DOI QR코드

DOI QR Code

The history and regulatory mechanism of the Hippo pathway

  • Kim, Wantae (Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB)) ;
  • Jho, Eek-hoon (Departement of Life Science, University of Seoul)
  • Received : 2018.01.08
  • Published : 2018.03.31

Abstract

How the organ size is adjusted to the proper size during development and how organs know that they reach the original size during regeneration remain long-standing questions. Based on studies using multiple model organisms and approaches for over 20 years, a consensus has been established that the Hippo pathway plays crucial roles in controlling organ size and maintaining tissue homeostasis. Given the significance of these processes, the dysregulation of the Hippo pathway has also implicated various diseases, such as tissue degeneration and cancer. By regulating the downstream transcriptional coactivators YAP and TAZ, the Hippo pathway coordinates cell proliferation and apoptosis in response to a variety of signals including cell contact inhibition, polarity, mechanical sensation and soluble factors. Since the core components and their functions of the Hippo pathway are evolutionarily conserved, this pathway serves as a global regulator of organ size control. Therefore, further investigation of the regulatory mechanisms will provide physiological insights to better understand tissue homeostasis. In this review, the historical developments and current understandings of the regulatory mechanism of Hippo signaling pathway are discussed.

Keywords

References

  1. Shingleton AW (2010) The regulation of organ size in Drosophila: physiology, plasticity, patterning and physical force. Organogenesis 6, 76-87 https://doi.org/10.4161/org.6.2.10375
  2. Gokhale RH and Shingleton AW (2015) Size control: the developmental physiology of body and organ size regulation. Wiley Interdiscip Rev Dev Biol 4, 335-356 https://doi.org/10.1002/wdev.181
  3. Halder G and Johnson RL (2011) Hippo signaling: growth control and beyond. Development 138, 9-22 https://doi.org/10.1242/dev.045500
  4. Twitty VCS, Josep L (1931) The growth of eyes and limbs transplanted heteroplastically between two species of Amblystoma. J Exp Zool 59, 61-86 https://doi.org/10.1002/jez.1400590105
  5. Michalopoulos GK and DeFrances MC (1997) Liver regeneration. Science 276, 60-66 https://doi.org/10.1126/science.276.5309.60
  6. Metcalf D (1964) Restricted Growth Capacity of Multiple Spleen Grafts. Transplantation 2, 387-392 https://doi.org/10.1097/00007890-196405000-00008
  7. Metcalf D (1963) The Autonomous Behaviour of Normal Thymus Grafts. Aust J Exp Biol Med Sci 41, SUPPL437-447 https://doi.org/10.1038/icb.1963.64
  8. Hilman D and Gat U (2011) The evolutionary history of YAP and the hippo/YAP pathway. Mol Biol Evol 28, 2403-2417 https://doi.org/10.1093/molbev/msr065
  9. Sebe-Pedros A, Zheng Y, Ruiz-Trillo I and Pan D (2012) Premetazoan origin of the hippo signaling pathway. Cell Rep 1, 13-20 https://doi.org/10.1016/j.celrep.2011.11.004
  10. Huang J, Wu S, Barrera J, Matthews K and Pan D (2005) The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila Homolog of YAP. Cell 122, 421-434 https://doi.org/10.1016/j.cell.2005.06.007
  11. Meng Z, Moroishi T and Guan KL (2016) Mechanisms of Hippo pathway regulation. Genes Dev 30, 1-17 https://doi.org/10.1101/gad.274027.115
  12. Piccolo S, Dupont S and Cordenonsi M (2014) The biology of YAP/TAZ: hippo signaling and beyond. Physiol Rev 94, 1287-1312 https://doi.org/10.1152/physrev.00005.2014
  13. Zhao B, Li L, Tumaneng K, Wang CY and Guan KL (2010) A coordinated phosphorylation by Lats and CK1 regulates YAP stability through SCF(beta-TRCP). Genes Dev 24, 72-85 https://doi.org/10.1101/gad.1843810
  14. Liu CY, Zha ZY, Zhou X et al (2010) The hippo tumor pathway promotes TAZ degradation by phosphorylating a phosphodegron and recruiting the SCF{beta}-TrCP E3 ligase. J Biol Chem 285, 37159-37169 https://doi.org/10.1074/jbc.M110.152942
  15. Huang W, Lv X, Liu C et al (2012) The N-terminal phosphodegron targets TAZ/WWTR1 protein for SCFbeta-TrCP-dependent degradation in response to phosphatidylinositol 3-kinase inhibition. J Biol Chem 287, 26245-26253 https://doi.org/10.1074/jbc.M112.382036
  16. Hansen CG, Moroishi T and Guan KL (2015) YAP and TAZ: a nexus for Hippo signaling and beyond. Trends Cell Biol 25, 499-513 https://doi.org/10.1016/j.tcb.2015.05.002
  17. Meng Z, Moroishi T, Mottier-Pavie V et al (2015) MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Nat Commun 6, 8357 https://doi.org/10.1038/ncomms9357
  18. Kim Y, Kim W, Song Y et al (2017) Deubiquitinase YOD1 potentiates YAP/TAZ activities through enhancing ITCH stability. Proc Natl Acad Sci U S A 114, 4691-4696 https://doi.org/10.1073/pnas.1620306114
  19. Yin F, Yu J, Zheng Y, Chen Q, Zhang N and Pan D (2013) Spatial organization of Hippo signaling at the plasma membrane mediated by the tumor suppressor Merlin/NF2. Cell 154, 1342-1355 https://doi.org/10.1016/j.cell.2013.08.025
  20. Fulford A, Tapon N and Ribeiro PS (2017) Upstairs, downstairs: spatial regulation of Hippo signalling. Curr Opin Cell Biol 51, 22-32
  21. Chaulk SG, Lattanzi VJ, Hiemer SE, Fahlman RP and Varelas X (2014) The Hippo pathway effectors TAZ/YAP regulate dicer expression and microRNA biogenesis through Let-7. J Biol Chem 289, 1886-1891 https://doi.org/10.1074/jbc.C113.529362
  22. Mori M, Triboulet R, Mohseni M et al (2014) Hippo signaling regulates microprocessor and links celldensity- dependent miRNA biogenesis to cancer. Cell 156, 893-906 https://doi.org/10.1016/j.cell.2013.12.043
  23. Sabine A, Bovay E, Demir CS et al (2015) FOXC2 and fluid shear stress stabilize postnatal lymphatic vasculature. J Clin Invest 125, 3861-3877 https://doi.org/10.1172/JCI80454
  24. Wang L, Luo JY, Li B et al (2016) Integrin-YAP/TAZ-JNK cascade mediates atheroprotective effect of unidirectional shear flow. Nature 540, 579-582 https://doi.org/10.1038/nature20602
  25. Liu B, Zheng Y, Yin F, Yu J, Silverman N and Pan D (2016) Toll Receptor-Mediated Hippo Signaling Controls Innate Immunity in Drosophila. Cell 164, 406-419 https://doi.org/10.1016/j.cell.2015.12.029
  26. Wang S, Xie F, Chu F et al (2017) YAP antagonizes innate antiviral immunity and is targeted for lysosomal degradation through IKKvarepsilon-mediated phosphorylation. Nat Immunol 18, 733-743 https://doi.org/10.1038/ni.3744
  27. Wilkinson DS, Jariwala JS, Anderson E et al (2015) Phosphorylation of LC3 by the Hippo kinases STK3/STK4 is essential for autophagy. Mol Cell 57, 55-68 https://doi.org/10.1016/j.molcel.2014.11.019
  28. Maejima Y, Kyoi S, Zhai P et al (2013) Mst1 inhibits autophagy by promoting the interaction between Beclin1 and Bcl-2. Nat Med 19, 1478-1488 https://doi.org/10.1038/nm.3322
  29. Zhang S, Chen Q, Liu Q et al (2017) Hippo Signaling Suppresses Cell Ploidy and Tumorigenesis through Skp2. Cancer Cell 31, 669-684 e7 https://doi.org/10.1016/j.ccell.2017.04.004
  30. Justice RW, Zilian O, Woods DF, Noll M and Bryant PJ (1995) The Drosophila tumor suppressor gene warts encodes a homolog of human myotonic dystrophy kinase and is required for the control of cell shape and proliferation. Genes Dev 9, 534-546 https://doi.org/10.1101/gad.9.5.534
  31. Xu T, Wang W, Zhang S, Stewart RA and Yu W (1995) Identifying tumor suppressors in genetic mosaics: the Drosophila lats gene encodes a putative protein kinase. Development 121, 1053-1063
  32. St John MA, Tao W, Fei X et al (1999) Mice deficient of Lats1 develop soft-tissue sarcomas, ovarian tumours and pituitary dysfunction. Nat Genet 21, 182-186 https://doi.org/10.1038/5965
  33. Tao W, Zhang S, Turenchalk GS et al (1999) Human homologue of the Drosophila melanogaster lats tumour suppressor modulates CDC2 activity. Nat Genet 21, 177-181 https://doi.org/10.1038/5960
  34. Tapon N, Harvey KF, Bell DW et al (2002) salvador Promotes both cell cycle exit and apoptosis in Drosophila and is mutated in human cancer cell lines. Cell 110, 467-478 https://doi.org/10.1016/S0092-8674(02)00824-3
  35. Kango-Singh M, Nolo R, Tao C et al (2002) Shar-pei mediates cell proliferation arrest during imaginal disc growth in Drosophila. Development 129, 5719-5730 https://doi.org/10.1242/dev.00168
  36. Harvey KF, Pfleger CM and Hariharan IK (2003) The Drosophila Mst ortholog, hippo, restricts growth and cell proliferation and promotes apoptosis. Cell 114, 457-467 https://doi.org/10.1016/S0092-8674(03)00557-9
  37. Udan RS, Kango-Singh M, Nolo R, Tao C and Halder G (2003) Hippo promotes proliferation arrest and apoptosis in the Salvador/Warts pathway. Nat Cell Biol 5, 914-920 https://doi.org/10.1038/ncb1050
  38. Pantalacci S, Tapon N and Leopold P (2003) The Salvador partner Hippo promotes apoptosis and cell-cycle exit in Drosophila. Nat Cell Biol 5, 921-927 https://doi.org/10.1038/ncb1051
  39. Jia J, Zhang W, Wang B, Trinko R and Jiang J (2003) The Drosophila Ste20 family kinase dMST functions as a tumor suppressor by restricting cell proliferation and promoting apoptosis. Genes Dev 17, 2514-2519 https://doi.org/10.1101/gad.1134003
  40. Wu S, Huang J, Dong J and Pan D (2003) hippo encodes a Ste-20 family protein kinase that restricts cell proliferation and promotes apoptosis in conjunction with salvador and warts. Cell 114, 445-456 https://doi.org/10.1016/S0092-8674(03)00549-X
  41. Lai ZC, Wei X, Shimizu T et al (2005) Control of cell proliferation and apoptosis by mob as tumor suppressor, mats. Cell 120, 675-685 https://doi.org/10.1016/j.cell.2004.12.036
  42. Wei X, Shimizu T and Lai ZC (2007) Mob as tumor suppressor is activated by Hippo kinase for growth inhibition in Drosophila. EMBO J 26, 1772-1781 https://doi.org/10.1038/sj.emboj.7601630
  43. Zhao B, Wei X, Li W et al (2007) Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev 21, 2747-2761 https://doi.org/10.1101/gad.1602907
  44. Zhang N, Bai H, David KK et al (2010) The Merlin/NF2 tumor suppressor functions through the YAP oncoprotein to regulate tissue homeostasis in mammals. Dev Cell 19, 27-38 https://doi.org/10.1016/j.devcel.2010.06.015
  45. Overholtzer M, Zhang J, Smolen GA et al (2006) Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc Natl Acad Sci U S A 103, 12405-12410 https://doi.org/10.1073/pnas.0605579103
  46. Harvey KF, Zhang X and Thomas DM (2013) The Hippo pathway and human cancer. Nat Rev Cancer 13, 246-257 https://doi.org/10.1038/nrc3458
  47. Johnson R and Halder G (2014) The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov 13, 63-79 https://doi.org/10.1038/nrd4161
  48. Moroishi T, Hansen CG and Guan KL (2015) The emerging roles of YAP and TAZ in cancer. Nat Rev Cancer 15, 73-79 https://doi.org/10.1038/nrc3876
  49. Sudol M (1994) Yes-associated protein (YAP65) is a proline-rich phosphoprotein that binds to the SH3 domain of the Yes proto-oncogene product. Oncogene 9, 2145-2152
  50. Komuro A, Nagai M, Navin NE and Sudol M (2003) WW domain-containing protein YAP associates with ErbB-4 and acts as a co-transcriptional activator for the carboxyl-terminal fragment of ErbB-4 that translocates to the nucleus. J Biol Chem 278, 33334-33341 https://doi.org/10.1074/jbc.M305597200
  51. Ferrigno O, Lallemand F, Verrecchia F et al (2002) Yes-associated protein (YAP65) interacts with Smad7 and potentiates its inhibitory activity against TGF-beta/Smad signaling. Oncogene 21, 4879-4884 https://doi.org/10.1038/sj.onc.1205623
  52. Strano S, Munarriz E, Rossi M et al (2001) Physical interaction with Yes-associated protein enhances p73 transcriptional activity. J Biol Chem 276, 15164-15173 https://doi.org/10.1074/jbc.M010484200
  53. Zaidi SK, Sullivan AJ, Medina R et al (2004) Tyrosine phosphorylation controls Runx2-mediated subnuclear targeting of YAP to repress transcription. EMBO J 23, 790-799 https://doi.org/10.1038/sj.emboj.7600073
  54. Vassilev A, Kaneko KJ, Shu H, Zhao Y and DePamphilis ML (2001) TEAD/TEF transcription factors utilize the activation domain of YAP65, a Src/Yes-associated protein localized in the cytoplasm. Genes Dev 15, 1229-1241 https://doi.org/10.1101/gad.888601
  55. Yagi R, Chen LF, Shigesada K, Murakami Y and Ito Y (1999) A WW domain-containing yes-associated protein (YAP) is a novel transcriptional co-activator. EMBO J 18, 2551-2562 https://doi.org/10.1093/emboj/18.9.2551
  56. Espanel X and Sudol M (2001) Yes-associated protein and p53-binding protein-2 interact through their WW and SH3 domains. J Biol Chem 276, 14514-14523 https://doi.org/10.1074/jbc.M008568200
  57. Ota M and Sasaki H (2008) Mammalian Tead proteins regulate cell proliferation and contact inhibition as transcriptional mediators of Hippo signaling. Development 135, 4059-4069 https://doi.org/10.1242/dev.027151
  58. Wu S, Liu Y, Zheng Y, Dong J and Pan D (2008) The TEAD/TEF family protein Scalloped mediates transcriptional output of the Hippo growth-regulatory pathway. Dev Cell 14, 388-398 https://doi.org/10.1016/j.devcel.2008.01.007
  59. Zhang L, Ren F, Zhang Q, Chen Y, Wang B and Jiang J (2008) The TEAD/TEF family of transcription factor Scalloped mediates Hippo signaling in organ size control. Dev Cell 14, 377-387 https://doi.org/10.1016/j.devcel.2008.01.006
  60. Zhao B, Ye X, Yu J et al (2008) TEAD mediates YAP-dependent gene induction and growth control. Genes Dev 22, 1962-1971 https://doi.org/10.1101/gad.1664408
  61. Schlegelmilch K, Mohseni M, Kirak O et al (2011) Yap1 acts downstream of alpha-catenin to control epidermal proliferation. Cell 144, 782-795 https://doi.org/10.1016/j.cell.2011.02.031
  62. von Gise A, Lin Z, Schlegelmilch K et al (2012) YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy. Proc Natl Acad Sci U S A 109, 2394-2399 https://doi.org/10.1073/pnas.1116136109
  63. Zhang W, Gao Y, Li P et al (2014) VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex. Cell Res 24, 331-343 https://doi.org/10.1038/cr.2014.10
  64. Koontz LM, Liu-Chittenden Y, Yin F et al (2013) The Hippo effector Yorkie controls normal tissue growth by antagonizing scalloped-mediated default repression. Dev Cell 25, 388-401 https://doi.org/10.1016/j.devcel.2013.04.021
  65. Hamaratoglu F, Willecke M, Kango-Singh M et al (2006) The tumour-suppressor genes NF2/Merlin and Expanded act through Hippo signalling to regulate cell proliferation and apoptosis. Nat Cell Biol 8, 27-36 https://doi.org/10.1038/ncb1339
  66. Willecke M, Hamaratoglu F, Kango-Singh M et al (2006) The fat cadherin acts through the hippo tumorsuppressor pathway to regulate tissue size. Curr Biol 16, 2090-2100 https://doi.org/10.1016/j.cub.2006.09.005
  67. Silva E, Tsatskis Y, Gardano L, Tapon N and McNeill H (2006) The tumor-suppressor gene fat controls tissue growth upstream of expanded in the hippo signaling pathway. Curr Biol 16, 2081-2089 https://doi.org/10.1016/j.cub.2006.09.004
  68. Cho E, Feng Y, Rauskolb C, Maitra S, Fehon R and Irvine KD (2006) Delineation of a Fat tumor suppressor pathway. Nat Genet 38, 1142-1150 https://doi.org/10.1038/ng1887
  69. Poon CL, Lin JI, Zhang X and Harvey KF (2011) The sterile 20-like kinase Tao-1 controls tissue growth by regulating the Salvador-Warts-Hippo pathway. Dev Cell 21, 896-906 https://doi.org/10.1016/j.devcel.2011.09.012
  70. Boggiano JC, Vanderzalm PJ and Fehon RG (2011) Tao-1 phosphorylates Hippo/MST kinases to regulate the Hippo-Salvador-Warts tumor suppressor pathway. Dev Cell 21, 888-895 https://doi.org/10.1016/j.devcel.2011.08.028
  71. Chung HL, Augustine GJ and Choi KW (2016) Drosophila Schip1 Links Expanded and Tao-1 to Regulate Hippo Signaling. Dev Cell 36, 511-524 https://doi.org/10.1016/j.devcel.2016.02.004
  72. Yu J, Zheng Y, Dong J, Klusza S, Deng WM and Pan D (2010) Kibra functions as a tumor suppressor protein that regulates Hippo signaling in conjunction with Merlin and Expanded. Dev Cell 18, 288-299 https://doi.org/10.1016/j.devcel.2009.12.012
  73. Genevet A, Wehr MC, Brain R, Thompson BJ and Tapon N (2010) Kibra is a regulator of the Salvador/Warts/Hippo signaling network. Dev Cell 18, 300-308 https://doi.org/10.1016/j.devcel.2009.12.011
  74. Baumgartner R, Poernbacher I, Buser N, Hafen E and Stocker H (2010) The WW domain protein Kibra acts upstream of Hippo in Drosophila. Dev Cell 18, 309-316 https://doi.org/10.1016/j.devcel.2009.12.013
  75. Su T, Ludwig MZ, Xu J and Fehon RG (2017) Kibra and Merlin Activate the Hippo Pathway Spatially Distinct from and Independent of Expanded. Dev Cell 40, 478-490 e473 https://doi.org/10.1016/j.devcel.2017.02.004
  76. Thompson BJ, Pichaud F and Roper K (2013) Sticking together the Crumbs - an unexpected function for an old friend. Nat Rev Mol Cell Biol 14, 307-314
  77. Chen CL, Gajewski KM, Hamaratoglu F et al (2010) The apical-basal cell polarity determinant Crumbs regulates Hippo signaling in Drosophila. Proc Natl Acad Sci U S A 107, 15810-15815 https://doi.org/10.1073/pnas.1004060107
  78. Ling C, Zheng Y, Yin F et al (2010) The apical transmembrane protein Crumbs functions as a tumor suppressor that regulates Hippo signaling by binding to Expanded. Proc Natl Acad Sci U S A 107, 10532-10537 https://doi.org/10.1073/pnas.1004279107
  79. Grzeschik NA, Parsons LM, Allott ML, Harvey KF and Richardson HE (2010) Lgl, aPKC, and Crumbs regulate the Salvador/Warts/Hippo pathway through two distinct mechanisms. Curr Biol 20, 573-581 https://doi.org/10.1016/j.cub.2010.01.055
  80. Varelas X, Samavarchi-Tehrani P, Narimatsu M et al (2010) The Crumbs complex couples cell density sensing to Hippo-dependent control of the TGF-beta- SMAD pathway. Dev Cell 19, 831-844 https://doi.org/10.1016/j.devcel.2010.11.012
  81. Hirano S, Kimoto N, Shimoyama Y, Hirohashi S and Takeichi M (1992) Identification of a neural alphacatenin as a key regulator of cadherin function and multicellular organization. Cell 70, 293-301 https://doi.org/10.1016/0092-8674(92)90103-J
  82. Herrenknecht K, Ozawa M, Eckerskorn C, Lottspeich F, Lenter M and Kemler R (1991) The uvomorulin-anchorage protein alpha catenin is a vinculin homologue. Proc Natl Acad Sci U S A 88, 9156-9160 https://doi.org/10.1073/pnas.88.20.9156
  83. Silvis MR, Kreger BT, Lien WH et al (2011) alpha-catenin is a tumor suppressor that controls cell accumulation by regulating the localization and activity of the transcriptional coactivator Yap1. Sci Signal 4, ra33
  84. Wei SY, Escudero LM, Yu F et al (2005) Echinoid is a component of adherens junctions that cooperates with DE-Cadherin to mediate cell adhesion. Dev Cell 8, 493-504 https://doi.org/10.1016/j.devcel.2005.03.015
  85. Yue T, Tian A and Jiang J (2012) The cell adhesion molecule echinoid functions as a tumor suppressor and upstream regulator of the Hippo signaling pathway. Dev Cell 22, 255-267 https://doi.org/10.1016/j.devcel.2011.12.011
  86. Nishioka N, Inoue K, Adachi K et al (2009) The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell 16, 398-410 https://doi.org/10.1016/j.devcel.2009.02.003
  87. Wang W, Huang J, Wang X et al (2012) PTPN14 is required for the density-dependent control of YAP1. Genes Dev 26, 1959-1971 https://doi.org/10.1101/gad.192955.112
  88. Liu X, Yang N, Figel SA et al (2013) PTPN14 interacts with and negatively regulates the oncogenic function of YAP. Oncogene 32, 1266-1273 https://doi.org/10.1038/onc.2012.147
  89. Michaloglou C, Lehmann W, Martin T et al (2013) The tyrosine phosphatase PTPN14 is a negative regulator of YAP activity. PLoS One 8, e61916 https://doi.org/10.1371/journal.pone.0061916
  90. Moon S, Kim W, Kim S et al (2017) Phosphorylation by NLK inhibits YAP-14-3-3-interactions and induces its nuclear localization. EMBO Rep 18, 61-71 https://doi.org/10.15252/embr.201642683
  91. Dupont S, Morsut L, Aragona M et al (2011) Role of YAP/TAZ in mechanotransduction. Nature 474, 179-183 https://doi.org/10.1038/nature10137
  92. Halder G, Dupont S and Piccolo S (2012) Transduction of mechanical and cytoskeletal cues by YAP and TAZ. Nat Rev Mol Cell Biol 13, 591-600 https://doi.org/10.1038/nrm3416
  93. Panciera T, Azzolin L, Cordenonsi M and Piccolo S (2017) Mechanobiology of YAP and TAZ in physiology and disease. Nat Rev Mol Cell Biol 12, 758-770
  94. Zhao B, Li L, Wang L, Wang CY, Yu J and Guan KL (2012) Cell detachment activates the Hippo pathway via cytoskeleton reorganization to induce anoikis. Genes Dev 26, 54-68 https://doi.org/10.1101/gad.173435.111
  95. Kim NG and Gumbiner BM (2015) Adhesion to fibronectin regulates Hippo signaling via the FAK-Src-PI3K pathway. J Cell Biol 210, 503-515 https://doi.org/10.1083/jcb.201501025
  96. Elosegui-Artola A, Andreu I, Beedle AEM et al (2017) Force Triggers YAP Nuclear Entry by Regulating Transport across Nuclear Pores. Cell 171, 1397-1410 https://doi.org/10.1016/j.cell.2017.10.008
  97. Mo JS (2017) The role of extracellular biophysical cues in modulating the Hippo-YAP pathway. BMB Rep 50, 71-78 https://doi.org/10.5483/BMBRep.2017.50.2.199
  98. Wang Z, Wu Y, Wang H et al (2014) Interplay of mevalonate and Hippo pathways regulates RHAMM transcription via YAP to modulate breast cancer cell motility. Proc Natl Acad Sci U S A 111, E89-98 https://doi.org/10.1073/pnas.1319190110
  99. Sorrentino G, Ruggeri N, Specchia V et al (2014) Metabolic control of YAP and TAZ by the mevalonate pathway. Nat Cell Biol 16, 357-366 https://doi.org/10.1038/ncb2936
  100. Mo JS, Meng Z, Kim YC et al (2015) Cellular energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nat Cell Biol 17, 500-510 https://doi.org/10.1038/ncb3111
  101. Wang W, Xiao ZD, Li X et al (2015) AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nat Cell Biol 17, 490-499 https://doi.org/10.1038/ncb3113
  102. Zhang X, Qiao Y, Wu Q et al (2017) The essential role of YAP O-GlcNAcylation in high-glucose-stimulated liver tumorigenesis. Nat Commun 8, 15280 https://doi.org/10.1038/ncomms15280
  103. Peng C, Zhu Y, Zhang W et al (2017) Regulation of the Hippo-YAP Pathway by Glucose Sensor O-GlcNAcylation. Mol Cell 68, 591-604 e595 https://doi.org/10.1016/j.molcel.2017.10.010
  104. Ma B, Chen Y, Chen L et al (2015) Hypoxia regulates Hippo signalling through the SIAH2 ubiquitin E3 ligase. Nat Cell Biol 17, 95-103
  105. Ma X, Zhang H, Xue X and Shah YM (2017) Hypoxia-inducible factor 2alpha (HIF-2alpha) promotes colon cancer growth by potentiating Yes-associated protein 1 (YAP1) activity. J Biol Chem 292, 17046-17056 https://doi.org/10.1074/jbc.M117.805655
  106. Xiang L, Gilkes DM, Hu H et al (2014) Hypoxiainducible factor 1 mediates TAZ expression and nuclear localization to induce the breast cancer stem cell phenotype. Oncotarget 5, 12509-12527
  107. Ma B, Cheng H, Gao R et al (2016) Zyxin-Siah2-Lats2 axis mediates cooperation between Hippo and TGF-beta signalling pathways. Nat Commun 7, 11123 https://doi.org/10.1038/ncomms11123
  108. Hong AW, Meng Z, Yuan HX et al (2017) Osmotic stress-induced phosphorylation by NLK at Ser128 activates YAP. EMBO Rep 18, 72-86 https://doi.org/10.15252/embr.201642681
  109. Lin KC, Moroishi T, Meng Z et al (2017) Regulation of Hippo pathway transcription factor TEAD by p38 MAPK-induced cytoplasmic translocation. Nat Cell Biol 19, 996-1002 https://doi.org/10.1038/ncb3581
  110. Taylor LK, Wang HC and Erikson RL (1996) Newly identified stress-responsive protein kinases, Krs-1 and Krs-2. Proc Natl Acad Sci U S A 93, 10099-10104 https://doi.org/10.1073/pnas.93.19.10099
  111. Lehtinen MK, Yuan Z, Boag PR et al (2006) A conserved MST-FOXO signaling pathway mediates oxidative-stress responses and extends life span. Cell 125, 987-1001 https://doi.org/10.1016/j.cell.2006.03.046
  112. Shao D, Zhai P, Del Re DP et al (2014) A functional interaction between Hippo-YAP signalling and FoxO1 mediates the oxidative stress response. Nat Commun 5, 3315
  113. Nusse R and Clevers H (2017) Wnt/beta-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 169, 985-999 https://doi.org/10.1016/j.cell.2017.05.016
  114. Kim W, Kim M and Jho EH (2013) Wnt/beta-catenin signalling: from plasma membrane to nucleus. Biochem J 450, 9-21 https://doi.org/10.1042/BJ20121284
  115. Varelas X, Miller BW, Sopko R et al (2010) The Hippo pathway regulates Wnt/beta-catenin signaling. Dev Cell 18, 579-591 https://doi.org/10.1016/j.devcel.2010.03.007
  116. Imajo M, Miyatake K, Iimura A, Miyamoto A and Nishida E (2012) A molecular mechanism that links Hippo signalling to the inhibition of Wnt/beta-catenin signalling. EMBO J 31, 1109-1122 https://doi.org/10.1038/emboj.2011.487
  117. Barry ER, Morikawa T, Butler BL et al (2013) Restriction of intestinal stem cell expansion and the regenerative response by YAP. Nature 493, 106-110
  118. Tsutsumi R, Masoudi M, Takahashi A et al (2013) YAP and TAZ, Hippo signaling targets, act as a rheostat for nuclear SHP2 function. Dev Cell 26, 658-665 https://doi.org/10.1016/j.devcel.2013.08.013
  119. Heallen T, Zhang M, Wang J et al (2011) Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science 332, 458-461 https://doi.org/10.1126/science.1199010
  120. Kim W, Khan SK, Gvozdenovic-Jeremic J et al (2017) Hippo signaling interactions with Wnt/beta-catenin and Notch signaling repress liver tumorigenesis. J Clin Invest 127, 137-152
  121. Fitamant J, Kottakis F, Benhamouche S et al (2015) YAP Inhibition Restores Hepatocyte Differentiation in Advanced HCC, Leading to Tumor Regression. Cell Rep 10, 1692-1707 https://doi.org/10.1016/j.celrep.2015.02.027
  122. Azzolin L, Zanconato F, Bresolin S et al (2012) Role of TAZ as mediator of Wnt signaling. Cell 151, 1443-1456 https://doi.org/10.1016/j.cell.2012.11.027
  123. Azzolin L, Panciera T, Soligo S et al (2014) YAP/TAZ incorporation in the beta-catenin destruction complex orchestrates the Wnt response. Cell 158, 157-170 https://doi.org/10.1016/j.cell.2014.06.013
  124. Cai J, Maitra A, Anders RA, Taketo MM and Pan D (2015) beta-Catenin destruction complex-independent regulation of Hippo-YAP signaling by APC in intestinal tumorigenesis. Genes Dev 29, 1493-1506 https://doi.org/10.1101/gad.264515.115
  125. Yu FX, Zhao B, Panupinthu N et al (2012) Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 150, 780-791 https://doi.org/10.1016/j.cell.2012.06.037
  126. Miller E, Yang J, DeRan M et al (2012) Identification of serum-derived sphingosine-1-phosphate as a small molecule regulator of YAP. Chem Biol 19, 955-962 https://doi.org/10.1016/j.chembiol.2012.07.005
  127. Park HW, Kim YC, Yu B et al (2015) Alternative Wnt Signaling Activates YAP/TAZ. Cell 162, 780-794 https://doi.org/10.1016/j.cell.2015.07.013
  128. Struhl G, Fitzgerald K and Greenwald I (1993) Intrinsic activity of the Lin-12 and Notch intracellular domains in vivo. Cell 74, 331-345 https://doi.org/10.1016/0092-8674(93)90424-O
  129. Schroeter EH, Kisslinger JA and Kopan R (1998) Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain. Nature 393, 382-386 https://doi.org/10.1038/30756
  130. Bray SJ (2016) Notch signalling in context. Nat Rev Mol Cell Biol 17, 722-735 https://doi.org/10.1038/nrm.2016.94
  131. Meignin C, Alvarez-Garcia I, Davis I and Palacios IM (2007) The salvador-warts-hippo pathway is required for epithelial proliferation and axis specification in Drosophila. Curr Biol 17, 1871-1878 https://doi.org/10.1016/j.cub.2007.09.062
  132. Polesello C and Tapon N (2007) Salvador-warts-hippo signaling promotes Drosophila posterior follicle cell maturation downstream of notch. Curr Biol 17, 1864-1870 https://doi.org/10.1016/j.cub.2007.09.049
  133. Camargo FD, Gokhale S, Johnnidis JB et al (2007) YAP1 increases organ size and expands undifferentiated progenitor cells. Curr Biol 17, 2054-2060 https://doi.org/10.1016/j.cub.2007.10.039
  134. Zhou D, Zhang Y, Wu H et al (2011) Mst1 and Mst2 protein kinases restrain intestinal stem cell proliferation and colonic tumorigenesis by inhibition of Yes-associated protein (Yap) overabundance. Proc Natl Acad Sci U S A 108, E1312-1320 https://doi.org/10.1073/pnas.1110428108
  135. Yimlamai D, Christodoulou C, Galli GG et al (2014) Hippo pathway activity influences liver cell fate. Cell 157, 1324-1338 https://doi.org/10.1016/j.cell.2014.03.060
  136. Manderfield LJ, Aghajanian H, Engleka KA et al (2015) Hippo signaling is required for Notch-dependent smooth muscle differentiation of neural crest. Development 142, 2962-2971 https://doi.org/10.1242/dev.125807
  137. Rayon T, Menchero S, Nieto A et al (2014) Notch and hippo converge on Cdx2 to specify the trophectoderm lineage in the mouse blastocyst. Dev Cell 30, 410-422 https://doi.org/10.1016/j.devcel.2014.06.019
  138. Tschaharganeh DF, Chen X, Latzko P et al (2013) Yes-associated protein up-regulates Jagged-1 and activates the Notch pathway in human hepatocellular carcinoma. Gastroenterology 144, 1530-1542 e1512 https://doi.org/10.1053/j.gastro.2013.02.009
  139. Massague J (2012) TGFbeta signalling in context. Nat Rev Mol Cell Biol 13, 616-630 https://doi.org/10.1038/nrm3434
  140. Luo K (2017) Signaling Cross Talk between TGF-beta/Smad and Other Signaling Pathways. Cold Spring Harb Perspect Biol 9, a022137 https://doi.org/10.1101/cshperspect.a022137
  141. Ayyaz A, Attisano L and Wrana JL (2017) Recent advances in understanding contextual TGFbeta signaling. F1000Res 6, 749 https://doi.org/10.12688/f1000research.11295.1
  142. Attisano L and Wrana JL (2013) Signal integration in TGF-beta, WNT, and Hippo pathways. F1000Prime Rep 5, 17
  143. Varelas X, Sakuma R, Samavarchi-Tehrani P et al (2008) TAZ controls Smad nucleocytoplasmic shuttling and regulates human embryonic stem-cell self-renewal. Nat Cell Biol 10, 837-848 https://doi.org/10.1038/ncb1748
  144. Nallet-Staub F, Yin X, Gilbert C et al (2015) Cell density sensing alters TGF-beta signaling in a cell-type-specific manner, independent from Hippo pathway activation. Dev Cell 32, 640-651 https://doi.org/10.1016/j.devcel.2015.01.011
  145. Mahoney JE, Mori M, Szymaniak AD, Varelas X and Cardoso WV (2014) The hippo pathway effector Yap controls patterning and differentiation of airway epithelial progenitors. Dev Cell 30, 137-150 https://doi.org/10.1016/j.devcel.2014.06.003
  146. Zhao R, Fallon TR, Saladi SV et al (2014) Yap tunes airway epithelial size and architecture by regulating the identity, maintenance, and self-renewal of stem cells. Dev Cell 30, 151-165 https://doi.org/10.1016/j.devcel.2014.06.004
  147. Lee DH, Park JO, Kim TS et al (2016) LATS-YAP/TAZ controls lineage specification by regulating TGFbeta signaling and Hnf4alpha expression during liver development. Nat Commun 7, 11961 https://doi.org/10.1038/ncomms11961
  148. Lai D and Yang X (2013) BMP4 is a novel transcriptional target and mediator of mammary cell migration downstream of the Hippo pathway component TAZ. Cell Signal 25, 1720-1728 https://doi.org/10.1016/j.cellsig.2013.05.002
  149. Nishio M, Sugimachi K, Goto H et al (2016) Dysregulated YAP1/TAZ and TGF-beta signaling mediate hepatocarcinogenesis in Mob1a/1b-deficient mice. Proc Natl Acad Sci U S A 113, E71-80 https://doi.org/10.1073/pnas.1517188113
  150. Pefani DE, Pankova D, Abraham AG et al (2016) TGF-beta Targets the Hippo Pathway Scaffold RASSF1A to Facilitate YAP/SMAD2 Nuclear Translocation. Mol Cell 63, 156-166 https://doi.org/10.1016/j.molcel.2016.05.012
  151. Stroschein SL, Wang W, Zhou S, Zhou Q and Luo K (1999) Negative feedback regulation of TGF-beta signaling by the SnoN oncoprotein. Science 286, 771-774 https://doi.org/10.1126/science.286.5440.771
  152. Zhu Q, Le Scolan E, Jahchan N, Ji X, Xu A and Luo K (2016) SnoN Antagonizes the Hippo Kinase Complex to Promote TAZ Signaling during Breast Carcinogenesis. Dev Cell 37, 399-412 https://doi.org/10.1016/j.devcel.2016.05.002

Cited by

  1. Repurposing of Drugs Targeting YAP-TEAD Functions vol.10, pp.9, 2018, https://doi.org/10.3390/cancers10090329
  2. Enhancement of connexin30.3 expression in mouse embryonic stem cell line EB3 in response to cell–cell contacts pp.1749-0774, 2019, https://doi.org/10.1007/s13577-018-00235-z