The RASopathy Family: Consequences of Germline Activation of the RAS/MAPK Pathway.

Noonan syndrome [NS; Mendelian Inheritance in Men (MIM) #163950] and related syndromes [Noonan syndrome with multiple lentigines (formerly called LEOPARD syndrome; MIM #151100), Noonan-like syndrome with loose anagen hair (MIM #607721), Costello syndrome (MIM #218040), cardio-facio-cutaneous syndrome (MIM #115150), type I neurofibromatosis (MIM #162200), and Legius syndrome (MIM #611431)] are a group of related genetic disorders associated with distinctive facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was clinically described more than 50 years ago, and disease genes have been identified throughout the last 3 decades, providing a molecular basis to better understand their physiopathology and identify targets for therapeutic strategies. Most of these genes encode proteins belonging to or regulating the so-called RAS/MAPK signaling pathway, so these syndromes have been gathered under the name RASopathies. In this review, we provide a clinical overview of RASopathies and an update on their genetics. We then focus on the functional and pathophysiological effects of RASopathy-causing mutations and discuss therapeutic perspectives and future directions.

[1]  B. Neel,et al.  Noonan syndrome-causing SHP2 mutants impair ERK-dependent chondrocyte differentiation during endochondral bone growth , 2018, Human molecular genetics.

[2]  B. Taylor,et al.  Allele-Specific Mechanisms of Activation of MEK1 Mutants Determine Their Properties. , 2018, Cancer discovery.

[3]  B. Neel,et al.  Gain-of-function mutations in the gene encoding the tyrosine phosphatase SHP2 induce hydrocephalus in a catalytically dependent manner , 2018, Science Signaling.

[4]  J. Charron,et al.  Mek1Y130C mice recapitulate aspects of human cardio-facio-cutaneous syndrome , 2018, Disease Models & Mechanisms.

[5]  J. Wu,et al.  Selective inhibition of leukemia-associated SHP2E69K mutant by the allosteric SHP2 inhibitor SHP099 , 2018, Leukemia.

[6]  A. Bennett,et al.  Mitogen-Activated Protein Kinase Regulation in Hepatic Metabolism , 2017, Trends in Endocrinology & Metabolism.

[7]  Lisa T. Emrick,et al.  Autosomal Recessive Noonan Syndrome Associated with Biallelic LZTR1Variants , 2017, Genetics in Medicine.

[8]  A. Yart,et al.  Noonan syndrome: an update on growth and development , 2017, Current opinion in endocrinology, diabetes, and obesity.

[9]  Rajika Roy,et al.  Heterozygous deletion of AKT1 rescues cardiac contractility, but not hypertrophy, in a mouse model of Noonan Syndrome with Multiple Lentigines. , 2017, Journal of molecular and cellular cardiology.

[10]  Y. Sako,et al.  Mutation-Specific Mechanisms of Hyperactivation of Noonan Syndrome SOS Molecules Detected with Single-molecule Imaging in Living Cells , 2017, Scientific Reports.

[11]  M. Digilio,et al.  Cardiac defects, morbidity and mortality in patients affected by RASopathies. CARNET study results. , 2017, International journal of cardiology.

[12]  P. Poulikakos,et al.  New perspectives for targeting RAF kinase in human cancer , 2017, Nature Reviews Cancer.

[13]  Olumide O. Aruwajoye,et al.  Targeted Disruption of NF1 in Osteocytes Increases FGF23 and Osteoid With Osteomalacia‐like Bone Phenotype , 2017, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[14]  A. Carè,et al.  Aberrant HRAS transcript processing underlies a distinctive phenotype within the RASopathy clinical spectrum , 2017, Human mutation.

[15]  Frank McCormick,et al.  RAS Proteins and Their Regulators in Human Disease , 2017, Cell.

[16]  B. Schwartz,et al.  In vivo efficacy of the AKT inhibitor ARQ 092 in Noonan Syndrome with multiple lentigines-associated hypertrophic cardiomyopathy , 2017, PloS one.

[17]  B. Neel,et al.  Cellular interplay via cytokine hierarchy causes pathological cardiac hypertrophy in RAF1-mutant Noonan syndrome , 2017, Nature Communications.

[18]  S. Kushner,et al.  Mechanisms underlying cognitive deficits in a mouse model for Costello Syndrome are distinct from other RASopathy mouse models , 2017, Scientific Reports.

[19]  Monia Magliozzi,et al.  Structural, Functional, and Clinical Characterization of a Novel PTPN11 Mutation Cluster Underlying Noonan Syndrome , 2017, Human mutation.

[20]  G. Ferrero,et al.  Constitutional bone impairment in Noonan syndrome , 2017, American journal of medical genetics. Part A.

[21]  M. Ahmadian,et al.  Aberrant neuronal activity-induced signaling and gene expression in a mouse model of RASopathy , 2017, PLoS genetics.

[22]  M. Baccarini,et al.  Deciphering the RAS/ERK pathway in vivo. , 2017, Biochemical Society transactions.

[23]  J. Skotheim,et al.  Spatial and temporal signal processing and decision making by MAPK pathways , 2017, The Journal of cell biology.

[24]  D. Tester,et al.  Elucidation of MRAS-mediated Noonan syndrome with cardiac hypertrophy. , 2016, JCI insight.

[25]  Gang Huang,et al.  Leukaemogenic effects of Ptpn11 activating mutations in the stem cell microenvironment , 2016, Nature.

[26]  W. Kolch,et al.  The spatiotemporal regulation of RAS signalling. , 2016, Biochemical Society transactions.

[27]  Oleksii S. Rukhlenko,et al.  The complexities and versatility of the RAS-to-ERK signalling system in normal and cancer cells. , 2016, Seminars in cell & developmental biology.

[28]  K. Rauen,et al.  Pathogenetics of the RASopathies. , 2016, Human molecular genetics.

[29]  K. Gripp,et al.  A novel rasopathy caused by recurrent de novo missense mutations in PPP1CB closely resembles Noonan syndrome with loose anagen hair , 2016, American journal of medical genetics. Part A.

[30]  B. Gelb,et al.  SHOC2 subcellular shuttling requires the KEKE motif-rich region and N-terminal leucine-rich repeat domain and impacts on ERK signalling. , 2016, Human molecular genetics.

[31]  L. Weiss,et al.  Autonomous and Non-autonomous Defects Underlie Hypertrophic Cardiomyopathy in BRAF-Mutant hiPSC-Derived Cardiomyocytes , 2016, Stem cell reports.

[32]  W. Jhang,et al.  Cardiac Manifestations and Associations with Gene Mutations in Patients Diagnosed with RASopathies , 2016, Pediatric Cardiology.

[33]  Kyu-Ho Lee,et al.  Developmental SHP2 dysfunction underlies cardiac hypertrophy in Noonan syndrome with multiple lentigines. , 2016, The Journal of clinical investigation.

[34]  K. Rauen,et al.  Expansion of the RASopathies , 2016, Current Genetic Medicine Reports.

[35]  Ping Zhu,et al.  Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases , 2016, Nature.

[36]  R. Buscà,et al.  ERK1 and ERK2 Map Kinases: Specific Roles or Functional Redundancy? , 2016, Front. Cell Dev. Biol..

[37]  Eun Ryoung Jang,et al.  The function of Shoc2: A scaffold and beyond , 2016, Communicative & integrative biology.

[38]  R. Kessels,et al.  Intellectual development in Noonan syndrome: a longitudinal study , 2016, Brain and behavior.

[39]  H. Cavé,et al.  Growth patterns of patients with Noonan syndrome: correlation with age and genotype. , 2016, European journal of endocrinology.

[40]  D. Mitter,et al.  Genotype and phenotype in patients with Noonan syndrome and a RIT1 mutation , 2016, Genetics in Medicine.

[41]  C. Kim,et al.  Nutritional aspects of Noonan syndrome and Noonan‐related disorders , 2016, American journal of medical genetics. Part A.

[42]  M. Tartaglia,et al.  Understanding Growth Failure in Costello Syndrome: Increased Resting Energy Expenditure. , 2016, The Journal of pediatrics.

[43]  Jian Zhong,et al.  RAS and downstream RAF-MEK and PI3K-AKT signaling in neuronal development, function and dysfunction , 2016, Biological chemistry.

[44]  O. Migita,et al.  Spectrum of mutations and genotype–phenotype analysis in Noonan syndrome patients with RIT1 mutations , 2016, Human Genetics.

[45]  P. Casey,et al.  Protein prenylation: unique fats make their mark on biology , 2016, Nature Reviews Molecular Cell Biology.

[46]  Kevin D. Costa,et al.  Human Engineered Cardiac Tissues Created Using Induced Pluripotent Stem Cells Reveal Functional Characteristics of BRAF-Mediated Hypertrophic Cardiomyopathy , 2016, PloS one.

[47]  H. Cavé,et al.  Mutations in RIT1 cause Noonan syndrome with possible juvenile myelomonocytic leukemia but are not involved in acute lymphoblastic leukemia , 2016, European Journal of Human Genetics.

[48]  B. Raught,et al.  Inhibition of SHP2-mediated dephosphorylation of Ras suppresses oncogenesis , 2015, Nature Communications.

[49]  M. Tartaglia,et al.  Response to long‐term growth hormone therapy in patients affected by RASopathies and growth hormone deficiency: Patterns of growth, puberty and final height data , 2015, American journal of medical genetics. Part A.

[50]  Trevor J Pugh,et al.  Activating Mutations Affecting the Dbl Homology Domain of SOS2 Cause Noonan Syndrome , 2015, Human mutation.

[51]  Z. Stark,et al.  Copy number variants including RAS pathway genes—How much RASopathy is in the phenotype? , 2015, American journal of medical genetics. Part A.

[52]  K. Dutton-Regester,et al.  Recurrent inactivating RASA2 mutations in melanoma , 2015, Nature Genetics.

[53]  H. Cavé,et al.  Myeloid Dysregulation in a Human Induced Pluripotent Stem Cell Model of PTPN11-Associated Juvenile Myelomonocytic Leukemia. , 2015, Cell reports.

[54]  P. Valet,et al.  SHP2 sails from physiology to pathology. , 2015, European journal of medical genetics.

[55]  Ying Liu,et al.  High Incidence of Noonan Syndrome Features Including Short Stature and Pulmonic Stenosis in Patients carrying NF1 Missense Mutations Affecting p.Arg1809: Genotype–Phenotype Correlation , 2015, Human mutation.

[56]  Wenjie Liu,et al.  Deletion of the tyrosine phosphatase Shp2 in Sertoli cells causes infertility in mice , 2015, Scientific Reports.

[57]  S. Shvartsman,et al.  RASopathies: unraveling mechanisms with animal models , 2015, Disease Models & Mechanisms.

[58]  M. Digilio,et al.  Molecular Diversity and Associated Phenotypic Spectrum of Germline CBL Mutations , 2015, Human mutation.

[59]  G. Ladds,et al.  Ras activation revisited: role of GEF and GAP systems , 2015, Biological chemistry.

[60]  E. I. Pierpont,et al.  Attention skills and executive functioning in children with Noonan syndrome and their unaffected siblings , 2015, Developmental medicine and child neurology.

[61]  B. Gelb,et al.  Rapidly progressive hypertrophic cardiomyopathy in an infant with Noonan syndrome with multiple lentigines: Palliative treatment with a rapamycin analog , 2015, American journal of medical genetics. Part A.

[62]  A. Pereira,et al.  Rare variants in SOS2 and LZTR1 are associated with Noonan syndrome , 2015, Journal of Medical Genetics.

[63]  K. Mohammad,et al.  Dystrophic Spinal Deformities in a Neurofibromatosis Type 1 Murine Model , 2015, PloS one.

[64]  M. Greene,et al.  Cancer spectrum and frequency among children with Noonan, Costello, and cardio-facio-cutaneous syndromes , 2015, British Journal of Cancer.

[65]  A. Deodati,et al.  The Impact of Growth Hormone Therapy on Adult Height in Noonan Syndrome: A Systematic Review , 2015, Hormone Research in Paediatrics.

[66]  T. Ogura,et al.  New BRAF knockin mice provide a pathogenetic mechanism of developmental defects and a therapeutic approach in cardio-facio-cutaneous syndrome. , 2014, Human molecular genetics.

[67]  J. Noonan,et al.  The Efficacy and Safety of Growth Hormone Therapy in Children with Noonan Syndrome: A Review of the Evidence , 2014, Hormone Research in Paediatrics.

[68]  Alcino J. Silva,et al.  Mechanism and treatment for the learning and memory deficits associated with mouse models of Noonan syndrome , 2014, Nature Neuroscience.

[69]  Manuel Desco,et al.  K-RasV14I recapitulates Noonan syndrome in mice , 2014, Proceedings of the National Academy of Sciences.

[70]  S. Fesik,et al.  Drugging the undruggable RAS: Mission Possible? , 2014, Nature Reviews Drug Discovery.

[71]  H. Cavé,et al.  LEOPARD syndrome-associated SHP2 mutation confers leanness and protection from diet-induced obesity , 2014, Proceedings of the National Academy of Sciences.

[72]  Alexander S. Banks,et al.  An Erk/Cdk5 axis controls the diabetogenic actions of PPARγ , 2014, Nature.

[73]  T. Jongens,et al.  Modulation of cAMP and Ras Signaling Pathways Improves Distinct Behavioral Deficits in a Zebrafish Model of Neurofibromatosis Type 1 , 2014, Cell reports.

[74]  Corinne Alberti,et al.  Juvenile myelomonocytic leukaemia and Noonan syndrome , 2014, Journal of Medical Genetics.

[75]  John D McPherson,et al.  Next-generation sequencing identifies rare variants associated with Noonan syndrome , 2014, Proceedings of the National Academy of Sciences.

[76]  M. Gos,et al.  Neurofibromin in neurofibromatosis type 1 - mutations in NF1gene as a cause of disease. , 2014, Developmental period medicine.

[77]  I. Östman-Smith Beta-Blockers in Pediatric Hypertrophic Cardiomyopathies , 2014, Reviews on recent clinical trials.

[78]  A. Heck,et al.  PZR Coordinates Shp2 Noonan and LEOPARD Syndrome Signaling in Zebrafish and Mice , 2014, Molecular and Cellular Biology.

[79]  J. den Hertog,et al.  Noonan and LEOPARD syndrome Shp2 variants induce heart displacement defects in zebrafish , 2014, Development.

[80]  I. Martinelli,et al.  Hemostatic Abnormalities in Noonan Syndrome , 2014, Pediatrics.

[81]  M. Loh,et al.  Activating mutations in RRAS underlie a phenotype within the RASopathy spectrum and contribute to leukaemogenesis , 2014, Human molecular genetics.

[82]  E. Pai,et al.  Structural insights into Noonan/LEOPARD syndrome-related mutants of protein-tyrosine phosphatase SHP2 (PTPN11) , 2014, BMC Structural Biology.

[83]  K. Gauvreau,et al.  Cardiovascular disease in Noonan syndrome , 2014, Archives of Disease in Childhood.

[84]  M. Bergo,et al.  Myocardial KRAS(G12D) expression does not cause cardiomyopathy in mice. , 2014, Cardiovascular research.

[85]  N. M. Powell,et al.  Co-targeting the MAPK and PI3K/AKT/mTOR pathways in two genetically engineered mouse models of schwann cell tumors reduces tumor grade and multiplicity , 2014, Oncotarget.

[86]  D. Viskochil,et al.  Hyperactive Ras/MAPK signaling is critical for tibial nonunion fracture in neurofibromin-deficient mice. , 2013, Human molecular genetics.

[87]  K. Mohammad,et al.  Hyperactive Transforming Growth Factor‐β1 Signaling Potentiates Skeletal Defects in a Neurofibromatosis Type 1 Mouse Model , 2013, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[88]  N. Mochizuki,et al.  Involvement of EphA2-mediated tyrosine phosphorylation of Shp2 in Shp2-regulated activation of extracellular signal-regulated kinase , 2013, Oncogene.

[89]  R. Bernards,et al.  Genetic and Functional Studies Implicate Synaptic Overgrowth and Ring Gland cAMP/PKA Signaling Defects in the Drosophila melanogaster Neurofibromatosis-1 Growth Deficiency , 2013, PLoS genetics.

[90]  C. Catsman-Berrevoets,et al.  Simvastatin for cognitive deficits and behavioural problems in patients with neurofibromatosis type 1 (NF1-SIMCODA): a randomised, placebo-controlled trial , 2013, The Lancet Neurology.

[91]  A. Straube,et al.  Lovastatin improves impaired synaptic plasticity and phasic alertness in patients with neurofibromatosis type 1 , 2013, BMC Neurology.

[92]  Eileen White,et al.  Exploiting the bad eating habits of Ras-driven cancers , 2013, Genes & development.

[93]  L. Garraway,et al.  C-RAF mutations confer resistance to RAF inhibitors. , 2013, Cancer research.

[94]  Jeffrey K. Mito,et al.  NF1 Deletion Generates Multiple Subtypes of Soft-Tissue Sarcoma That Respond to MEK Inhibition , 2013, Molecular Cancer Therapeutics.

[95]  Toshihiko Ogura,et al.  Gain-of-function mutations in RIT1 cause Noonan syndrome, a RAS/MAPK pathway syndrome. , 2013, American journal of human genetics.

[96]  Hong Zheng,et al.  Activating Mutations in Protein Tyrosine Phosphatase Ptpn11 (Shp2) Enhance Reactive Oxygen Species Production That Contributes to Myeloproliferative Disorder , 2013, PloS one.

[97]  Christian M. Metallo,et al.  Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells , 2013, Nature.

[98]  P. Lapunzina,et al.  LEOPARD syndrome: a variant of Noonan syndrome strongly associated with hypertrophic cardiomyopathy. , 2013, Revista espanola de cardiologia.

[99]  Matthew J. Wolf,et al.  Raf-mediated cardiac hypertrophy in adult Drosophila , 2013, Disease Models & Mechanisms.

[100]  M. Mizuguchi,et al.  A novel SOS1 mutation in Costello/CFC syndrome affects signaling in both RAS and PI3K pathways , 2013, Journal of receptor and signal transduction research.

[101]  John M. Asara,et al.  Glutamine supports pancreatic cancer growth through a Kras-regulated metabolic pathway , 2013, Nature.

[102]  Shenmin Zhang,et al.  Structural and Mechanistic Insights into LEOPARD Syndrome-Associated SHP2 Mutations* , 2013, The Journal of Biological Chemistry.

[103]  G. Page,et al.  MEK inhibition exhibits efficacy in human and mouse neurofibromatosis tumors. , 2013, The Journal of clinical investigation.

[104]  Jin Xu,et al.  Sustained MEK inhibition abrogates myeloproliferative disease in Nf1 mutant mice. , 2013, The Journal of clinical investigation.

[105]  P. King,et al.  Nonreceptor tyrosine phosphatase Shp2 promotes adipogenesis through inhibition of p38 MAP kinase , 2012, Proceedings of the National Academy of Sciences.

[106]  E. Pasmant,et al.  Review and update of SPRED1 mutations causing legius syndrome , 2012, Human mutation.

[107]  A. Pereira,et al.  Growth standards of patients with Noonan and Noonan‐like syndromes with mutations in the RAS/MAPK pathway , 2012, American journal of medical genetics. Part A.

[108]  C. Pritchard,et al.  The intermediate-activity (L597V)BRAF mutant acts as an epistatic modifier of oncogenic RAS by enhancing signaling through the RAF/MEK/ERK pathway. , 2012, Genes & development.

[109]  G. Binder,et al.  Health and quality of life in adults with Noonan syndrome. , 2012, The Journal of pediatrics.

[110]  Long-Sheng Chang,et al.  ERK Inhibition Rescues Defects in Fate Specification of Nf1-Deficient Neural Progenitors and Brain Abnormalities , 2012, Cell.

[111]  Kyoung-Han Kim,et al.  Increased BRAF Heterodimerization Is the Common Pathogenic Mechanism for Noonan Syndrome-Associated RAF1 Mutants , 2012, Molecular and Cellular Biology.

[112]  Seok-Hyung Kim,et al.  Zebrafish neurofibromatosis type 1 genes have redundant functions in tumorigenesis and embryonic development , 2012, Disease Models & Mechanisms.

[113]  N. Duesbery,et al.  MEK genomics in development and disease , 2012, Briefings in functional genomics.

[114]  Alyssa A. Tran,et al.  Decreased bone mineralization in children with Noonan syndrome: another consequence of dysregulated RAS MAPKinase pathway? , 2012, Molecular genetics and metabolism.

[115]  Gerald C. Chu,et al.  Oncogenic Kras Maintains Pancreatic Tumors through Regulation of Anabolic Glucose Metabolism , 2012, Cell.

[116]  N. Normanno,et al.  The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: role in cancer pathogenesis and implications for therapeutic approaches , 2012, Expert opinion on therapeutic targets.

[117]  G. Feng,et al.  Shp2 Controls Female Body Weight and Energy Balance by Integrating Leptin and Estrogen Signals , 2012, Molecular and Cellular Biology.

[118]  M. Furutani,et al.  Characterization of a novel KRAS mutation identified in Noonan syndrome , 2012, American journal of medical genetics. Part A.

[119]  B. Neel,et al.  Noonan syndrome-causing SHP2 mutants inhibit insulin-like growth factor 1 release via growth hormone-induced ERK hyperactivation, which contributes to short stature , 2012, Proceedings of the National Academy of Sciences.

[120]  C. Anastasaki,et al.  Continual low-level MEK inhibition ameliorates cardio-facio-cutaneous phenotypes in zebrafish , 2012, Disease Models & Mechanisms.

[121]  Scott M Lippman,et al.  Targeting the MAPK–RAS–RAF signaling pathway in cancer therapy , 2012, Expert opinion on therapeutic targets.

[122]  S. Cohen,et al.  MAPK/ERK Signaling Regulates Insulin Sensitivity to Control Glucose Metabolism in Drosophila , 2011, PLoS genetics.

[123]  Beom Hee Lee,et al.  Spectrum of mutations in Noonan syndrome and their correlation with phenotypes. , 2011, The Journal of pediatrics.

[124]  J. Nyman,et al.  Mice lacking Nf1 in osteochondroprogenitor cells display skeletal dysplasia similar to patients with neurofibromatosis type I. , 2011, Human molecular genetics.

[125]  H. Steller,et al.  A Gain-of-Function Germline Mutation in Drosophila ras1 Affects Apoptosis and Cell Fate during Development , 2011, PloS one.

[126]  D. Largaespada,et al.  Nf1 mutant mice with p19ARF gene loss develop accelerated hematopoietic disease resembling acute leukemia with a variable phenotype , 2011, American journal of hematology.

[127]  C. Ankarberg-Lindgren,et al.  Testicular size development and reproductive hormones in boys and adult males with Noonan syndrome: a longitudinal study. , 2011, European journal of endocrinology.

[128]  B. Gelb,et al.  Cyclosporine attenuates cardiomyocyte hypertrophy induced by RAF1 mutants in Noonan and LEOPARD syndromes. , 2011, Journal of molecular and cellular cardiology.

[129]  Yingze Zhang,et al.  Primary osteopathy of vertebrae in a neurofibromatosis type 1 murine model. , 2011, Bone.

[130]  S. Nadaf,et al.  Costello and cardio‐facio‐cutaneous syndromes: Moving toward clinical trials in RASopathies , 2011, American journal of medical genetics. Part C, Seminars in medical genetics.

[131]  L. Kiemeney,et al.  Cancer risk in patients with Noonan syndrome carrying a PTPN11 mutation , 2011, European Journal of Human Genetics.

[132]  O. Gabrielli,et al.  SOS1 Mutations in Noonan Syndrome: Molecular Spectrum, Structural Insights on Pathogenic Effects, and Genotype–Phenotype Correlations , 2011, Human mutation.

[133]  M. Barbacid,et al.  Constitutive activation of B-Raf in the mouse germ line provides a model for human cardio-facio-cutaneous syndrome , 2011, Proceedings of the National Academy of Sciences.

[134]  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.

[135]  B. Neel,et al.  Rapamycin reverses hypertrophic cardiomyopathy in a mouse model of LEOPARD syndrome-associated PTPN11 mutation. , 2011, The Journal of clinical investigation.

[136]  H. Coller,et al.  Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. , 2011, Genes & development.

[137]  B. Gelb,et al.  Noonan syndrome and clinically related disorders. , 2011, Best practice & research. Clinical endocrinology & metabolism.

[138]  M. Tartaglia,et al.  PTPN11 (protein tyrosine phosphatase, non-receptor type, 11) , 2011 .

[139]  J. den Hertog,et al.  Noonan syndrome gain-of-function mutations in NRAS cause zebrafish gastrulation defects , 2011, Disease Models & Mechanisms.

[140]  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.

[141]  Gordon Chan,et al.  A germline gain-of-function mutation in Ptpn11 (Shp-2) phosphatase induces myeloproliferative disease by aberrant activation of hematopoietic stem cells. , 2010, Blood.

[142]  J. Allanson,et al.  Noonan Syndrome: Clinical Features, Diagnosis, and Management Guidelines , 2010, Pediatrics.

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

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

[145]  M. Mizuguchi,et al.  Comprehensive genetic analysis of overlapping syndromes of RAS/RAF/MEK/ERK pathway , 2010, Pediatrics international : official journal of the Japan Pediatric Society.

[146]  Ronald L. Davis,et al.  A Distinct Set of Drosophila Brain Neurons Required for Neurofibromatosis Type 1-Dependent Learning and Memory , 2010, The Journal of Neuroscience.

[147]  Vandana Sachdev,et al.  Cardiomyopathy in Congenital and Acquired Generalized Lipodystrophy: A Clinical Assessment , 2010, Medicine.

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

[149]  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.

[150]  P. Pfluger,et al.  A functional role for the p62–ERK1 axis in the control of energy homeostasis and adipogenesis , 2010, EMBO reports.

[151]  M. Ahmadian,et al.  Duplication of Glu37 in the switch I region of HRAS impairs effector/GAP binding and underlies Costello syndrome by promoting enhanced growth factor-dependent MAPK and AKT activation. , 2010, Human molecular genetics.

[152]  Gabriele Gillessen-Kaesbach,et al.  Molecular and clinical analysis of RAF1 in Noonan syndrome and related disorders: dephosphorylation of serine 259 as the essential mechanism for mutant activation , 2010, Human mutation.

[153]  R. Kucherlapati,et al.  A suggested role for mitochondria in Noonan syndrome. , 2010, Biochimica et biophysica acta.

[154]  J. Nyman,et al.  Local Low-Dose Lovastatin Delivery Improves the Bone-Healing Defect Caused by Nf1 Loss of Function in Osteoblasts , 2010, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

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

[156]  A. Look,et al.  Cardiac and vascular functions of the zebrafish orthologues of the type I neurofibromatosis gene NFI , 2009, Proceedings of the National Academy of Sciences.

[157]  Bruce D. Gelb,et al.  The Phosphatase SHP2 Regulates the Spacing Effect for Long-Term Memory Induction , 2009, Cell.

[158]  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.

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

[160]  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.

[161]  B. Bakker,et al.  Growth response, near-adult height, and patterns of growth and puberty in patients with noonan syndrome treated with growth hormone. , 2009, The Journal of clinical endocrinology and metabolism.

[162]  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.

[163]  Stephen M. Hedrick,et al.  MAPK3/1 (ERK1/2) in Ovarian Granulosa Cells Are Essential for Female Fertility , 2009, Science.

[164]  J. Fagin,et al.  Endogenous expression of HrasG12V induces developmental defects and neoplasms with copy number imbalances of the oncogene , 2009, Proceedings of the National Academy of Sciences.

[165]  G. Feng,et al.  Coordinated regulation by Shp2 tyrosine phosphatase of signaling events controlling insulin biosynthesis in pancreatic β-cells , 2009, Proceedings of the National Academy of Sciences.

[166]  R. Marais,et al.  Kinase-activating and kinase-impaired cardio-facio-cutaneous syndrome alleles have activity during zebrafish development and are sensitive to small molecule inhibitors , 2009, Human molecular genetics.

[167]  M. Digilio,et al.  Germline BRAF mutations in Noonan, LEOPARD, and cardiofaciocutaneous syndromes: Molecular diversity and associated phenotypic spectrum , 2009, Human mutation.

[168]  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.

[169]  A. Mark,et al.  Hypothalamic ERK Mediates the Anorectic and Thermogenic Sympathetic Effects of Leptin , 2009, Diabetes.

[170]  G. Rosenberger,et al.  Oncogenic HRAS mutations cause prolonged PI3K signaling in response to epidermal growth factor in fibroblasts of patients with Costello syndrome , 2009, Human mutation.

[171]  E. Mercuri,et al.  Cognitive profile of disorders associated with dysregulation of the RAS/MAPK signaling cascade , 2009, American journal of medical genetics. Part A.

[172]  M. Digilio,et al.  Spectrum of MEK1 and MEK2 gene mutations in cardio-facio-cutaneous syndrome and genotype–phenotype correlations , 2009, European Journal of Human Genetics.

[173]  Nils Z. Borgesius,et al.  Spred1 Is Required for Synaptic Plasticity and Hippocampus-Dependent Learning , 2008, The Journal of Neuroscience.

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

[175]  C. Kahn,et al.  Deletion of Shp2 Tyrosine Phosphatase in Muscle Leads to Dilated Cardiomyopathy, Insulin Resistance, and Premature Death , 2008, Molecular and Cellular Biology.

[176]  C. Noordam,et al.  Long-term GH treatment improves adult height in children with Noonan syndrome with and without mutations in protein tyrosine phosphatase, non-receptor-type 11. , 2008, European journal of endocrinology.

[177]  Alcino J. Silva,et al.  Effect of Simvastatin on Cognitive Functioning in Children with Neurofibromatosis Type 1 a Randomized Controlled Trial , 2022 .

[178]  J. Epstein,et al.  Cardiomyocyte-Specific Loss of Neurofibromin Promotes Cardiac Hypertrophy and Dysfunction , 2008, Circulation research.

[179]  L. Castagnoli,et al.  Diverse driving forces underlie the invariant occurrence of the T42A, E139D, I282V and T468M SHP2 amino acid substitutions causing Noonan and LEOPARD syndromes. , 2008, Human molecular genetics.

[180]  M. Barbacid,et al.  A mouse model for Costello syndrome reveals an Ang II-mediated hypertensive condition. , 2008, The Journal of clinical investigation.

[181]  A. Bennett,et al.  Noonan Syndrome-associated SHP-2/Ptpn11 Mutants Enhance SIRPα and PZR Tyrosyl Phosphorylation and Promote Adhesion-mediated ERK Activation* , 2008, Journal of Biological Chemistry.

[182]  M. Digilio,et al.  Leopard syndrome , 2008, Orphanet journal of rare diseases.

[183]  G. Di Salvo,et al.  Genotype–phenotype analysis and natural history of left ventricular hypertrophy in LEOPARD syndrome , 2008, American journal of medical genetics. Part A.

[184]  A. Stemmer-Rachamimov,et al.  Plexiform and dermal neurofibromas and pigmentation are caused by Nf1 loss in desert hedgehog-expressing cells. , 2008, Cancer cell.

[185]  F. Conlon,et al.  SHP-2 is required for the maintenance of cardiac progenitors , 2007, Development.

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

[187]  Gideon Bollag,et al.  Biochemical and Functional Characterization of Germ Line KRAS Mutations , 2007, Molecular and Cellular Biology.

[188]  G. Di Salvo,et al.  Prevalence and clinical significance of cardiovascular abnormalities in patients with the LEOPARD syndrome. , 2007, The American journal of cardiology.

[189]  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.

[190]  I. Hakker,et al.  Distinct Functional Domains of Neurofibromatosis Type 1 Regulate Immediate versus Long-Term Memory Formation , 2007, The Journal of Neuroscience.

[191]  David F. McCleary,et al.  Life extension through neurofibromin mitochondrial regulation and antioxidant therapy for neurofibromatosis-1 in Drosophila melanogaster , 2007, Nature Genetics.

[192]  Peter T. McKenney,et al.  Reduced growth of Drosophila neurofibromatosis 1 mutants reflects a non-cell-autonomous requirement for GTPase-Activating Protein activity in larval neurons. , 2006, Genes & development.

[193]  M. Patton,et al.  The natural history of Noonan syndrome: a long-term follow-up study , 2006, Archives of Disease in Childhood.

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

[195]  J. Epstein,et al.  The neurofibromin GAP-related domain rescues endothelial but not neural crest development in Nf1 mice. , 2006, The Journal of clinical investigation.

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

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

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

[199]  Pablo Rodriguez-Viciana,et al.  Germline Mutations in Genes Within the MAPK Pathway Cause Cardio-facio-cutaneous Syndrome , 2006, Science.

[200]  L. Pick,et al.  Transgenic Drosophila models of Noonan syndrome causing PTPN11 gain-of-function mutations. , 2006, Human molecular genetics.

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

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

[203]  Alcino J. Silva,et al.  The HMG-CoA Reductase Inhibitor Lovastatin Reverses the Learning and Attention Deficits in a Mouse Model of Neurofibromatosis Type 1 , 2005, Current Biology.

[204]  K. Yutzey,et al.  Noonan Syndrome Mutation Q79R in Shp2 Increases Proliferation of Valve Primordia Mesenchymal Cells via Extracellular Signal–Regulated Kinase 1/2 Signaling , 2005, Circulation research.

[205]  J. Dahlgren,et al.  Improved final height with long‐term growth hormone treatment in Noonan syndrome , 2005 .

[206]  Dawen Zhao,et al.  Early inactivation of p53 tumor suppressor gene cooperating with NF1 loss induces malignant astrocytoma. , 2005, Cancer cell.

[207]  D. Gutmann,et al.  Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors. , 2005, Cancer research.

[208]  J. Salles,et al.  A Novel Role for Gab1 and SHP2 in Epidermal Growth Factor-induced Ras Activation* , 2005, Journal of Biological Chemistry.

[209]  C. Dani,et al.  The extracellular signal-regulated kinase isoform ERK1 is specifically required for in vitro and in vivo adipogenesis. , 2005, Diabetes.

[210]  M. Kubo,et al.  Spred-1 negatively regulates allergen-induced airway eosinophilia and hyperresponsiveness , 2005, The Journal of experimental medicine.

[211]  G. Feng,et al.  Neuronal Shp2 tyrosine phosphatase controls energy balance and metabolism. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[212]  M. Patton,et al.  Paternal germline origin and sex-ratio distortion in transmission of PTPN11 mutations in Noonan syndrome. , 2004, American journal of human genetics.

[213]  R. Weksberg,et al.  Growth hormone deficiency in Costello syndrome , 2004, American journal of medical genetics. Part A.

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

[215]  P. Arner,et al.  Targets for TNF-α-induced lipolysis in human adipocytes , 2004 .

[216]  M. Digilio,et al.  Clinical and molecular analysis of 30 patients with multiple lentigines LEOPARD syndrome , 2004, Journal of Medical Genetics.

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

[218]  M. Hayman,et al.  Molecular Mechanism for a Role of SHP2 in Epidermal Growth Factor Receptor Signaling , 2003, Molecular and Cellular Biology.

[219]  Naoya Nakai,et al.  Essential role for ERK2 mitogen‐activated protein kinase in placental development , 2003, Genes to cells : devoted to molecular & cellular mechanisms.

[220]  B. Neel,et al.  Tyrosyl Phosphorylation of Shp2 Is Required for Normal ERK Activation in Response to Some, but Not All, Growth Factors* , 2003, Journal of Biological Chemistry.

[221]  Wei Li,et al.  Extracellular signal-regulated kinase 2 is necessary for mesoderm differentiation , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[222]  C. Noordam,et al.  Effects of Growth Hormone Treatment on Left Ventricular Dimensions in Children with Noonan’s Syndrome , 2002, Hormone Research in Paediatrics.

[223]  Alcino J. Silva,et al.  Mechanism for the learning deficits in a mouse model of neurofibromatosis type 1 , 2002, Nature.

[224]  R. Roth,et al.  Stimulation of Lipolysis and Hormone-sensitive Lipase via the Extracellular Signal-regulated Kinase Pathway* , 2001, The Journal of Biological Chemistry.

[225]  A. Sehgal,et al.  A Circadian Output in Drosophila Mediated by Neurofibromatosis-1 and Ras/MAPK , 2001, Science.

[226]  D. Dunger,et al.  Short stature in Noonan syndrome: response to growth hormone therapy , 2001, Archives of disease in childhood.

[227]  J. Marth,et al.  Ablation of NF1 function in neurons induces abnormal development of cerebral cortex and reactive gliosis in the brain. , 2001, Genes & development.

[228]  Y. Zhong,et al.  A neurofibromatosis-1-regulated pathway is required for learning in Drosophila , 2000, Nature.

[229]  J. Charron,et al.  Embryonic death of Mek1-deficient mice reveals a role for this kinase in angiogenesis in the labyrinthine region of the placenta , 1999, Current Biology.

[230]  J A Epstein,et al.  Neurofibromin modulation of ras activity is required for normal endocardial-mesenchymal transformation in the developing heart. , 1998, Development.

[231]  U. Wiegand,et al.  Cardiomyopathic lentiginosis/LEOPARD syndrome presenting as sudden cardiac arrest. , 1998, Chest.

[232]  M. Hengartner,et al.  The Caenorhabditis elegans SH2 domain-containing protein tyrosine phosphatase PTP-2 participates in signal transduction during oogenesis and vulval development. , 1998, Genes & development.

[233]  Jun Miyoshi,et al.  K-Ras is essential for the development of the mouse embryo , 1997, Oncogene.

[234]  U. Rapp,et al.  Endothelial apoptosis in Braf-deficient mice , 1997, Nature Genetics.

[235]  J. Gusella,et al.  Rescue of a Drosophila NF1 mutant phenotype by protein kinase A. , 1997, Science.

[236]  T. Pawson,et al.  Abnormal mesoderm patterning in mouse embryos mutant for the SH2 tyrosine phosphatase Shp‐2 , 1997, The EMBO journal.

[237]  D. Dunger,et al.  The short-term effects of growth hormone therapy on height velocity and cardiac ventricular wall thickness in children with Noonan's syndrome. , 1996, The Journal of clinical endocrinology and metabolism.

[238]  J. Smith,et al.  Mesoderm induction in Xenopus caused by activation of MAP kinase , 1995, Nature.

[239]  E. Nishida,et al.  Involvement of the MAP kinase cascade in Xenopus mesoderm induction. , 1995, The EMBO journal.

[240]  Min Han,et al.  MEK-2, a Caenorhabditis elegans MAP kinase kinase, functions in Ras-mediated vulval induction and other developmental events. , 1995, Genes & development.

[241]  Robert A. Weinberg,et al.  Tumour predisposition in mice heterozygous for a targeted mutation in Nf1 , 1994, Nature Genetics.

[242]  N. Copeland,et al.  Targeted disruption of the neurofibromatosis type-1 gene leads to developmental abnormalities in heart and various neural crest-derived tissues. , 1994, Genes & development.

[243]  M. Whitman,et al.  Mesoderm induction by activin requires FGF-mediated intracellular signals. , 1994, Development.

[244]  M. Burch,et al.  A clinical study of Noonan syndrome. , 1992, Archives of disease in childhood.

[245]  M. Patton,et al.  Coagulation-factor deficiencies and abnormal bleeding in Noonan's syndrome , 1992, The Lancet.

[246]  H. Horvitz,et al.  Caenorhabditis elegans ras gene let-60 acts as a switch in the pathway of vulval induction , 1990, Nature.

[247]  M. Ranke,et al.  Noonan syndrome: growth and clinical manifestations in 144 cases , 1988, European Journal of Pediatrics.

[248]  J E Allanson,et al.  Growth curves for height in Noonan syndrome , 1986, Clinical genetics.

[249]  J. Noonan Hypertelorism with Turner phenotype. A new syndrome with associated congenital heart disease. , 1968, American journal of diseases of children.

[250]  Vaibhav Sidarala,et al.  The Regulatory Roles of Mitogen-Activated Protein Kinase (MAPK) Pathways in Health and Diabetes: Lessons Learned from the Pancreatic β-Cell. , 2017, Recent patents on endocrine, metabolic & immune drug discovery.

[251]  C. Guerra,et al.  Modeling RASopathies with Genetically Modified Mouse Models. , 2017, Methods in molecular biology.

[252]  M. Edwards,et al.  The Q510E mutation in Shp2 perturbs heart valve development by increasing cell migration. , 2015, Journal of applied physiology.

[253]  Jessica Lauriol,et al.  The role of the protein tyrosine phosphatase SHP2 in cardiac development and disease. , 2015, Seminars in cell & developmental biology.

[254]  O. Ishikawa,et al.  Pathogenesis of multiple lentigines in LEOPARD syndrome with PTPN11 gene mutation. , 2015, Acta dermato-venereologica.

[255]  C. Kahn,et al.  Insulin receptor signaling in normal and insulin-resistant states. , 2014, Cold Spring Harbor perspectives in biology.

[256]  M. Edwards,et al.  The PTPN11 loss-of-function mutation Q510E-Shp2 causes hypertrophic cardiomyopathy by dysregulating mTOR signaling. , 2012, American journal of physiology. Heart and circulatory physiology.

[257]  W. Williams,et al.  Survival implications: hypertrophic cardiomyopathy in Noonan syndrome. , 2011, Congenital heart disease.

[258]  Yongchao Ge,et al.  Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome , 2010 .

[259]  Rony Seger,et al.  The MAP kinase signaling cascades: a system of hundreds of components regulates a diverse array of physiological functions. , 2010, Methods in molecular biology.

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

[261]  J. Tanti,et al.  Deficiency in the extracellular signal-regulated kinase 1 (ERK1) protects leptin-deficient mice from insulin resistance without affecting obesity , 2010, Diabetologia.

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

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

[264]  K. Kawakami,et al.  Expression of H-RASV12 in a zebrafish model of Costello syndrome causes cellular senescence in adult proliferating cells , 2009, Disease Models & Mechanisms.

[265]  大石 公彦 Transgenic Drosophila models of Noonan syndrome causing PTPN11 gain-of-function mutations , 2008 .

[266]  J. Gregory,et al.  Response to Growth Hormone Treatment and Final Height in Noonan Syndrome in a Large Cohort of Patients in the KIGS Database , 2008, Journal of pediatric endocrinology & metabolism : JPEM.

[267]  C. Sweep,et al.  Impaired Sertoli Cell Function in Males Diagnosed with Noonan Syndrome , 2008, Journal of pediatric endocrinology & metabolism : JPEM.

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

[269]  M. Vidaud,et al.  Noonan syndrome: relationships between genotype, growth, and growth factors. , 2006, The Journal of clinical endocrinology and metabolism.

[270]  C. Dani,et al.  The Extracellular Signal-Regulated Kinase Isoform ERK1 Is Specifically Required for In Vitro and In Vivo , 2005 .

[271]  J. Epstein,et al.  Nf1 has an essential role in endothelial cells , 2003, Nature Genetics.