Ubiquitin Ligases Involved in the Regulation of Wnt, TGF-β, and Notch Signaling Pathways and Their Roles in Mouse Development and Homeostasis
Jazyk angličtina Země Švýcarsko Médium electronic
Typ dokumentu časopisecké články, práce podpořená grantem, přehledy
PubMed
31623112
PubMed Central
PMC6826584
DOI
10.3390/genes10100815
PII: genes10100815
Knihovny.cz E-zdroje
- Klíčová slova
- cancer, gene inactivation, mouse model, ubiquitin–proteasome system,
- MeSH
- beta-katenin metabolismus MeSH
- buněčná diferenciace fyziologie MeSH
- homeostáza genetika MeSH
- ligasy metabolismus MeSH
- myši embryologie genetika MeSH
- proliferace buněk fyziologie MeSH
- proteiny Wnt metabolismus MeSH
- receptory Notch metabolismus MeSH
- signální dráha Wnt fyziologie MeSH
- transformující růstový faktor beta metabolismus MeSH
- transkripční faktory metabolismus MeSH
- ubikvitin metabolismus MeSH
- ubikvitinligasy metabolismus fyziologie MeSH
- vývojová regulace genové exprese genetika MeSH
- zvířata MeSH
- Check Tag
- myši embryologie genetika MeSH
- zvířata MeSH
- Publikační typ
- časopisecké články MeSH
- práce podpořená grantem MeSH
- přehledy MeSH
- Názvy látek
- beta-katenin MeSH
- ligasy MeSH
- proteiny Wnt MeSH
- receptory Notch MeSH
- transformující růstový faktor beta MeSH
- transkripční faktory MeSH
- ubikvitin MeSH
- ubikvitinligasy MeSH
The Wnt, TGF-β, and Notch signaling pathways are essential for the regulation of cellular polarity, differentiation, proliferation, and migration. Differential activation and mutual crosstalk of these pathways during animal development are crucial instructive forces in the initiation of the body axis and the development of organs and tissues. Due to the ability to initiate cell proliferation, these pathways are vulnerable to somatic mutations selectively producing cells, which ultimately slip through cellular and organismal checkpoints and develop into cancer. The architecture of the Wnt, TGF-β, and Notch signaling pathways is simple. The transmembrane receptor, activated by the extracellular stimulus, induces nuclear translocation of the transcription factor, which subsequently changes the expression of target genes. Nevertheless, these pathways are regulated by a myriad of factors involved in various feedback mechanisms or crosstalk. The most prominent group of regulators is the ubiquitin-proteasome system (UPS). To open the door to UPS-based therapeutic manipulations, a thorough understanding of these regulations at a molecular level and rigorous confirmation in vivo are required. In this quest, mouse models are exceptional and, thanks to the progress in genetic engineering, also an accessible tool. Here, we reviewed the current understanding of how the UPS regulates the Wnt, TGF-β, and Notch pathways and we summarized the knowledge gained from related mouse models.
Zobrazit více v PubMed
Sanchez-Vega F., Mina M., Armenia J., Chatila W.K., Luna A., La K.C., Dimitriadoy S., Liu D.L., Kantheti H.S., Saghafinia S., et al. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell. 2018;173:321–337. doi: 10.1016/j.cell.2018.03.035. PubMed DOI PMC
Kinzler K.W., Nilbert M.C., Su L.K., Vogelstein B., Bryan T.M., Levy D.B., Smith K.J., Preisinger A.C., Hedge P., McKechnie D., et al. Identification of FAP locus genes from chromosome 5q21. Science. 1991;253:661–665. doi: 10.1126/science.1651562. PubMed DOI
Ellisen L.W., Bird J., West D.C., Soreng A.L., Reynolds T.C., Smith S.D., Sklar J. TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. Cell. 1991;66:649–661. doi: 10.1016/0092-8674(91)90111-B. PubMed DOI
Yakicier M.C., Irmak M.B., Romano A., Kew M., Ozturk M. Smad2 and Smad4 gene mutations in hepatocellular carcinoma. Oncogene. 1999;18:4879–4883. doi: 10.1038/sj.onc.1202866. PubMed DOI
Manasanch E.E., Orlowski R.Z. Proteasome inhibitors in cancer therapy. Nat. Rev. Clin. Oncol. 2017;14:417–433. doi: 10.1038/nrclinonc.2016.206. PubMed DOI PMC
Skaar J.R., Pagan J.K., Pagano M. SCF ubiquitin ligase-targeted therapies. Nat. Rev. Drug Discov. 2014;13:889–903. doi: 10.1038/nrd4432. PubMed DOI PMC
Kumari N., Jaynes P.W., Saei A., Iyengar P.V., Richard J.L.C., Eichhorn P.J.A. The roles of ubiquitin modifying enzymes in neoplastic disease. Biochim. Biophys. Acta Rev. Cancer. 2017;1868:456–483. doi: 10.1016/j.bbcan.2017.09.002. PubMed DOI
Nalepa G., Rolfe M., Harper J.W. Drug discovery in the ubiquitin-proteasome system. Nat. Rev. Drug Discov. 2006;5:596–613. doi: 10.1038/nrd2056. PubMed DOI
Buetow L., Huang D.T. Structural insights into the catalysis and regulation of E3 ubiquitin ligases. Nat. Rev. Mol. Cell Biol. 2016;17:626–642. doi: 10.1038/nrm.2016.91. PubMed DOI PMC
Hershko A., Heller H., Elias S., Ciechanover A. Components of ubiquitin-protein ligase system. Resolution, affinity purification, and role in protein breakdown. J. Biol. Chem. 1983;258:8206–8214. PubMed
Deol K.K., Lorenz S., Strieter E.R. Enzymatic Logic of Ubiquitin Chain Assembly. Front. Physiol. 2019;10:835. doi: 10.3389/fphys.2019.00835. PubMed DOI PMC
Stewart M.D., Ritterhoff T., Klevit R.E., Brzovic P.S. E2 enzymes: More than just middle men. Cell Res. 2016;26:423–440. doi: 10.1038/cr.2016.35. PubMed DOI PMC
Rittinger K., Ikeda F. Linear ubiquitin chains: Enzymes, mechanisms and biology. Open Biol. 2017:7. doi: 10.1098/rsob.170026. PubMed DOI PMC
Akutsu M., Dikic I., Bremm A. Ubiquitin chain diversity at a glance. J. Cell Sci. 2016;129:875–880. doi: 10.1242/jcs.183954. PubMed DOI
Nakayama K.I., Nakayama K. Ubiquitin ligases: Cell-cycle control and cancer. Nat. Rev. Cancer. 2006;6:369–381. doi: 10.1038/nrc1881. PubMed DOI
Zheng N., Shabek N. Ubiquitin Ligases: Structure, Function, and Regulation. Annu. Rev. Biochem. 2017;86:129–157. doi: 10.1146/annurev-biochem-060815-014922. PubMed DOI
Lilley C.E., Chaurushiya M.S., Boutell C., Landry S., Suh J., Panier S., Everett R.D., Stewart G.S., Durocher D., Weitzman M.D. A viral E3 ligase targets RNF8 and RNF168 to control histone ubiquitination and DNA damage responses. EMBO J. 2010;29:943–955. doi: 10.1038/emboj.2009.400. PubMed DOI PMC
Huen M.S., Grant R., Manke I., Minn K., Yu X., Yaffe M.B., Chen J. RNF8 transduces the DNA-damage signal via histone ubiquitylation and checkpoint protein assembly. Cell. 2007;131:901–914. doi: 10.1016/j.cell.2007.09.041. PubMed DOI PMC
Skaar J.R., Pagan J.K., Pagano M. Mechanisms and function of substrate recruitment by F-box proteins. Nat. Rev. Mol. Cell Biol. 2013;14:369–381. doi: 10.1038/nrm3582. PubMed DOI PMC
Honda R., Tanaka H., Yasuda H. Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett. 1997;420:25–27. doi: 10.1016/S0014-5793(97)01480-4. PubMed DOI
de Toledo S.M., Azzam E.I., Dahlberg W.K., Gooding T.B., Little J.B. ATM complexes with HDM2 and promotes its rapid phosphorylation in a p53-independent manner in normal and tumor human cells exposed to ionizing radiation. Oncogene. 2000;19:6185–6193. doi: 10.1038/sj.onc.1204020. PubMed DOI
Khosravi R., Maya R., Gottlieb T., Oren M., Shiloh Y., Shkedy D. Rapid ATM-dependent phosphorylation of MDM2 precedes p53 accumulation in response to DNA damage. Proc. Natl. Acad. Sci. USA. 1999;96:14973–14977. doi: 10.1073/pnas.96.26.14973. PubMed DOI PMC
Frescas D., Pagano M. Deregulated proteolysis by the F-box proteins SKP2 and beta-TrCP: Tipping the scales of cancer. Nat. Rev. Cancer. 2008;8:438–449. doi: 10.1038/nrc2396. PubMed DOI PMC
Bulatov E., Ciulli A. Targeting Cullin-RING E3 ubiquitin ligases for drug discovery: Structure, assembly and small-molecule modulation. Biochem. J. 2015;467:365–386. doi: 10.1042/BJ20141450. PubMed DOI PMC
Aravind L., Koonin E.V. The U box is a modified RING finger—A common domain in ubiquitination. Curr. Biol. 2000;10:R132–R134. doi: 10.1016/S0960-9822(00)00398-5. PubMed DOI
Wang T., Wang W., Wang Q., Xie R., Landay A., Chen D. The E3 ubiquitin ligase CHIP in normal cell function and in disease conditions. Ann. N. Y. Acad. Sci. 2019 doi: 10.1111/nyas.14206. PubMed DOI PMC
Rotin D., Kumar S. Physiological functions of the HECT family of ubiquitin ligases. Nat. Rev. Mol. Cell Biol. 2009;10:398–409. doi: 10.1038/nrm2690. PubMed DOI
Podos S.D., Hanson K.K., Wang Y.C., Ferguson E.L. The DSmurf ubiquitin-protein ligase restricts BMP signaling spatially and temporally during Drosophila embryogenesis. Dev. Cell. 2001;1:567–578. doi: 10.1016/S1534-5807(01)00057-0. PubMed DOI
Staub O., Dho S., Henry P., Correa J., Ishikawa T., McGlade J., Rotin D. WW domains of Nedd4 bind to the proline-rich PY motifs in the epithelial Na+ channel deleted in Liddle’s syndrome. EMBO J. 1996;15:2371–2380. doi: 10.1002/j.1460-2075.1996.tb00593.x. PubMed DOI PMC
Ebisawa T., Fukuchi M., Murakami G., Chiba T., Tanaka K., Imamura T., Miyazono K. Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and induces receptor degradation. J. Biol. Chem. 2001;276:12477–12480. doi: 10.1074/jbc.C100008200. PubMed DOI
Spratt D.E., Walden H., Shaw G.S. RBR E3 ubiquitin ligases: New structures, new insights, new questions. Biochem. J. 2014;458:421–437. doi: 10.1042/BJ20140006. PubMed DOI PMC
Marin I., Ferrus A. Comparative genomics of the RBR family, including the Parkinson’s disease-related gene parkin and the genes of the ariadne subfamily. Mol. Biol. Evol. 2002;19:2039–2050. doi: 10.1093/oxfordjournals.molbev.a004029. PubMed DOI
Yoshii S.R., Kishi C., Ishihara N., Mizushima N. Parkin mediates proteasome-dependent protein degradation and rupture of the outer mitochondrial membrane. J. Biol. Chem. 2011;286:19630–19640. doi: 10.1074/jbc.M110.209338. PubMed DOI PMC
Lee J.Y., Nagano Y., Taylor J.P., Lim K.L., Yao T.P. Disease-causing mutations in parkin impair mitochondrial ubiquitination, aggregation, and HDAC6-dependent mitophagy. J. Cell Biol. 2010;189:671–679. doi: 10.1083/jcb.201001039. PubMed DOI PMC
Zhan T., Rindtorff N., Boutros M. Wnt signaling in cancer. Oncogene. 2017;36:1461–1473. doi: 10.1038/onc.2016.304. PubMed DOI PMC
Nusse R., Clevers H. Wnt/beta-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell. 2017;169:985–999. doi: 10.1016/j.cell.2017.05.016. PubMed DOI
Tolwinski N.S., Wehrli M., Rives A., Erdeniz N., DiNardo S., Wieschaus E. Wg/Wnt signal can be transmitted through arrow/LRP5,6 and Axin independently of Zw3/Gsk3beta activity. Dev. Cell. 2003;4:407–418. doi: 10.1016/S1534-5807(03)00063-7. PubMed DOI
Hao H.X., Xie Y., Zhang Y., Charlat O., Oster E., Avello M., Lei H., Mickanin C., Liu D., Ruffner H., et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature. 2012;485:195–200. doi: 10.1038/nature11019. PubMed DOI
Koo B.K., Spit M., Jordens I., Low T.Y., Stange D.E., van de Wetering M., van Es J.H., Mohammed S., Heck A.J., Maurice M.M., et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature. 2012;488:665–669. doi: 10.1038/nature11308. PubMed DOI
Jiang X., Charlat O., Zamponi R., Yang Y., Cong F. Dishevelled promotes Wnt receptor degradation through recruitment of ZNRF3/RNF43 E3 ubiquitin ligases. Mol. Cell. 2015;58:522–533. doi: 10.1016/j.molcel.2015.03.015. PubMed DOI
Peters J.M., McKay R.M., McKay J.P., Graff J.M. Casein kinase I transduces Wnt signals. Nature. 1999;401:345–350. doi: 10.1038/43830. PubMed DOI
Ikeda S., Kishida S., Yamamoto H., Murai H., Koyama S., Kikuchi A. Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3beta and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J. 1998;17:1371–1384. doi: 10.1093/emboj/17.5.1371. PubMed DOI PMC
Doble B.W., Patel S., Wood G.A., Kockeritz L.K., Woodgett J.R. Functional redundancy of GSK-3alpha and GSK-3beta in Wnt/beta-catenin signaling shown by using an allelic series of embryonic stem cell lines. Dev. Cell. 2007;12:957–971. doi: 10.1016/j.devcel.2007.04.001. PubMed DOI PMC
Morin P.J., Sparks A.B., Korinek V., Barker N., Clevers H., Vogelstein B., Kinzler K.W. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science. 1997;275:1787–1790. doi: 10.1126/science.275.5307.1787. PubMed DOI
Korinek V., Barker N., Morin P.J., van Wichen D., de Weger R., Kinzler K.W., Vogelstein B., Clevers H. Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC-/- colon carcinoma. Science. 1997;275:1784–1787. doi: 10.1126/science.275.5307.1784. PubMed DOI
Marikawa Y., Elinson R.P. beta-TrCP is a negative regulator of Wnt/beta-catenin signaling pathway and dorsal axis formation in Xenopus embryos. Mech. Dev. 1998;77:75–80. doi: 10.1016/S0925-4773(98)00134-8. PubMed DOI
Hart M., Concordet J.P., Lassot I., Albert I., del los Santos R., Durand H., Perret C., Rubinfeld B., Margottin F., Benarous R., et al. The F-box protein beta-TrCP associates with phosphorylated beta-catenin and regulates its activity in the cell. Curr. Biol. 1999;9:207–210. doi: 10.1016/S0960-9822(99)80091-8. PubMed DOI
Zeng X., Huang H., Tamai K., Zhang X., Harada Y., Yokota C., Almeida K., Wang J., Doble B., Woodgett J., et al. Initiation of Wnt signaling: Control of Wnt coreceptor Lrp6 phosphorylation/activation via frizzled, dishevelled and axin functions. Development. 2008;135:367–375. doi: 10.1242/dev.013540. PubMed DOI PMC
Pan W., Choi S.C., Wang H., Qin Y., Volpicelli-Daley L., Swan L., Lucast L., Khoo C., Zhang X., Li L., et al. Wnt3a-mediated formation of phosphatidylinositol 4,5-bisphosphate regulates LRP6 phosphorylation. Science. 2008;321:1350–1353. doi: 10.1126/science.1160741. PubMed DOI PMC
Li V.S., Ng S.S., Boersema P.J., Low T.Y., Karthaus W.R., Gerlach J.P., Mohammed S., Heck A.J., Maurice M.M., Mahmoudi T., et al. Wnt signaling through inhibition of beta-catenin degradation in an intact Axin1 complex. Cell. 2012;149:1245–1256. doi: 10.1016/j.cell.2012.05.002. PubMed DOI
Ji L., Jiang B., Jiang X., Charlat O., Chen A., Mickanin C., Bauer A., Xu W., Yan X., Cong F. The SIAH E3 ubiquitin ligases promote Wnt/beta-catenin signaling through mediating Wnt-induced Axin degradation. Genes Dev. 2017;31:904–915. doi: 10.1101/gad.300053.117. PubMed DOI PMC
Callow M.G., Tran H., Phu L., Lau T., Lee J., Sandoval W.N., Liu P.S., Bheddah S., Tao J., Lill J.R., et al. Ubiquitin ligase RNF146 regulates tankyrase and Axin to promote Wnt signaling. PLoS ONE. 2011;6:e22595. doi: 10.1371/journal.pone.0022595. PubMed DOI PMC
Zhang Y., Liu S., Mickanin C., Feng Y., Charlat O., Michaud G.A., Schirle M., Shi X., Hild M., Bauer A., et al. RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nat. Cell Biol. 2011;13:623–629. doi: 10.1038/ncb2222. PubMed DOI
DaRosa P.A., Klevit R.E., Xu W. Structural basis for tankyrase-RNF146 interaction reveals noncanonical tankyrase-binding motifs. Protein Sci. A Publ. Protein Soc. 2018;27:1057–1067. doi: 10.1002/pro.3413. PubMed DOI PMC
Matsumoto Y., La Rose J., Lim M., Adissu H.A., Law N., Mao X., Cong F., Mera P., Karsenty G., Goltzman D., et al. Ubiquitin ligase RNF146 coordinates bone dynamics and energy metabolism. J. Clin. Investig. 2017;127:2612–2625. doi: 10.1172/JCI92233. PubMed DOI PMC
Zhu X., Xing R., Tan R., Dai R., Tao Q. The RNF146 E3 ubiquitin ligase is required for the control of Wnt signaling and body pattern formation in Xenopus. Mech. Dev. 2017;147:28–36. doi: 10.1016/j.mod.2017.08.001. PubMed DOI
Wang Z., Tacchelly-Benites O., Noble G.P., Johnson M.K., Gagne J.P., Poirier G.G., Ahmed Y. A Context-Dependent Role for the RNF146 Ubiquitin Ligase in Wingless/Wnt Signaling in Drosophila. Genetics. 2019;211:913–923. doi: 10.1534/genetics.118.301393. PubMed DOI PMC
Fei C., He X., Xie S., Miao H., Zhou Z., Li L. Smurf1-mediated axin ubiquitination requires Smurf1 C2 domain and is cell cycle-dependent. J. Biol. Chem. 2014;289:14170–14177. doi: 10.1074/jbc.M113.536714. PubMed DOI PMC
Fei C., Li Z., Li C., Chen Y., Chen Z., He X., Mao L., Wang X., Zeng R., Li L. Smurf1-mediated Lys29-linked nonproteolytic polyubiquitination of axin negatively regulates Wnt/beta-catenin signaling. Mol. Cell. Biol. 2013;33:4095–4105. doi: 10.1128/MCB.00418-13. PubMed DOI PMC
Kim S., Jho E.H. The protein stability of Axin, a negative regulator of Wnt signaling, is regulated by Smad ubiquitination regulatory factor 2 (Smurf2) J. Biol. Chem. 2010;285:36420–36426. doi: 10.1074/jbc.M110.137471. PubMed DOI PMC
Lee H.K., Lee E.W., Seo J., Jeong M., Lee S.H., Kim S.Y., Jho E.H., Choi C.H., Chung J.Y., Song J. Ubiquitylation and degradation of adenomatous polyposis coli by MKRN1 enhances Wnt/beta-catenin signaling. Oncogene. 2018;37:4273–4286. doi: 10.1038/s41388-018-0267-3. PubMed DOI
Ding Y., Zhang Y., Xu C., Tao Q.H., Chen Y.G. HECT domain-containing E3 ubiquitin ligase NEDD4L negatively regulates Wnt signaling by targeting dishevelled for proteasomal degradation. J. Biol. Chem. 2013;288:8289–8298. doi: 10.1074/jbc.M112.433185. PubMed DOI PMC
Wei W., Li M., Wang J., Nie F., Li L. The E3 ubiquitin ligase ITCH negatively regulates canonical Wnt signaling by targeting dishevelled protein. Mol. Cell. Biol. 2012;32:3903–3912. doi: 10.1128/MCB.00251-12. PubMed DOI PMC
Nethe M., de Kreuk B.J., Tauriello D.V., Anthony E.C., Snoek B., Stumpel T., Salinas P.C., Maurice M.M., Geerts D., Deelder A.M., et al. Rac1 acts in conjunction with Nedd4 and dishevelled-1 to promote maturation of cell-cell contacts. J. Cell Sci. 2012;125:3430–3442. doi: 10.1242/jcs.100925. PubMed DOI PMC
Zhang Y., Ding Y., Chen Y.G., Tao Q. NEDD4L regulates convergent extension movements in Xenopus embryos via Disheveled-mediated non-canonical Wnt signaling. Dev. Biol. 2014;392:15–25. doi: 10.1016/j.ydbio.2014.05.003. PubMed DOI
Tsukiyama T., Fukui A., Terai S., Fujioka Y., Shinada K., Takahashi H., Yamaguchi T.P., Ohba Y., Hatakeyama S. Molecular Role of RNF43 in Canonical and Noncanonical Wnt Signaling. Mol. Cell. Biol. 2015;35:2007–2023. doi: 10.1128/MCB.00159-15. PubMed DOI PMC
Angers S., Thorpe C.J., Biechele T.L., Goldenberg S.J., Zheng N., MacCoss M.J., Moon R.T. The KLHL12-Cullin-3 ubiquitin ligase negatively regulates the Wnt-beta-catenin pathway by targeting Dishevelled for degradation. Nat. Cell Biol. 2006;8:348–357. doi: 10.1038/ncb1381. PubMed DOI
Lee H., Cheong S.M., Han W., Koo Y., Jo S.B., Cho G.S., Yang J.S., Kim S., Han J.K. Head formation requires Dishevelled degradation that is mediated by March2 in concert with Dapper1. Development. 2018;145 doi: 10.1242/dev.143107. PubMed DOI
Zhou M.I., Wang H., Foy R.L., Ross J.J., Cohen H.T. Tumor suppressor von Hippel-Lindau (VHL) stabilization of Jade-1 protein occurs through plant homeodomains and is VHL mutation dependent. Cancer Res. 2004;64:1278–1286. doi: 10.1158/0008-5472.CAN-03-0884. PubMed DOI
Shafique S., Rashid S. Structural basis for renal cancer by the dynamics of pVHL-dependent JADE1 stabilization and beta-catenin regulation. Prog. Biophys. Mol. Biol. 2019;145:65–77. doi: 10.1016/j.pbiomolbio.2018.12.005. PubMed DOI
Chitalia V.C., Foy R.L., Bachschmid M.M., Zeng L., Panchenko M.V., Zhou M.I., Bharti A., Seldin D.C., Lecker S.H., Dominguez I., et al. Jade-1 inhibits Wnt signalling by ubiquitylating beta-catenin and mediates Wnt pathway inhibition by pVHL. Nat. Cell Biol. 2008;10:1208–1216. doi: 10.1038/ncb1781. PubMed DOI PMC
Shivanna S., Harrold I., Shashar M., Meyer R., Kiang C., Francis J., Zhao Q., Feng H., Edelman E.R., Rahimi N., et al. The c-Cbl ubiquitin ligase regulates nuclear beta-catenin and angiogenesis by its tyrosine phosphorylation mediated through the Wnt signaling pathway. J. Biol. Chem. 2015;290:12537–12546. doi: 10.1074/jbc.M114.616623. PubMed DOI PMC
Chitalia V., Shivanna S., Martorell J., Meyer R., Edelman E., Rahimi N. c-Cbl, a ubiquitin E3 ligase that targets active beta-catenin: A novel layer of Wnt signaling regulation. J. Biol. Chem. 2013;288:23505–23517. doi: 10.1074/jbc.M113.473801. PubMed DOI PMC
Flack J.E., Mieszczanek J., Novcic N., Bienz M. Wnt-Dependent Inactivation of the Groucho/TLE Co-repressor by the HECT E3 Ubiquitin Ligase Hyd/UBR5. Mol. Cell. 2017;67:181–193. doi: 10.1016/j.molcel.2017.06.009. PubMed DOI PMC
Hay-Koren A., Caspi M., Zilberberg A., Rosin-Arbesfeld R. The EDD E3 ubiquitin ligase ubiquitinates and up-regulates beta-catenin. Mol. Biol. Cell. 2011;22:399–411. doi: 10.1091/mbc.e10-05-0440. PubMed DOI PMC
Liu L., Zhang Y., Wong C.C., Zhang J., Dong Y., Li X., Kang W., Chan F.K.L., Sung J.J.Y., Yu J. RNF6 Promotes Colorectal Cancer by Activating the Wnt/beta-Catenin Pathway via Ubiquitination of TLE3. Cancer Res. 2018;78:1958–1971. doi: 10.1158/0008-5472.CAN-17-2683. PubMed DOI
Glaeser K., Urban M., Fenech E., Voloshanenko O., Kranz D., Lari F., Christianson J.C., Boutros M. ERAD-dependent control of the Wnt secretory factor Evi. EMBO J. 2018;37 doi: 10.15252/embj.201797311. PubMed DOI PMC
Perrody E., Abrami L., Feldman M., Kunz B., Urbe S., van der Goot F.G. Ubiquitin-dependent folding of the Wnt signaling coreceptor LRP6. Elife. 2016;5 doi: 10.7554/eLife.19083. PubMed DOI PMC
Feldman M., van der Goot F.G. Novel ubiquitin-dependent quality control in the endoplasmic reticulum. Trends Cell Biol. 2009;19:357–363. doi: 10.1016/j.tcb.2009.05.005. PubMed DOI
Abrami L., Kunz B., Iacovache I., van der Goot F.G. Palmitoylation and ubiquitination regulate exit of the Wnt signaling protein LRP6 from the endoplasmic reticulum. Proc. Natl. Acad. Sci. USA. 2008;105:5384–5389. doi: 10.1073/pnas.0710389105. PubMed DOI PMC
Joshi V., Amanullah A., Upadhyay A., Mishra R., Kumar A., Mishra A. A Decade of Boon or Burden: What Has the CHIP Ever Done for Cellular Protein Quality Control Mechanism Implicated in Neurodegeneration and Aging? Front. Mol. Neurosci. 2016;9:93. doi: 10.3389/fnmol.2016.00093. PubMed DOI PMC
Bhuripanyo K., Wang Y., Liu X., Zhou L., Liu R., Duong D., Zhao B., Bi Y., Zhou H., Chen G., et al. Identifying the substrate proteins of U-box E3s E4B and CHIP by orthogonal ubiquitin transfer. Sci. Adv. 2018;4:e1701393. doi: 10.1126/sciadv.1701393. PubMed DOI PMC
Rudloff S., Kemler R. Differential requirements for beta-catenin during mouse development. Development. 2012;139:3711–3721. doi: 10.1242/dev.085597. PubMed DOI
Wang Y., Guo N., Nathans J. The role of Frizzled3 and Frizzled6 in neural tube closure and in the planar polarity of inner-ear sensory hair cells. J. Neurosci. 2006;26:2147–2156. doi: 10.1523/JNEUROSCI.4698-05.2005. PubMed DOI PMC
Korinek V., Barker N., Moerer P., van Donselaar E., Huls G., Peters P.J., Clevers H. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat. Genet. 1998;19:379–383. doi: 10.1038/1270. PubMed DOI
Nakayama K., Hatakeyama S., Maruyama S., Kikuchi A., Onoe K., Good R.A., Nakayama K.I. Impaired degradation of inhibitory subunit of NF-kappa B (I kappa B) and beta-catenin as a result of targeted disruption of the beta-TrCP1 gene. Proc. Natl. Acad. Sci. USA. 2003;100:8752–8757. doi: 10.1073/pnas.1133216100. PubMed DOI PMC
Narimatsu M., Bose R., Pye M., Zhang L., Miller B., Ching P., Sakuma R., Luga V., Roncari L., Attisano L., et al. Regulation of planar cell polarity by Smurf ubiquitin ligases. Cell. 2009;137:295–307. doi: 10.1016/j.cell.2009.02.025. PubMed DOI
Lee M.S., Han H.J., Han S.Y., Kim I.Y., Chae S., Lee C.S., Kim S.E., Yoon S.G., Park J.W., Kim J.H., et al. Loss of the E3 ubiquitin ligase MKRN1 represses diet-induced metabolic syndrome through AMPK activation. Nat. Commun. 2018;9:3404. doi: 10.1038/s41467-018-05721-4. PubMed DOI PMC
Lu C., Thoeni C., Connor A., Kawabe H., Gallinger S., Rotin D. Intestinal knockout of Nedd4 enhances growth of Apc(min) tumors. Oncogene. 2016;35:5839–5849. doi: 10.1038/onc.2016.125. PubMed DOI
Grima B., Lamouroux A., Chelot E., Papin C., Limbourg-Bouchon B., Rouyer F. The F-box protein slimb controls the levels of clock proteins period and timeless. Nature. 2002;420:178–182. doi: 10.1038/nature01122. PubMed DOI
Nakagawa T., Zhang T., Kushi R., Nakano S., Endo T., Nakagawa M., Yanagihara N., Zarkower D., Nakayama K. Regulation of mitosis-meiosis transition by the ubiquitin ligase beta-TrCP in male germ cells. Development. 2017;144:4137–4147. doi: 10.1242/dev.158485. PubMed DOI PMC
Guardavaccaro D., Kudo Y., Boulaire J., Barchi M., Busino L., Donzelli M., Margottin-Goguet F., Jackson P.K., Yamasaki L., Pagano M. Control of meiotic and mitotic progression by the F box protein beta-Trcp1 in vivo. Dev. Cell. 2003;4:799–812. doi: 10.1016/S1534-5807(03)00154-0. PubMed DOI
Ohsaki K., Oishi K., Kozono Y., Nakayama K., Nakayama K.I., Ishida N. The role of {beta}-TrCP1 and {beta}-TrCP2 in circadian rhythm generation by mediating degradation of clock protein PER2. J. Biochem. 2008;144:609–618. doi: 10.1093/jb/mvn112. PubMed DOI
Kanarek N., Horwitz E., Mayan I., Leshets M., Cojocaru G., Davis M., Tsuberi B.Z., Pikarsky E., Pagano M., Ben-Neriah Y. Spermatogenesis rescue in a mouse deficient for the ubiquitin ligase SCF{beta}-TrCP by single substrate depletion. Genes Dev. 2010;24:470–477. doi: 10.1101/gad.551610. PubMed DOI PMC
Kudo Y., Guardavaccaro D., Santamaria P.G., Koyama-Nasu R., Latres E., Bronson R., Yamasaki L., Pagano M. Role of F-box protein betaTrcp1 in mammary gland development and tumorigenesis. Mol. Cell. Biol. 2004;24:8184–8194. doi: 10.1128/MCB.24.18.8184-8194.2004. PubMed DOI PMC
Baguma-Nibasheka M., Kablar B. Abnormal retinal development in the Btrc null mouse. Dev. Dyn. 2009;238:2680–2687. doi: 10.1002/dvdy.22081. PubMed DOI PMC
Kanarek N., Grivennikov S.I., Leshets M., Lasry A., Alkalay I., Horwitz E., Shaul Y.D., Stachler M., Voronov E., Apte R.N., et al. Critical role for IL-1beta in DNA damage-induced mucositis. Proc. Natl. Acad. Sci. USA. 2014;111:E702–E711. doi: 10.1073/pnas.1322691111. PubMed DOI PMC
Nakagawa T., Araki T., Nakagawa M., Hirao A., Unno M., Nakayama K. S6 Kinase- and beta-TrCP2-Dependent Degradation of p19Arf Is Required for Cell Proliferation. Mol. Cell. Biol. 2015;35:3517–3527. doi: 10.1128/MCB.00343-15. PubMed DOI PMC
Bond C.E., McKeone D.M., Kalimutho M., Bettington M.L., Pearson S.A., Dumenil T.D., Wockner L.F., Burge M., Leggett B.A., Whitehall V.L. RNF43 and ZNRF3 are commonly altered in serrated pathway colorectal tumorigenesis. Oncotarget. 2016;7:70589–70600. doi: 10.18632/oncotarget.12130. PubMed DOI PMC
Giannakis M., Hodis E., Jasmine Mu X., Yamauchi M., Rosenbluh J., Cibulskis K., Saksena G., Lawrence M.S., Qian Z.R., Nishihara R., et al. RNF43 is frequently mutated in colorectal and endometrial cancers. Nat. Genet. 2014;46:1264–1266. doi: 10.1038/ng.3127. PubMed DOI PMC
Koo B.K., van Es J.H., van den Born M., Clevers H. Porcupine inhibitor suppresses paracrine Wnt-driven growth of Rnf43;Znrf3-mutant neoplasia. Proc. Natl. Acad. Sci. USA. 2015;112:7548–7550. doi: 10.1073/pnas.1508113112. PubMed DOI PMC
Basham K.J., Rodriguez S., Turcu A.F., Lerario A.M., Logan C.Y., Rysztak M.R., Gomez-Sanchez C.E., Breault D.T., Koo B.K., Clevers H., et al. A ZNRF3-dependent Wnt/beta-catenin signaling gradient is required for adrenal homeostasis. Genes Dev. 2019;33:209–220. doi: 10.1101/gad.317412.118. PubMed DOI PMC
Jameson S.A., Lin Y.T., Capel B. Testis development requires the repression of Wnt4 by Fgf signaling. Dev. Biol. 2012;370:24–32. doi: 10.1016/j.ydbio.2012.06.009. PubMed DOI PMC
Harris A., Siggers P., Corrochano S., Warr N., Sagar D., Grimes D.T., Suzuki M., Burdine R.D., Cong F., Koo B.K., et al. ZNRF3 functions in mammalian sex determination by inhibiting canonical WNT signaling. Proc. Natl. Acad. Sci. USA. 2018;115:5474–5479. doi: 10.1073/pnas.1801223115. PubMed DOI PMC
Shen J., Yu Z., Li N. The E3 ubiquitin ligase RNF146 promotes colorectal cancer by activating the Wnt/beta-catenin pathway via ubiquitination of Axin1. Biochem. Biophys. Res. Commun. 2018;503:991–997. doi: 10.1016/j.bbrc.2018.06.107. PubMed DOI
Wei W., Zeve D., Suh J.M., Wang X., Du Y., Zerwekh J.E., Dechow P.C., Graff J.M., Wan Y. Biphasic and dosage-dependent regulation of osteoclastogenesis by beta-catenin. Mol. Cell. Biol. 2011;31:4706–4719. doi: 10.1128/MCB.05980-11. PubMed DOI PMC
Matsumoto Y., Larose J., Kent O.A., Lim M., Changoor A., Zhang L., Storozhuk Y., Mao X., Grynpas M.D., Cong F., et al. RANKL coordinates multiple osteoclastogenic pathways by regulating expression of ubiquitin ligase RNF146. J. Clin. Investig. 2017;127:1303–1315. doi: 10.1172/JCI90527. PubMed DOI PMC
Ueki Y., Tiziani V., Santanna C., Fukai N., Maulik C., Garfinkle J., Ninomiya C., doAmaral C., Peters H., Habal M., et al. Mutations in the gene encoding c-Abl-binding protein SH3BP2 cause cherubism. Nat. Genet. 2001;28:125–126. doi: 10.1038/88832. PubMed DOI
Levaot N., Voytyuk O., Dimitriou I., Sircoulomb F., Chandrakumar A., Deckert M., Krzyzanowski P.M., Scotter A., Gu S., Janmohamed S., et al. Loss of Tankyrase-mediated destruction of 3BP2 is the underlying pathogenic mechanism of cherubism. Cell. 2011;147:1324–1339. doi: 10.1016/j.cell.2011.10.045. PubMed DOI PMC
Zou Y.F., Xie C.W., Yang S.X., Xiong J.P. AMPK activators suppress breast cancer cell growth by inhibiting DVL3-facilitated Wnt/beta-catenin signaling pathway activity. Mol. Med. Rep. 2017;15:899–907. doi: 10.3892/mmr.2016.6094. PubMed DOI
Lee H., Kang R., Bae S., Yoon Y. AICAR, an activator of AMPK, inhibits adipogenesis via the WNT/beta-catenin pathway in 3T3-L1 adipocytes. Int. J. Mol. Med. 2011;28:65–71. doi: 10.3892/ijmm.2011.674. PubMed DOI
Inoki K., Ouyang H., Zhu T., Lindvall C., Wang Y., Zhang X., Yang Q., Bennett C., Harada Y., Stankunas K., et al. TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell. 2006;126:955–968. doi: 10.1016/j.cell.2006.06.055. PubMed DOI
Park S.Y., Kim D., Kee S.H. Metformin-activated AMPK regulates beta-catenin to reduce cell proliferation in colon carcinoma RKO cells. Oncol. Lett. 2019;17:2695–2702. doi: 10.3892/ol.2019.9892. PubMed DOI PMC
Kondo S., Seo S.Y., Yoshizaki T., Wakisaka N., Furukawa M., Joab I., Jang K.L., Pagano J.S. EBV latent membrane protein 1 up-regulates hypoxia-inducible factor 1alpha through Siah1-mediated down-regulation of prolyl hydroxylases 1 and 3 in nasopharyngeal epithelial cells. Cancer Res. 2006;66:9870–9877. doi: 10.1158/0008-5472.CAN-06-1679. PubMed DOI
Dickins R.A., Frew I.J., House C.M., O’Bryan M.K., Holloway A.J., Haviv I., Traficante N., de Kretser D.M., Bowtell D.D. The ubiquitin ligase component Siah1a is required for completion of meiosis I in male mice. Mol. Cell. Biol. 2002;22:2294–2303. doi: 10.1128/MCB.22.7.2294-2303.2002. PubMed DOI PMC
Frew I.J., Sims N.A., Quinn J.M., Walkley C.R., Purton L.E., Bowtell D.D., Gillespie M.T. Osteopenia in Siah1a mutant mice. J. Biol. Chem. 2004;279:29583–29588. doi: 10.1074/jbc.M312755200. PubMed DOI
Frew I.J., Hammond V.E., Dickins R.A., Quinn J.M., Walkley C.R., Sims N.A., Schnall R., Della N.G., Holloway A.J., Digby M.R., et al. Generation and analysis of Siah2 mutant mice. Mol. Cell. Biol. 2003;23:9150–9161. doi: 10.1128/MCB.23.24.9150-9161.2003. PubMed DOI PMC
Scortegagna M., Kim H., Li J.L., Yao H., Brill L.M., Han J., Lau E., Bowtell D., Haddad G., Kaufman R.J., et al. Fine tuning of the UPR by the ubiquitin ligases Siah1/2. PLoS Genet. 2014;10:e1004348. doi: 10.1371/journal.pgen.1004348. PubMed DOI PMC
Wang D., Wang Y., Kong T., Fan F., Jiang Y. Hypoxia-induced beta-catenin downregulation involves p53-dependent activation of Siah-1. Cancer Sci. 2011;102:1322–1328. doi: 10.1111/j.1349-7006.2011.01950.x. PubMed DOI PMC
Simon M.C. Siah proteins, HIF prolyl hydroxylases, and the physiological response to hypoxia. Cell. 2004;117:851–853. doi: 10.1016/j.cell.2004.06.010. PubMed DOI
Gudjonsson T., Altmeyer M., Savic V., Toledo L., Dinant C., Grofte M., Bartkova J., Poulsen M., Oka Y., Bekker-Jensen S., et al. TRIP12 and UBR5 suppress spreading of chromatin ubiquitylation at damaged chromosomes. Cell. 2012;150:697–709. doi: 10.1016/j.cell.2012.06.039. PubMed DOI
Shearer R.F., Iconomou M., Watts C.K., Saunders D.N. Functional Roles of the E3 Ubiquitin Ligase UBR5 in Cancer. Mol. Cancer Res. 2015;13:1523–1532. doi: 10.1158/1541-7786.MCR-15-0383. PubMed DOI
Saunders D.N., Hird S.L., Withington S.L., Dunwoodie S.L., Henderson M.J., Biben C., Sutherland R.L., Ormandy C.J., Watts C.K. Edd, the murine hyperplastic disc gene, is essential for yolk sac vascularization and chorioallantoic fusion. Mol. Cell. Biol. 2004;24:7225–7234. doi: 10.1128/MCB.24.16.7225-7234.2004. PubMed DOI PMC
Ishikawa T., Tamai Y., Zorn A.M., Yoshida H., Seldin M.F., Nishikawa S., Taketo M.M. Mouse Wnt receptor gene Fzd5 is essential for yolk sac and placental angiogenesis. Development. 2001;128:25–33. PubMed
Kinsella E., Dora N., Mellis D., Lettice L., Deveney P., Hill R., Ditzel M. Use of a Conditional Ubr5 Mutant Allele to Investigate the Role of an N-End Rule Ubiquitin-Protein Ligase in Hedgehog Signalling and Embryonic Limb Development. PLoS ONE. 2016;11:e0157079. doi: 10.1371/journal.pone.0157079. PubMed DOI PMC
Ye X., Wang L., Shang B., Wang Z., Wei W. NEDD4: A promising target for cancer therapy. Curr. Cancer Drug Targets. 2014;14:549–556. doi: 10.2174/1568009614666140725092430. PubMed DOI PMC
Li J.J., Ferry R.J., Jr., Diao S., Xue B., Bahouth S.W., Liao F.F. Nedd4 haploinsufficient mice display moderate insulin resistance, enhanced lipolysis, and protection against high-fat diet-induced obesity. Endocrinology. 2015;156:1283–1291. doi: 10.1210/en.2014-1909. PubMed DOI PMC
Liu Y., Oppenheim R.W., Sugiura Y., Lin W. Abnormal development of the neuromuscular junction in Nedd4-deficient mice. Dev. Biol. 2009;330:153–166. doi: 10.1016/j.ydbio.2009.03.023. PubMed DOI PMC
Kawabe H., Neeb A., Dimova K., Young S.M., Jr., Takeda M., Katsurabayashi S., Mitkovski M., Malakhova O.A., Zhang D.E., Umikawa M., et al. Regulation of Rap2A by the ubiquitin ligase Nedd4-1 controls neurite development. Neuron. 2010;65:358–372. doi: 10.1016/j.neuron.2010.01.007. PubMed DOI PMC
Cao X.R., Lill N.L., Boase N., Shi P.P., Croucher D.R., Shan H., Qu J., Sweezer E.M., Place T., Kirby P.A., et al. Nedd4 controls animal growth by regulating IGF-1 signaling. Sci. Signal. 2008;1 doi: 10.1126/scisignal.1160940. PubMed DOI PMC
Fouladkou F., Lu C., Jiang C., Zhou L., She Y., Walls J.R., Kawabe H., Brose N., Henkelman R.M., Huang A., et al. The ubiquitin ligase Nedd4-1 is required for heart development and is a suppressor of thrombospondin-1. J. Biol. Chem. 2010;285:6770–6780. doi: 10.1074/jbc.M109.082347. PubMed DOI PMC
Lee J.H., Jeon S.A., Kim B.G., Takeda M., Cho J.J., Kim D.I., Kawabe H., Cho J.Y. Nedd4 Deficiency in Vascular Smooth Muscle Promotes Vascular Calcification by Stabilizing pSmad1. J. Bone Min. Res. 2017;32:927–938. doi: 10.1002/jbmr.3073. PubMed DOI
Jeon S.A., Lee J.H., Kim D.W., Cho J.Y. E3-ubiquitin ligase NEDD4 enhances bone formation by removing TGFbeta1-induced pSMAD1 in immature osteoblast. Bone. 2018;116:248–258. doi: 10.1016/j.bone.2018.08.012. PubMed DOI
Wiszniak S., Harvey N., Schwarz Q. Cell autonomous roles of Nedd4 in craniofacial bone formation. Dev. Biol. 2016;410:98–107. doi: 10.1016/j.ydbio.2015.12.001. PubMed DOI
Wiszniak S., Kabbara S., Lumb R., Scherer M., Secker G., Harvey N., Kumar S., Schwarz Q. The ubiquitin ligase Nedd4 regulates craniofacial development by promoting cranial neural crest cell survival and stem-cell like properties. Dev. Biol. 2013;383:186–200. doi: 10.1016/j.ydbio.2013.09.024. PubMed DOI
Yang B., Gay D.L., MacLeod M.K., Cao X., Hala T., Sweezer E.M., Kappler J., Marrack P., Oliver P.M. Nedd4 augments the adaptive immune response by promoting ubiquitin-mediated degradation of Cbl-b in activated T cells. Nat. Immunol. 2008;9:1356–1363. doi: 10.1038/ni.1670. PubMed DOI PMC
Guo H., Qiao G., Ying H., Li Z., Zhao Y., Liang Y., Yang L., Lipkowitz S., Penninger J.M., Langdon W.Y., et al. E3 ubiquitin ligase Cbl-b regulates Pten via Nedd4 in T cells independently of its ubiquitin ligase activity. Cell Rep. 2012;1:472–482. doi: 10.1016/j.celrep.2012.04.008. PubMed DOI PMC
Russo C.J., Melista E., Cui J., DeStefano A.L., Bakris G.L., Manolis A.J., Gavras H., Baldwin C.T. Association of NEDD4L ubiquitin ligase with essential hypertension. Hypertension. 2005;46:488–491. doi: 10.1161/01.HYP.0000178594.63193.c0. PubMed DOI
Yanpallewar S., Wang T., Koh D.C., Quarta E., Fulgenzi G., Tessarollo L. Nedd4-2 haploinsufficiency causes hyperactivity and increased sensitivity to inflammatory stimuli. Sci. Rep. 2016;6:32957. doi: 10.1038/srep32957. PubMed DOI PMC
Harvey K.F., Dinudom A., Cook D.I., Kumar S. The Nedd4-like protein KIAA0439 is a potential regulator of the epithelial sodium channel. J. Biol. Chem. 2001;276:8597–8601. doi: 10.1074/jbc.C000906200. PubMed DOI
Zhou R., Patel S.V., Snyder P.M. Nedd4-2 catalyzes ubiquitination and degradation of cell surface ENaC. J. Biol. Chem. 2007;282:20207–20212. doi: 10.1074/jbc.M611329200. PubMed DOI
Hanukoglu I., Hanukoglu A. Epithelial sodium channel (ENaC) family: Phylogeny, structure-function, tissue distribution, and associated inherited diseases. Gene. 2016;579:95–132. doi: 10.1016/j.gene.2015.12.061. PubMed DOI PMC
Sun C., Hummler E., Hill D.L. Selective Deletion of Sodium Salt Taste during Development Leads to Expanded Terminal Fields of Gustatory Nerves in the Adult Mouse Nucleus of the Solitary Tract. J. Neurosci. Off. J. Soc. Neurosci. 2017;37:660–672. doi: 10.1523/JNEUROSCI.2913-16.2016. PubMed DOI PMC
Shi P.P., Cao X.R., Sweezer E.M., Kinney T.S., Williams N.R., Husted R.F., Nair R., Weiss R.M., Williamson R.A., Sigmund C.D., et al. Salt-sensitive hypertension and cardiac hypertrophy in mice deficient in the ubiquitin ligase Nedd4-2. Am. J. Physiol. Ren. Physiol. 2008;295:F462–F470. doi: 10.1152/ajprenal.90300.2008. PubMed DOI PMC
Boase N.A., Rychkov G.Y., Townley S.L., Dinudom A., Candi E., Voss A.K., Tsoutsman T., Semsarian C., Melino G., Koentgen F., et al. Respiratory distress and perinatal lethality in Nedd4-2-deficient mice. Nat. Commun. 2011;2:287. doi: 10.1038/ncomms1284. PubMed DOI PMC
Henshall T.L., Manning J.A., Alfassy O.S., Goel P., Boase N.A., Kawabe H., Kumar S. Deletion of Nedd4-2 results in progressive kidney disease in mice. Cell Death Differ. 2017;24:2150–2160. doi: 10.1038/cdd.2017.137. PubMed DOI PMC
Yip K.H., Kolesnikoff N., Hauschild N., Biggs L., Lopez A.F., Galli S.J., Kumar S., Grimbaldeston M.A. The Nedd4-2/Ndfip1 axis is a negative regulator of IgE-mediated mast cell activation. Nat. Commun. 2016;7:13198. doi: 10.1038/ncomms13198. PubMed DOI PMC
Infante P., Lospinoso Severini L., Bernardi F., Bufalieri F., Di Marcotullio L. Targeting Hedgehog Signalling through the Ubiquitylation Process: The Multiple Roles of the HECT-E3 Ligase Itch. Cells. 2019;8 doi: 10.3390/cells8020098. PubMed DOI PMC
Liu Y.C. The E3 ubiquitin ligase Itch in T cell activation, differentiation, and tolerance. Semin. Immunol. 2007;19:197–205. doi: 10.1016/j.smim.2007.02.003. PubMed DOI PMC
Gao M., Labuda T., Xia Y., Gallagher E., Fang D., Liu Y.C., Karin M. Jun turnover is controlled through JNK-dependent phosphorylation of the E3 ligase Itch. Science. 2004;306:271–275. doi: 10.1126/science.1099414. PubMed DOI
Lohr N.J., Molleston J.P., Strauss K.A., Torres-Martinez W., Sherman E.A., Squires R.H., Rider N.L., Chikwava K.R., Cummings O.W., Morton D.H., et al. Human ITCH E3 ubiquitin ligase deficiency causes syndromic multisystem autoimmune disease. Am. J. Hum. Genet. 2010;86:447–453. doi: 10.1016/j.ajhg.2010.01.028. PubMed DOI PMC
Perry W.L., Hustad C.M., Swing D.A., O’Sullivan T.N., Jenkins N.A., Copeland N.G. The itchy locus encodes a novel ubiquitin protein ligase that is disrupted in a18H mice. Nat. Genet. 1998;18:143–146. doi: 10.1038/ng0298-143. PubMed DOI
Hustad C.M., Perry W.L., Siracusa L.D., Rasberry C., Cobb L., Cattanach B.M., Kovatch R., Copeland N.G., Jenkins N.A. Molecular genetic characterization of six recessive viable alleles of the mouse agouti locus. Genetics. 1995;140:255–265. PubMed PMC
Fang D., Elly C., Gao B., Fang N., Altman Y., Joazeiro C., Hunter T., Copeland N., Jenkins N., Liu Y.C. Dysregulation of T lymphocyte function in itchy mice: A role for Itch in TH2 differentiation. Nat. Immunol. 2002;3:281–287. doi: 10.1038/ni763. PubMed DOI
Jin H.S., Park Y., Elly C., Liu Y.C. Itch expression by Treg cells controls Th2 inflammatory responses. J. Clin. Investig. 2013;123:4923–4934. doi: 10.1172/JCI69355. PubMed DOI PMC
Xiao N., Eto D., Elly C., Peng G., Crotty S., Liu Y.C. The E3 ubiquitin ligase Itch is required for the differentiation of follicular helper T cells. Nat. Immunol. 2014;15:657–666. doi: 10.1038/ni.2912. PubMed DOI PMC
Huang H., Jeon M.S., Liao L., Yang C., Elly C., Yates J.R., 3rd, Liu Y.C. K33-linked polyubiquitination of T cell receptor-zeta regulates proteolysis-independent T cell signaling. Immunity. 2010;33:60–70. doi: 10.1016/j.immuni.2010.07.002. PubMed DOI PMC
Giamboi-Miraglia A., Cianfarani F., Cattani C., Lena A.M., Serra V., Campione E., Terrinoni A., Zambruno G., Odorisio T., Di Daniele N., et al. The E3 ligase Itch knockout mice show hyperproliferation and wound healing alteration. FEBS J. 2015;282:4435–4449. doi: 10.1111/febs.13514. PubMed DOI
Stermer A.R., Myers J.L., Murphy C.J., Di Bona K.R., Matesic L., Richburg J.H. Female mice with loss-of-function ITCH display an altered reproductive phenotype. Exp. Biol. Med. (Maywood) 2016;241:367–374. doi: 10.1177/1535370215610656. PubMed DOI PMC
Marino A., Menghini R., Fabrizi M., Casagrande V., Mavilio M., Stoehr R., Candi E., Mauriello A., Moreno-Navarrete J.M., Gomez-Serrano M., et al. ITCH deficiency protects from diet-induced obesity. Diabetes. 2014;63:550–561. doi: 10.2337/db13-0802. PubMed DOI
Chang L., Kamata H., Solinas G., Luo J.L., Maeda S., Venuprasad K., Liu Y.C., Karin M. The E3 ubiquitin ligase itch couples JNK activation to TNFalpha-induced cell death by inducing c-FLIP(L) turnover. Cell. 2006;124:601–613. doi: 10.1016/j.cell.2006.01.021. PubMed DOI
Rathinam C., Matesic L.E., Flavell R.A. The E3 ligase Itch is a negative regulator of the homeostasis and function of hematopoietic stem cells. Nat. Immunol. 2011;12:399–407. doi: 10.1038/ni.2021. PubMed DOI PMC
Thien C.B., Langdon W.Y. c-Cbl and Cbl-b ubiquitin ligases: Substrate diversity and the negative regulation of signalling responses. Biochem. J. 2005;391:153–166. doi: 10.1042/BJ20050892. PubMed DOI PMC
Mohapatra B., Ahmad G., Nadeau S., Zutshi N., An W., Scheffe S., Dong L., Feng D., Goetz B., Arya P., et al. Protein tyrosine kinase regulation by ubiquitination: Critical roles of Cbl-family ubiquitin ligases. Biochim. Biophys. Acta. 2013;1833:122–139. doi: 10.1016/j.bbamcr.2012.10.010. PubMed DOI PMC
Tanaka S., Neff L., Baron R., Levy J.B. Tyrosine phosphorylation and translocation of the c-cbl protein after activation of tyrosine kinase signaling pathways. J. Biol. Chem. 1995;270:14347–14351. doi: 10.1074/jbc.270.24.14347. PubMed DOI
Naramura M., Kole H.K., Hu R.J., Gu H. Altered thymic positive selection and intracellular signals in Cbl-deficient mice. Proc. Natl. Acad. Sci. USA. 1998;95:15547–15552. doi: 10.1073/pnas.95.26.15547. PubMed DOI PMC
Murphy M.A., Schnall R.G., Venter D.J., Barnett L., Bertoncello I., Thien C.B., Langdon W.Y., Bowtell D.D. Tissue hyperplasia and enhanced T-cell signalling via ZAP-70 in c-Cbl-deficient mice. Mol. Cell. Biol. 1998;18:4872–4882. doi: 10.1128/MCB.18.8.4872. PubMed DOI PMC
Naramura M., Jang I.K., Kole H., Huang F., Haines D., Gu H. c-Cbl and Cbl-b regulate T cell responsiveness by promoting ligand-induced TCR down-modulation. Nat. Immunol. 2002;3:1192–1199. doi: 10.1038/ni855. PubMed DOI
Bachmaier K., Krawczyk C., Kozieradzki I., Kong Y.Y., Sasaki T., Oliveira-dos-Santos A., Mariathasan S., Bouchard D., Wakeham A., Itie A., et al. Negative regulation of lymphocyte activation and autoimmunity by the molecular adaptor Cbl-b. Nature. 2000;403:211–216. doi: 10.1038/35003228. PubMed DOI
Wang H.Y., Altman Y., Fang D., Elly C., Dai Y., Shao Y., Liu Y.C. Cbl promotes ubiquitination of the T cell receptor zeta through an adaptor function of Zap-70. J. Biol. Chem. 2001;276:26004–26011. doi: 10.1074/jbc.M010738200. PubMed DOI
D’Oro U., Munitic I., Chacko G., Karpova T., McNally J., Ashwell J.D. Regulation of constitutive TCR internalization by the zeta-chain. J. Immunol. 2002;169:6269–6278. doi: 10.4049/jimmunol.169.11.6269. PubMed DOI
Lutz-Nicoladoni C., Wolf D., Sopper S. Modulation of Immune Cell Functions by the E3 Ligase Cbl-b. Front. Oncol. 2015;5:58. doi: 10.3389/fonc.2015.00058. PubMed DOI PMC
Adams C.O., Housley W.J., Bhowmick S., Cone R.E., Rajan T.V., Forouhar F., Clark R.B. Cbl-b(-/-) T cells demonstrate in vivo resistance to regulatory T cells but a context-dependent resistance to TGF-beta. J. Immunol. 2010;185:2051–2058. doi: 10.4049/jimmunol.1001171. PubMed DOI
Chiusaroli R., Sanjay A., Henriksen K., Engsig M.T., Horne W.C., Gu H., Baron R. Deletion of the gene encoding c-Cbl alters the ability of osteoclasts to migrate, delaying resorption and ossification of cartilage during the development of long bones. Dev. Biol. 2003;261:537–547. doi: 10.1016/S0012-1606(03)00299-9. PubMed DOI
Molero J.C., Jensen T.E., Withers P.C., Couzens M., Herzog H., Thien C.B., Langdon W.Y., Walder K., Murphy M.A., Bowtell D.D., et al. c-Cbl-deficient mice have reduced adiposity, higher energy expenditure, and improved peripheral insulin action. J. Clin. Investig. 2004;114:1326–1333. doi: 10.1172/JCI21480. PubMed DOI PMC
Li X., Gadzinsky A., Gong L., Tong H., Calderon V., Li Y., Kitamura D., Klein U., Langdon W.Y., Hou F., et al. Cbl Ubiquitin Ligases Control B Cell Exit from the Germinal-Center Reaction. Immunity. 2018;48:530–541.e536. doi: 10.1016/j.immuni.2018.03.006. PubMed DOI
Gustin S.E., Thien C.B., Langdon W.Y. Cbl-b is a negative regulator of inflammatory cytokines produced by IgE-activated mast cells. J. Immunol. 2006;177:5980–5989. doi: 10.4049/jimmunol.177.9.5980. PubMed DOI
Naramura M., Nandwani N., Gu H., Band V., Band H. Rapidly fatal myeloproliferative disorders in mice with deletion of Casitas B-cell lymphoma (Cbl) and Cbl-b in hematopoietic stem cells. Proc. Natl. Acad. Sci. USA. 2010;107:16274–16279. doi: 10.1073/pnas.1007575107. PubMed DOI PMC
Mohapatra B., Zutshi N., An W., Goetz B., Arya P., Bielecki T.A., Mushtaq I., Storck M.D., Meza J.L., Band V., et al. An essential role of CBL and CBL-B ubiquitin ligases in mammary stem cell maintenance. Development. 2017;144:1072–1086. doi: 10.1242/dev.138164. PubMed DOI PMC
Meng X.M., Nikolic-Paterson D.J., Lan H.Y. TGF-beta: The master regulator of fibrosis. Nat. Rev. Nephrol. 2016;12:325–338. doi: 10.1038/nrneph.2016.48. PubMed DOI
Taipale J., Miyazono K., Heldin C.H., Keski-Oja J. Latent transforming growth factor-beta 1 associates to fibroblast extracellular matrix via latent TGF-beta binding protein. J. Cell Biol. 1994;124:171–181. doi: 10.1083/jcb.124.1.171. PubMed DOI PMC
Massague J. TGFbeta signalling in context. Nat. Rev. Mol. Cell Biol. 2012;13:616–630. doi: 10.1038/nrm3434. PubMed DOI PMC
Itoh F., Divecha N., Brocks L., Oomen L., Janssen H., Calafat J., Itoh S., Dijke Pt P. The FYVE domain in Smad anchor for receptor activation (SARA) is sufficient for localization of SARA in early endosomes and regulates TGF-beta/Smad signalling. Genes Cells Devoted Mol. Cell. Mech. 2002;7:321–331. doi: 10.1046/j.1365-2443.2002.00519.x. PubMed DOI
Miura S., Takeshita T., Asao H., Kimura Y., Murata K., Sasaki Y., Hanai J.I., Beppu H., Tsukazaki T., Wrana J.L., et al. Hgs (Hrs), a FYVE domain protein, is involved in Smad signaling through cooperation with SARA. Mol. Cell. Biol. 2000;20:9346–9355. doi: 10.1128/MCB.20.24.9346-9355.2000. PubMed DOI PMC
Massague J., Seoane J., Wotton D. Smad transcription factors. Genes Dev. 2005;19:2783–2810. doi: 10.1101/gad.1350705. PubMed DOI
Janknecht R., Wells N.J., Hunter T. TGF-beta-stimulated cooperation of smad proteins with the coactivators CBP/p300. Genes Dev. 1998;12:2114–2119. doi: 10.1101/gad.12.14.2114. PubMed DOI PMC
Pei D., Shu X., Gassama-Diagne A., Thiery J.P. Mesenchymal-epithelial transition in development and reprogramming. Nat. Cell Biol. 2019;21:44–53. doi: 10.1038/s41556-018-0195-z. PubMed DOI
Moustakas A., Kardassis D. Regulation of the human p21/WAF1/Cip1 promoter in hepatic cells by functional interactions between Sp1 and Smad family members. Proc. Natl. Acad. Sci. USA. 1998;95:6733–6738. doi: 10.1073/pnas.95.12.6733. PubMed DOI PMC
Nakao A., Afrakhte M., Moren A., Nakayama T., Christian J.L., Heuchel R., Itoh S., Kawabata M., Heldin N.E., Heldin C.H., et al. Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling. Nature. 1997;389:631–635. doi: 10.1038/39369. PubMed DOI
Chong P.A., Lin H., Wrana J.L., Forman-Kay J.D. An expanded WW domain recognition motif revealed by the interaction between Smad7 and the E3 ubiquitin ligase Smurf2. J. Biol. Chem. 2006;281:17069–17075. doi: 10.1074/jbc.M601493200. PubMed DOI
Kavsak P., Rasmussen R.K., Causing C.G., Bonni S., Zhu H., Thomsen G.H., Wrana J.L. Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF beta receptor for degradation. Mol. Cell. 2000;6:1365–1375. doi: 10.1016/S1097-2765(00)00134-9. PubMed DOI
Zhou F., Li F., Xie F., Zhang Z., Huang H., Zhang L. TRAF4 mediates activation of TGF-beta signaling and is a biomarker for oncogenesis in breast cancer. Sci. China Life Sci. 2014;57:1172–1176. doi: 10.1007/s11427-014-4727-x. PubMed DOI
Wang X., Jin C., Tang Y., Tang L.Y., Zhang Y.E. Ubiquitination of tumor necrosis factor receptor-associated factor 4 (TRAF4) by Smad ubiquitination regulatory factor 1 (Smurf1) regulates motility of breast epithelial and cancer cells. J. Biol. Chem. 2013;288:21784–21792. doi: 10.1074/jbc.M113.472704. PubMed DOI PMC
Zhang L., Zhou F., Garcia de Vinuesa A., de Kruijf E.M., Mesker W.E., Hui L., Drabsch Y., Li Y., Bauer A., Rousseau A., et al. TRAF4 promotes TGF-beta receptor signaling and drives breast cancer metastasis. Mol. Cell. 2013;51:559–572. doi: 10.1016/j.molcel.2013.07.014. PubMed DOI
Zhang Y., Chang C., Gehling D.J., Hemmati-Brivanlou A., Derynck R. Regulation of Smad degradation and activity by Smurf2, an E3 ubiquitin ligase. Proc. Natl. Acad. Sci. USA. 2001;98:974–979. doi: 10.1073/pnas.98.3.974. PubMed DOI PMC
Tang L.Y., Yamashita M., Coussens N.P., Tang Y., Wang X., Li C., Deng C.X., Cheng S.Y., Zhang Y.E. Ablation of Smurf2 reveals an inhibition in TGF-beta signalling through multiple mono-ubiquitination of Smad3. EMBO J. 2011;30:4777–4789. doi: 10.1038/emboj.2011.393. PubMed DOI PMC
Bonni S., Wang H.R., Causing C.G., Kavsak P., Stroschein S.L., Luo K., Wrana J.L. TGF-beta induces assembly of a Smad2-Smurf2 ubiquitin ligase complex that targets SnoN for degradation. Nat. Cell Biol. 2001;3:587–595. doi: 10.1038/35078562. PubMed DOI
Alarcon C., Zaromytidou A.I., Xi Q., Gao S., Yu J., Fujisawa S., Barlas A., Miller A.N., Manova-Todorova K., Macias M.J., et al. Nuclear CDKs drive Smad transcriptional activation and turnover in BMP and TGF-beta pathways. Cell. 2009;139:757–769. doi: 10.1016/j.cell.2009.09.035. PubMed DOI PMC
Aragon E., Goerner N., Zaromytidou A.I., Xi Q., Escobedo A., Massague J., Macias M.J. A Smad action turnover switch operated by WW domain readers of a phosphoserine code. Genes Dev. 2011;25:1275–1288. doi: 10.1101/gad.2060811. PubMed DOI PMC
Bai Y., Yang C., Hu K., Elly C., Liu Y.C. Itch E3 ligase-mediated regulation of TGF-beta signaling by modulating smad2 phosphorylation. Mol. Cell. 2004;15:825–831. doi: 10.1016/j.molcel.2004.07.021. PubMed DOI
Park S.H., Jung E.H., Kim G.Y., Kim B.C., Lim J.H., Woo C.H. Itch E3 ubiquitin ligase positively regulates TGF-beta signaling to EMT via Smad7 ubiquitination. Mol. Cells. 2015;38:20–25. doi: 10.14348/molcells.2015.2120. PubMed DOI PMC
Pefani D.E., Pankova D., Abraham A.G., Grawenda A.M., Vlahov N., Scrace S., O’Neill E. TGF-beta Targets the Hippo Pathway Scaffold RASSF1A to Facilitate YAP/SMAD2 Nuclear Translocation. Mol. Cell. 2016;63:156–166. doi: 10.1016/j.molcel.2016.05.012. PubMed DOI
Koinuma D., Shinozaki M., Komuro A., Goto K., Saitoh M., Hanyu A., Ebina M., Nukiwa T., Miyazawa K., Imamura T., et al. Arkadia amplifies TGF-beta superfamily signalling through degradation of Smad7. EMBO J. 2003;22:6458–6470. doi: 10.1093/emboj/cdg632. PubMed DOI PMC
Liu F.Y., Li X.Z., Peng Y.M., Liu H., Liu Y.H. Arkadia-Smad7-mediated positive regulation of TGF-beta signaling in a rat model of tubulointerstitial fibrosis. Am. J. Nephrol. 2007;27:176–183. doi: 10.1159/000100518. PubMed DOI
Liu W., Rui H., Wang J., Lin S., He Y., Chen M., Li Q., Ye Z., Zhang S., Chan S.C., et al. Axin is a scaffold protein in TGF-beta signaling that promotes degradation of Smad7 by Arkadia. EMBO J. 2006;25:1646–1658. doi: 10.1038/sj.emboj.7601057. PubMed DOI PMC
Zhang L., Huang H., Zhou F., Schimmel J., Pardo C.G., Zhang T., Barakat T.S., Sheppard K.A., Mickanin C., Porter J.A., et al. RNF12 controls embryonic stem cell fate and morphogenesis in zebrafish embryos by targeting Smad7 for degradation. Mol. Cell. 2012;46:650–661. doi: 10.1016/j.molcel.2012.04.003. PubMed DOI
Nagano Y., Mavrakis K.J., Lee K.L., Fujii T., Koinuma D., Sase H., Yuki K., Isogaya K., Saitoh M., Imamura T., et al. Arkadia induces degradation of SnoN and c-Ski to enhance transforming growth factor-beta signaling. J. Biol. Chem. 2007;282:20492–20501. doi: 10.1074/jbc.M701294200. PubMed DOI
Inoue Y., Imamura T. Regulation of TGF-beta family signaling by E3 ubiquitin ligases. Cancer Sci. 2008;99:2107–2112. doi: 10.1111/j.1349-7006.2008.00925.x. PubMed DOI PMC
Le Scolan E., Zhu Q., Wang L., Bandyopadhyay A., Javelaud D., Mauviel A., Sun L., Luo K. Transforming growth factor-beta suppresses the ability of Ski to inhibit tumor metastasis by inducing its degradation. Cancer Res. 2008;68:3277–3285. doi: 10.1158/0008-5472.CAN-07-6793. PubMed DOI
Huang Y., Yang Y., Gao R., Yang X., Yan X., Wang C., Jiang S., Yu L. RLIM interacts with Smurf2 and promotes TGF-beta induced U2OS cell migration. Biochem. Biophys. Res. Commun. 2011;414:181–185. doi: 10.1016/j.bbrc.2011.09.053. PubMed DOI
Gruber T., Hinterleitner R., Hermann-Kleiter N., Meisel M., Kleiter I., Wang C.M., Viola A., Pfeifhofer-Obermair C., Baier G. Cbl-b mediates TGFbeta sensitivity by downregulating inhibitory SMAD7 in primary T cells. J. Mol. Cell Biol. 2013;5:358–368. doi: 10.1093/jmcb/mjt017. PubMed DOI
Xie F., Jin K., Shao L., Fan Y., Tu Y., Li Y., Yang B., van Dam H., Ten Dijke P., Weng H., et al. FAF1 phosphorylation by AKT accumulates TGF-beta type II receptor and drives breast cancer metastasis. Nat. Commun. 2017;8:15021. doi: 10.1038/ncomms15021. PubMed DOI PMC
He W., Dorn D.C., Erdjument-Bromage H., Tempst P., Moore M.A., Massague J. Hematopoiesis controlled by distinct TIF1gamma and Smad4 branches of the TGFbeta pathway. Cell. 2006;125:929–941. doi: 10.1016/j.cell.2006.03.045. PubMed DOI
Morsut L., Yan K.P., Enzo E., Aragona M., Soligo S.M., Wendling O., Mark M., Khetchoumian K., Bressan G., Chambon P., et al. Negative control of Smad activity by ectodermin/Tif1gamma patterns the mammalian embryo. Development. 2010;137:2571–2578. doi: 10.1242/dev.053801. PubMed DOI
Xi Q., Wang Z., Zaromytidou A.I., Zhang X.H., Chow-Tsang L.F., Liu J.X., Kim H., Barlas A., Manova-Todorova K., Kaartinen V., et al. A poised chromatin platform for TGF-beta access to master regulators. Cell. 2011;147:1511–1524. doi: 10.1016/j.cell.2011.11.032. PubMed DOI PMC
Agricola E., Randall R.A., Gaarenstroom T., Dupont S., Hill C.S. Recruitment of TIF1gamma to chromatin via its PHD finger-bromodomain activates its ubiquitin ligase and transcriptional repressor activities. Mol. Cell. 2011;43:85–96. doi: 10.1016/j.molcel.2011.05.020. PubMed DOI
Tracy Cai X., Li H., Safyan A., Gawlik J., Pyrowolakis G., Jasper H. AWD regulates timed activation of BMP signaling in intestinal stem cells to maintain tissue homeostasis. Nat. Commun. 2019;10:2988. doi: 10.1038/s41467-019-10926-2. PubMed DOI PMC
McCabe B.D., Hom S., Aberle H., Fetter R.D., Marques G., Haerry T.E., Wan H., O’Connor M.B., Goodman C.S., Haghighi A.P. Highwire regulates presynaptic BMP signaling essential for synaptic growth. Neuron. 2004;41:891–905. doi: 10.1016/S0896-6273(04)00073-X. PubMed DOI
Liang M., Liang Y.Y., Wrighton K., Ungermannova D., Wang X.P., Brunicardi F.C., Liu X., Feng X.H., Lin X. Ubiquitination and proteolysis of cancer-derived Smad4 mutants by SCFSkp2. Mol. Cell. Biol. 2004;24:7524–7537. doi: 10.1128/MCB.24.17.7524-7537.2004. PubMed DOI PMC
Yang X., Li C., Xu X., Deng C. The tumor suppressor SMAD4/DPC4 is essential for epiblast proliferation and mesoderm induction in mice. Proc. Natl. Acad. Sci. USA. 1998;95:3667–3672. doi: 10.1073/pnas.95.7.3667. PubMed DOI PMC
Shull M.M., Ormsby I., Kier A.B., Pawlowski S., Diebold R.J., Yin M., Allen R., Sidman C., Proetzel G., Calvin D., et al. Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature. 1992;359:693–699. doi: 10.1038/359693a0. PubMed DOI PMC
Mavrakis K.J., Andrew R.L., Lee K.L., Petropoulou C., Dixon J.E., Navaratnam N., Norris D.P., Episkopou V. Arkadia enhances Nodal/TGF-beta signaling by coupling phospho-Smad2/3 activity and turnover. PLoS Biol. 2007;5:e67. doi: 10.1371/journal.pbio.0050067. PubMed DOI PMC
Yamashita M., Ying S.X., Zhang G.M., Li C., Cheng S.Y., Deng C.X., Zhang Y.E. Ubiquitin ligase Smurf1 controls osteoblast activity and bone homeostasis by targeting MEKK2 for degradation. Cell. 2005;121:101–113. doi: 10.1016/j.cell.2005.01.035. PubMed DOI PMC
Ramkumar C., Kong Y., Trabucco S.E., Gerstein R.M., Zhang H. Smurf2 regulates hematopoietic stem cell self-renewal and aging. Aging Cell. 2014;13:478–486. doi: 10.1111/acel.12195. PubMed DOI PMC
Zhao L., Huang J., Guo R., Wang Y., Chen D., Xing L. Smurf1 inhibits mesenchymal stem cell proliferation and differentiation into osteoblasts through JunB degradation. J. Bone Min. Res. 2010;25:1246–1256. doi: 10.1002/jbmr.28. PubMed DOI PMC
Guo R., Yamashita M., Zhang Q., Zhou Q., Chen D., Reynolds D.G., Awad H.A., Yanoso L., Zhao L., Schwarz E.M., et al. Ubiquitin ligase Smurf1 mediates tumor necrosis factor-induced systemic bone loss by promoting proteasomal degradation of bone morphogenetic signaling proteins. J. Biol. Chem. 2008;283:23084–23092. doi: 10.1074/jbc.M709848200. PubMed DOI PMC
Zhu K., Tang Y., Xu X., Dang H., Tang L.Y., Wang X., Wang X.W., Zhang Y.E. Non-proteolytic ubiquitin modification of PPARgamma by Smurf1 protects the liver from steatosis. PLoS Biol. 2018;16:e3000091. doi: 10.1371/journal.pbio.3000091. PubMed DOI PMC
Ramkumar C., Kong Y., Cui H., Hao S., Jones S.N., Gerstein R.M., Zhang H. Smurf2 regulates the senescence response and suppresses tumorigenesis in mice. Cancer Res. 2012;72:2714–2719. doi: 10.1158/0008-5472.CAN-11-3773. PubMed DOI PMC
Ramkumar C., Cui H., Kong Y., Jones S.N., Gerstein R.M., Zhang H. Smurf2 suppresses B-cell proliferation and lymphomagenesis by mediating ubiquitination and degradation of YY1. Nat. Commun. 2013;4:2598. doi: 10.1038/ncomms3598. PubMed DOI PMC
Sriramachandran A.M., Meyer-Teschendorf K., Pabst S., Ulrich H.D., Gehring N.H., Hofmann K., Praefcke G.J.K., Dohmen R.J. Arkadia/RNF111 is a SUMO-targeted ubiquitin ligase with preference for substrates marked with SUMO1-capped SUMO2/3 chain. Nat. Commun. 2019;10:3678. doi: 10.1038/s41467-019-11549-3. PubMed DOI PMC
Sharma V., Antonacopoulou A.G., Tanaka S., Panoutsopoulos A.A., Bravou V., Kalofonos H.P., Episkopou V. Enhancement of TGF-beta signaling responses by the E3 ubiquitin ligase Arkadia provides tumor suppression in colorectal cancer. Cancer Res. 2011;71:6438–6449. doi: 10.1158/0008-5472.CAN-11-1645. PubMed DOI PMC
Episkopou V., Arkell R., Timmons P.M., Walsh J.J., Andrew R.L., Swan D. Induction of the mammalian node requires Arkadia function in the extraembryonic lineages. Nature. 2001;410:825–830. doi: 10.1038/35071095. PubMed DOI
Carrano A.C., Eytan E., Hershko A., Pagano M. SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27. Nat. Cell. Biol. 1999;1:193–199. doi: 10.1038/12013. PubMed DOI
Tsvetkov L.M., Yeh K.H., Lee S.J., Sun H., Zhang H. p27(Kip1) ubiquitination and degradation is regulated by the SCF(Skp2) complex through phosphorylated Thr187 in p27. Curr. Biol. 1999;9:661–664. doi: 10.1016/S0960-9822(99)80290-5. PubMed DOI
Ganoth D., Bornstein G., Ko T.K., Larsen B., Tyers M., Pagano M., Hershko A. The cell-cycle regulatory protein Cks1 is required for SCF(Skp2)-mediated ubiquitinylation of p27. Nat. Cell. Biol. 2001;3:321–324. doi: 10.1038/35060126. PubMed DOI
Nakayama K., Nagahama H., Minamishima Y.A., Matsumoto M., Nakamichi I., Kitagawa K., Shirane M., Tsunematsu R., Tsukiyama T., Ishida N., et al. Targeted disruption of Skp2 results in accumulation of cyclin E and p27(Kip1), polyploidy and centrosome overduplication. EMBO J. 2000;19:2069–2081. doi: 10.1093/emboj/19.9.2069. PubMed DOI PMC
Kossatz U., Dietrich N., Zender L., Buer J., Manns M.P., Malek N.P. Skp2-dependent degradation of p27kip1 is essential for cell cycle progression. Genes Dev. 2004;18:2602–2607. doi: 10.1101/gad.321004. PubMed DOI PMC
Minamishima Y.A., Nakayama K., Nakayama K. Recovery of liver mass without proliferation of hepatocytes after partial hepatectomy in Skp2-deficient mice. Cancer Res. 2002;62:995–999. PubMed
Yoshida K., Nakayama K., Nagahama H., Harada T., Harada C., Imaki J., Matsuda A., Yamamoto K., Ito M., Ohno S., et al. Involvement of p27(KIP1) degradation by Skp2 in the regulation of proliferation in response to wounding of corneal epithelium. Investig. Ophthalmol. Vis. Sci. 2002;43:364–370. PubMed
Ge Z., Guo X., Li J., Hartman M., Kawasawa Y.I., Dovat S., Song C. Clinical significance of high c-MYC and low MYCBP2 expression and their association with Ikaros dysfunction in adult acute lymphoblastic leukemia. Oncotarget. 2015;6:42300–42311. doi: 10.18632/oncotarget.5982. PubMed DOI PMC
Lewcock J.W., Genoud N., Lettieri K., Pfaff S.L. The ubiquitin ligase Phr1 regulates axon outgrowth through modulation of microtubule dynamics. Neuron. 2007;56:604–620. doi: 10.1016/j.neuron.2007.09.009. PubMed DOI
Bloom A.J., Miller B.R., Sanes J.R., DiAntonio A. The requirement for Phr1 in CNS axon tract formation reveals the corticostriatal boundary as a choice point for cortical axons. Genes Dev. 2007;21:2593–2606. doi: 10.1101/gad.1592107. PubMed DOI PMC
Han S., Kim S., Bahl S., Li L., Burande C.F., Smith N., James M., Beauchamp R.L., Bhide P., DiAntonio A., et al. The E3 ubiquitin ligase protein associated with Myc (Pam) regulates mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling in vivo through N- and C-terminal domains. J. Biol. Chem. 2012;287:30063–30072. doi: 10.1074/jbc.M112.353987. PubMed DOI PMC
Han S., Witt R.M., Santos T.M., Polizzano C., Sabatini B.L., Ramesh V. Pam (Protein associated with Myc) functions as an E3 ubiquitin ligase and regulates TSC/mTOR signaling. Cell Signal. 2008;20:1084–1091. doi: 10.1016/j.cellsig.2008.01.020. PubMed DOI PMC
Babetto E., Beirowski B., Russler E.V., Milbrandt J., DiAntonio A. The Phr1 ubiquitin ligase promotes injury-induced axon self-destruction. Cell Rep. 2013;3:1422–1429. doi: 10.1016/j.celrep.2013.04.013. PubMed DOI PMC
Chretien M.L., Legouge C., Martin R.Z., Hammann A., Trad M., Aucagne R., Largeot A., Bastie J.N., Delva L., Quere R. Trim33/Tif1gamma is involved in late stages of granulomonopoiesis in mice. Exp. Hematol. 2016;44:727–739.e726. doi: 10.1016/j.exphem.2016.04.009. PubMed DOI
Kim J., Kaartinen V. Generation of mice with a conditional allele for Trim33. Genesis. 2008;46:329–333. doi: 10.1002/dvg.20401. PubMed DOI PMC
Ferri F., Parcelier A., Petit V., Gallouet A.S., Lewandowski D., Dalloz M., van den Heuvel A., Kolovos P., Soler E., Squadrito M.L., et al. TRIM33 switches off Ifnb1 gene transcription during the late phase of macrophage activation. Nat. Commun. 2015;6:8900. doi: 10.1038/ncomms9900. PubMed DOI PMC
Kusy S., Gault N., Ferri F., Lewandowski D., Barroca V., Jaracz-Ros A., Losson R., Romeo P.H. Adult hematopoiesis is regulated by TIF1gamma, a repressor of TAL1 and PU.1 transcriptional activity. Cell Stem Cell. 2011;8:412–425. doi: 10.1016/j.stem.2011.02.005. PubMed DOI
Bai X., Trowbridge J.J., Riley E., Lee J.A., DiBiase A., Kaartinen V.M., Orkin S.H., Zon L.I. TiF1-gamma plays an essential role in murine hematopoiesis and regulates transcriptional elongation of erythroid genes. Dev. Biol. 2013;373:422–430. doi: 10.1016/j.ydbio.2012.10.008. PubMed DOI PMC
Quere R., Saint-Paul L., Carmignac V., Martin R.Z., Chretien M.L., Largeot A., Hammann A., Pais de Barros J.P., Bastie J.N., Delva L. Tif1gamma regulates the TGF-beta1 receptor and promotes physiological aging of hematopoietic stem cells. Proc. Natl. Acad. Sci. USA. 2014;111:10592–10597. doi: 10.1073/pnas.1405546111. PubMed DOI PMC
Gontan C., Achame E.M., Demmers J., Barakat T.S., Rentmeester E., van I.W., Grootegoed J.A., Gribnau J. RNF12 initiates X-chromosome inactivation by targeting REX1 for degradation. Nature. 2012;485:386–390. doi: 10.1038/nature11070. PubMed DOI
Goodrich L., Panning B., Leung K.N. Activators and repressors: A balancing act for X-inactivation. Semin. Cell Dev. Biol. 2016;56:3–8. doi: 10.1016/j.semcdb.2016.05.005. PubMed DOI
Shin J., Bossenz M., Chung Y., Ma H., Byron M., Taniguchi-Ishigaki N., Zhu X., Jiao B., Hall L.L., Green M.R., et al. Maternal Rnf12/RLIM is required for imprinted X-chromosome inactivation in mice. Nature. 2010;467:977–981. doi: 10.1038/nature09457. PubMed DOI PMC
Kammoun M., Maas E., Criem N., Gribnau J., Zwijsen A., Vermeesch J.R. RLIM enhances BMP signalling mediated fetal lung development in mice. bioRxiv. 2018 doi: 10.1101/507921. DOI
Shin J., Wallingford M.C., Gallant J., Marcho C., Jiao B., Byron M., Bossenz M., Lawrence J.B., Jones S.N., Mager J., et al. RLIM is dispensable for X-chromosome inactivation in the mouse embryonic epiblast. Nature. 2014;511:86–89. doi: 10.1038/nature13286. PubMed DOI PMC
Wang F., Shin J., Shea J.M., Yu J., Boskovic A., Byron M., Zhu X., Shalek A.K., Regev A., Lawrence J.B., et al. Regulation of X-linked gene expression during early mouse development by Rlim. Elife. 2016:5. doi: 10.7554/eLife.19127. PubMed DOI PMC
Jiao B., Ma H., Shokhirev M.N., Drung A., Yang Q., Shin J., Lu S., Byron M., Kalantry S., Mercurio A.M., et al. Paternal RLIM/Rnf12 is a survival factor for milk-producing alveolar cells. Cell. 2012;149:630–641. doi: 10.1016/j.cell.2012.02.056. PubMed DOI PMC
Chen W., Jiang X., Luo Z. WWP2: A multifunctional ubiquitin ligase gene. Pathol. Oncol. Res. 2014;20:799–803. doi: 10.1007/s12253-014-9838-y. PubMed DOI
Zou W., Chen X., Shim J.H., Huang Z., Brady N., Hu D., Drapp R., Sigrist K., Glimcher L.H., Jones D. The E3 ubiquitin ligase Wwp2 regulates craniofacial development through mono-ubiquitylation of Goosecoid. Nat. Cell Biol. 2011;13:59–65. doi: 10.1038/ncb2134. PubMed DOI PMC
Yang Y., Liao B., Wang S., Yan B., Jin Y., Shu H.B., Wang Y.Y. E3 ligase WWP2 negatively regulates TLR3-mediated innate immune response by targeting TRIF for ubiquitination and degradation. Proc. Natl. Acad. Sci. USA. 2013;110:5115–5120. doi: 10.1073/pnas.1220271110. PubMed DOI PMC
Li H., Zhang P., Zhang Q., Li C., Zou W., Chang Z., Cui C.P., Zhang L. WWP2 is a physiological ubiquitin ligase for phosphatase and tensin homolog (PTEN) in mice. J. Biol. Chem. 2018;293:8886–8899. doi: 10.1074/jbc.RA117.001060. PubMed DOI PMC
Ambrozkiewicz M.C., Schwark M., Kishimoto-Suga M., Borisova E., Hori K., Salazar-Lazaro A., Rusanova A., Altas B., Piepkorn L., Bessa P., et al. Polarity Acquisition in Cortical Neurons Is Driven by Synergistic Action of Sox9-Regulated Wwp1 and Wwp2 E3 Ubiquitin Ligases and Intronic miR-140. Neuron. 2018;100:1097–1115.e1015. doi: 10.1016/j.neuron.2018.10.008. PubMed DOI
Guruharsha K.G., Kankel M.W., Artavanis-Tsakonas S. The Notch signalling system: Recent insights into the complexity of a conserved pathway. Nat. Rev. Genet. 2012;13:654–666. doi: 10.1038/nrg3272. PubMed DOI PMC
Nowell C.S., Radtke F. Notch as a tumour suppressor. Nat. Rev. Cancer. 2017;17:145–159. doi: 10.1038/nrc.2016.145. PubMed DOI
Zavadil J., Cermak L., Soto-Nieves N., Bottinger E.P. Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J. 2004;23:1155–1165. doi: 10.1038/sj.emboj.7600069. PubMed DOI PMC
Niimi H., Pardali K., Vanlandewijck M., Heldin C.H., Moustakas A. Notch signaling is necessary for epithelial growth arrest by TGF-beta. J. Cell Biol. 2007;176:695–707. doi: 10.1083/jcb.200612129. PubMed DOI PMC
Bray S.J. Notch signalling in context. Nat. Rev. Mol. Cell Biol. 2016;17:722–735. doi: 10.1038/nrm.2016.94. PubMed DOI
Haines N., Irvine K.D. Glycosylation regulates Notch signalling. Nat. Rev. Mol. Cell Biol. 2003;4:786–797. doi: 10.1038/nrm1228. PubMed DOI
Logeat F., Bessia C., Brou C., LeBail O., Jarriault S., Seidah N.G., Israel A. The Notch1 receptor is cleaved constitutively by a furin-like convertase. Proc. Natl. Acad. Sci. USA. 1998;95:8108–8112. doi: 10.1073/pnas.95.14.8108. PubMed DOI PMC
Blobel C.P. Metalloprotease-disintegrins: Links to cell adhesion and cleavage of TNF alpha and Notch. Cell. 1997;90:589–592. doi: 10.1016/S0092-8674(00)80519-X. PubMed DOI
Pan D., Rubin G.M. Kuzbanian controls proteolytic processing of Notch and mediates lateral inhibition during Drosophila and vertebrate neurogenesis. Cell. 1997;90:271–280. doi: 10.1016/S0092-8674(00)80335-9. PubMed DOI
Itoh M., Kim C.H., Palardy G., Oda T., Jiang Y.J., Maust D., Yeo S.Y., Lorick K., Wright G.J., Ariza-McNaughton L., et al. Mind bomb is a ubiquitin ligase that is essential for efficient activation of Notch signaling by Delta. Dev. Cell. 2003;4:67–82. doi: 10.1016/S1534-5807(02)00409-4. PubMed DOI
De Strooper B., Annaert W., Cupers P., Saftig P., Craessaerts K., Mumm J.S., Schroeter E.H., Schrijvers V., Wolfe M.S., Ray W.J., et al. A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain. Nature. 1999;398:518–522. doi: 10.1038/19083. PubMed DOI
Wu L., Aster J.C., Blacklow S.C., Lake R., Artavanis-Tsakonas S., Griffin J.D. MAML1, a human homologue of Drosophila mastermind, is a transcriptional co-activator for NOTCH receptors. Nat. Genet. 2000;26:484–489. doi: 10.1038/82644. PubMed DOI
Tamura K., Taniguchi Y., Minoguchi S., Sakai T., Tun T., Furukawa T., Honjo T. Physical interaction between a novel domain of the receptor Notch and the transcription factor RBP-J kappa/Su(H) Curr. Biol. 1995;5:1416–1423. doi: 10.1016/S0960-9822(95)00279-X. PubMed DOI
Nagel A.C., Krejci A., Tenin G., Bravo-Patino A., Bray S., Maier D., Preiss A. Hairless-mediated repression of notch target genes requires the combined activity of Groucho and CtBP corepressors. Mol. Cell. Biol. 2005;25:10433–10441. doi: 10.1128/MCB.25.23.10433-10441.2005. PubMed DOI PMC
Oswald F., Winkler M., Cao Y., Astrahantseff K., Bourteele S., Knochel W., Borggrefe T. RBP-Jkappa/SHARP recruits CtIP/CtBP corepressors to silence Notch target genes. Mol. Cell. Biol. 2005;25:10379–10390. doi: 10.1128/MCB.25.23.10379-10390.2005. PubMed DOI PMC
Qiu L., Joazeiro C., Fang N., Wang H.Y., Elly C., Altman Y., Fang D., Hunter T., Liu Y.C. Recognition and ubiquitination of Notch by Itch, a hect-type E3 ubiquitin ligase. J. Biol. Chem. 2000;275:35734–35737. doi: 10.1074/jbc.M007300200. PubMed DOI
Puca L., Chastagner P., Meas-Yedid V., Israel A., Brou C. Alpha-arrestin 1 (ARRDC1) and beta-arrestins cooperate to mediate Notch degradation in mammals. J. Cell Sci. 2013;126:4457–4468. doi: 10.1242/jcs.130500. PubMed DOI
McGill M.A., McGlade C.J. Mammalian numb proteins promote Notch1 receptor ubiquitination and degradation of the Notch1 intracellular domain. J. Biol. Chem. 2003;278:23196–23203. doi: 10.1074/jbc.M302827200. PubMed DOI
Nie J., McGill M.A., Dermer M., Dho S.E., Wolting C.D., McGlade C.J. LNX functions as a RING type E3 ubiquitin ligase that targets the cell fate determinant Numb for ubiquitin-dependent degradation. EMBO J. 2002;21:93–102. doi: 10.1093/emboj/21.1.93. PubMed DOI PMC
Mund T., Graeb M., Mieszczanek J., Gammons M., Pelham H.R., Bienz M. Disinhibition of the HECT E3 ubiquitin ligase WWP2 by polymerized Dishevelled. Open Biol. 2015;5:150185. doi: 10.1098/rsob.150185. PubMed DOI PMC
Jung J.G., Stoeck A., Guan B., Wu R.C., Zhu H., Blackshaw S., Shih Ie M., Wang T.L. Notch3 interactome analysis identified WWP2 as a negative regulator of Notch3 signaling in ovarian cancer. PLoS Genet. 2014;10:e1004751. doi: 10.1371/journal.pgen.1004751. PubMed DOI PMC
Sorensen E.B., Conner S.D. gamma-secretase-dependent cleavage initiates notch signaling from the plasma membrane. Traffic. 2010;11:1234–1245. doi: 10.1111/j.1600-0854.2010.01090.x. PubMed DOI PMC
Zheng L., Conner S.D. PI5P4Kgamma functions in DTX1-mediated Notch signaling. Proc. Natl. Acad. Sci. USA. 2018;115:E1983–E1990. doi: 10.1073/pnas.1712142115. PubMed DOI PMC
Fryer C.J., White J.B., Jones K.A. Mastermind recruits CycC:CDK8 to phosphorylate the Notch ICD and coordinate activation with turnover. Mol. Cell. 2004;16:509–520. doi: 10.1016/j.molcel.2004.10.014. PubMed DOI
Rallis C., Pinchin S.M., Ish-Horowicz D. Cell-autonomous integrin control of Wnt and Notch signalling during somitogenesis. Development. 2010;137:3591–3601. doi: 10.1242/dev.050070. PubMed DOI
Ruel L., Bourouis M., Heitzler P., Pantesco V., Simpson P. Drosophila shaggy kinase and rat glycogen synthase kinase-3 have conserved activities and act downstream of Notch. Nature. 1993;362:557–560. doi: 10.1038/362557a0. PubMed DOI
Espinosa L., Ingles-Esteve J., Aguilera C., Bigas A. Phosphorylation by glycogen synthase kinase-3 beta down-regulates Notch activity, a link for Notch and Wnt pathways. J. Biol. Chem. 2003;278:32227–32235. doi: 10.1074/jbc.M304001200. PubMed DOI
Zhou T., Yi F., Wang Z., Guo Q., Liu J., Bai N., Li X., Dong X., Ren L., Cao L., et al. The Functions of DNA Damage Factor RNF8 in the Pathogenesis and Progression of Cancer. Int. J. Biol. Sci. 2019;15:909–918. doi: 10.7150/ijbs.31972. PubMed DOI PMC
Pettersson S., Sczaniecka M., McLaren L., Russell F., Gladstone K., Hupp T., Wallace M. Non-degradative ubiquitination of the Notch1 receptor by the E3 ligase MDM2 activates the Notch signalling pathway. Biochem. J. 2013;450:523–536. doi: 10.1042/BJ20121249. PubMed DOI
Kim H., Ronai Z.A. Rewired Notch/p53 by Numb’ing Mdm2. J. Cell Biol. 2018;217:445–446. doi: 10.1083/jcb.201712007. PubMed DOI PMC
Deblandre G.A., Lai E.C., Kintner C. Xenopus neuralized is a ubiquitin ligase that interacts with XDelta1 and regulates Notch signaling. Dev. Cell. 2001;1:795–806. doi: 10.1016/S1534-5807(01)00091-0. PubMed DOI
Kramer H. Neuralized: Regulating notch by putting away delta. Dev. Cell. 2001;1:725–726. doi: 10.1016/S1534-5807(01)00095-8. PubMed DOI
Fischer A., Schumacher N., Maier M., Sendtner M., Gessler M. The Notch target genes Hey1 and Hey2 are required for embryonic vascular development. Genes Dev. 2004;18:901–911. doi: 10.1101/gad.291004. PubMed DOI PMC
Zhang N., Gridley T. Defects in somite formation in lunatic fringe-deficient mice. Nature. 1998;394:374–377. doi: 10.1038/28625. PubMed DOI
Swiatek P.J., Lindsell C.E., del Amo F.F., Weinmaster G., Gridley T. Notch1 is essential for postimplantation development in mice. Genes Dev. 1994;8:707–719. doi: 10.1101/gad.8.6.707. PubMed DOI
Koo B.K., Yoon M.J., Yoon K.J., Im S.K., Kim Y.Y., Kim C.H., Suh P.G., Jan Y.N., Kong Y.Y. An obligatory role of mind bomb-1 in notch signaling of mammalian development. PLoS ONE. 2007;2:e1221. doi: 10.1371/journal.pone.0001221. PubMed DOI PMC
Koo B.K., Lim H.S., Song R., Yoon M.J., Yoon K.J., Moon J.S., Kim Y.W., Kwon M.C., Yoo K.W., Kong M.P., et al. Mind bomb 1 is essential for generating functional Notch ligands to activate Notch. Development. 2005;132:3459–3470. doi: 10.1242/dev.01922. PubMed DOI
Barsi J.C., Rajendra R., Wu J.I., Artzt K. Mind bomb1 is a ubiquitin ligase essential for mouse embryonic development and Notch signaling. Mech. Dev. 2005;122:1106–1117. doi: 10.1016/j.mod.2005.06.005. PubMed DOI
Yeh C.H., Bellon M., Nicot C. FBXW7: A critical tumor suppressor of human cancers. Mol. Cancer. 2018;17:115. doi: 10.1186/s12943-018-0857-2. PubMed DOI PMC
Mao J.H., Perez-Losada J., Wu D., Delrosario R., Tsunematsu R., Nakayama K.I., Brown K., Bryson S., Balmain A. Fbxw7/Cdc4 is a p53-dependent, haploinsufficient tumour suppressor gene. Nature. 2004;432:775–779. doi: 10.1038/nature03155. PubMed DOI
Busino L., Millman S.E., Scotto L., Kyratsous C.A., Basrur V., O’Connor O., Hoffmann A., Elenitoba-Johnson K.S., Pagano M. Fbxw7alpha- and GSK3-mediated degradation of p100 is a pro-survival mechanism in multiple myeloma. Nat. Cell Biol. 2012;14:375–385. doi: 10.1038/ncb2463. PubMed DOI PMC
Sato M., Rodriguez-Barrueco R., Yu J., Do C., Silva J.M., Gautier J. MYC is a critical target of FBXW7. Oncotarget. 2015;6:3292–3305. doi: 10.18632/oncotarget.3203. PubMed DOI PMC
Tetzlaff M.T., Yu W., Li M., Zhang P., Finegold M., Mahon K., Harper J.W., Schwartz R.J., Elledge S.J. Defective cardiovascular development and elevated cyclin E and Notch proteins in mice lacking the Fbw7 F-box protein. Proc. Natl. Acad. Sci. USA. 2004;101:3338–3345. doi: 10.1073/pnas.0307875101. PubMed DOI PMC
Tsunematsu R., Nakayama K., Oike Y., Nishiyama M., Ishida N., Hatakeyama S., Bessho Y., Kageyama R., Suda T., Nakayama K.I. Mouse Fbw7/Sel-10/Cdc4 is required for notch degradation during vascular development. J. Biol. Chem. 2004;279:9417–9423. doi: 10.1074/jbc.M312337200. PubMed DOI
Onoyama I., Tsunematsu R., Matsumoto A., Kimura T., de Alboran I.M., Nakayama K., Nakayama K.I. Conditional inactivation of Fbxw7 impairs cell-cycle exit during T cell differentiation and results in lymphomatogenesis. J. Exp. Med. 2007;204:2875–2888. doi: 10.1084/jem.20062299. PubMed DOI PMC
Matsuoka S., Oike Y., Onoyama I., Iwama A., Arai F., Takubo K., Mashimo Y., Oguro H., Nitta E., Ito K., et al. Fbxw7 acts as a critical fail-safe against premature loss of hematopoietic stem cells and development of T-ALL. Genes Dev. 2008;22:986–991. doi: 10.1101/gad.1621808. PubMed DOI PMC
Sancho R., Jandke A., Davis H., Diefenbacher M.E., Tomlinson I., Behrens A. F-box and WD repeat domain-containing 7 regulates intestinal cell lineage commitment and is a haploinsufficient tumor suppressor. Gastroenterology. 2010;139:929–941. doi: 10.1053/j.gastro.2010.05.078. PubMed DOI
Matsumoto A., Onoyama I., Sunabori T., Kageyama R., Okano H., Nakayama K.I. Fbxw7-dependent degradation of Notch is required for control of “stemness” and neuronal-glial differentiation in neural stem cells. J. Biol. Chem. 2011;286:13754–13764. doi: 10.1074/jbc.M110.194936. PubMed DOI PMC
Onoyama I., Suzuki A., Matsumoto A., Tomita K., Katagiri H., Oike Y., Nakayama K., Nakayama K.I. Fbxw7 regulates lipid metabolism and cell fate decisions in the mouse liver. J. Clin. Investig. 2011;121:342–354. doi: 10.1172/JCI40725. PubMed DOI PMC
Krebs L.T., Iwai N., Nonaka S., Welsh I.C., Lan Y., Jiang R., Saijoh Y., O’Brien T.P., Hamada H., Gridley T. Notch signaling regulates left-right asymmetry determination by inducing Nodal expression. Genes Dev. 2003;17:1207–1212. doi: 10.1101/gad.1084703. PubMed DOI PMC
Yoon M.J., Koo B.K., Song R., Jeong H.W., Shin J., Kim Y.W., Kong Y.Y., Suh P.G. Mind bomb-1 is essential for intraembryonic hematopoiesis in the aortic endothelium and the subaortic patches. Mol. Cell. Biol. 2008;28:4794–4804. doi: 10.1128/MCB.00436-08. PubMed DOI PMC
Kang K., Lee D., Hong S., Park S.G., Song M.R. The E3 ligase Mind bomb-1 (Mib1) modulates Delta-Notch signaling to control neurogenesis and gliogenesis in the developing spinal cord. J. Biol. Chem. 2013;288:2580–2592. doi: 10.1074/jbc.M112.398263. PubMed DOI PMC
Horn S., Kobberup S., Jorgensen M.C., Kalisz M., Klein T., Kageyama R., Gegg M., Lickert H., Lindner J., Magnuson M.A., et al. Mind bomb 1 is required for pancreatic beta-cell formation. Proc. Natl. Acad. Sci. USA. 2012;109:7356–7361. doi: 10.1073/pnas.1203605109. PubMed DOI PMC
Luxan G., Casanova J.C., Martinez-Poveda B., Prados B., D’Amato G., MacGrogan D., Gonzalez-Rajal A., Dobarro D., Torroja C., Martinez F., et al. Mutations in the NOTCH pathway regulator MIB1 cause left ventricular noncompaction cardiomyopathy. Nat. Med. 2013;19:193–201. doi: 10.1038/nm.3046. PubMed DOI
Kim Y.W., Koo B.K., Jeong H.W., Yoon M.J., Song R., Shin J., Jeong D.C., Kim S.H., Kong Y.Y. Defective Notch activation in microenvironment leads to myeloproliferative disease. Blood. 2008;112:4628–4638. doi: 10.1182/blood-2008-03-148999. PubMed DOI
Song R., Kim Y.W., Koo B.K., Jeong H.W., Yoon M.J., Yoon K.J., Jun D.J., Im S.K., Shin J., Kong M.P., et al. Mind bomb 1 in the lymphopoietic niches is essential for T and marginal zone B cell development. J. Exp. Med. 2008;205:2525–2536. doi: 10.1084/jem.20081344. PubMed DOI PMC
Hsu T.S., Mo S.T., Hsu P.N., Lai M.Z. c-FLIP is a target of the E3 ligase deltex1 in gastric cancer. Cell Death Dis. 2018;9:135. doi: 10.1038/s41419-017-0165-6. PubMed DOI PMC
Hsiao H.W., Liu W.H., Wang C.J., Lo Y.H., Wu Y.H., Jiang S.T., Lai M.Z. Deltex1 is a target of the transcription factor NFAT that promotes T cell anergy. Immunity. 2009;31:72–83. doi: 10.1016/j.immuni.2009.04.017. PubMed DOI
Storck S., Delbos F., Stadler N., Thirion-Delalande C., Bernex F., Verthuy C., Ferrier P., Weill J.C., Reynaud C.A. Normal immune system development in mice lacking the Deltex-1 RING finger domain. Mol. Cell. Biol. 2005;25:1437–1445. doi: 10.1128/MCB.25.4.1437-1445.2005. PubMed DOI PMC
Hsiao H.W., Hsu T.S., Liu W.H., Hsieh W.C., Chou T.F., Wu Y.J., Jiang S.T., Lai M.Z. Deltex1 antagonizes HIF-1alpha and sustains the stability of regulatory T cells in vivo. Nat. Commun. 2015;6:6353. doi: 10.1038/ncomms7353. PubMed DOI PMC
Ou-Yang H.F., Zhang H.W., Wu C.G., Zhang P., Zhang J., Li J.C., Hou L.H., He F., Ti X.Y., Song L.Q., et al. Notch signaling regulates the FOXP3 promoter through RBP-J- and Hes1-dependent mechanisms. Mol. Cell Biochem. 2009;320:109–114. doi: 10.1007/s11010-008-9912-4. PubMed DOI
Burghardt S., Claass B., Erhardt A., Karimi K., Tiegs G. Hepatocytes induce Foxp3(+) regulatory T cells by Notch signaling. J. Leukoc. Biol. 2014;96:571–577. doi: 10.1189/jlb.2AB0613-342RR. PubMed DOI
Panier S., Durocher D. Regulatory ubiquitylation in response to DNA double-strand breaks. DNA Repair. 2009;8:436–443. doi: 10.1016/j.dnarep.2009.01.013. PubMed DOI
Kolas N.K., Chapman J.R., Nakada S., Ylanko J., Chahwan R., Sweeney F.D., Panier S., Mendez M., Wildenhain J., Thomson T.M., et al. Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase. Science. 2007;318:1637–1640. doi: 10.1126/science.1150034. PubMed DOI PMC
Li L., Halaby M.J., Hakem A., Cardoso R., El Ghamrasni S., Harding S., Chan N., Bristow R., Sanchez O., Durocher D., et al. Rnf8 deficiency impairs class switch recombination, spermatogenesis, and genomic integrity and predisposes for cancer. J. Exp. Med. 2010;207:983–997. doi: 10.1084/jem.20092437. PubMed DOI PMC
Lu L.Y., Wu J., Ye L., Gavrilina G.B., Saunders T.L., Yu X. RNF8-dependent histone modifications regulate nucleosome removal during spermatogenesis. Dev. Cell. 2010;18:371–384. doi: 10.1016/j.devcel.2010.01.010. PubMed DOI PMC
Ouyang S., Song Y., Tian Y., Chen Y., Yu X., Wang D. RNF8 deficiency results in neurodegeneration in mice. Neurobiol. Aging. 2015;36:2850–2860. doi: 10.1016/j.neurobiolaging.2015.07.010. PubMed DOI PMC
Valnegri P., Huang J., Yamada T., Yang Y., Mejia L.A., Cho H.Y., Oldenborg A., Bonni A. RNF8/UBC13 ubiquitin signaling suppresses synapse formation in the mammalian brain. Nat. Commun. 2017;8:1271. doi: 10.1038/s41467-017-01333-6. PubMed DOI PMC
Senturk E., Manfredi J.J. Mdm2 and tumorigenesis: Evolving theories and unsolved mysteries. Genes Cancer. 2012;3:192–198. doi: 10.1177/1947601912457368. PubMed DOI PMC
Mendrysa S.M., McElwee M.K., Michalowski J., O’Leary K.A., Young K.M., Perry M.E. mdm2 Is critical for inhibition of p53 during lymphopoiesis and the response to ionizing irradiation. Mol. Cell. Biol. 2003;23:462–472. doi: 10.1128/MCB.23.2.462-473.2003. PubMed DOI PMC
Jones S.N., Roe A.E., Donehower L.A., Bradley A. Rescue of embryonic lethality in Mdm2-deficient mice by absence of p53. Nature. 1995;378:206–208. doi: 10.1038/378206a0. PubMed DOI
Mendrysa S.M., O’Leary K.A., McElwee M.K., Michalowski J., Eisenman R.N., Powell D.A., Perry M.E. Tumor suppression and normal aging in mice with constitutively high p53 activity. Genes Dev. 2006;20:16–21. doi: 10.1101/gad.1378506. PubMed DOI PMC
Bult C.J., Blake J.A., Smith C.L., Kadin J.A., Richardson J.E., Mouse Genome Database G. Mouse Genome Database (MGD) 2019. Nucleic Acids Res. 2019;47:D801–D806. doi: 10.1093/nar/gky1056. PubMed DOI PMC
Amberger J.S., Bocchini C.A., Schiettecatte F., Scott A.F., Hamosh A. OMIM.org: Online Mendelian Inheritance in Man (OMIM(R)), an online catalog of human genes and genetic disorders. Nucleic Acids Res. 2015;43:D789–D798. doi: 10.1093/nar/gku1205. PubMed DOI PMC