Skip to main content

Role of PTPN11 (SHP2) in Cancer

  • Chapter
  • First Online:
Protein Tyrosine Phosphatases in Cancer

Abstract

Src homology-2 domain-containing phosphatase 2 (SHP2), encoded by the PTPN11 gene, is a highly conserved, non-transmembrane protein-tyrosine phosphatase (PTP), found in all metazoans. The molecular details of SHP2 regulation by phosphotyrosyl (pTyr) peptide ligand binding are well-understood, and knowledge of these details is critical to understanding SHP2 function in health and disease. Studies using mice with gain- or loss-of-function alleles of Ptpn11 have provided much detail about the physiological functions and signaling pathways regulated by SHP2 at the cellular and whole organism levels. Germline mutations in PTPN11 cause Noonan syndrome, Noonan syndrome with multiple lentigines (previously, LEOPARD syndrome), as well as the cartilage tumor syndrome, metachondromatosis. Somatic PTPN11 mutations occur in several types of hematologic malignancy, most notably juvenile myelomonocytic leukemia and, more rarely, in neuroblastoma and other solid tumors. PTPN11 is crucial for transformation initiated by mutant receptor-tyrosine kinases (RTKs) and is an important effector of H. pylori virulence. However, the direct target(s) of SHP2 responsible for its physiological and pathological effects remain controversial and their identification remains a major goal for the future research.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Bonetti M, Rodriguez-Martinez V, Paardekooper Overman J, Overvoorde J, van Eekelen M, Jopling C, Hertog J. Distinct and overlapping functions of ptpn11 genes in Zebrafish development. PLoS One. 2014;9(4), e94884. doi:10.1371/journal.pone.0094884.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Neel BG, Chan G, Dhanji S. SH2 domain-containing protein-tyrosine phosphatases. In: Neel BG, editor. Handbook of Cell Signaling. 2nd ed. Oxford: Academic; 2009. p. 771–810.

    Google Scholar 

  3. Tsutsumi R, Masoudi M, Takahashi A, Fujii Y, Hayashi T, Kikuchi I, Satou Y, Taira M, Hatakeyama M. YAP and TAZ, Hippo signaling targets, act as a rheostat for nuclear SHP2 function. Dev Cell. 2013;26(6):658–65. doi:10.1016/j.devcel.2013.08.013.

    Article  CAS  PubMed  Google Scholar 

  4. Freeman Jr RM, Plutzky J, Neel BG. Identification of a human src homology 2-containing protein-tyrosine-phosphatase: a putative homolog of Drosophila corkscrew. Proc Natl Acad Sci U S A. 1992;89(23):11239–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Feng GS, Hui CC, Pawson T. SH2-containing phosphotyrosine phosphatase as a target of protein-tyrosine kinases. Science. 1993;259(5101):1607–11.

    Article  CAS  PubMed  Google Scholar 

  6. Ahmad S, Banville D, Zhao Z, Fischer EH, Shen SH. A widely expressed human protein-tyrosine phosphatase containing src homology 2 domains. Proc Natl Acad Sci U S A. 1993;90(6):2197–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Perkins LA, Larsen I, Perrimon N. Corkscrew encodes a putative protein tyrosine phosphatase that functions to transduce the terminal signal from the receptor tyrosine kinase torso. Cell. 1992;70(2):225–36.

    Article  CAS  PubMed  Google Scholar 

  8. Ravetch JV, Lanier LL. Immune inhibitory receptors. Science. 2000;290(5489):84–9.

    Article  CAS  PubMed  Google Scholar 

  9. Hof P, Pluskey S, Dhe-Paganon S, Eck MJ, Shoelson SE. Crystal structure of the tyrosine phosphatase SHP-2. Cell. 1998;92(4):441–50.

    Article  CAS  PubMed  Google Scholar 

  10. Barford D, Neel BG. Revealing mechanisms for SH2 domain mediated regulation of the protein tyrosine phosphatase SHP-2. Structure. 1998;6(3):249–54.

    Article  CAS  PubMed  Google Scholar 

  11. Neel BG, Gu H, Pao L. The Shping news: SH2 domain-containing tyrosine phosphatases in cell signaling. Trends Biochem Sci. 2003;28(6):284–93.

    Article  CAS  PubMed  Google Scholar 

  12. O’Reilly AM, Pluskey S, Shoelson SE, Neel BG. Activated mutants of SHP-2 preferentially induce elongation of Xenopus animal caps. Mol Cell Biol. 2000;20(1):299–311.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Shi ZQ, Lu W, Feng GS. The Shp-2 tyrosine phosphatase has opposite effects in mediating the activation of extracellular signal-regulated and c-Jun NH2-terminal mitogen-activated protein kinases. J Biol Chem. 1998;273(9):4904–8.

    Article  CAS  PubMed  Google Scholar 

  14. Zhang SQ, Tsiaras WG, Araki T, Wen G, Minichiello L, Klein R, Neel BG. Receptor-specific regulation of phosphatidylinositol 3’-kinase activation by the protein tyrosine phosphatase Shp2. Mol Cell Biol. 2002;22(12):4062–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Saxton TM, Henkemeyer M, Gasca S, Shen R, Rossi DJ, Shalaby F, Feng GS, Pawson T. Abnormal mesoderm patterning in mouse embryos mutant for the SH2 tyrosine phosphatase Shp-2. EMBO J. 1997;16(9):2352–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Shi ZQ, Yu DH, Park M, Marshall M, Feng GS. Molecular mechanism for the Shp-2 tyrosine phosphatase function in promoting growth factor stimulation of Erk activity. Mol Cell Biol. 2000;20(5):1526–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Holgado-Madruga M, Emlet DR, Moscatello DK, Godwin AK, Wong AJ. A Grb2-associated docking protein in EGF- and insulin-receptor signalling. Nature. 1996;379(6565):560–4. doi:10.1038/379560a0.

    Article  CAS  PubMed  Google Scholar 

  18. Weidner KM, Di Cesare S, Sachs M, Brinkmann V, Behrens J, Birchmeier W. Interaction between Gab1 and the c-Met receptor tyrosine kinase is responsible for epithelial morphogenesis. Nature. 1996;384(6605):173–6. doi:10.1038/384173a0.

    Article  CAS  PubMed  Google Scholar 

  19. White MF, Yenush L. The IRS-signaling system: a network of docking proteins that mediate insulin and cytokine action. Curr Top Microbiol Immunol. 1998;228:179–208.

    CAS  PubMed  Google Scholar 

  20. Kouhara H, Hadari YR, Spivak-Kroizman T, Schilling J, Bar-Sagi D, Lax I, Schlessinger J. A lipid-anchored Grb2-binding protein that links FGF-receptor activation to the Ras/MAPK signaling pathway. Cell. 1997;89(5):693–702.

    Article  CAS  PubMed  Google Scholar 

  21. Itoh M, Yoshida Y, Nishida K, Narimatsu M, Hibi M, Hirano T. Role of Gab1 in heart, placenta, and skin development and growth factor- and cytokine-induced extracellular signal-regulated kinase mitogen-activated protein kinase activation. Mol Cell Biol. 2000;20(10):3695–704.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sachs M, Brohmann H, Zechner D, Muller T, Hulsken J, Walther I, Schaeper U, Birchmeier C, Birchmeier W. Essential role of Gab1 for signaling by the c-Met receptor in vivo. J Cell Biol. 2000;150(6):1375–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hadari YR, Gotoh N, Kouhara H, Lax I, Schlessinger J. Critical role for the docking-protein FRS2 alpha in FGF receptor-mediated signal transduction pathways. Proc Natl Acad Sci U S A. 2001;98(15):8578–83. doi:10.1073/pnas.161259898.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Maroun CR, Naujokas MA, Holgado-Madruga M, Wong AJ, Park M. The tyrosine phosphatase SHP-2 is required for sustained activation of extracellular signal-regulated kinase and epithelial morphogenesis downstream from the met receptor tyrosine kinase. Mol Cell Biol. 2000;20(22):8513–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Schaeper U, Gehring NH, Fuchs KP, Sachs M, Kempkes B, Birchmeier W. Coupling of Gab1 to c-Met, Grb2, and Shp2 mediates biological responses. J Cell Biol. 2000;149(7):1419–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Noguchi T, Matozaki T, Horita K, Fujioka Y, Kasuga M. Role of SH-PTP2, a protein-tyrosine phosphatase with Src homology 2 domains, in insulin-stimulated Ras activation. Mol Cell Biol. 1994;14(10):6674–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang SQ, Yang W, Kontaridis MI, Bivona TG, Wen G, Araki T, Luo J, Thompson JA, Schraven BL, Philips MR, Neel BG. Shp2 regulates SRC family kinase activity and Ras/Erk activation by controlling Csk recruitment. Mol Cell. 2004;13(3):341–55.

    Article  PubMed  Google Scholar 

  28. Yang W, Klaman LD, Chen B, Araki T, Harada H, Thomas SM, George EL, Neel BG. An Shp2/SFK/Ras/Erk signaling pathway controls trophoblast stem cell survival. Dev Cell. 2006;10(3):317–27.

    Article  CAS  PubMed  Google Scholar 

  29. Bennett AM, Tang TL, Sugimoto S, Walsh CT, Neel BG. Protein-tyrosine-phosphatase SHPTP2 couples platelet-derived growth factor receptor beta to Ras. Proc Natl Acad Sci U S A. 1994;91(15):7335–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Li W, Nishimura R, Kashishian A, Batzer AG, Kim WJ, Cooper JA, Schlessinger J. A new function for a phosphotyrosine phosphatase: linking GRB2-Sos to a receptor tyrosine kinase. Mol Cell Biol. 1994;14(1):509–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Welham MJ, Dechert U, Leslie KB, Jirik F, Schrader JW. Interleukin (IL)-3 and granulocyte/macrophage colony-stimulating factor, but not IL-4, induce tyrosine phosphorylation, activation, and association of SHPTP2 with Grb2 and phosphatidylinositol 3'-kinase. J Biol Chem. 1994;269(38):23764–8.

    CAS  PubMed  Google Scholar 

  32. Lu W, Gong D, Bar-Sagi D, Cole PA. Site-specific incorporation of a phosphotyrosine mimetic reveals a role for tyrosine phosphorylation of SHP-2 in cell signaling. Mol Cell. 2001;8(4):759–69.

    Article  CAS  PubMed  Google Scholar 

  33. Sun J, Lu S, Ouyang M, Lin LJ, Zhuo Y, Liu B, Chien S, Neel BG, Wang Y. Antagonism between binding site affinity and conformational dynamics tunes alternative cis-interactions within Shp2. Nat Commun. 2013;4:2037. doi:10.1038/ncomms3037.

    PubMed  PubMed Central  Google Scholar 

  34. Vogel W, Ullrich A. Multiple in vivo phosphorylated tyrosine phosphatase SHP-2 engages binding to Grb2 via tyrosine 584. Cell Growth Differ. 1996;7(12):1589–97.

    CAS  PubMed  Google Scholar 

  35. Klinghoffer RA, Kazlauskas A. Identification of a putative Syp substrate, the PDGF beta receptor. J Biol Chem. 1995;270(38):22208–17.

    Article  CAS  PubMed  Google Scholar 

  36. Cleghon V, Feldmann P, Ghiglione C, Copeland TD, Perrimon N, Hughes DA, Morrison DK. Opposing actions of CSW and RasGAP modulate the strength of Torso RTK signaling in the Drosophila terminal pathway. Mol Cell. 1998;2(6):719–27.

    Article  CAS  PubMed  Google Scholar 

  37. Agazie YM, Hayman MJ. Molecular mechanism for a role of SHP2 in epidermal growth factor receptor signaling. Mol Cell Biol. 2003;23(21):7875–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhou X, Agazie YM. Molecular mechanism for SHP2 in promoting HER2-induced signaling and transformation. J Biol Chem. 2009;284(18):12226–34. doi:10.1074/jbc.M900020200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dankort DL, Wang Z, Blackmore V, Moran MF, Muller WJ. Distinct tyrosine autophosphorylation sites negatively and positively modulate neu-mediated transformation. Mol Cell Biol. 1997;17(9):5410–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Montagner A, Yart A, Dance M, Perret B, Salles JP, Raynal P. A novel role for Gab1 and SHP2 in epidermal growth factor-induced Ras activation. J Biol Chem. 2005;280(7):5350–60.

    Article  CAS  PubMed  Google Scholar 

  41. Yamasaki S, Nishida K, Yoshida Y, Itoh M, Hibi M, Hirano T. Gab1 is required for EGF receptor signaling and the transformation by activated ErbB2. Oncogene. 2003;22(10):1546–56. doi:10.1038/sj.onc.1206284.

    Article  CAS  PubMed  Google Scholar 

  42. Herbst R, Zhang X, Qin J, Simon MA. Recruitment of the protein tyrosine phosphatase CSW by DOS is an essential step during signaling by the sevenless receptor tyrosine kinase. EMBO J. 1999;18(24):6950–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Mattoon DR, Lamothe B, Lax I, Schlessinger J. The docking protein Gab1 is the primary mediator of EGF-stimulated activation of the PI-3K/Akt cell survival pathway. BMC Biol. 2004;2:24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Bunda S, Heir P, Srikumar T, Cook JD, Burrell K, Kano Y, Lee JE, Zadeh G, Raught B, Ohh M. Src promotes GTPase activity of Ras via tyrosine 32 phosphorylation. Proc Natl Acad Sci U S A. 2014;111(36):E3785–94. doi:10.1073/pnas.1406559111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Cunnick JM, Meng S, Ren Y, Desponts C, Wang HG, Djeu JY, Wu J. Regulation of the mitogen-activated protein kinase signaling pathway by SHP2. J Biol Chem. 2002;277(11):9498–504. doi:10.1074/jbc.M110547200.

    Article  CAS  PubMed  Google Scholar 

  46. Wu CJ, O’Rourke DM, Feng GS, Johnson GR, Wang Q, Greene MI. The tyrosine phosphatase SHP-2 is required for mediating phosphatidylinositol 3-kinase/Akt activation by growth factors. Oncogene. 2001;20(42):6018–25.

    Article  CAS  PubMed  Google Scholar 

  47. Bertotti A, Comoglio PM, Trusolino L. Beta4 integrin activates a Shp2-Src signaling pathway that sustains HGF-induced anchorage-independent growth. J Cell Biol. 2006;175(6):993–1003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ren Y, Meng S, Mei L, Zhao ZJ, Jove R, Wu J. Roles of Gab1 and SHP2 in paxillin tyrosine dephosphorylation and Src activation in response to epidermal growth factor. J Biol Chem. 2004;279(9):8497–505.

    Article  CAS  PubMed  Google Scholar 

  49. Hanafusa H, Torii S, Yasunaga T, Matsumoto K, Nishida E. Shp2, an SH2-containing protein-tyrosine phosphatase, positively regulates receptor tyrosine kinase signaling by dephosphorylating and inactivating the inhibitor Sprouty. J Biol Chem. 2004;279(22):22992–5.

    Article  CAS  PubMed  Google Scholar 

  50. Jarvis LA, Toering SJ, Simon MA, Krasnow MA, Smith-Bolton RK. Sprouty proteins are in vivo targets of Corkscrew/SHP-2 tyrosine phosphatases. Development. 2006;133(6):1133–42.

    Article  CAS  PubMed  Google Scholar 

  51. Pan Y, Carbe C, Powers A, Feng GS, Zhang X. Sprouty2-modulated Kras signaling rescues Shp2 deficiency during lens and lacrimal gland development. Development. 2010;137(7):1085–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mason JM, Morrison DJ, Basson MA, Licht JD. Sprouty proteins: multifaceted negative-feedback regulators of receptor tyrosine kinase signaling. Trends Cell Biol. 2006;16(1):45–54.

    Article  CAS  PubMed  Google Scholar 

  53. Wakioka T, Sasaki A, Kato R, Shouda T, Matsumoto A, Miyoshi K, Tsuneoka M, Komiya S, Baron R, Yoshimura A. Spred is a Sprouty-related suppressor of Ras signalling. Nature. 2001;412(6847):647–51. doi:10.1038/35088082.

    Article  CAS  PubMed  Google Scholar 

  54. Kato R, Nonami A, Taketomi T, Wakioka T, Kuroiwa A, Matsuda Y, Yoshimura A. Molecular cloning of mammalian Spred-3 which suppresses tyrosine kinase-mediated Erk activation. Biochem Biophys Res Commun. 2003;302(4):767–72.

    Article  CAS  PubMed  Google Scholar 

  55. Nonami A, Kato R, Taniguchi K, Yoshiga D, Taketomi T, Fukuyama S, Harada M, Sasaki A, Yoshimura A. Spred-1 negatively regulates interleukin-3-mediated ERK/mitogen-activated protein (MAP) kinase activation in hematopoietic cells. J Biol Chem. 2004;279(50):52543–51. doi:10.1074/jbc.M405189200.

    Article  CAS  PubMed  Google Scholar 

  56. Taniguchi K, Kohno R, Ayada T, Kato R, Ichiyama K, Morisada T, Oike Y, Yonemitsu Y, Maehara Y, Yoshimura A. Spreds are essential for embryonic lymphangiogenesis by regulating vascular endothelial growth factor receptor 3 signaling. Mol Cell Biol. 2007;27(12):4541–50. doi:10.1128/MCB.01600-06.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Quintanar-Audelo M, Yusoff P, Sinniah S, Chandramouli S, Guy GR. Sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein (SPRED1), a tyrosine-protein phosphatase non-receptor type 11 (SHP2) substrate in the Ras/extracellular signal-regulated kinase (ERK) pathway. J Biol Chem. 2011;286(26):23102–12. doi:10.1074/jbc.M110.212662.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Brems H, Chmara M, Sahbatou M, Denayer E, Taniguchi K, Kato R, Somers R, Messiaen L, De Schepper S, Fryns JP, Cools J, Marynen P, Thomas G, Yoshimura A, Legius E. Germline loss-of-function mutations in SPRED1 cause a neurofibromatosis 1-like phenotype. Nat Genet. 2007;39(9):1120–6.

    Article  CAS  PubMed  Google Scholar 

  59. Spurlock G, Bennett E, Chuzhanova N, Thomas N, Jim HP, Side L, Davies S, Haan E, Kerr B, Huson SM, Upadhyaya M. SPRED1 mutations (Legius syndrome): another clinically useful genotype for dissecting the neurofibromatosis type 1 phenotype. J Med Genet. 2009;46(7):431–7. doi:10.1136/jmg.2008.065474.

    Article  CAS  PubMed  Google Scholar 

  60. Zito CI, Qin H, Blenis J, Bennett AM. SHP-2 regulates cell growth by controlling the mTOR/S6 kinase 1 pathway. J Biol Chem. 2007;282(10):6946–53.

    Article  CAS  PubMed  Google Scholar 

  61. Yu M, Luo J, Yang W, Wang Y, Mizuki M, Kanakura Y, Besmer P, Neel BG, Gu H. The scaffolding adapter Gab2, via Shp-2, regulates kit-evoked mast cell proliferation by activating the Rac/JNK pathway. J Biol Chem. 2006;281(39):28615–26.

    Article  CAS  PubMed  Google Scholar 

  62. Yang Z, Li Y, Yin F, Chan RJ. Activating PTPN11 mutants promote hematopoietic progenitor cell-cycle progression and survival. Exp Hematol. 2008;36(10):1285–96. doi:10.1016/j.exphem.2008.04.016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. You M, Flick LM, Yu D, Feng GS. Modulation of the nuclear factor kappa B pathway by Shp-2 tyrosine phosphatase in mediating the induction of interleukin (IL)-6 by IL-1 or tumor necrosis factor. J Exp Med. 2001;193(1):101–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Schoenwaelder SM, Petch LA, Williamson D, Shen R, Feng GS, Burridge K. The protein tyrosine phosphatase Shp-2 regulates RhoA activity. Curr Biol. 2000;10(23):1523–6.

    Article  CAS  PubMed  Google Scholar 

  65. Kontaridis MI, Eminaga S, Fornaro M, Zito CI, Sordella R, Settleman J, Bennett AM. SHP-2 positively regulates myogenesis by coupling to the Rho GTPase signaling pathway. Mol Cell Biol. 2004;24(12):5340–52. doi:10.1128/MCB.24.12.5340-5352.2004.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Uhlen P, Burch PM, Zito CI, Estrada M, Ehrlich BE, Bennett AM. Gain-of-function/Noonan syndrome SHP-2/Ptpn11 mutants enhance calcium oscillations and impair NFAT signaling. Proc Natl Acad Sci U S A. 2006;103(7):2160–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Paardekooper Overman J, Preisinger C, Prummel K, Bonetti M, Giansanti P, Heck A, den Hertog J. Phosphoproteomics-mediated identification of Fer kinase as a target of mutant Shp2 in Noonan and LEOPARD syndrome. PLoS One. 2014;9(9), e106682. doi:10.1371/journal.pone.0106682.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Mitra S, Beach C, Feng GS, Plattner R. SHP-2 is a novel target of Abl kinases during cell proliferation. J Cell Sci. 2008;121(Pt 20):3335–46. doi:10.1242/jcs.035691.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kontaridis MI, Swanson KD, David FS, Barford D, Neel BG. PTPN11 (Shp2) mutations in LEOPARD syndrome have dominant negative, not activating, effects. J Biol Chem. 2006;281(10):6785–92.

    Article  CAS  PubMed  Google Scholar 

  70. Tartaglia M, Martinelli S, Stella L, Bocchinfuso G, Flex E, Cordeddu V, Zampino G, Burgt I, Palleschi A, Petrucci TC, Sorcini M, Schoch C, Foa R, Emanuel PD, Gelb BD. Diversity and functional consequences of germline and somatic PTPN11 mutations in human disease. Am J Hum Genet. 2006;78(2):279–90.

    Article  CAS  PubMed  Google Scholar 

  71. Hanna N, Montagner A, Lee WH, Miteva M, Vidal M, Vidaud M, Parfait B, Raynal P. Reduced phosphatase activity of SHP-2 in LEOPARD syndrome: consequences for PI3K binding on Gab1. FEBS Lett. 2006;580(10):2477–82.

    Article  CAS  PubMed  Google Scholar 

  72. Zou GM, Chan RJ, Shelley WC, Yoder MC. Reduction of Shp-2 expression by small interfering RNA reduces murine embryonic stem cell-derived in vitro hematopoietic differentiation. Stem Cells. 2006;24(3):587–94.

    Article  CAS  PubMed  Google Scholar 

  73. Chan RJ, Johnson SA, Li Y, Yoder MC, Feng GS. A definitive role of Shp-2 tyrosine phosphatase in mediating embryonic stem cell differentiation and hematopoiesis. Blood. 2003;102(6):2074–80.

    Article  CAS  PubMed  Google Scholar 

  74. Qu CK, Shi ZQ, Shen R, Tsai FY, Orkin SH, Feng GS. A deletion mutation in the SH2-N domain of Shp-2 severely suppresses hematopoietic cell development. Mol Cell Biol. 1997;17(9):5499–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Qu CK, Yu WM, Azzarelli B, Cooper S, Broxmeyer HE, Feng GS. Biased suppression of hematopoiesis and multiple developmental defects in chimeric mice containing Shp-2 mutant cells. Mol Cell Biol. 1998;18(10):6075–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Qu CK, Nguyen S, Chen J, Feng GS. Requirement of Shp-2 tyrosine phosphatase in lymphoid and hematopoietic cell development. Blood. 2001;97(4):911–4.

    Article  CAS  PubMed  Google Scholar 

  77. Chan RJ, Li Y, Hass MN, Walter A, Voorhorst CS, Shelley WC, Yang Z, Orschell CM, Yoder MC. Shp-2 heterozygous hematopoietic stem cells have deficient repopulating ability due to diminished self-renewal. Exp Hematol. 2006;34(9):1230–9.

    Article  CAS  PubMed  Google Scholar 

  78. Chan G, Cheung LS, Yang W, Milyavsky M, Sanders AD, Gu S, Hong WX, Liu AX, Wang X, Barbara M, Sharma T, Gavin J, Kutok JL, Iscove NN, Shannon KM, Dick JE, Neel BG, Braun BS. Essential role for Ptpn11 in survival of hematopoietic stem and progenitor cells. Blood. 2011;117(16):4253–61. doi:10.1182/blood-2010-11-319517.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhu HH, Ji K, Alderson N, He Z, Li S, Liu W, Zhang DE, Li L, Feng GS. Kit-Shp2-Kit signaling acts to maintain a functional hematopoietic stem and progenitor cell pool. Blood. 2011;117(20):5350–61. doi:10.1182/blood-2011-01-333476.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Li L, Modi H, McDonald T, Rossi J, Yee JK, Bhatia R. A critical role for SHP2 in STAT5 activation and growth factor-mediated proliferation, survival, and differentiation of human CD34+ cells. Blood. 2011;118(6):1504–15. doi:10.1182/blood-2010-06-288910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Huang W, Saberwal G, Horvath E, Zhu C, Lindsey S, Eklund EA. Leukemia-associated, constitutively active mutants of SHP2 protein tyrosine phosphatase inhibit NF1 transcriptional activation by the interferon consensus sequence binding protein. Mol Cell Biol. 2006;26(17):6311–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Huang H, Woo AJ, Waldon Z, Schindler Y, Moran TB, Zhu HH, Feng GS, Steen H, Cantor AB. A Src family kinase-Shp2 axis controls RUNX1 activity in megakaryocyte and T-lymphocyte differentiation. Genes Dev. 2012;26(14):1587–601. doi:10.1101/gad.192054.112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Holtschke T, Lohler J, Kanno Y, Fehr T, Giese N, Rosenbauer F, Lou J, Knobeloch KP, Gabriele L, Waring JF, Bachmann MF, Zinkernagel RM, Morse 3rd HC, Ozato K, Horak I. Immunodeficiency and chronic myelogenous leukemia-like syndrome in mice with a targeted mutation of the ICSBP gene. Cell. 1996;87(2):307–17.

    Article  CAS  PubMed  Google Scholar 

  84. Konieczna I, Horvath E, Wang H, Lindsey S, Saberwal G, Bei L, Huang W, Platanias L, Eklund EA. Constitutive activation of SHP2 in mice cooperates with ICSBP deficiency to accelerate progression to acute myeloid leukemia. J Clin Invest. 2008;118(3):853–67.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Growney JD, Shigematsu H, Li Z, Lee BH, Adelsperger J, Rowan R, Curley DP, Kutok JL, Akashi K, Williams IR, Speck NA, Gilliland DG. Loss of Runx1 perturbs adult hematopoiesis and is associated with a myeloproliferative phenotype. Blood. 2005;106(2):494–504. doi:10.1182/blood-2004-08-3280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ichikawa M, Asai T, Saito T, Seo S, Yamazaki I, Yamagata T, Mitani K, Chiba S, Ogawa S, Kurokawa M, Hirai H. AML-1 is required for megakaryocytic maturation and lymphocytic differentiation, but not for maintenance of hematopoietic stem cells in adult hematopoiesis. Nat Med. 2004;10(3):299–304. doi:10.1038/nm997.

    Article  CAS  PubMed  Google Scholar 

  87. Cai X, Gaudet JJ, Mangan JK, Chen MJ, De Obaldia ME, Oo Z, Ernst P, Speck NA. Runx1 loss minimally impacts long-term hematopoietic stem cells. PLoS One. 2011;6(12), e28430. doi:10.1371/journal.pone.0028430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Staser K, Park SJ, Rhodes SD, Zeng Y, He YZ, Shew MA, Gehlhausen JR, Cerabona D, Menon K, Chen S, Sun Z, Yuan J, Ingram DA, Nalepa G, Yang FC, Clapp DW. Normal hematopoiesis and neurofibromin-deficient myeloproliferative disease require Erk. J Clin Invest. 2012. doi:10.1172/JCI66167.

    PubMed  PubMed Central  Google Scholar 

  89. Chan G, Gu S, Neel BG. Erk1 and Erk2 are required for maintenance of hematopoietic stem cells and adult hematopoiesis. Blood. 2013;121(18):3594–8. doi:10.1182/blood-2012-12-476200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Takahashi A, Tsutsumi R, Kikuchi I, Obuse C, Saito Y, Seidi A, Karisch R, Fernandez M, Cho T, Ohnishi N, Rozenblatt-Rosen O, Meyerson M, Neel BG, Hatakeyama M. SHP2 tyrosine phosphatase converts parafibromin/Cdc73 from a tumor suppressor to an oncogenic driver. Mol Cell. 2011;43(1):45–56. doi:10.1016/j.molcel.2011.05.014.

    Article  CAS  PubMed  Google Scholar 

  91. Roberts AE, Allanson JE, Tartaglia M, Gelb BD. Noonan syndrome. Lancet. 2013;381(9863):333–42. doi:10.1016/S0140-6736(12)61023-X.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Noonan JA. Hypertelorism with Turner phenotype. A new syndrome with associated congenital heart disease. Am J Dis Child. 1968;116(4):373–80.

    Article  CAS  PubMed  Google Scholar 

  93. Noonan JA. Noonan syndrome. An update and review for the primary pediatrician. Clin Pediatr. 1994;33(9):548–55.

    Article  CAS  Google Scholar 

  94. Noonan JA. Noonan syndrome revisited. J Pediatr. 1999;135(6):667–8.

    Article  CAS  PubMed  Google Scholar 

  95. Choong K, Freedman MH, Chitayat D, Kelly EN, Taylor G, Zipursky A. Juvenile myelomonocytic leukemia and Noonan syndrome. J Pediatr Hematol Oncol. 1999;21(6):523–7.

    Article  CAS  PubMed  Google Scholar 

  96. Bader-Meunier B, Tchernia G, Mielot F, Fontaine JL, Thomas C, Lyonnet S, Lavergne JM, Dommergues JP. Occurrence of myeloproliferative disorder in patients with Noonan syndrome. J Pediatr. 1997;130(6):885–9.

    Article  CAS  PubMed  Google Scholar 

  97. Attard-Montalto SP, Kingston JE, Eden T. Noonan’s syndrome and acute lymphoblastic leukaemia. Med Pediatr Oncol. 1994;23(4):391–2.

    Article  CAS  PubMed  Google Scholar 

  98. Klopfenstein KJ, Sommer A, Ruymann FB. Neurofibromatosis-Noonan syndrome and acute lymphoblastic leukemia: a report of two cases. J Pediatr Hematol Oncol. 1999;21(2):158–60.

    Article  CAS  PubMed  Google Scholar 

  99. Schubbert S, Zenker M, Rowe SL, Boll S, Klein C, Bollag G, van der Burgt I, Musante L, Kalscheuer V, Wehner LE, Nguyen H, West B, Zhang KY, Sistermans E, Rauch A, Niemeyer CM, Shannon K, Kratz CP. Germline KRAS mutations cause Noonan syndrome. Nat Genet. 2006;38(3):331–6.

    Article  CAS  PubMed  Google Scholar 

  100. Carta C, Pantaleoni F, Bocchinfuso G, Stella L, Vasta I, Sarkozy A, Digilio C, Palleschi A, Pizzuti A, Grammatico P, Zampino G, Dallapiccola B, Gelb BD, Tartaglia M. Germline missense mutations affecting KRAS Isoform B are associated with a severe Noonan syndrome phenotype. Am J Hum Genet. 2006;79(1):129–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Kraoua L, Journel H, Bonnet P, Amiel J, Pouvreau N, Baumann C, Verloes A, Cave H. Constitutional NRAS mutations are rare among patients with Noonan syndrome or juvenile myelomonocytic leukemia. Am J Med Genet. 2012;158A(10):2407–11. doi:10.1002/ajmg.a.35513.

    Article  PubMed  CAS  Google Scholar 

  102. Cirstea IC, Kutsche K, Dvorsky R, Gremer L, Carta C, Horn D, Roberts AE, Lepri F, Merbitz-Zahradnik T, Konig R, Kratz CP, Pantaleoni F, Dentici ML, Joshi VA, Kucherlapati RS, Mazzanti L, Mundlos S, Patton MA, Silengo MC, Rossi C, Zampino G, Digilio C, Stuppia L, Seemanova E, Pennacchio LA, Gelb BD, Dallapiccola B, Wittinghofer A, Ahmadian MR, Tartaglia M, Zenker M. A restricted spectrum of NRAS mutations causes Noonan syndrome. Nat Genet. 2010;42(1):27–9. doi:10.1038/ng.497.

    Article  CAS  PubMed  Google Scholar 

  103. Roberts AE, Araki T, Swanson KD, Montgomery KT, Schiripo TA, Joshi VA, Li L, Yassin Y, Tamburino AM, Neel BG, Kucherlapati RS. Germline gain-of-function mutations in SOS1 cause Noonan syndrome. Nat Genet. 2007;39(1):70–4.

    Article  CAS  PubMed  Google Scholar 

  104. Tartaglia M, Pennacchio LA, Zhao C, Yadav KK, Fodale V, Sarkozy A, Pandit B, Oishi K, Martinelli S, Schackwitz W, Ustaszewska A, Martin J, Bristow J, Carta C, Lepri F, Neri C, Vasta I, Gibson K, Curry CJ, Siguero JP, Digilio MC, Zampino G, Dallapiccola B, Bar-Sagi D, Gelb BD. Gain-of-function SOS1 mutations cause a distinctive form of Noonan syndrome. Nat Genet. 2007;39(1):75–9.

    Article  CAS  PubMed  Google Scholar 

  105. Razzaque MA, Nishizawa T, Komoike Y, Yagi H, Furutani M, Amo R, Kamisago M, Momma K, Katayama H, Nakagawa M, Fujiwara Y, Matsushima M, Mizuno K, Tokuyama M, Hirota H, Muneuchi J, Higashinakagawa T, Matsuoka R. Germline gain-of-function mutations in RAF1 cause Noonan syndrome. 2007. Nat Genet.

    Google Scholar 

  106. Pandit B, Sarkozy A, Pennacchio LA, Carta C, Oishi K, Martinelli S, Pogna EA, Schackwitz W, Ustaszewska A, Landstrom A, Bos JM, Ommen SR, Esposito G, Lepri F, Faul C, Mundel P, Lopez Siguero JP, Tenconi R, Selicorni A, Rossi C, Mazzanti L, Torrente I, Marino B, Digilio MC, Zampino G, Ackerman MJ, Dallapiccola B, Tartaglia M, Gelb BD. Gain-of-function RAF1 mutations cause Noonan and LEOPARD syndromes with hypertrophic cardiomyopathy. 2007. Nat Genet.

    Google Scholar 

  107. Aoki Y, Niihori T, Banjo T, Okamoto N, Mizuno S, Kurosawa K, Ogata T, Takada F, Yano M, Ando T, Hoshika T, Barnett C, Ohashi H, Kawame H, Hasegawa T, Okutani T, Nagashima T, Hasegawa S, Funayama R, Nagashima T, Nakayama K, Inoue S, Watanabe Y, Ogura T, Matsubara Y. Gain-of-Function Mutations in RIT1 Cause Noonan Syndrome, a RAS/MAPK Pathway Syndrome. Am J Hum Genet. 2013;93(1):173–80. doi:10.1016/j.ajhg.2013.05.021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Chen PC, Yin J, Yu HW, Yuan T, Fernandez M, Yung CK, Trinh QM, Peltekova VD, Reid JG, Tworog-Dube E, Morgan MB, Muzny DM, Stein L, McPherson JD, Roberts AE, Gibbs RA, Neel BG, Kucherlapati R. Next-generation sequencing identifies rare variants associated with Noonan syndrome. Proc Natl Acad Sci U S A. 2014;111(31):11473–8. doi:10.1073/pnas.1324128111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Cordeddu V, Di Schiavi E, Pennacchio LA, Ma’ayan A, Sarkozy A, Fodale V, Cecchetti S, Cardinale A, Martin J, Schackwitz W, Lipzen A, Zampino G, Mazzanti L, Digilio MC, Martinelli S, Flex E, Lepri F, Bartholdi D, Kutsche K, Ferrero GB, Anichini C, Selicorni A, Rossi C, Tenconi R, Zenker M, Merlo D, Dallapiccola B, Iyengar R, Bazzicalupo P, Gelb BD, Tartaglia M. Mutation of SHOC2 promotes aberrant protein N-myristoylation and causes Noonan-like syndrome with loose anagen hair. Nat Genet. 2009;41(9):1022–6. doi:10.1038/ng.425.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Emanuel PD. Juvenile myelomonocytic leukemia and chronic myelomonocytic leukemia. Leukemia. 2008;22(7):1335–42.

    Article  CAS  PubMed  Google Scholar 

  111. Loh ML. Recent advances in the pathogenesis and treatment of juvenile myelomonocytic leukaemia. Br J Haematol. 2011;152(6):677–87. doi:10.1111/j.1365-2141.2010.08525.x.

    Article  CAS  PubMed  Google Scholar 

  112. Chang TY, Dvorak CC, Loh ML. Bedside to bench in juvenile myelomonocytic leukemia: insights into leukemogenesis from a rare pediatric leukemia. Blood. 2014;124(16):2487–97. doi:10.1182/blood-2014-03-300319.

    Article  CAS  PubMed  Google Scholar 

  113. Tartaglia M, Niemeyer CM, Fragale A, Song X, Buechner J, Jung A, Hahlen K, Hasle H, Licht JD, Gelb BD. Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat Genet. 2003;34(2):148–50.

    Article  CAS  PubMed  Google Scholar 

  114. Loh ML, Vattikuti S, Schubbert S, Reynolds MG, Carlson E, Lieuw KH, Cheng JW, Lee CM, Stokoe D, Bonifas JM, Curtiss NP, Gotlib J, Meshinchi S, Le Beau MM, Emanuel PD, Shannon KM. Mutations in PTPN11 implicate the SHP-2 phosphatase in leukemogenesis. Blood. 2004;103(6):2325–31.

    Article  CAS  PubMed  Google Scholar 

  115. Loh ML, Reynolds MG, Vattikuti S, Gerbing RB, Alonzo TA, Carlson E, Cheng JW, Lee CM, Lange BJ, Meshinchi S. PTPN11 mutations in pediatric patients with acute myeloid leukemia: results from the Children’s Cancer Group. Leukemia. 2004;18(11):1831–4.

    Article  CAS  PubMed  Google Scholar 

  116. Bentires-Alj M, Paez JG, David FS, Keilhack H, Halmos B, Naoki K, Maris JM, Richardson A, Bardelli A, Sugarbaker DJ, Richards WG, Du J, Girard L, Minna JD, Loh ML, Fisher DE, Velculescu VE, Vogelstein B, Meyerson M, Sellers WR, Neel BG. Activating mutations of the Noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia. Cancer Res. 2004;64(24):8816–20.

    Article  CAS  PubMed  Google Scholar 

  117. Tartaglia M, Niemeyer CM, Shannon KM, Loh ML. SHP-2 and myeloid malignancies. Curr Opin Hematol. 2004;11(1):44–50.

    Article  CAS  PubMed  Google Scholar 

  118. Yamamoto T, Isomura M, Xu Y, Liang J, Yagasaki H, Kamachi Y, Kudo K, Kiyoi H, Naoe T, Kojma S. PTPN11, RAS and FLT3 mutations in childhood acute lymphoblastic leukemia. Leuk Res. 2006;30(9):1085–9.

    Article  CAS  PubMed  Google Scholar 

  119. Martinelli S, Carta C, Flex E, Binni F, Cordisco EL, Moretti S, Puxeddu E, Tonacchera M, Pinchera A, McDowell HP, Dominici C, Rosolen A, Di Rocco C, Riccardi R, Celli P, Picardo M, Genuardi M, Grammatico P, Sorcini M, Tartaglia M. Activating PTPN11 mutations play a minor role in pediatric and adult solid tumors. Cancer Genet Cytogenet. 2006;166(2):124–9.

    Article  CAS  PubMed  Google Scholar 

  120. Pugh TJ, Morozova O, Attiyeh EF, Asgharzadeh S, Wei JS, Auclair D, Carter SL, Cibulskis K, Hanna M, Kiezun A, Kim J, Lawrence MS, Lichenstein L, McKenna A, Pedamallu CS, Ramos AH, Shefler E, Sivachenko A, Sougnez C, Stewart C, Ally A, Birol I, Chiu R, Corbett RD, Hirst M, Jackman SD, Kamoh B, Khodabakshi AH, Krzywinski M, Lo A, Moore RA, Mungall KL, Qian J, Tam A, Thiessen N, Zhao Y, Cole KA, Diamond M, Diskin SJ, Mosse YP, Wood AC, Ji L, Sposto R, Badgett T, London WB, Moyer Y, Gastier-Foster JM, Smith MA, Guidry Auvil JM, Gerhard DS, Hogarty MD, Jones SJ, Lander ES, Gabriel SB, Getz G, Seeger RC, Khan J, Marra MA, Meyerson M, Maris JM. The genetic landscape of high-risk neuroblastoma. Nat Genet. 2013;45(3):279–84. doi:10.1038/ng.2529.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Keilhack H, David FS, McGregor M, Cantley LC, Neel BG. Diverse biochemical properties of Shp2 mutants. Implications for disease phenotypes. J Biol Chem. 2005;280(35):30984–93.

    Article  CAS  PubMed  Google Scholar 

  122. Niihori T, Aoki Y, Ohashi H, Kurosawa K, Kondoh T, Ishikiriyama S, Kawame H, Kamasaki H, Yamanaka T, Takada F, Nishio K, Sakurai M, Tamai H, Nagashima T, Suzuki Y, Kure S, Fujii K, Imaizumi M, Matsubara Y. Functional analysis of PTPN11/SHP-2 mutants identified in Noonan syndrome and childhood leukemia. J Hum Genet. 2005;50(4):192–202.

    Article  CAS  PubMed  Google Scholar 

  123. Chan RJ, Leedy MB, Munugalavadla V, Voorhorst CS, Li Y, Yu M, Kapur R. Human somatic PTPN11 mutations induce hematopoietic-cell hypersensitivity to granulocyte-macrophage colony-stimulating factor. Blood. 2005;105(9):3737–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Mohi MG, Williams IR, Dearolf CR, Chan G, Kutok JL, Cohen S, Morgan K, Boulton C, Shigematsu H, Keilhack H, Akashi K, Gilliland DG, Neel BG. Prognostic, therapeutic, and mechanistic implications of a mouse model of leukemia evoked by Shp2 (PTPN11) mutations. Cancer Cell. 2005;7(2):179–91.

    Article  CAS  PubMed  Google Scholar 

  125. Schubbert S, Lieuw K, Rowe SL, Lee CM, Li X, Loh ML, Clapp DW, Shannon KM. Functional analysis of leukemia-associated PTPN11 mutations in primary hematopoietic cells. Blood. 2005;106(1):311–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Gandre-Babbe S, Paluru P, Aribeana C, Chou ST, Bresolin S, Lu L, Sullivan SK, Tasian SK, Weng J, Favre H, Choi JK, French DL, Loh ML, Weiss MJ. Patient-derived induced pluripotent stem cells recapitulate hematopoietic abnormalities of juvenile myelomonocytic leukemia. Blood. 2013;121(24):4925–9. doi:10.1182/blood-2013-01-478412.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Schubbert S, Shannon K, Bollag G. Hyperactive Ras in developmental disorders and cancer. Nat Rev Cancer. 2007;7(4):295–308.

    Article  CAS  PubMed  Google Scholar 

  128. Ward AF, Braun BS, Shannon KM. Targeting oncogenic Ras signaling in hematologic malignancies. Blood. 2012;120(17):3397–406. doi:10.1182/blood-2012-05-378596.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Zhang YY, Vik TA, Ryder JW, Srour EF, Jacks T, Shannon K, Clapp DW. Nf1 regulates hematopoietic progenitor cell growth and Ras signaling in response to multiple cytokines. J Exp Med. 1998;187(11):1893–902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Le DT, Kong N, Zhu Y, Lauchle JO, Aiyigari A, Braun BS, Wang E, Kogan SC, Le Beau MM, Parada L, Shannon KM. Somatic inactivation of Nf1 in hematopoietic cells results in a progressive myeloproliferative disorder. Blood. 2004;103(11):4243–50.

    Article  CAS  PubMed  Google Scholar 

  131. Donovan S, See W, Bonifas J, Stokoe D, Shannon KM. Hyperactivation of protein kinase B and ERK have discrete effects on survival, proliferation, and cytokine expression in Nf1-deficient myeloid cells. Cancer Cell. 2002;2(6):507–14.

    Article  CAS  PubMed  Google Scholar 

  132. Largaespada DA, Brannan CI, Jenkins NA, Copeland NG. Nf1 deficiency causes Ras-mediated granulocyte/macrophage colony stimulating factor hypersensitivity and chronic myeloid leukaemia. Nat Genet. 1996;12(2):137–43.

    Article  CAS  PubMed  Google Scholar 

  133. Bollag G, Clapp DW, Shih S, Adler F, Zhang YY, Thompson P, Lange BJ, Freedman MH, McCormick F, Jacks T, Shannon K. Loss of NF1 results in activation of the Ras signaling pathway and leads to aberrant growth in haematopoietic cells. Nat Genet. 1996;12(2):144–8.

    Article  CAS  PubMed  Google Scholar 

  134. Braun BS, Tuveson DA, Kong N, Le DT, Kogan SC, Rozmus J, Le Beau MM, Jacks TE, Shannon KM. Somatic activation of oncogenic Kras in hematopoietic cells initiates a rapidly fatal myeloproliferative disorder. Proc Natl Acad Sci U S A. 2004;101(2):597–602.

    Article  CAS  PubMed  Google Scholar 

  135. Chan IT, Kutok JL, Williams IR, Cohen S, Kelly L, Shigematsu H, Johnson L, Akashi K, Tuveson DA, Jacks T, Gilliland DG. Conditional expression of oncogenic K-ras from its endogenous promoter induces a myeloproliferative disease. J Clin Invest. 2004;113(4):528–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Sabnis AJ, Cheung LS, Dail M, Kang HC, Santaguida M, Hermiston ML, Passegue E, Shannon K, Braun BS. Oncogenic Kras initiates leukemia in hematopoietic stem cells. PLoS Biol. 2009;7(3), e59. doi:10.1371/journal.pbio.1000059.

    Article  PubMed  CAS  Google Scholar 

  137. Zhang J, Wang J, Liu Y, Sidik H, Young KH, Lodish HF, Fleming MD. Oncogenic Kras-induced leukemogeneis: hematopoietic stem cells as the initial target and lineage-specific progenitors as the potential targets for final leukemic transformation. Blood. 2009;113(6):1304–14. doi:10.1182/blood-2008-01-134262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Li Q, Haigis KM, McDaniel A, Harding-Theobald E, Kogan SC, Akagi K, Wong JC, Braun BS, Wolff L, Jacks T, Shannon K. Hematopoiesis and leukemogenesis in mice expressing oncogenic NrasG12D from the endogenous locus. Blood. 2011;117(6):2022–32. doi:10.1182/blood-2010-04-280750.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Wang J, Liu Y, Li Z, Du J, Ryu MJ, Taylor PR, Fleming MD, Young KH, Pitot H, Zhang J. Endogenous oncogenic Nras mutation promotes aberrant GM-CSF signaling in granulocytic/monocytic precursors in a murine model of chronic myelomonocytic leukemia. Blood. 2010;116(26):5991–6002. doi:10.1182/blood-2010-04-281527.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Wang J, Liu Y, Li Z, Wang Z, Tan LX, Ryu MJ, Meline B, Du J, Young KH, Ranheim E, Chang Q, Zhang J. Endogenous oncogenic Nras mutation initiates hematopoietic malignancies in a dose- and cell type-dependent manner. Blood. 2011;118(2):368–79. doi:10.1182/blood-2010-12-326058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Lyubynska N, Gorman MF, Lauchle JO, Hong WX, Akutagawa JK, Shannon K, Braun BS. A MEK inhibitor abrogates myeloproliferative disease in Kras mutant mice. Sci Transl Med. 2011;3(76):76ra27. doi:10.1126/scitranslmed.3001069

    Google Scholar 

  142. Burgess MR, Hwang E, Firestone AJ, Huang T, Xu J, Zuber J, Bohin N, Wen T, Kogan SC, Haigis KM, Sampath D, Lowe S, Shannon K, Li Q. Preclinical efficacy of MEK inhibition in Nras mutant acute myeloid leukemia. Blood. 2014. doi:10.1182/blood-2014-05-574582.

    PubMed  PubMed Central  Google Scholar 

  143. Chang T, Krisman K, Theobald EH, Xu J, Akutagawa J, Lauchle JO, Kogan S, Braun BS, Shannon K. Sustained MEK inhibition abrogates myeloproliferative disease in Nf1 mutant mice. J Clin Invest. 2012. doi:10.1172/JCI63193.

    Google Scholar 

  144. Sakaguchi H, Okuno Y, Muramatsu H, Yoshida K, Shiraishi Y, Takahashi M, Kon A, Sanada M, Chiba K, Tanaka H, Makishima H, Wang X, Xu Y, Doisaki S, Hama A, Nakanishi K, Takahashi Y, Yoshida N, Maciejewski JP, Miyano S, Ogawa S, Kojima S. Exome sequencing identifies secondary mutations of SETBP1 and JAK3 in juvenile myelomonocytic leukemia. Nat Genet. 2013;45(8):937–41. doi:10.1038/ng.2698.

    Article  CAS  PubMed  Google Scholar 

  145. Araki T, Mohi MG, Ismat FA, Bronson RT, Williams IR, Kutok JL, Yang W, Pao LI, Gilliland DG, Epstein JA, Neel BG. Mouse model of Noonan syndrome reveals cell type- and gene dosage-dependent effects of Ptpn11 mutation. Nat Med. 2004;10(8):849–57.

    Article  CAS  PubMed  Google Scholar 

  146. Chan G, Kalaitzidis D, Usenko T, Kutok JL, Yang W, Mohi MG, Neel BG. Leukemogenic Ptpn11 causes fatal myeloproliferative disorder via cell-autonomous effects on multiple stages of hematopoiesis. Blood. 2009;113(18):4414–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Xu D, Liu X, Yu WM, Meyerson HJ, Guo C, Gerson SL, Qu CK. Non-lineage/stage-restricted effects of a gain-of-function mutation in tyrosine phosphatase Ptpn11 (Shp2) on malignant transformation of hematopoietic cells. J Exp Med. 2011;208(10):1977–88. doi:10.1084/jem.20110450.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Usenko T, Chan G, Torlakovic E, Klingmuller U, Neel BG. Leukemogenic ptpn11 allele causes defective erythropoiesis in mice. PLoS One. 2014;9(10), e109682. doi:10.1371/journal.pone.0109682.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Rampal R, Mascarenhas J. Pathogenesis and management of acute myeloid leukemia that has evolved from a myeloproliferative neoplasm. Curr Opin Hematol. 2014;21(2):65–71. doi:10.1097/MOH.0000000000000017.

    Article  CAS  PubMed  Google Scholar 

  150. Xu D, Zheng H, Yu WM, Qu CK. Activating mutations in protein tyrosine phosphatase Ptpn11 (Shp2) enhance reactive oxygen species production that contributes to myeloproliferative disorder. PLoS One. 2013;8(5), e63152. doi:10.1371/journal.pone.0063152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Zheng H, Li S, Hsu P, Qu CK. Induction of a tumor-associated activating mutation in protein tyrosine phosphatase Ptpn11 (Shp2) enhances mitochondrial metabolism. Leading to oxidative stress and senescence. J Biol Chem. 2013. doi:10.1074/jbc.M113.462291.

    Google Scholar 

  152. Tuveson DA, Shaw AT, Willis NA, Silver DP, Jackson EL, Chang S, Mercer KL, Grochow R, Hock H, Crowley D, Hingorani SR, Zaks T, King C, Jacobetz MA, Wang L, Bronson RT, Orkin SH, DePinho RA, Jacks T. Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell. 2004;5(4):375–87.

    Article  CAS  PubMed  Google Scholar 

  153. Yang Z, Kondo T, Voorhorst CS, Nabinger SC, Ndong L, Yin F, Chan EM, Yu M, Wurstlin O, Kratz CP, Niemeyer CM, Flotho C, Hashino E, Chan RJ. Increased c-Jun expression and reduced GATA2 expression promote aberrant monocytic differentiation induced by activating PTPN11 mutants. Mol Cell Biol. 2009;29(16):4376–93. doi:10.1128/MCB.01330-08.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Zhang W, Chan RJ, Chen H, Yang Z, He Y, Zhang X, Luo Y, Yin F, Moh A, Miller LC, Payne RM, Zhang ZY, Fu XY, Shou W. Negative regulation of Stat3 by activating PTPN11 mutants contributes to the pathogenesis of Noonan syndrome and juvenile myelomonocytic leukemia. J Biol Chem. 2009;284(33):22353–63. doi:10.1074/jbc.M109.020495.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Goodwin CB, Li XJ, Mali RS, Chan G, Kang M, Liu Z, Vanhaesebroeck B, Neel BG, Loh ML, Lannutti BJ, Kapur R, Chan RJ. PI3K p110delta uniquely promotes gain-of-function Shp2-induced GM-CSF hypersensitivity in a model of JMML. Blood. 2014;123(18):2838–42. doi:10.1182/blood-2013-10-535104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Yu WM, Hawley TS, Hawley RG, Qu CK. Catalytic-dependent and -independent roles of SHP-2 tyrosine phosphatase in interleukin-3 signaling. Oncogene. 2003;22(38):5995–6004.

    Article  CAS  PubMed  Google Scholar 

  157. Yu WM, Daino H, Chen J, Bunting KD, Qu CK. Effects of a leukemia-associated gain-of-function mutation of SHP-2 phosphatase on interleukin-3 signaling. J Biol Chem. 2006;281(9):5426–34.

    Article  CAS  PubMed  Google Scholar 

  158. Kotecha N, Flores NJ, Irish JM, Simonds EF, Sakai DS, Archambeault S, Diaz-Flores E, Coram M, Shannon KM, Nolan GP, Loh ML. Single-cell profiling identifies aberrant STAT5 activation in myeloid malignancies with specific clinical and biologic correlates. Cancer Cell. 2008;14(4):335–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Hasegawa D, Bugarin C, Giordan M, Bresolin S, Longoni D, Micalizzi C, Ramenghi U, Bertaina A, Basso G, Locatelli F, Biondi A, Te Kronnie G, Gaipa G. Validation of flow cytometric phospho-STAT5 as a diagnostic tool for juvenile myelomonocytic leukemia. Blood Cancer J. 2013;3, e160. doi:10.1038/bcj.2013.56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Gaipa G, Bugarin C, Longoni D, Cesana S, Molteni C, Faini A, Timeus F, Zecca M, Biondi A. Aberrant GM-CSF signal transduction pathway in juvenile myelomonocytic leukemia assayed by flow cytometric intracellular STAT5 phosphorylation measurement. Leukemia. 2009;23(4):791–3. doi:10.1038/leu.2008.265.

    Article  CAS  PubMed  Google Scholar 

  161. Kong G, Wunderlich M, Yang D, Ranheim EA, Young KH, Wang J, Chang YI, Du J, Liu Y, Tey SR, Zhang X, Juckett M, Mattison R, Damnernsawad A, Zhang J, Mulloy JC, Zhang J. Combined MEK and JAK inhibition abrogates murine myeloproliferative neoplasm. J Clin Invest. 2014;124(6):2762–73. doi:10.1172/JCI74182.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Mali RS, Ma P, Zeng LF, Martin H, Ramdas B, He Y, Sims E, Nabinger S, Ghosh J, Sharma N, Munugalavadla V, Chatterjee A, Li S, Sandusky G, Craig AW, Bunting KD, Feng GS, Chan RJ, Zhang ZY, Kapur R. Role of SHP2 phosphatase in KIT-induced transformation: identification of SHP2 as a druggable target in diseases involving oncogenic KIT. Blood. 2012;120(13):2669–78. doi:10.1182/blood-2011-08-375873.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Sharma N, Everingham S, Zeng LF, Zhang ZY, Kapur R, Craig AW. Oncogenic KIT-induced aggressive systemic mastocytosis requires SHP2/PTPN11 phosphatase for disease progression in mice. Oncotarget. 2014;5(15):6130–41.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Nabinger SC, Li XJ, Ramdas B, He Y, Zhang X, Zeng L, Richine B, Bowling JD, Fukuda S, Goenka S, Liu Z, Feng GS, Yu M, Sandusky GE, Boswell HS, Zhang ZY, Kapur R, Chan RJ. The protein tyrosine phosphatase, Shp2, positively contributes to FLT3-ITD-induced hematopoietic progenitor hyperproliferation and malignant disease in vivo. Leukemia. 2013;27(2):398–408. doi:10.1038/leu.2012.308.

    Article  CAS  PubMed  Google Scholar 

  165. Cancer Genome Atlas Research N. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–74. doi:10.1056/NEJMoa1301689.

    Article  CAS  Google Scholar 

  166. Chan G, Kalaitzidis D, Neel BG. The tyrosine phosphatase Shp2 (PTPN11) in cancer. Cancer Metastasis Rev. 2008;27(2):179–92.

    Article  CAS  PubMed  Google Scholar 

  167. Sattler M, Mohi MG, Pride YB, Quinnan LR, Malouf NA, Podar K, Gesbert F, Iwasaki H, Li S, Van Etten RA, Gu H, Griffin JD, Neel BG. Critical role for Gab2 in transformation by BCR/ABL. Cancer Cell. 2002;1(5):479–92.

    Article  CAS  PubMed  Google Scholar 

  168. Sha F, Gencer EB, Georgeon S, Koide A, Yasui N, Koide S, Hantschel O. Dissection of the BCR-ABL signaling network using highly specific monobody inhibitors to the SHP2 SH2 domains. Proc Natl Acad Sci U S A. 2013;110(37):14924–9. doi:10.1073/pnas.1303640110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Chen J, Yu WM, Daino H, Broxmeyer HE, Druker BJ, Qu CK. SHP-2 phosphatase is required for hematopoietic cell transformation by Bcr-Abl. Blood. 2007;109(2):778–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Scherr M, Chaturvedi A, Battmer K, Dallmann I, Schultheis B, Ganser A, Eder M. Enhanced sensitivity to inhibition of SHP2, STAT5, and Gab2 expression in chronic myeloid leukemia (CML). Blood. 2006;107(8):3279–87.

    Article  CAS  PubMed  Google Scholar 

  171. Teal HE, Ni S, Xu J, Finkelstein LD, Cheng AM, Paulson RF, Feng GS, Correll PH. GRB2-mediated recruitment of GAB2, but not GAB1, to SF-STK supports the expansion of Friend virus-infected erythroid progenitor cells. Oncogene. 2006;25(17):2433–43.

    Article  CAS  PubMed  Google Scholar 

  172. Ischenko I, Petrenko O, Gu H, Hayman MJ. Scaffolding protein Gab2 mediates fibroblast transformation by the SEA tyrosine kinase. Oncogene. 2003;22(41):6311–8.

    Article  CAS  PubMed  Google Scholar 

  173. Higuchi M, Tsutsumi R, Higashi H, Hatakeyama M. Conditional gene silencing utilizing the lac repressor reveals a role of SHP-2 in cagA-positive Helicobacter pylori pathogenicity. Cancer Sci. 2004;95(5):442–7.

    Article  CAS  PubMed  Google Scholar 

  174. Higashi H, Tsutsumi R, Muto S, Sugiyama T, Azuma T, Asaka M, Hatakeyama M. SHP-2 tyrosine phosphatase as an intracellular target of Helicobacter pylori CagA protein. Science. 2002;295(5555):683–6.

    Article  CAS  PubMed  Google Scholar 

  175. Tsutsumi R, Higashi H, Higuchi M, Okada M, Hatakeyama M. Attenuation of Helicobacter pylori CagA x SHP-2 signaling by interaction between CagA and C-terminal Src kinase. J Biol Chem. 2003;278(6):3664–70.

    Article  CAS  PubMed  Google Scholar 

  176. Ohnishi N, Yuasa H, Tanaka S, Sawa H, Miura M, Matsui A, Higashi H, Musashi M, Iwabuchi K, Suzuki M, Yamada G, Azuma T, Hatakeyama M. Transgenic expression of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in mouse. Proc Natl Acad Sci U S A. 2008;105(3):1003–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Mimuro H, Suzuki T, Tanaka J, Asahi M, Haas R, Sasakawa C. Grb2 is a key mediator of helicobacter pylori CagA protein activities. Mol Cell. 2002;10(4):745–55.

    Article  CAS  PubMed  Google Scholar 

  178. Suzuki M, Mimuro H, Suzuki T, Park M, Yamamoto T, Sasakawa C. Interaction of CagA with Crk plays an important role in Helicobacter pylori-induced loss of gastric epithelial cell adhesion. J Exp Med. 2005;202(9):1235–47. doi:10.1084/jem.20051027.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Higashi H, Nakaya A, Tsutsumi R, Yokoyama K, Fujii Y, Ishikawa S, Higuchi M, Takahashi A, Kurashima Y, Teishikata Y, Tanaka S, Azuma T, Hatakeyama M. Helicobacter pylori CagA induces Ras-independent morphogenetic response through SHP-2 recruitment and activation. J Biol Chem. 2004;279(17):17205–16.

    Article  CAS  PubMed  Google Scholar 

  180. Tsutsumi R, Takahashi A, Azuma T, Higashi H, Hatakeyama M. Focal adhesion kinase is a substrate and downstream effector of SHP-2 complexed with Helicobacter pylori CagA. Mol Cell Biol. 2006;26(1):261–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Saadat I, Higashi H, Obuse C, Umeda M, Murata-Kamiya N, Saito Y, Lu H, Ohnishi N, Azuma T, Suzuki A, Ohno S, Hatakeyama M. Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt epithelial cell polarity. Nature. 2007;447(7142):330–3.

    Article  CAS  PubMed  Google Scholar 

  182. Lu H, Murata-Kamiya N, Saito Y, Hatakeyama M. Role of partitioning-defective 1/microtubule affinity-regulating kinases in the morphogenetic activity of Helicobacter pylori CagA. J Biol Chem. 2009;284(34):23024–36. doi:10.1074/jbc.M109.001008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Yang Z, Xue B, Umitsu M, Ikura M, Muthuswamy SK, Neel BG. The signaling adaptor GAB1 regulates cell polarity by acting as a PAR protein scaffold. Mol Cell. 2012;47(3):469–83. doi:10.1016/j.molcel.2012.06.037.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Ormandy CJ, Musgrove EA, Hui R, Daly RJ, Sutherland RL. Cyclin D1, EMS1 and 11q13 amplification in breast cancer. Breast Cancer Res Treat. 2003;78(3):323–35.

    Article  CAS  PubMed  Google Scholar 

  185. Bekri S, Adelaide J, Merscher S, Grosgeorge J, Caroli-Bosc F, Perucca-Lostanlen D, Kelley PM, Pebusque MJ, Theillet C, Birnbaum D, Gaudray P. Detailed map of a region commonly amplified at 11q13-- > q14 in human breast carcinoma. Cytogenet Cell Genet. 1997;79(1-2):125–31.

    Article  CAS  PubMed  Google Scholar 

  186. Bentires-Alj M, Gil SG, Chan R, Wang ZC, Wang Y, Imanaka N, Harris LN, Richardson A, Neel BG, Gu H. A role for the scaffolding adapter GAB2 in breast cancer. Nat Med. 2006;12(1):114–21.

    Article  CAS  PubMed  Google Scholar 

  187. Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70. doi:10.1038/nature11412.

    Article  CAS  Google Scholar 

  188. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, Yates LR, Papaemmanuil E, Beare D, Butler A, Cheverton A, Gamble J, Hinton J, Jia M, Jayakumar A, Jones D, Latimer C, Lau KW, McLaren S, McBride DJ, Menzies A, Mudie L, Raine K, Rad R, Chapman MS, Teague J, Easton D, Langerod A, Oslo Breast Cancer C, Lee MT, Shen CY, Tee BT, Huimin BW, Broeks A, Vargas AC, Turashvili G, Martens J, Fatima A, Miron P, Chin SF, Thomas G, Boyault S, Mariani O, Lakhani SR, van de Vijver M, van’t Veer L, Foekens J, Desmedt C, Sotiriou C, Tutt A, Caldas C, Reis-Filho JS, Aparicio SA, Salomon AV, Borresen-Dale AL, Richardson AL, Campbell PJ, Futreal PA, Stratton MR. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486(7403):400–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, Lawrence MS, Sivachenko AY, Sougnez C, Zou L, Cortes ML, Fernandez-Lopez JC, Peng S, Ardlie KG, Auclair D, Bautista-Pina V, Duke F, Francis J, Jung J, Maffuz-Aziz A, Onofrio RC, Parkin M, Pho NH, Quintanar-Jurado V, Ramos AH, Rebollar-Vega R, Rodriguez-Cuevas S, Romero-Cordoba SL, Schumacher SE, Stransky N, Thompson KM, Uribe-Figueroa L, Baselga J, Beroukhim R, Polyak K, Sgroi DC, Richardson AL, Jimenez-Sanchez G, Lander ES, Gabriel SB, Garraway LA, Golub TR, Melendez-Zajgla J, Toker A, Getz G, Hidalgo-Miranda A, Meyerson M. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature. 2012;486(7403):405–9. doi:10.1038/nature11154.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, Turashvili G, Ding J, Tse K, Haffari G, Bashashati A, Prentice LM, Khattra J, Burleigh A, Yap D, Bernard V, McPherson A, Shumansky K, Crisan A, Giuliany R, Heravi-Moussavi A, Rosner J, Lai D, Birol I, Varhol R, Tam A, Dhalla N, Zeng T, Ma K, Chan SK, Griffith M, Moradian A, Cheng SW, Morin GB, Watson P, Gelmon K, Chia S, Chin SF, Curtis C, Rueda OM, Pharoah PD, Damaraju S, Mackey J, Hoon K, Harkins T, Tadigotla V, Sigaroudinia M, Gascard P, Tlsty T, Costello JF, Meyer IM, Eaves CJ, Wasserman WW, Jones S, Huntsman D, Hirst M, Caldas C, Marra MA, Aparicio S. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature. 2012;486(7403):395–9. doi:10.1038/nature10933.

    CAS  PubMed  Google Scholar 

  191. Brummer T, Schramek D, Hayes VM, Bennett HL, Caldon CE, Musgrove EA, Daly RJ. Increased proliferation and altered growth factor dependence of human mammary epithelial cells overexpressing the Gab2 docking protein. J Biol Chem. 2006;281(1):626–37.

    Article  CAS  PubMed  Google Scholar 

  192. Aceto N, Sausgruber N, Brinkhaus H, Gaidatzis D, Martiny-Baron G, Mazzarol G, Confalonieri S, Quarto M, Hu G, Balwierz PJ, Pachkov M, Elledge SJ, van Nimwegen E, Stadler MB, Bentires-Alj M. Tyrosine phosphatase SHP2 promotes breast cancer progression and maintains tumor-initiating cells via activation of key transcription factors and a positive feedback signaling loop. Nat Med. 2012;18(4):529–37. doi:10.1038/nm.2645.

    Article  CAS  PubMed  Google Scholar 

  193. Sausgruber N, Coissieux MM, Britschgi A, Wyckoff J, Aceto N, Leroy C, Stadler MB, Voshol H, Bonenfant D, Bentires-Alj M. Tyrosine phosphatase SHP2 increases cell motility in triple-negative breast cancer through the activation of SRC-family kinases. Oncogene. 2014. doi:10.1038/onc.2014.170.

    Google Scholar 

  194. Wang Y, Sheng Q, Spillman MA, Behbakht K, Gu H. Gab2 regulates the migratory behaviors and E-cadherin expression via activation of the PI3K pathway in ovarian cancer cells. Oncogene. 2012;31(20):2512–20. doi:10.1038/onc.2011.435.

    Article  CAS  PubMed  Google Scholar 

  195. Dunn GP, Cheung HW, Agarwalla PK, Thomas S, Zektser Y, Karst AM, Boehm JS, Weir BA, Berlin AM, Zou L, Getz G, Liu JF, Hirsch M, Vazquez F, Root DE, Beroukhim R, Drapkin R, Hahn WC. In vivo multiplexed interrogation of amplified genes identifies GAB2 as an ovarian cancer oncogene. Proc Natl Acad Sci U S A. 2014;111(3):1102–7. doi:10.1073/pnas.1311909111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Schneeberger VE, Luetteke N, Ren Y, Berns H, Chen L, Foroutan P, Martinez GV, Haura EB, Chen J, Coppola D, Wu J. SHP2E76K mutant promotes lung tumorigenesis in transgenic mice. Carcinogenesis. 2014;35(8):1717–25. doi:10.1093/carcin/bgu025.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Jones DT, Hutter B, Jager N, Korshunov A, Kool M, Warnatz HJ, Zichner T, Lambert SR, Ryzhova M, Quang DA, Fontebasso AM, Stutz AM, Hutter S, Zuckermann M, Sturm D, Gronych J, Lasitschka B, Schmidt S, Seker-Cin H, Witt H, Sultan M, Ralser M, Northcott PA, Hovestadt V, Bender S, Pfaff E, Stark S, Faury D, Schwartzentruber J, Majewski J, Weber UD, Zapatka M, Raeder B, Schlesner M, Worth CL, Bartholomae CC, von Kalle C, Imbusch CD, Radomski S, Lawerenz C, van Sluis P, Koster J, Volckmann R, Versteeg R, Lehrach H, Monoranu C, Winkler B, Unterberg A, Herold-Mende C, Milde T, Kulozik AE, Ebinger M, Schuhmann MU, Cho YJ, Pomeroy SL, von Deimling A, Witt O, Taylor MD, Wolf S, Karajannis MA, Eberhart CG, Scheurlen W, Hasselblatt M, Ligon KL, Kieran MW, Korbel JO, Yaspo ML, Brors B, Felsberg J, Reifenberger G, Collins VP, Jabado N, Eils R, Lichter P, Pfister SM, International Cancer Genome Consortium PedBrain Tumor P. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat Genet. 2013;45(8):927–32. doi:10.1038/ng.2682.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Fryssira H, Leventopoulos G, Psoni S, Kitsiou-Tzeli S, Stavrianeas N, Kanavakis E. Tumor development in three patients with Noonan syndrome. Eur J Pediatr. 2008;167(9):1025–31. doi:10.1007/s00431-007-0636-3.

    Article  PubMed  Google Scholar 

  199. Schuettpelz LG, McDonald S, Whitesell K, Desruisseau DM, Grange DK, Gurnett CA, Wilson DB. Pilocytic astrocytoma in a child with Noonan syndrome. Pediatr Blood Cancer. 2009;53(6):1147–9. doi:10.1002/pbc.22193.

    Article  PubMed  Google Scholar 

  200. Voena C, Conte C, Ambrogio C, Boeri Erba E, Boccalatte F, Mohammed S, Jensen ON, Palestro G, Inghirami G, Chiarle R. The tyrosine phosphatase Shp2 interacts with NPM-ALK and regulates anaplastic lymphoma cell growth and migration. Cancer Res. 2007;67(9):4278–86.

    Article  CAS  PubMed  Google Scholar 

  201. Bard-Chapeau EA, Li S, Ding J, Zhang SS, Zhu HH, Princen F, Fang DD, Han T, Bailly-Maitre B, Poli V, Varki NM, Wang H, Feng GS. Ptpn11/Shp2 acts as a tumor suppressor in hepatocellular carcinogenesis. Cancer Cell. 2011;19(5):629–39. doi:10.1016/j.ccr.2011.03.023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Fisher TJ, Williams N, Morris L, Cundy PJ. Metachondromatosis: more than just multiple osteochondromas. J Child Orthop. 2013;7(6):455–64. doi:10.1007/s11832-013-0526-3.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Sobreira NL, Cirulli ET, Avramopoulos D, Wohler E, Oswald GL, Stevens EL, Ge D, Shianna KV, Smith JP, Maia JM, Gumbs CE, Pevsner J, Thomas G, Valle D, Hoover-Fong JE, Goldstein DB. Whole-genome sequencing of a single proband together with linkage analysis identifies a Mendelian disease gene. PLoS Genet. 2010;6(6), e1000991. doi:10.1371/journal.pgen.1000991.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Bowen ME, Boyden ED, Holm IA, Campos-Xavier B, Bonafe L, Superti-Furga A, Ikegawa S, Cormier-Daire V, Bovee JV, Pansuriya TC, de Sousa SB, Savarirayan R, Andreucci E, Vikkula M, Garavelli L, Pottinger C, Ogino T, Sakai A, Regazzoni BM, Wuyts W, Sangiorgi L, Pedrini E, Zhu M, Kozakewich HP, Kasser JR, Seidman JG, Kurek KC, Warman ML. Loss-of-function mutations in PTPN11 cause metachondromatosis, but not Ollier disease or Maffucci syndrome. PLoS Genet. 2011;7(4), e1002050. doi:10.1371/journal.pgen.1002050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Yang W, Wang J, Moore DC, Liang H, Dooner M, Wu Q, Terek R, Chen Q, Ehrlich MG, Quesenberry PJ, Neel BG. Ptpn11 deletion in a novel progenitor causes metachondromatosis by inducing hedgehog signalling. Nature. 2013;499(7459):491–5. doi:10.1038/nature12396.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Kim HK, Feng GS, Chen D, King PD, Kamiya N. Targeted disruption of Shp2 in chondrocytes leads to metachondromatosis with multiple cartilaginous protrusions. J Bone Mineral Res. 2014;29(3):761–9. doi:10.1002/jbmr.2062.

    Article  CAS  Google Scholar 

  207. Digilio MC, Conti E, Sarkozy A, Mingarelli R, Dottorini T, Marino B, Pizzuti A, Dallapiccola B. Grouping of multiple-lentigines/LEOPARD and Noonan syndromes on the PTPN11 gene. Am J Hum Genet. 2002;71(2):389–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Legius E, Schrander-Stumpel C, Schollen E, Pulles-Heintzberger C, Gewillig M, Fryns JP. PTPN11 mutations in LEOPARD syndrome. J Med Genet. 2002;39(8):571–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Yu ZH, Xu J, Walls CD, Chen L, Zhang S, Zhang R, Wu L, Wang L, Liu S, Zhang ZY. Structural and mechanistic insights into LEOPARD syndrome-associated SHP2 mutations. J Biol Chem. 2013;288(15):10472–82. doi:10.1074/jbc.M113.450023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Yu ZH, Zhang RY, Walls CD, Chen L, Zhang S, Wu L, Liu S, Zhang ZY. Molecular basis of gain-of-function LEOPARD syndrome-associated SHP2 mutations. Biochemistry. 2014;53(25):4136–51. doi:10.1021/bi5002695.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2016 Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Chan, G., Neel, B.G. (2016). Role of PTPN11 (SHP2) in Cancer. In: Neel, B., Tonks, N. (eds) Protein Tyrosine Phosphatases in Cancer. Springer, New York, NY. https://doi.org/10.1007/978-1-4939-3649-6_4

Download citation

Publish with us

Policies and ethics