Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The duality of human oncoproteins: drivers of cancer and congenital disorders

Abstract

Human oncoproteins promote transformation of cells into tumours by dysregulating the signalling pathways that are involved in cell growth, proliferation and death. Although oncoproteins were discovered many years ago and have been widely studied in the context of cancer, the recent use of high-throughput sequencing techniques has led to the identification of cancer-associated mutations in other conditions, including many congenital disorders. These syndromes offer an opportunity to study oncoprotein signalling and its biology in the absence of additional driver or passenger mutations, as a result of their monogenic nature. Moreover, their expression in multiple tissue lineages provides insight into the biology of the proto-oncoprotein at the physiological level, in both transformed and unaffected tissues. Given the recent paradigm shift in regard to how oncoproteins promote transformation, we review the fundamentals of genetics, signalling and pathogenesis underlying oncoprotein duality.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2: Factors that influence the ability of oncoproteins to cause transformation.
Fig. 3: Genetic transmission of oncoproteins.
Fig. 4: The pathways of oncoprotein signalling in congenital disorders.

Similar content being viewed by others

References

  1. Adams, D. R. & Eng, C. M. Next-generation sequencing to diagnose suspected genetic disorders. N. Engl. J. Med. 379, 1353–1362 (2018).

    CAS  PubMed  Google Scholar 

  2. Bamshad, M. J. et al. Exome sequencing as a tool for Mendelian disease gene discovery. Nat. Rev. Genet. 12, 745–755 (2011).

    CAS  PubMed  Google Scholar 

  3. Erickson, R. P. Somatic gene mutation and human disease other than cancer: an update. Mutat. Res. 705, 96–106 (2010).

    CAS  PubMed  Google Scholar 

  4. Martincorena, I. et al. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348, 880–886 (2015). In this study, ultradeep sequencing of skin eyelids reveals the presence of an elevated number of somatic mutations, including many cancer-associated mutations.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Martincorena, I. et al. Somatic mutant clones colonize the human esophagus with age. Science 362, 911–917 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Yizhak, K. et al. RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues. Science 364, eaaw0726 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Haigis, K. M., Cichowski, K. & Elledge, S. J. Tissue-specificity in cancer: the rule, not the exception. Science 363, 1150–1151 (2019).

    CAS  PubMed  Google Scholar 

  8. Haigis, K. M. KRAS alleles: the devil is in the detail. Trends Cancer 3, 686–697 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Li, S., Balmain, A. & Counter, C. M. A model for RAS mutation patterns in cancers: finding the sweet spot. Nat. Rev. Cancer 18, 767–777 (2018).

    CAS  PubMed  Google Scholar 

  10. Schneider, G., Schmidt-Supprian, M., Rad, R. & Saur, D. Tissue-specific tumorigenesis: context matters. Nat. Rev. Cancer 17, 239–253 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Mueller, S. et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature 554, 62–68 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Estep, A. L., Tidyman, W. E., Teitell, M. A., Cotter, P. D. & Rauen, K. A. HRAS mutations in Costello syndrome: detection of constitutional activating mutations in codon 12 and 13 and loss of wild-type allele in malignancy. Am. J. Med. Genet. A 140, 8–16 (2006).

    PubMed  Google Scholar 

  13. Zhao, L. & Vogt, P. K. Helical domain and kinase domain mutations in p110α of phosphatidylinositol 3-kinase induce gain of function by different mechanisms. Proc. Natl Acad. Sci. USA 105, 2652–2657 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Burke, J. E., Perisic, O., Masson, G. R., Vadas, O. & Williams, R. L. Oncogenic mutations mimic and enhance dynamic events in the natural activation of phosphoinositide 3-kinase p110α (PIK3CA). Proc. Natl Acad. Sci. USA 109, 15259–15264 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hobbs, G. A. et al. Atypical KRASG12R mutant is impaired in PI3K signaling and macropinocytosis in pancreatic cancer. Cancer Discov. 10, 104–123 (2020).

    PubMed  Google Scholar 

  16. Poulin, E. J. et al. Tissue-specific oncogenic activity of KRASA146T. Cancer Discov. 9, 738–755 (2019).

    PubMed  PubMed Central  Google Scholar 

  17. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    PubMed  PubMed Central  Google Scholar 

  18. Iwahara, T. et al. Molecular characterization of ALK, a receptor tyrosine kinase expressed specifically in the nervous system. Oncogene 14, 439–449 (1997).

    CAS  PubMed  Google Scholar 

  19. Vernersson, E. et al. Characterization of the expression of the ALK receptor tyrosine kinase in mice. Gene Expr. Patterns 6, 448–461 (2006).

    CAS  PubMed  Google Scholar 

  20. Sherr, C. J. Principles of tumor suppression. Cell 116, 235–246 (2004).

    CAS  PubMed  Google Scholar 

  21. Schwartz, R. & Schäffer, A. A. The evolution of tumour phylogenetics: principles and practice. Nat. Rev. Genet. 18, 213–229 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Merino, M. M., Levayer, R. & Moreno, E. Survival of the fittest: essential roles of cell competition in development, aging, and cancer. Trends Cell Biol. 26, 776–788 (2016).

    PubMed  Google Scholar 

  23. Bowling, S., Lawlor, K. & Rodríguez, T. A. Cell competition: the winners and losers of fitness selection. Development 146, dev167486 (2019).

    CAS  PubMed  Google Scholar 

  24. Hogan, C. et al. Characterization of the interface between normal and transformed epithelial cells. Nat. Cell Biol. 11, 460–467 (2009).

    CAS  PubMed  Google Scholar 

  25. Nelson, C. M. & Bissell, M. J. Of extracellular matrix, scaffolds, and signaling: tissue architecture regulates development, homeostasis, and cancer. Annu. Rev. Cell Dev. Biol. 22, 287–309 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Brown, S. et al. Correction of aberrant growth preserves tissue homeostasis. Nature 548, 334–337 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Pineda, C. M. et al. Hair follicle regeneration suppresses Ras-driven oncogenic growth. J. Cell Biol. 218, 3212–3222 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Greten, F. R. & Grivennikov, S. I. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity 51, 27–41 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Swann, J. B. & Smyth, M. J. Immune surveillance of tumors. J. Clin. Invest. 117, 1137–1146 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Little, J. B. Radiation carcinogenesis. Carcinogenesis 21, 397–404 (2000).

    CAS  PubMed  Google Scholar 

  31. Loeb, L. A. & Harris, C. C. Advances in chemical carcinogenesis: a historical review and prospective. Cancer Res. 68, 6863–6872 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Steen, H. B. The origin of oncogenic mutations: where is the primary damage? Carcinogenesis 21, 1773–1776 (2000).

    CAS  PubMed  Google Scholar 

  33. Petljak, M. et al. Characterizing mutational signatures in human cancer cell lines reveals episodic APOBEC mutagenesis. Cell 176, 1282–1294.e20 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Aoki, Y. et al. Germline mutations in HRAS proto-oncogene cause Costello syndrome. Nat. Genet. 37, 1038–1040 (2005). This study identifies germline HRAS mutations as the causative factor for Costello syndrome, a RASopathy. The authors show that mutations in codon 12 are frequent in these patients, similar to HRAS mutations found in certain human sporadic tumours.

    CAS  PubMed  Google Scholar 

  35. Cancer Genome Atlas Research Network. Integrated genomic characterization of papillary thyroid carcinoma. Cell 159, 676–690 (2014).

    Google Scholar 

  36. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    PubMed  PubMed Central  Google Scholar 

  37. Goriely, A. & Wilkie, A. O. M. Paternal age effect mutations and selfish spermatogonial selection: causes and consequences for human disease. Am. J. Hum. Genet. 90, 175–200 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Maher, G. J. et al. Visualizing the origins of selfish de novo mutations in individual seminiferous tubules of human testes. Proc. Natl Acad. Sci. USA 113, 2454–2459 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Maher, G. J. et al. Selfish mutations dysregulating RAS-MAPK signaling are pervasive in aged human testes. Genome Res. 28, 1779–1790 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Shiang, R. et al. Mutations in the transmembrane domain of FGFR3 cause the most common genetic form of dwarfism, achondroplasia. Cell 78, 335–342 (1994).

    CAS  PubMed  Google Scholar 

  41. Hare, L. M. et al. Heterozygous expression of the oncogenic Pik3caH1047R mutation during murine development results in fatal embryonic and extraembryonic defects. Dev. Biol. 404, 14–26 (2015).

    CAS  PubMed  Google Scholar 

  42. Guerra, C. et al. Tumor induction by an endogenous K-ras oncogene is highly dependent on cellular context. Cancer Cell 4, 111–120 (2003).

    CAS  PubMed  Google Scholar 

  43. Tuveson, D. A. et al. Endogenous oncogenic K-rasG12D stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell 5, 375–387 (2004).

    CAS  PubMed  Google Scholar 

  44. Schubbert, S. et al. Germline KRAS mutations cause Noonan syndrome. Nat. Genet. 38, 331–336 (2006).

    CAS  PubMed  Google Scholar 

  45. Rauen, K. A. et al. Molecular and functional analysis of a novel MEK2 mutation in cardio-facio-cutaneous syndrome: transmission through four generations. Am. J. Med. Genet. A 152A, 807–814 (2010).

    CAS  PubMed  Google Scholar 

  46. Rodriguez-Laguna, L. et al. Somatic activating mutations in PIK3CA cause generalized lymphatic anomaly. J. Exp. Med. 216, 407–418 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Jacobs, K. B. et al. Detectable clonal mosaicism and its relationship to aging and cancer. Nat. Genet. 44, 651–658 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Laurie, C. C. et al. Detectable clonal mosaicism from birth to old age and its relationship to cancer. Nat. Genet. 44, 642–650 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Forsberg, L. A. et al. Mosaic loss of chromosome Y in peripheral blood is associated with shorter survival and higher risk of cancer. Nat. Genet. 46, 624–628 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Happle, R. Lethal genes surviving by mosaicism: a possible explanation for sporadic birth defects involving the skin. J. Am. Acad. Dermatol. 16, 899–906 (1987). In this article, the author hypothesizes the reason for which certain dermatological conditions are mosaicisms driven by mutations that do not follow a Mendelian inheritance pattern.

    CAS  PubMed  Google Scholar 

  51. Groesser, L. et al. Postzygotic HRAS and KRAS mutations cause nevus sebaceous and Schimmelpenning syndrome. Nat. Genet. 44, 783–787 (2012). In this article, oncogenic HRAS and KRAS mutations are identified to be the cause of a syndromic mosaicism that is characterized by the presence of sebaceous nevi.

    CAS  PubMed  Google Scholar 

  52. Bolognia, J. L., Orlow, S. J. & Glick, S. A. Lines of Blaschko. J. Am. Acad. Dermatol. 31, 157–190 (1994).

    CAS  PubMed  Google Scholar 

  53. Hafner, C. et al. Oncogenic PIK3CA mutations occur in epidermal nevi and seborrheic keratoses with a characteristic mutation pattern. Proc. Natl Acad. Sci. USA 104, 13450–13454 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Hafner, C. et al. Mosaicism of activating FGFR3 mutations in human skin causes epidermal nevi. J. Clin. Invest. 116, 2201–2207 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Hafner, C. et al. Keratinocytic epidermal nevi are associated with mosaic RAS mutations. J. Med. Genet. 49, 249–253 (2012).

    CAS  PubMed  Google Scholar 

  56. Biesecker, L. G. & Spinner, N. B. A genomic view of mosaicism and human disease. Nat. Rev. Genet. 14, 307–320 (2013).

    CAS  PubMed  Google Scholar 

  57. Fernández, L. C., Torres, M. & Real, F. X. Somatic mosaicism: on the road to cancer. Nat. Rev. Cancer 16, 43–55 (2016).

    PubMed  Google Scholar 

  58. Adams, J. R. et al. Cooperation between Pik3ca and p53 mutations in mouse mammary tumor formation. Cancer Res. 71, 2706–2717 (2011).

    CAS  PubMed  Google Scholar 

  59. Jackson, E. L. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev. 15, 3243–3248 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Kirsch, D. G. et al. A spatially and temporally restricted mouse model of soft tissue sarcoma. Nat. Med. 13, 992–997 (2007).

    CAS  PubMed  Google Scholar 

  61. Cavenee, W. K. et al. Expression of recessive alleles by chromosomal mechanisms in retinoblastoma. Nature 305, 779–784 (1983). This study identifies the acquisition of secondary hits in the RB1 gene in patients with retinoblastoma, confirming the Knudson hypothesis.

    CAS  PubMed  Google Scholar 

  62. Guerra, C. et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 11, 291–302 (2007).

    CAS  PubMed  Google Scholar 

  63. Ayturk, U. M. et al. Somatic activating mutations in GNAQ and GNA11 are associated with congenital hemangioma. Am. J. Hum. Genet. 98, 789–795 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Shirley, M. D. et al. Sturge-Weber syndrome and port-wine stains caused by somatic mutation in GNAQ. N. Engl. J. Med. 368, 1971–1979 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Cai, X. et al. Single-cell, genome-wide sequencing identifies clonal somatic copy-number variation in the human brain. Cell Rep. 8, 1280–1289 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    PubMed  PubMed Central  Google Scholar 

  67. Yokoyama, A. et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565, 312–317 (2019).

    CAS  PubMed  Google Scholar 

  68. Anglesio, M. S. et al. Cancer-associated mutations in endometriosis without cancer. N. Engl. J. Med. 376, 1835–1848 (2017).

    PubMed  PubMed Central  Google Scholar 

  69. Suda, K. et al. Clonal expansion and diversification of cancer-associated mutations in endometriosis and normal endometrium. Cell Rep. 24, 1777–1789 (2018).

    CAS  PubMed  Google Scholar 

  70. Hafner, C., Toll, A. & Real, F. X. HRAS mutation mosaicism causing urothelial cancer and epidermal nevus. N. Engl. J. Med. 365, 1940–1942 (2011). In this case report, the authors describe the presence of FGFR3-mutant mosaicism giving rise to both urothelial cancer and epidermal nevi.

    CAS  PubMed  Google Scholar 

  71. Doucet, M. E., Bloomhardt, H. M., Moroz, K., Lindhurst, M. J. & Biesecker, L. G. Lack of mutation-histopathology correlation in a patient with Proteus syndrome. Am. J. Med. Genet. A 170, 1422–1432 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Sanchez-Vega, F. et al. Oncogenic signaling pathways in the cancer genome atlas. Cell 173, 321–337.e10 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Lemmon, M. A. & Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 141, 1117–1134 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Robertson, S. C., Tynan, J. A. & Donoghue, D. J. RTK mutations and human syndromes when good receptors turn bad. Trends Genet. 16, 265–271 (2000).

    CAS  PubMed  Google Scholar 

  75. Bell, D. W. et al. Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR. Nat. Genet. 37, 1315–1316 (2005).

    CAS  PubMed  Google Scholar 

  76. Oxnard, G. R., Nguyen, K.-S. H. & Costa, D. B. Germline mutations in driver oncogenes and inherited lung cancer risk independent of smoking history. J. Natl Cancer Inst. 106, djt361 (2014).

    PubMed  Google Scholar 

  77. Yamamoto, H. et al. Novel germline mutation in the transmembrane domain of HER2 in familial lung adenocarcinomas. J. Natl Cancer Inst. 106, djt338 (2013).

    PubMed  PubMed Central  Google Scholar 

  78. Janoueix-Lerosey, I. et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 455, 967–970 (2008).

    CAS  PubMed  Google Scholar 

  79. Mossé, Y. P. et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 455, 930–935 (2008).

    PubMed  PubMed Central  Google Scholar 

  80. Morris, S. et al. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin’s lymphoma. Science 263, 1281 (1994).

    CAS  PubMed  Google Scholar 

  81. Mulligan, L. M. RET revisited: expanding the oncogenic portfolio. Nat. Rev. Cancer 14, 173–186 (2014).

    CAS  PubMed  Google Scholar 

  82. Chompret, A. et al. PDGFRA germline mutation in a family with multiple cases of gastrointestinal stromal tumor. Gastroenterology 126, 318–321 (2004).

    CAS  PubMed  Google Scholar 

  83. Hirota, S. et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 279, 577–580 (1998).

    CAS  PubMed  Google Scholar 

  84. Nishida, T. et al. Familial gastrointestinal stromal tumours with germline mutation of the KIT gene. Nat. Genet. 19, 323–324 (1998).

    CAS  PubMed  Google Scholar 

  85. Farrell, B. & Breeze, A. L. Structure, activation and dysregulation of fibroblast growth factor receptor kinases: perspectives for clinical targeting. Biochem. Soc. Trans. 46, 1753–1770 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Muenke, M. et al. A unique point mutation in the fibroblast growth factor receptor 3 gene (FGFR3) defines a new craniosynostosis syndrome. Am. J. Hum. Genet. 60, 555–564 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Tavormina, P. L. et al. Thanatophoric dysplasia (types I and II) caused by distinct mutations in fibroblast growth factor receptor 3. Nat. Genet. 9, 321–328 (1995).

    CAS  PubMed  Google Scholar 

  88. Reardon, W. et al. Mutations in the fibroblast growth factor receptor 2 gene cause Crouzon syndrome. Nat. Genet. 8, 98–103 (1994).

    CAS  PubMed  Google Scholar 

  89. Hafner, C. et al. High frequency of FGFR3 mutations in adenoid seborrheic keratoses. J. Invest. Dermatol. 126, 2404–2407 (2006).

    CAS  PubMed  Google Scholar 

  90. Muenke, M. et al. A common mutation in the fibroblast growth factor receptor 1 gene in Pfeiffer syndrome. Nat. Genet. 8, 269–274 (1994).

    CAS  PubMed  Google Scholar 

  91. Wilkie, A. O. et al. Apert syndrome results from localized mutations of FGFR2 and is allelic with Crouzon syndrome. Nat. Genet. 9, 165–172 (1995).

    CAS  PubMed  Google Scholar 

  92. Simanshu, D. K., Nissley, D. V. & McCormick, F. RAS proteins and their regulators in human disease. Cell 170, 17–33 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Burd, C. E. et al. Mutation-specific RAS oncogenicity explains NRAS codon 61 selection in melanoma. Cancer Discov. 4, 1418–1429 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Nikolaev, S. I. et al. Somatic activating KRAS mutations in arteriovenous malformations of the brain. N. Engl. J. Med. 378, 250–261 (2018). In this work, the authors identify the presence of cancer-associated KRAS mutations in arteriovenous malformations of the brain.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Al-Olabi, L. et al. Mosaic RAS/MAPK variants cause sporadic vascular malformations which respond to targeted therapy. J. Clin. Invest. 128, 1496–1508 (2018).

    PubMed  PubMed Central  Google Scholar 

  96. Peacock, J. D. et al. Oculoectodermal syndrome is a mosaic RASopathy associated with KRAS alterations. Am. J. Med. Genet. A 167, 1429–1435 (2015).

    CAS  PubMed  Google Scholar 

  97. Hafner, C. & Groesser, L. Mosaic RASopathies. Cell Cycle 12, 43–50 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Groesser, L. et al. Phacomatosis pigmentokeratotica is caused by a postzygotic HRAS mutation in a multipotent progenitor cell. J. Invest. Dermatol. 133, 1998–2003 (2013).

    CAS  PubMed  Google Scholar 

  99. Levinsohn, J. L. et al. Somatic HRAS p.G12S mutation causes woolly hair and epidermal Nevi. J. Invest. Dermatol. 134, 1149–1152 (2014).

    CAS  PubMed  Google Scholar 

  100. Kinsler, V. A. et al. Multiple congenital melanocytic nevi and neurocutaneous melanosis are caused by postzygotic mutations in codon 61 of NRAS. J. Invest. Dermatol. 133, 2229–2236 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Charbel, C. et al. NRAS mutation is the sole recurrent somatic mutation in large congenital melanocytic nevi. J. Invest. Dermatol. 134, 1067–1074 (2014).

    CAS  PubMed  Google Scholar 

  102. Lim, Y. H. et al. Multilineage somatic activating mutations in HRAS and NRAS cause mosaic cutaneous and skeletal lesions, elevated FGF23 and hypophosphatemia. Hum. Mol. Genet. 23, 397–407 (2014).

    CAS  PubMed  Google Scholar 

  103. Lim, Y. H. et al. Cutaneous skeletal hypophosphatemia syndrome (CSHS) is a multilineage somatic mosaic RASopathy. J. Am. Acad. Dermatol. 75, 420–427 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Florenzano, P., Gafni, R. I. & Collins, M. T. Tumor-induced osteomalacia. Bone Rep. 7, 90–97 (2017).

    PubMed  PubMed Central  Google Scholar 

  105. Kang, H. et al. Somatic activating mutations in MAP2K1 cause melorheostosis. Nat. Commun. 9, 1–12 (2018).

    Google Scholar 

  106. Tidyman, W. E. & Rauen, K. A. The RASopathies: developmental syndromes of Ras/MAPK pathway dysregulation. Curr. Opin. Genet. Dev. 19, 230–236 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Goodwin, A. F. et al. Craniofacial and dental development in Costello syndrome. Am. J. Med. Genet. A 164A, 1425–1430 (2014).

    PubMed  Google Scholar 

  108. Gripp, K. W. et al. Costello syndrome: clinical phenotype, genotype, and management guidelines. Am. J. Med. Genet. A 179, 1725–1744 (2019).

    PubMed  PubMed Central  Google Scholar 

  109. Gripp, K. W. Tumor predisposition in Costello syndrome. Am. J. Med. Genet. C. Semin. Med Genet 137C, 72–77 (2005).

    PubMed  Google Scholar 

  110. Tartaglia, M. et al. Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome. Nat. Genet. 29, 465–468 (2001).

    CAS  PubMed  Google Scholar 

  111. Tartaglia, M. et al. Gain-of-function SOS1 mutations cause a distinctive form of Noonan syndrome. Nat. Genet. 39, 75–79 (2007).

    CAS  PubMed  Google Scholar 

  112. Aoki, Y. et al. Gain-of-function mutations in RIT1 cause Noonan syndrome, a RAS/MAPK pathway syndrome. Am. J. Hum. Genet. 93, 173–180 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Razzaque, M. A. et al. Germline gain-of-function mutations in RAF1 cause Noonan syndrome. Nat. Genet. 39, 1013–1017 (2007).

    CAS  PubMed  Google Scholar 

  114. Berger, A. H. et al. Oncogenic RIT1 mutations in lung adenocarcinoma. Oncogene 33, 4418–4423 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Lyons, J. et al. Two G protein oncogenes in human endocrine tumors. Science 249, 655–659 (1990).

    CAS  PubMed  Google Scholar 

  116. O’Hayre, M. et al. The emerging mutational landscape of G proteins and G-protein-coupled receptors in cancer. Nat. Rev. Cancer 13, 412–424 (2013).

    PubMed  PubMed Central  Google Scholar 

  117. Weinstein, L. S. et al. Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. N. Engl. J. Med. 325, 1688–1695 (1991). In this article, GNAS is identified as the causative gene for McCune–Albright syndrome. The mutations in this gene are similar to those in endocrine tumours.

    CAS  PubMed  Google Scholar 

  118. Albright, F., Butler, A. M., Hampton, A. O. & Smith, P. Syndrome characterized by osteitis fibrosa disseminata, areas of pigmentation and endocrine dysfunction, with precocious puberty in females. N. Engl. J. Med. 216, 727–746 (1937).

    Google Scholar 

  119. Song, Z.-J. et al. The genome-wide mutational landscape of pituitary adenomas. Cell Res. 26, 1255–1259 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Wu, J. et al. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci. Transl. Med. 3, 92ra66 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Hu, Q. & Shokat, K. M. Disease-causing mutations in the G protein Gαs subvert the roles of GDP and GTP. Cell 173, 1254–1264.e11 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Van Raamsdonk, C. D. et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue nevi. Nature 457, 599–602 (2009).

    PubMed  Google Scholar 

  123. Van Raamsdonk, C. D. et al. Mutations in GNA11 in uveal melanoma. N. Engl. J. Med. 363, 2191–2199 (2010).

    PubMed  PubMed Central  Google Scholar 

  124. Klebanov, N. et al. Use of targeted next-generation sequencing to identify activating hot spot mutations in cherry angiomas. JAMA Dermatol. 155, 211–215 (2019).

    PubMed  PubMed Central  Google Scholar 

  125. Bilanges, B., Posor, Y. & Vanhaesebroeck, B. PI3K isoforms in cell signalling and vesicle trafficking. Nat. Rev. Mol. Cell Biol. 20, 515–534 (2019).

    CAS  PubMed  Google Scholar 

  126. Fruman, D. A. et al. The PI3K pathway in human disease. Cell 170, 605–635 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Manning, B. D. & Toker, A. AKT/PKB signaling: navigating the network. Cell 169, 381–405 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 168, 960–976 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Carson, J. D. et al. Effects of oncogenic p110α subunit mutations on the lipid kinase activity of phosphoinositide 3-kinase. Biochem. J. 409, 519–524 (2008).

    CAS  PubMed  Google Scholar 

  130. Samuels, Y. et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 304, 554 (2004).

    CAS  PubMed  Google Scholar 

  131. Madsen, R. R., Vanhaesebroeck, B. & Semple, R. K. Cancer-associated PIK3CA mutations in overgrowth disorders. Trends Mol. Med. 24, 856–870 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Kurek, K. C. et al. Somatic mosaic activating mutations in PIK3CA cause CLOVES syndrome. Am. J. Hum. Genet. 90, 1108–1115 (2012). In this work, the authors identify mosaic cancer-associated PIK3CA mutations in patients with CLOVES, a syndrome that is now part of PROS.

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Rivière, J.-B. et al. De novo germline and postzygotic mutations in AKT3, PIK3R2 and PIK3CA cause a spectrum of related megalencephaly syndromes. Nat. Genet. 44, 934–940 (2012).

    PubMed  PubMed Central  Google Scholar 

  134. Lindhurst, M. J. et al. Mosaic overgrowth with fibroadipose hyperplasia is caused by somatic activating mutations in PIK3CA. Nat. Genet. 44, 928–933 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Keppler-Noreuil, K. M. et al. Clinical delineation and natural history of the PIK3CA-related overgrowth spectrum. Am. J. Med. Genet. A 164A, 1713–1733 (2014).

    PubMed  Google Scholar 

  136. Castel, P. et al. Somatic PIK3CA mutations as a driver of sporadic venous malformations. Sci. Transl. Med. 8, 332ra42 (2016).

    PubMed  PubMed Central  Google Scholar 

  137. Castillo, S. D. et al. Somatic activating mutations in Pik3ca cause sporadic venous malformations in mice and humans. Sci. Transl. Med. 8, 332ra43 (2016).

    PubMed  PubMed Central  Google Scholar 

  138. Luks, V. L. et al. Lymphatic and other vascular malformative/overgrowth disorders are caused by somatic mutations in PIK3CA. J. Pediatr. 166, 1048–1054.e1–5 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Orloff, M. S. et al. Germline PIK3CA and AKT1 mutations in Cowden and Cowden-like syndromes. Am. J. Hum. Genet. 92, 76–80 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Liaw, D. et al. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat. Genet. 16, 64–67 (1997).

    CAS  PubMed  Google Scholar 

  141. Angulo, I. et al. Phosphoinositide 3-kinase δ gene mutation predisposes to respiratory infection and airway damage. Science 342, 866–871 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Kracker, S. et al. Occurrence of B-cell lymphomas in patients with activated phosphoinositide 3-kinase δ syndrome. J. Allergy Clin. Immunol. 134, 233–236.e3 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Carpten, J. D. et al. A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature 448, 439–444 (2007).

    CAS  PubMed  Google Scholar 

  144. Lindhurst, M. J. et al. A mosaic activating mutation in AKT1 associated with the Proteus syndrome. N. Engl. J. Med. 365, 611–619 (2011). Sequencing of samples from a patient with Proteus syndrome reveals the presence of mosaic cancer-associated hotspot mutation AKT1 E17K, elucidating the causative gene in this syndrome.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Hussain, K. et al. An activating mutation of AKT2 and human hypoglycemia. Science 334, 474 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Khan, S. K. et al. Induced Gnas R201H expression from the endogenous Gnas locus causes fibrous dysplasia by up-regulating Wnt/β-catenin signaling. Proc. Natl Acad. Sci. USA 115, E418–E427 (2018).

    CAS  PubMed  Google Scholar 

  147. Lindhurst, M. J. et al. A mouse model of proteus syndrome. Hum. Mol. Genet. 28, 2920–2936 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Lieschke, G. J. & Currie, P. D. Animal models of human disease: zebrafish swim into view. Nat. Rev. Genet. 8, 353–367 (2007).

    CAS  PubMed  Google Scholar 

  149. Anastasaki, C., Rauen, K. A. & Patton, E. E. Continual low-level MEK inhibition ameliorates cardio-facio-cutaneous phenotypes in zebrafish. Dis. Model. Mech. 5, 546–552 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Hernández-Porras, I. et al. K-RasV14I recapitulates Noonan syndrome in mice. Proc. Natl Acad. Sci. USA 111, 16395–16400 (2014).

    PubMed  PubMed Central  Google Scholar 

  151. Wu, X. et al. MEK-ERK pathway modulation ameliorates disease phenotypes in a mouse model of Noonan syndrome associated with the Raf1 L613V mutation. J. Clin. Invest. 121, 1009–1025 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Komla-Ebri, D. et al. Tyrosine kinase inhibitor NVP-BGJ398 functionally improves FGFR3-related dwarfism in mouse model. J. Clin. Invest. 126, 1871–1884 (2016).

    PubMed  PubMed Central  Google Scholar 

  153. Venot, Q. et al. Targeted therapy in patients with PIK3CA-related overgrowth syndrome. Nature 558, 540–546 (2018). In this work, the authors describe 19 patients with PROS treated under compassionate use with an inhibitor targeting PI3Kα, which was developed and approved to treat metastatic breast cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Pauli, R. M. Achondroplasia: a comprehensive clinical review. Orphanet J. Rare Dis. 14, 1 (2019).

    PubMed  PubMed Central  Google Scholar 

  155. Wendt, D. J. et al. Neutral endopeptidase-resistant C-type natriuretic peptide variant represents a new therapeutic approach for treatment of fibroblast growth factor receptor 3-related dwarfism. J. Pharmacol. Exp. Ther. 353, 132–149 (2015).

    CAS  PubMed  Google Scholar 

  156. Savarirayan, R. et al. C-type natriuretic peptide analogue therapy in children with achondroplasia. N. Engl. J. Med. 381, 25–35 (2019).

    CAS  PubMed  Google Scholar 

  157. Inoue, S.-I., Morozumi, N., Yoshikiyo, K., Maeda, H. & Aoki, Y. C-type natriuretic peptide improves growth retardation in a mouse model of cardio-facio-cutaneous syndrome. Hum. Mol. Genet. 28, 74–83 (2019).

    CAS  PubMed  Google Scholar 

  158. Javle, M. et al. Phase II study of BGJ398 in patients with FGFR-altered advanced cholangiocarcinoma. J. Clin. Oncol. 36, 276–282 (2018).

    CAS  PubMed  Google Scholar 

  159. Dombi, E. et al. Activity of selumetinib in neurofibromatosis type 1–related plexiform neurofibromas. N. Engl. J. Med. 375, 2550–2560 (2016). In this study, and in reference 160, the authors report the safety and efficacy profiles of the MEK1/MEK2 inhibitor selumetinib for the treatment of plexiform neurofibromas in patients with neurofibromatosis type 1.

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Gross, A. M. et al. Selumetinib in children with inoperable plexiform neurofibromas. N. Engl. J. Med. 382, 1430–1442 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Rauen, K. A. et al. Proceedings of the fifth international RASopathies symposium: when development and cancer intersect. Am. J. Med. Genet. A 176, 2924–2929 (2018).

    PubMed  PubMed Central  Google Scholar 

  162. Andelfinger, G. et al. Hypertrophic cardiomyopathy in Noonan syndrome treated by MEK-inhibition. J. Am. Coll. Cardiol. 73, 2237–2239 (2019).

    PubMed  PubMed Central  Google Scholar 

  163. Rauen, K. A. The RASopathies. Annu. Rev. Genomics Hum. Genet. 14, 355–369 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Keppler-Noreuil, K. M. et al. Pharmacodynamic study of miransertib in individuals with Proteus syndrome. Am. J. Hum. Genet. 104, 484–491 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Leoni, C. et al. First evidence of a therapeutic effect of miransertib in a teenager with Proteus syndrome and ovarian carcinoma. Am. J. Med. Genet. A 179, 1319–1324 (2019).

    CAS  PubMed  Google Scholar 

  166. Rodon, J. & Tabernero, J. Improving the armamentarium of PI3K inhibitors with isoform-selective agents: a new light in the darkness. Cancer Discov. 7, 666–669 (2017).

    CAS  PubMed  Google Scholar 

  167. Grippo, P. J., Nowlin, P. S., Demeure, M. J., Longnecker, D. S. & Sandgren, E. P. Preinvasive pancreatic neoplasia of ductal phenotype induced by acinar cell targeting of mutant Kras in transgenic mice. Cancer Res. 63, 2016–2019 (2003).

    CAS  PubMed  Google Scholar 

  168. Gidekel Friedlander, S. Y. et al. Context-dependent transformation of adult pancreatic cells by oncogenic K-Ras. Cancer Cell 16, 379–389 (2009).

    PubMed  PubMed Central  Google Scholar 

  169. Johnson, L. et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature 410, 1111–1116 (2001).

    CAS  PubMed  Google Scholar 

  170. Braun, B. S. et al. Somatic activation of oncogenic Kras in hematopoietic cells initiates a rapidly fatal myeloproliferative disorder. Proc. Natl Acad. Sci. USA 101, 597–602 (2004).

    CAS  PubMed  Google Scholar 

  171. Haigis, K. M. et al. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat. Genet. 40, 600–608 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Johnson, C. W. et al. Isoform-specific destabilization of the active site reveals a molecular mechanism of intrinsic activation of KRas G13D. Cell Rep. 28, 1538–1550.e7 (2019).

    PubMed  PubMed Central  Google Scholar 

  173. Zhang, Z.-T. et al. Role of Ha- ras activation in superficial papillary pathway of urothelial tumor formation. Oncogene 20, 1973–1980 (2001).

    CAS  PubMed  Google Scholar 

  174. Schuhmacher, A. J. et al. A mouse model for Costello syndrome reveals an Ang II-mediated hypertensive condition. J. Clin. Invest. 118, 2169–2179 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Oba, D. et al. Mice with an oncogenic HRAS mutation are resistant to high-fat diet-induced obesity and exhibit impaired hepatic energy homeostasis. EBioMedicine 27, 138–150 (2017).

    PubMed  PubMed Central  Google Scholar 

  176. Li, Q.-F., Decker-Rockefeller, B., Bajaj, A. & Pumiglia, K. Activation of Ras in the vascular endothelium induces brain vascular malformations and hemorrhagic stroke. Cell Rep. 24, 2869–2882 (2018).

    CAS  PubMed  Google Scholar 

  177. Chin, L. et al. Cooperative effects of INK4a and ras in melanoma susceptibility in vivo. Genes Dev. 11, 2822–2834 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Ackermann, J. et al. Metastasizing melanoma formation caused by expression of activated N-RasQ61K on an INK4a-deficient background. Cancer Res. 65, 4005–4011 (2005).

    CAS  PubMed  Google Scholar 

  179. Li, Q. et al. Hematopoiesis and leukemogenesis in mice expressing oncogenic NrasG12D from the endogenous locus. Blood 117, 2022–2032 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Wang, Y. et al. Mutant N-RAS protects colorectal cancer cells from stress-induced apoptosis and contributes to cancer development and progression. Cancer Discov. 3, 294–307 (2013).

    CAS  PubMed  Google Scholar 

  181. Takahara, S. et al. New Noonan syndrome model mice with RIT1 mutation exhibit cardiac hypertrophy and susceptibility to β-adrenergic stimulation-induced cardiac fibrosis. EBioMedicine 42, 43–53 (2019).

    PubMed  PubMed Central  Google Scholar 

  182. Huang, J. L.-Y., Urtatiz, O. & Van Raamsdonk, C. D. Oncogenic G protein GNAQ induces uveal melanoma and intravasation in mice. Cancer Res. 75, 3384–3397 (2015).

    CAS  PubMed  Google Scholar 

  183. Moore, A. R. et al. GNA11 Q209L mouse model reveals RasGRP3 as an essential signaling node in uveal melanoma. Cell Rep. 22, 2455–2468 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Saggio, I. et al. Constitutive expression of GsαR201C in mice produces a heritable, direct replica of human fibrous dysplasia bone pathology and demonstrates its natural history. J. Bone Miner. Res. 29, 2357–2368 (2014).

    CAS  PubMed  Google Scholar 

  185. Dankort, D. et al. A new mouse model to explore the initiation, progression, and therapy of BRAF V600E-induced lung tumors. Genes Dev. 21, 379–384 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Dankort, D. et al. BrafV600E cooperates with Pten loss to induce metastatic melanoma. Nat. Genet. 41, 544–552 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Charles, R.-P., Iezza, G., Amendola, E., Dankort, D. & McMahon, M. Mutationally activated BRAFV600E elicits papillary thyroid cancer in the adult mouse. Cancer Res. 71, 3863–3871 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Inoue, S.-I. et al. Activated Braf induces esophageal dilation and gastric epithelial hyperplasia in mice. Hum. Mol. Genet. 26, 4715–4727 (2017).

    CAS  PubMed  Google Scholar 

  189. Collisson, E. A. et al. A central role for RAF→MEK→ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov. 2, 685–693 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Rad, R. et al. A genetic progression model of BrafV600E-induced intestinal tumorigenesis reveals targets for therapeutic intervention. Cancer Cell 24, 15–29 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Aoidi, R. et al. Mek1 Y130C mice recapitulate aspects of human cardio-facio-cutaneous syndrome. Dis. Model. Mech. 11, dmm031278 (2018).

    PubMed  PubMed Central  Google Scholar 

  192. Roy, A. et al. Mouse models of human PIK3CA-related brain overgrowth have acutely treatable epilepsy. eLife 4, e12703 (2015).

    PubMed  PubMed Central  Google Scholar 

  193. Stratikopoulos, E. E. et al. Mouse ER+/PIK3CAH1047R breast cancers caused by exogenous estrogen are heterogeneously dependent on estrogen and undergo BIM-dependent apoptosis with BH3 and PI3K agents. Oncogene 38, 47–59 (2019).

    CAS  PubMed  Google Scholar 

  194. Eser, S. et al. Selective requirement of PI3K/PDK1 signaling for Kras oncogene-driven pancreatic cell plasticity and cancer. Cancer Cell 23, 406–420 (2013).

    CAS  PubMed  Google Scholar 

  195. Avery, D. T. et al. Germline-activating mutations in PIK3CD compromise B cell development and function. J. Exp. Med. 215, 2073–2095 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Mancini, M. L., Lien, E. C. & Toker, A. Oncogenic AKT1E17K mutation induces mammary hyperplasia but prevents HER2-driven tumorigenesis. Oncotarget 7, 17301–17313 (2016).

    PubMed  PubMed Central  Google Scholar 

  197. Wang, Y. et al. A mouse model for achondroplasia produced by targeting fibroblast growth factor receptor 3. Proc. Natl Acad. Sci. USA 96, 4455–4460 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Logié, A. et al. Activating mutations of the tyrosine kinase receptor FGFR3 are associated with benign skin tumors in mice and humans. Hum. Mol. Genet. 14, 1153–1160 (2005).

    PubMed  Google Scholar 

  199. Ahmad, I. et al. K-Ras and β-catenin mutations cooperate with Fgfr3 mutations in mice to promote tumorigenesis in the skin and lung, but not in the bladder. Dis. Model. Mech. 4, 548–555 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Sommer, G. et al. Gastrointestinal stromal tumors in a mouse model by targeted mutation of the Kit receptor tyrosine kinase. Proc. Natl Acad. Sci. USA 100, 6706–6711 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Ono, S., Saito, T., Terui, K., Yoshida, H. & Enomoto, H. Generation of conditional ALK F1174L mutant mouse models for the study of neuroblastoma pathogenesis. Genesis 57, e23323 (2019).

    PubMed  Google Scholar 

  202. Berry, T. et al. The ALKF1174L mutation potentiates the oncogenic Activity of MYCN in Neuroblastoma. Cancer Cell 22, 117–130 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Chiarle, R. et al. NPM-ALK transgenic mice spontaneously develop T-cell lymphomas and plasma cell tumors. Blood 101, 1919–1927 (2003).

    CAS  PubMed  Google Scholar 

  204. Soda, M. et al. A mouse model for EML4-ALK-positive lung cancer. Proc. Natl Acad. Sci. USA 105, 19893–19897 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Politi, K. et al. Lung adenocarcinomas induced in mice by mutant EGF receptors found in human lung cancers respond to a tyrosine kinase inhibitor or to down-regulation of the receptors. Genes Dev. 20, 1496–1510 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Rous, P. A transmissible avian neoplasm. (Sarcoma of the common fowl.). J. Exp. Med. 12, 696–705 (1910).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Halberstaedter, L., Doljanski, L. & Tenenbaum, E. Experiments on the cancerization of cells in vitro by means of Rous sarcoma agent. Br. J. Exp. Pathol. 22, 179–187 (1941).

    CAS  PubMed Central  Google Scholar 

  208. Harvey, J. J. An unidentified virus which causes the rapid production of tumours in mice. Nature 204, 1104–1105 (1964).

    CAS  PubMed  Google Scholar 

  209. Stehelin, D., Varmus, H. E., Bishop, J. M. & Vogt, P. K. DNA related to the transforming gene(s) of avian sarcoma viruses is present in normal avian DNA. Nature 260, 170–173 (1976).

    CAS  PubMed  Google Scholar 

  210. Santos, E., Tronick, S. R., Aaronson, S. A., Pulciani, S. & Barbacid, M. T24 human bladder carcinoma oncogene is an activated form of the normal human homologue of BALB- and Harvey-MSV transforming genes. Nature 298, 343–347 (1982).

    CAS  PubMed  Google Scholar 

  211. Parada, L. F., Tabin, C. J., Shih, C. & Weinberg, R. A. Human EJ bladder carcinoma oncogene is homologue of Harvey sarcoma virus ras gene. Nature 297, 474–478 (1982).

    CAS  PubMed  Google Scholar 

  212. Der, C. J., Krontiris, T. G. & Cooper, G. M. Transforming genes of human bladder and lung carcinoma cell lines are homologous to the ras genes of Harvey and Kirsten sarcoma viruses. Proc. Natl Acad. Sci. USA 79, 3637–3640 (1982).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Whitman, M., Kaplan, D. R., Schaffhausen, B., Cantley, L. & Roberts, T. M. Association of phosphatidylinositol kinase activity with polyoma middle-T competent for transformation. Nature 315, 239–242 (1985).

    CAS  PubMed  Google Scholar 

  214. Gurumurthy, C. B. & Lloyd, K. C. K. Generating mouse models for biomedical research: technological advances. Dis. Model. Mech. 12, dmm029462 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Kersten, K., de Visser, K. E., van Miltenburg, M. H. & Jonkers, J. Genetically engineered mouse models in oncology research and cancer medicine. EMBO Mol. Med. 9, 137–153 (2017).

    CAS  PubMed  Google Scholar 

  216. Heyer, J., Kwong, L. N., Lowe, S. W. & Chin, L. Non-germline genetically engineered mouse models for translational cancer research. Nat. Rev. Cancer 10, 470–480 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  217. Zambrowicz, B. P. et al. Disruption of overlapping transcripts in the ROSA beta geo 26 gene trap strain leads to widespread expression of beta-galactosidase in mouse embryos and hematopoietic cells. Proc. Natl Acad. Sci. USA 94, 3789–3794 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Soriano, P. Generalized lacZ expression with the ROSA26 Cre reporter strain. Nat. Genet. 21, 70–71 (1999).

    CAS  PubMed  Google Scholar 

  219. Kinross, K. M. et al. An activating Pik3ca mutation coupled with Pten loss is sufficient to initiate ovarian tumorigenesis in mice. J. Clin. Invest. 122, 553–557 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. McLellan, M. A., Rosenthal, N. A. & Pinto, A. R. Cre-loxP-mediated recombination: general principles and experimental considerations. Curr. Protoc. Mouse Biol. 7, 1–12 (2017).

    PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank all the scientists who have contributed to this exciting field and apologize to those colleagues they were unable to cite. P.C. is a fellow of the Jane Coffin Childs Memorial Fund for Medical Research. This research was supported by the Thrasher Research Fund Early Career Award programme (to P.C.), the University of California, San Francisco Program for Breakthrough Biomedical Research Independent Postdoctoral Research Fellow (to P.C.) and the NIH/NCI grant R35CA197709-01 (to F.M.).

Author information

Authors and Affiliations

Authors

Contributions

P.C. conceived the ideas for this article and structured the manuscript. All authors contributed equally to writing and reviewing the manuscript.

Corresponding author

Correspondence to Pau Castel.

Ethics declarations

Competing interests

P.C. is a co-founder and advisory board member of Venthera. F.M. is a consultant for Aduro Biotech, Amgen, Daiichi, Ideaya Biosciences, Kura Oncology, Leidos Biomedical Research, PellePharm, Pfizer, PMV Pharma, Portola Pharmaceuticals and Quanta Therapeutics, has received research grants from Daiichi and Gilead Sciences and is a consultant for and cofounder of BridgeBio Pharma, DNAtrix, Olema Pharmaceuticals, and Quartz.

Additional information

Peer review information

Nature Reviews Cancer thanks S. Chanock, K. Haigis and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

cBioPortal: https://www.cbioportal.org/

GeneReviews: https://www.ncbi.nlm.nih.gov/books/NBK1116/

NSEuronet: https://nseuronet.com/php/index.php

Online Mendelian Inheritance in Man (OMIM): https://www.omim.org

Glossary

Allele bias

In the context of clinical genetics, this is when a specific mutation in a gene is far more frequent than expected.

Modifying alleles

Single-nucleotide polymorphisms that can either decrease or exacerbate a clinical phenotype driven by a pathogenic mutation.

Mosaicism

Characterized by the presence of cells with at least two distinct genetic make-ups.

Schimmelpenning–Feuerstein–Mims syndrome

Neuro-oculocutaneous mosaicism characterized by the presence of skin lesions and pigmentation abnormalities, epilepsy, epibulbar dermoids, cloudy cornea, eyelid colobomas and arteriovascular defects, among other manifestations.

Blaschko lines

Skin patterns found in adults that recapitulate the normal cell development during embryogenesis. These can be often appreciated in individuals with genetically driven skin stains.

Field cancerization

The presence of large areas of tissue affected by carcinogenic mutations, which often contribute to malignant transformation. It is generally the result of a genotoxic exposure during a prolonged time and can lead to the presence of low-grade and high-grade tumours.

Achondroplasia

An autosomal dominant syndrome that is the most common form of skeletal dysplasia in humans and is caused by the FGFR3 mutation G380R. Patients exhibit macrocephaly and short limbs.

Acanthosis nigricans

A hyperpigmentation and hyperkeratosis of the skin.

Arteriovenous malformations

Abnormal blood vessels that tangle and allow direct connection between veins and arteries and can cause pain and severe haemorrhage if ruptured.

G protein-coupled receptor-associated GTPases

Gα proteins are bound to Gβγ, forming an inactive trimeric complex that associates with G protein-coupled receptors (GPCRs). On GPCR stimulation, conformational changes in the receptor lead to Gβγ dissociation and Gα GTP loading and activation, resulting in the production of second messengers; for Gαs (encoded by GNAS) adenylate cyclase and production of cAMP, and for Gαq and Gα11 (encoded by GNAQ and GNA11, respectively) phospholipase C, resulting in diacylglycerol and inositol trisphosphate.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Castel, P., Rauen, K.A. & McCormick, F. The duality of human oncoproteins: drivers of cancer and congenital disorders. Nat Rev Cancer 20, 383–397 (2020). https://doi.org/10.1038/s41568-020-0256-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-0256-z

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer