DOI QR코드

DOI QR Code

Regulation of the Hippo signaling pathway by ubiquitin modification

  • Kim, Youngeun (Department of Life Science, University of Seoul) ;
  • Jho, Eek-hoon (Department of Life Science, University of Seoul)
  • Received : 2017.12.31
  • Published : 2018.03.31

Abstract

The Hippo signaling pathway plays an essential role in adult tissue homeostasis and organ size control. Abnormal regulation of Hippo signaling can be a cause for multiple types of human cancers. Since the awareness of the importance of the Hippo signaling in a wide range of biological fields has been continually grown, it is also understood that a thorough and well-rounded comprehension of the precise dynamics could provide fundamental insights for therapeutic applications. Several components in the Hippo signaling pathway are known to be targeted for proteasomal degradation via ubiquitination by E3 ligases. ${\beta}-TrCP$ is a well-known E3 ligase of YAP/TAZ, which leads to the reduction of YAP/TAZ levels. The Hippo signaling pathway can also be inhibited by the E3 ligases (such as ITCH) which target LATS1/2 for degradation. Regulation via ubiquitination involves not only complex network of E3 ligases but also deubiquitinating enzymes (DUBs), which remove ubiquitin from its targets. Interestingly, non-degradative ubiquitin modifications are also known to play important roles in the regulation of Hippo signaling. Although there has been much advanced progress in the investigation of ubiquitin modifications acting as regulators of the Hippo signaling pathway, research done to date still remains inadequate due to the sheer complexity and diversity of the subject. Herein, we review and discuss recent developments that implicate ubiquitin-mediated regulatory mechanisms at multiple steps of the Hippo signaling pathway.

Keywords

References

  1. 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
  2. Zanconato F, Cordenonsi M and Piccolo S (2016) YAP/TAZ at the Roots of Cancer. Cancer Cell 29, 783-803 https://doi.org/10.1016/j.ccell.2016.05.005
  3. Hershko A, Ciechanover A (1998) The ubiquitin system. Annu Rev Biochem 67, 425-479 https://doi.org/10.1146/annurev.biochem.67.1.425
  4. Koegl M, Hoppe T, Schlenker S, Ulrich HD, Mayer TU and Jentsch S (1999) A novel ubiquitination factor, E4, is involved in multiubiquitin chain assembly. Cell 96, 635-644 https://doi.org/10.1016/S0092-8674(00)80574-7
  5. Ikeda F and Dikic I (2008) Atypical ubiquitin chains: new molecular signals. 'Protein Modifications: Beyond the Usual Suspects' review series. EMBO Rep 9, 536-542 https://doi.org/10.1038/embor.2008.93
  6. Kirisako T, Kamei K, Murata S et al (2006) A ubiquitin ligase complex assembles linear polyubiquitin chains. EMBO J 25, 4877-4887 https://doi.org/10.1038/sj.emboj.7601360
  7. Li W, Bengtson MH, Ulbrich A et al (2008) Genome-wide and functional annotation of human E3 ubiquitin ligases identifies MULAN, a mitochondrial E3 that regulates the organelle's dynamics and signaling. PLoS One 3, e1487 https://doi.org/10.1371/journal.pone.0001487
  8. Nijman SM, Luna-Vargas MP, Velds A et al (2005) A genomic and functional inventory of deubiquitinating enzymes. Cell 123, 773-786 https://doi.org/10.1016/j.cell.2005.11.007
  9. Canning M, Boutell C, Parkinson J and Everett RD (2004) A RING finger ubiquitin ligase is protected from autocatalyzed ubiquitination and degradation by binding to ubiquitin-specific protease USP7. J Biol Chem 279, 38160-38168 https://doi.org/10.1074/jbc.M402885200
  10. Wu X, Yen L, Irwin L, Sweeney C and Carraway KL 3rd (2004) Stabilization of the E3 ubiquitin ligase Nrdp1 by the deubiquitinating enzyme USP8. Mol Cell Biol 24, 7748-7757 https://doi.org/10.1128/MCB.24.17.7748-7757.2004
  11. Hetfeld BK, Helfrich A, Kapelari B et al (2005) The zinc finger of the CSN-associated deubiquitinating enzyme USP15 is essential to rescue the E3 ligase Rbx1. Curr Biol 15, 1217-1221 https://doi.org/10.1016/j.cub.2005.05.059
  12. Brooks CL, Li M, Hu M, Shi Y and Gu W (2007) The p53--Mdm2--HAUSP complex is involved in p53 stabilization by HAUSP. Oncogene 26, 7262-7266 https://doi.org/10.1038/sj.onc.1210531
  13. Trompouki E, Hatzivassiliou E, Tsichritzis T, Farmer H, Ashworth A and Mosialos G (2003) CYLD is a deubiquitinating enzyme that negatively regulates NF-kappaB activation by TNFR family members. Nature 424, 793-796 https://doi.org/10.1038/nature01803
  14. Wertz IE, O'Rourke KM, Zhou H et al (2004) De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature 430, 694-699 https://doi.org/10.1038/nature02794
  15. 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
  16. 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
  17. 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
  18. Tu K, Yang W, Li C et al (2014) Fbxw7 is an independent prognostic marker and induces apoptosis and growth arrest by regulating YAP abundance in hepatocellular carcinoma. Mol Cancer 13, 110 https://doi.org/10.1186/1476-4598-13-110
  19. 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
  20. Chan SW, Lim CJ, Chong YF, Pobbati AV, Huang C and Hong W (2011) Hippo pathway-independent restriction of TAZ and YAP by angiomotin. J Biol Chem 286, 7018-7026 https://doi.org/10.1074/jbc.C110.212621
  21. Wang W, Huang J and Chen J (2011) Angiomotin-like proteins associate with and negatively regulate YAP1. J Biol Chem 286, 4364-4370 https://doi.org/10.1074/jbc.C110.205401
  22. Zhao B, Li L, Lu Q et al (2011) Angiomotin is a novel Hippo pathway component that inhibits YAP oncoprotein. Genes Dev 25, 51-63 https://doi.org/10.1101/gad.2000111
  23. Wang C, An J, Zhang P et al (2015) The Nedd4-like ubiquitin E3 ligases target angiomotin/p130 to ubiquitindependent degradation. Biochem J 444, 279-289
  24. Wang W, Li N, Li X, Tran MK, Han X and Chen J (2015) Tankyrase Inhibitors Target YAP by Stabilizing Angiomotin Family Proteins. Cell Rep 13, 524-532 https://doi.org/10.1016/j.celrep.2015.09.014
  25. Zhang Y, Liu S, Mickanin C et al (2011) RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nat Cell Biol 13, 623-629 https://doi.org/10.1038/ncb2222
  26. Thanh Nguyen H, Andrejeva D, Gupta R et al (2016) Deubiquitylating enzyme USP9x regulates hippo pathway activity by controlling angiomotin protein turnover. Cell Discov 2, 16001
  27. 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
  28. Lei QY, Zhang H, Zhao B et al (2008) TAZ promotes cell proliferation and epithelial-mesenchymal transition and is inhibited by the hippo pathway. Mol Cell Biol 28, 2426-36 https://doi.org/10.1128/MCB.01874-07
  29. 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
  30. 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
  31. 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
  32. Visser-Grieve S, Zhou Z, She YM et al (2011) LATS1 tumor suppressor is a novel actin-binding protein and negative regulator of actin polymerization. Cell Res 21, 1513-1516 https://doi.org/10.1038/cr.2011.122
  33. Mo JS, Yu FX, Gong R, Brown JH and Guan KL (2012) Regulation of the Hippo-YAP pathway by protease-activated receptors (PARs). Genes Dev 26, 2138-2143 https://doi.org/10.1101/gad.197582.112
  34. 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
  35. 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
  36. Wada K, Itoga K, Okano T, Yonemura S and Sasaki H (2011) Hippo pathway regulation by cell morphology and stress fibers. Development 138, 3907-3914 https://doi.org/10.1242/dev.070987
  37. Chen HI and Sudol M (1995) The WW domain of Yes-associated protein binds a proline-rich ligand that differs from the consensus established for Src homology 3-binding modules. Proc Natl Acad Sci U S A 92, 7819-7823 https://doi.org/10.1073/pnas.92.17.7819
  38. Ho KC, Zhou Z, She YM, Chun A, Cyr TD and Yang X (2011) Itch E3 ubiquitin ligase regulates large tumor suppressor 1 stability [corrected]. Proc Natl Acad Sci U S A 108, 4870-5 https://doi.org/10.1073/pnas.1101273108
  39. Salah Z, Melino G and Aqeilan RI (2011) Negative regulation of the Hippo pathway by E3 ubiquitin ligase ITCH is sufficient to promote tumorigenicity. Cancer Res 71, 2010-2020 https://doi.org/10.1158/0008-5472.CAN-10-3516
  40. Salah Z, Cohen S, Itzhaki E and Aqeilan RI (2013) NEDD4 E3 ligase inhibits the activity of the Hippo pathway by targeting LATS1 for degradation. Cell Cycle 12, 3817-3823 https://doi.org/10.4161/cc.26672
  41. Bae SJ, Kim M, Kim SH et al (2015) NEDD4 controls intestinal stem cell homeostasis by regulating the Hippo signalling pathway. Nat Commun 6, 6314 https://doi.org/10.1038/ncomms7314
  42. Yeung B, Ho KC and Yang X (2013) WWP1 E3 ligase targets LATS1 for ubiquitin-mediated degradation in breast cancer cells. PLoS One 8, e61027 https://doi.org/10.1371/journal.pone.0061027
  43. Cao L, Wang P, Gao Y, Lin X, Wang F and Wu S (2014) Ubiquitin E3 ligase dSmurf is essential for Wts protein turnover and Hippo signaling. Biochem Biophys Res Commun 454, 167-171 https://doi.org/10.1016/j.bbrc.2014.10.058
  44. Li W, You L, Cooper J et al (2010) Merlin/NF2 suppresses tumorigenesis by inhibiting the E3 ubiquitin ligase CRL4(DCAF1) in the nucleus. Cell 140, 477-490 https://doi.org/10.1016/j.cell.2010.01.029
  45. Li W, Cooper J, Zhou L et al (2014) Merlin/NF2 loss-driven tumorigenesis linked to CRL4(DCAF1)-mediated inhibition of the hippo pathway kinases Lats1 and 2 in the nucleus. Cancer Cell 26, 48-60 https://doi.org/10.1016/j.ccr.2014.05.001
  46. Deng J, Lei W, Xiang X et al (2016) Cullin 4A (CUL4A), a direct target of miR-9 and miR-137, promotes gastric cancer proliferation and invasion by regulating the Hippo signaling pathway. Oncotarget 7, 10037-10050
  47. Sang Y, Yan F and Ren X (2015) The role and mechanism of CRL4 E3 ubiquitin ligase in cancer and its potential therapy implications. Oncotarget 6, 42590-42602
  48. 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
  49. Kim M, Kim M, Park SJ, Lee C and Lim DS (2016) Role of Angiomotin-like 2 mono-ubiquitination on YAP inhibition. EMBO Rep 17, 64-78 https://doi.org/10.15252/embr.201540809
  50. 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
  51. Praskova M, Xia F and Avruch J (2008) MOBKL1A/MOBKL1B phosphorylation by MST1 and MST2 inhibits cell proliferation. Curr Biol 18, 311-321 https://doi.org/10.1016/j.cub.2008.02.006
  52. 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
  53. Chan EH, Nousiainen M, Chalamalasetty RB, Schafer A, Nigg EA and Sillje HH (2005) The Ste20-like kinase Mst2 activates the human large tumor suppressor kinase Lats1. Oncogene 24, 2076-2086 https://doi.org/10.1038/sj.onc.1208445
  54. Lignitto L, Arcella A, Sepe M et al (2013) Proteolysis of MOB1 by the ubiquitin ligase praja2 attenuates Hippo signalling and supports glioblastoma growth. Nat Commun 4, 1822 https://doi.org/10.1038/ncomms2791
  55. 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
  56. Callus BA, Verhagen AM and Vaux DL (2006) Vaux, Association of mammalian sterile twenty kinases, Mst1 and Mst2, with hSalvador via C-terminal coiled-coil domains, leads to its stabilization and phosphorylation. FEBS J 273, 4264-4276 https://doi.org/10.1111/j.1742-4658.2006.05427.x
  57. Lee JH, Kim TS, Yang TH et al (2008) A crucial role of WW45 in developing epithelial tissues in the mouse. EMBO J 27, 1231-1242 https://doi.org/10.1038/emboj.2008.63
  58. Lu L, Li Y, Kim SM et al (2010) Hippo signaling is a potent in vivo growth and tumor suppressor pathway in the mammalian liver. Proc Natl Acad Sci U S A 107, 1437-1442 https://doi.org/10.1073/pnas.0911427107
  59. Aerne BL, Gailite I, Sims D and Tapon N (2015) Hippo Stabilises Its Adaptor Salvador by Antagonising the HECT Ubiquitin Ligase Herc4. PLoS One 10, e0131113 https://doi.org/10.1371/journal.pone.0131113
  60. 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
  61. 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
  62. Praskova M, Khoklatchev A, Ortiz-Vega S and Avruch J (2004) Regulation of the MST1 kinase by autophosphorylation, by the growth inhibitory proteins, RASSF1 and NORE1, and by Ras. Biochem J 381(Pt 2), 453-462 https://doi.org/10.1042/BJ20040025
  63. Yuan Z, Kim D, Shu S et al (2010) Phosphoinositide 3-kinase/Akt inhibits MST1-mediated pro-apoptotic signaling through phosphorylation of threonine 120. J Biol Chem 285, 3815-3824 https://doi.org/10.1074/jbc.M109.059675
  64. Collak FK, Yagiz K, Luthringer DJ, Erkaya B and Cinar B (2012) Threonine-120 phosphorylation regulated by phosphoinositide-3-kinase/Akt and mammalian target of rapamycin pathway signaling limits the antitumor activity of mammalian sterile 20-like kinase 1. J Biol Chem 287, 23698-23709 https://doi.org/10.1074/jbc.M112.358713
  65. Xiao L, Chen D, Hu P et al (2011) The c-Abl-MST1 signaling pathway mediates oxidative stress-induced neuronal cell death. J Neurosci 31, 9611-9619 https://doi.org/10.1523/JNEUROSCI.0035-11.2011
  66. Chen Y, Wang Z, Wang P, Li D, Zhou J and Wu S (2014) CYLD negatively regulates Hippo signaling by limiting Hpo phosphorylation in Drosophila. Biochem Biophys Res Commun 452, 808-812 https://doi.org/10.1016/j.bbrc.2014.09.005
  67. Rodriguez-Boulan E and Nelson WJ (1989) Morphogenesis of the polarized epithelial cell phenotype. Science 245, 718-725 https://doi.org/10.1126/science.2672330
  68. Gumbiner BM (1996) Cell adhesion: the molecular basis of tissue architecture and morphogenesis. Cell 84, 345-357 https://doi.org/10.1016/S0092-8674(00)81279-9
  69. 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
  70. 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
  71. 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
  72. Robinson BS, Huang J, Hong Y and Moberg KH (2010) Crumbs regulates Salvador/Warts/Hippo signaling in Drosophila via the FERM-domain protein Expanded. Curr Biol 20, 582-590 https://doi.org/10.1016/j.cub.2010.03.019
  73. Ribeiro P, Holder M, Frith D, Snijders AP and Tapon N (2014) Crumbs promotes expanded recognition and degradation by the SCF(Slimb/beta-TrCP) ubiquitin ligase. Proc Natl Acad Sci U S A 111, E1980- E19809 https://doi.org/10.1073/pnas.1315508111