The Journal of Biological Chemistry.

[92]  S. Shoelson,et al.  Crystal Structure of the Tyrosine Phosphatase SHP-2 , 1998, Cell.

[93]  B. Neel,et al.  Structural Determinants of SHP-2 Function and Specificity in Xenopus Mesoderm Induction , 1998, Molecular and Cellular Biology.

[94]  E. Lakatta,et al.  Rapamycin inhibits alpha 1-adrenergic receptor-stimulated cardiac myocyte hypertrophy but not activation of hypertrophy-associated genes. Evidence for involvement of p70 S6 kinase. , 1997, Circulation research.

[95]  B. A. French,et al.  Gene recombination in postmitotic cells. Targeted expression of Cre recombinase provokes cardiac-restricted, site-specific rearrangement in adult ventricular muscle in vivo. , 1997, The Journal of clinical investigation.

[96]  M. C. Johnson,et al.  The genetic basis of pediatric cardiovascular disease. , 1996, Seminars in perinatology.

[97]  T. Nishikawa,et al.  Hypertrophic cardiomyopathy in Noonan syndrome , 1996, Acta paediatrica Japonica : Overseas edition.

[98]  J. Sadoshima,et al.  Rapamycin selectively inhibits angiotensin II-induced increase in protein synthesis in cardiac myocytes in vitro. Potential role of 70-kD S6 kinase in angiotensin II-induced cardiac hypertrophy. , 1995, Circulation research.

[99]  K. Nakao,et al.  Rapid transcriptional activation and early mRNA turnover of brain natriuretic peptide in cardiocyte hypertrophy. Evidence for brain natriuretic peptide as an "emergency" cardiac hormone against ventricular overload. , 1995, The Journal of clinical investigation.

[100]  A. Marian,et al.  Recent advances in the molecular genetics of hypertrophic cardiomyopathy. , 1995, Circulation.

[101]  R R Markwald,et al.  Molecular regulation of atrioventricular valvuloseptal morphogenesis. , 1995, Circulation research.

[102]  M. Komajda,et al.  Molecular basis of familial cardiomyopathies. , 1995, Circulation.

[103]  J. G. García García,et al.  [Noonan's syndrome and aortic coarctation]. , 1994, Anales de medicina interna.

[104]  D. Rees,et al.  Three-dimensional structures of acidic and basic fibroblast growth factors. , 1993, Science.

[105]  M. Patton,et al.  Cardiologic Abnormalities in Noonan Syndrome: Phenotypic Diagnosis and Echocardiographic Assessment of 118 Patients , 1993, Journal of the American College of Cardiology.

[106]  T. Pawson,et al.  SH2-containing phosphotyrosine phosphatase as a target of protein-tyrosine kinases. , 1993, Science.

[107]  J. Moller,et al.  The leopard (multiple lentigines) syndrome revisited , 1971, Birth defects original article series.

[108]  R. Schrier,et al.  Potential Pharmacological Interventions in Polycystic Kidney Disease , 2012, Drugs.

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

[110]  B. Gelb,et al.  Phosphatase-defective LEOPARD syndrome mutations in PTPN11 gene have gain-of-function effects during Drosophila development. , 2009, Human molecular genetics.

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

[112]  W. Dassen,et al.  Cardiac remodelling: concentric versus eccentric hypertrophy in strength and endurance athletes , 2008, Netherlands heart journal : monthly journal of the Netherlands Society of Cardiology and the Netherlands Heart Foundation.

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

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

[115]  D. Sabatini,et al.  Raptor and mTOR: subunits of a nutrient-sensitive complex. , 2004, Current topics in microbiology and immunology.

[116]  J. Terwilliger,et al.  Roles of growth hormone and insulin-like growth factor 1 in mouse postnatal growth. , 2001, Developmental biology.

[117]  E. Raymond,et al.  [Rapamycin and CCI-779]. , 1999, Bulletin du cancer.

[118]  S. Solomon,et al.  Prognostic implications of novel beta cardiac myosin heavy chain gene mutations that cause familial hypertrophic cardiomyopathy. , 1994, The Journal of clinical investigation.