Deubiquitinating Enzyme-Mediated Signaling Networks in Cancer Stem Cells
Abstract
:Simple Summary
Abstract
1. Introduction
1.1. Ubiquitination and Deubiquitination
1.2. DUBs and Stem Cell Fate Determinants
2. DUBs Involved in Stemness-signaling Pathways Related to CSCs
2.1. Hedgehog Pathway
2.2. Wnt/β-catenin
2.3. Notch Pathway
2.4. TGF-β/BMP Signaling
2.5. Hippo Pathway
2.6. CSC Resistance-signaling Pathways
2.7. The CSC Microenvironment and Crosstalk between Signaling Pathways
3. DUB Inhibitors in CSC-targeted Therapy
4. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
References
- Fuchs, E.; Segre, J.A. Stem Cells: A New Lease on Life. Cell 2000, 100, 143–155. [Google Scholar] [CrossRef] [Green Version]
- Colter, D.C.; Sekiya, I.; Prockop, D.J. Identification of a subpopulation of rapidly self-renewing and multipotential adult stem cells in colonies of human marrow stromal cells. Proc. Natl. Acad. Sci. USA 2001, 98, 7841–7845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tata, P.R.; Mou, H.; Pardo-Saganta, A.; Zhao, R.; Prabhu, M.; Law, B.M.; Vinarsky, V.; Cho, J.L.; Breton, S.; Sahay, A.; et al. Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature 2013, 503, 218–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schatton, T.; Murphy, G.F.; Frank, N.Y.; Yamaura, K.; Waaga-Gasser, A.M.; Gasser, M.; Zhan, Q.; Jordan, S.; Duncan, L.M.; Weishaupt, C.; et al. Identification of cells initiating human melanomas. Nature 2008, 451, 345–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef] [PubMed]
- Lee, G.; Hall, R.R., 3rd; Ahmed, A.U. Cancer Stem Cells: Cellular Plasticity, Niche, and its Clinical Relevance. J. Stem Cell Res. Ther. 2016, 6, 363. [Google Scholar] [CrossRef]
- Fuchs, E. Skin stem cells: Rising to the surface. J. Cell Biol. 2008, 180, 273–284. [Google Scholar] [CrossRef] [Green Version]
- Sancho, E.; Batlle, E.; Clevers, H. Signaling pathways in intestinal development and cancer. Annu. Rev. Cell Dev. Biol. 2004, 20, 695–723. [Google Scholar] [CrossRef] [PubMed]
- Seita, J.; Weissman, I.L. Hematopoietic stem cell: Self-renewal versus differentiation. Wiley Interdiscip. Rev. Syst. Biol. Med. 2010, 2, 640–653. [Google Scholar] [CrossRef] [Green Version]
- Visvader, J.E.; Lindeman, G.J. Cancer stem cells: Current status and evolving complexities. Cell Stem Cell 2012, 10, 717–728. [Google Scholar] [CrossRef] [Green Version]
- Strikoudis, A.; Guillamot, M.; Aifantis, I. Regulation of stem cell function by protein ubiquitylation. EMBO Rep. 2014, 15, 365–382. [Google Scholar] [CrossRef] [PubMed]
- Okita, K.; Ichisaka, T.; Yamanaka, S. Generation of germline-competent induced pluripotent stem cells. Nature 2007, 448, 313–317. [Google Scholar] [CrossRef]
- Catic, A.; Ploegh, H.L. Ubiquitin--conserved protein or selfish gene? Trends Biochem. Sci. 2005, 30, 600–604. [Google Scholar] [CrossRef] [PubMed]
- Pickart, C.M.; Eddins, M.J. Ubiquitin: Structures, functions, mechanisms. Biochim. Biophys. Acta 2004, 1695, 55–72. [Google Scholar] [CrossRef] [Green Version]
- Varadan, R.; Walker, O.; Pickart, C.; Fushman, D. Structural properties of polyubiquitin chains in solution. J. Mol. Biol. 2002, 324, 637–647. [Google Scholar] [CrossRef]
- Husnjak, K.; Dikic, I. Ubiquitin-binding proteins: Decoders of ubiquitin-mediated cellular functions. Annu. Rev. Biochem. 2012, 81, 291–322. [Google Scholar] [CrossRef]
- Khaminets, A.; Behl, C.; Dikic, I. Ubiquitin-Dependent And Independent Signals In Selective Autophagy. Trends Cell Biol. 2016, 26, 6–16. [Google Scholar] [CrossRef]
- Akutsu, M.; Dikic, I.; Bremm, A. Ubiquitin chain diversity at a glance. J. Cell Sci. 2016, 129, 875–880. [Google Scholar] [CrossRef] [Green Version]
- Herhaus, L.; Dikic, I. Expanding the ubiquitin code through post-translational modification. EMBO Rep. 2015, 16, 1071–1083. [Google Scholar] [CrossRef] [Green Version]
- Wagner, S.A.; Beli, P.; Weinert, B.T.; Nielsen, M.L.; Cox, J.; Mann, M.; Choudhary, C. A proteome-wide, quantitative survey of in vivo ubiquitylation sites reveals widespread regulatory roles. Mol. Cell. Proteom. 2011, 10, M111.013284. [Google Scholar] [CrossRef] [Green Version]
- Matsumoto, M.L.; Wickliffe, K.E.; Dong, K.C.; Yu, C.; Bosanac, I.; Bustos, D.; Phu, L.; Kirkpatrick, D.S.; Hymowitz, S.G.; Rape, M.; et al. K11-linked polyubiquitination in cell cycle control revealed by a K11 linkage-specific antibody. Mol. Cell 2010, 39, 477–484. [Google Scholar] [CrossRef] [PubMed]
- Michel, M.A.; Elliott, P.R.; Swatek, K.N.; Simicek, M.; Pruneda, J.N.; Wagstaff, J.L.; Freund, S.M.; Komander, D. Assembly and specific recognition of k29- and k33-linked polyubiquitin. Mol. Cell 2015, 58, 95–109. [Google Scholar] [CrossRef] [Green Version]
- Kristariyanto, Y.A.; Abdul Rehman, S.A.; Campbell, D.G.; Morrice, N.A.; Johnson, C.; Toth, R.; Kulathu, Y. K29-selective ubiquitin binding domain reveals structural basis of specificity and heterotypic nature of k29 polyubiquitin. Mol. Cell 2015, 58, 83–94. [Google Scholar] [CrossRef] [Green Version]
- Nishikawa, H.; Ooka, S.; Sato, K.; Arima, K.; Okamoto, J.; Klevit, R.E.; Fukuda, M.; Ohta, T. Mass spectrometric and mutational analyses reveal Lys-6-linked polyubiquitin chains catalyzed by BRCA1-BARD1 ubiquitin ligase. J. Biol. Chem. 2004, 279, 3916–3924. [Google Scholar] [CrossRef] [Green Version]
- Gatti, M.; Pinato, S.; Maiolica, A.; Rocchio, F.; Prato, M.G.; Aebersold, R.; Penengo, L. RNF168 promotes noncanonical K27 ubiquitination to signal DNA damage. Cell Rep. 2015, 10, 226–238. [Google Scholar] [CrossRef] [Green Version]
- Amerik, A.Y.; Hochstrasser, M. Mechanism and function of deubiquitinating enzymes. Biochim. Biophys. Acta 2004, 1695, 189–207. [Google Scholar] [CrossRef] [Green Version]
- Fraile, J.M.; Quesada, V.; Rodriguez, D.; Freije, J.M.; Lopez-Otin, C. Deubiquitinases in cancer: New functions and therapeutic options. Oncogene 2012, 31, 2373–2388. [Google Scholar] [CrossRef] [Green Version]
- Fortelny, N.; Cox, J.H.; Kappelhoff, R.; Starr, A.E.; Lange, P.F.; Pavlidis, P.; Overall, C.M. Network analyses reveal pervasive functional regulation between proteases in the human protease web. PLoS Biol. 2014, 12, e1001869. [Google Scholar] [CrossRef]
- Abdul Rehman, S.A.; Kristariyanto, Y.A.; Choi, S.Y.; Nkosi, P.J.; Weidlich, S.; Labib, K.; Hofmann, K.; Kulathu, Y. MINDY-1 Is a Member of an Evolutionarily Conserved and Structurally Distinct New Family of Deubiquitinating Enzymes. Mol. Cell 2016, 63, 146–155. [Google Scholar] [CrossRef] [Green Version]
- Hermanns, T.; Pichlo, C.; Woiwode, I.; Klopffleisch, K.; Witting, K.F.; Ovaa, H.; Baumann, U.; Hofmann, K. A family of unconventional deubiquitinases with modular chain specificity determinants. Nat. Commun. 2018, 9, 799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reyes-Turcu, F.E.; Ventii, K.H.; Wilkinson, K.D. Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu. Rev. Biochem. 2009, 78, 363–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramakrishna, S.; Suresh, B.; Baek, K.H. The role of deubiquitinating enzymes in apoptosis. Cell. Mol. Life Sci. 2011, 68, 15–26. [Google Scholar] [CrossRef]
- Cao, P.D.; Cheung, W.K.; Nguyen, D.X. Cell lineage specification in tumor progression and metastasis. Discov. Med. 2011, 12, 329–340. [Google Scholar]
- Schmidt, R.; Plath, K. The roles of the reprogramming factors Oct4, Sox2 and Klf4 in resetting the somatic cell epigenome during induced pluripotent stem cell generation. Genome Biol. 2012, 13, 251. [Google Scholar] [CrossRef] [Green Version]
- Zhao, W.; Li, Y.; Zhang, X. Stemness-Related Markers in Cancer. Cancer Transl. Med. 2017, 3, 87–95. [Google Scholar] [CrossRef] [Green Version]
- Liao, B.; Jin, Y. Wwp2 mediates Oct4 ubiquitination and its own auto-ubiquitination in a dosage-dependent manner. Cell Res. 2010, 20, 332–344. [Google Scholar] [CrossRef] [PubMed]
- Fang, L.; Zhang, L.; Wei, W.; Jin, X.; Wang, P.; Tong, Y.; Li, J.; Du, J.X.; Wong, J. A methylation-phosphorylation switch determines Sox2 stability and function in ESC maintenance or differentiation. Mol. Cell 2014, 55, 537–551. [Google Scholar] [CrossRef] [Green Version]
- Nichols, J.; Zevnik, B.; Anastassiadis, K.; Niwa, H.; Klewe-Nebenius, D.; Chambers, I.; Scholer, H.; Smith, A. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 1998, 95, 379–391. [Google Scholar] [CrossRef] [Green Version]
- Du, Z.; Jia, D.; Liu, S.; Wang, F.; Li, G.; Zhang, Y.; Cao, X.; Ling, E.A.; Hao, A. Oct4 is expressed in human gliomas and promotes colony formation in glioma cells. Glia 2009, 57, 724–733. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Wang, W.; Li, C.; Yu, H.; Yang, A.; Wang, B.; Jin, Y. WWP2 promotes degradation of transcription factor OCT4 in human embryonic stem cells. Cell Res. 2009, 19, 561–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buckley, S.M.; Aranda-Orgilles, B.; Strikoudis, A.; Apostolou, E.; Loizou, E.; Moran-Crusio, K.; Farnsworth, C.L.; Koller, A.A.; Dasgupta, R.; Silva, J.C.; et al. Regulation of pluripotency and cellular reprogramming by the ubiquitin-proteasome system. Cell Stem Cell 2012, 11, 783–798. [Google Scholar] [CrossRef] [Green Version]
- Hagerstrand, D.; He, X.; Bradic Lindh, M.; Hoefs, S.; Hesselager, G.; Ostman, A.; Nister, M. Identification of a SOX2-dependent subset of tumor- and sphere-forming glioblastoma cells with a distinct tyrosine kinase inhibitor sensitivity profile. Neuro Oncol. 2011, 13, 1178–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cox, J.L.; Wilder, P.J.; Gilmore, J.M.; Wuebben, E.L.; Washburn, M.P.; Rizzino, A. The SOX2-interactome in brain cancer cells identifies the requirement of MSI2 and USP9X for the growth of brain tumor cells. PLoS ONE 2013, 8, e62857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sussman, R.T.; Stanek, T.J.; Esteso, P.; Gearhart, J.D.; Knudsen, K.E.; McMahon, S.B. The epigenetic modifier ubiquitin-specific protease 22 (USP22) regulates embryonic stem cell differentiation via transcriptional repression of sex-determining region Y-box 2 (SOX2). J. Biol. Chem. 2013, 288, 24234–24246. [Google Scholar] [CrossRef] [Green Version]
- Boyer, L.A.; Lee, T.I.; Cole, M.F.; Johnstone, S.E.; Levine, S.S.; Zucker, J.P.; Guenther, M.G.; Kumar, R.M.; Murray, H.L.; Jenner, R.G.; et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 2005, 122, 947–956. [Google Scholar] [CrossRef] [Green Version]
- Hu, D.; Zhou, Z.; Davidson, N.E.; Huang, Y.; Wan, Y. Novel insight into KLF4 proteolytic regulation in estrogen receptor signaling and breast carcinogenesis. J. Biol. Chem. 2012, 287, 13584–13597. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.O.; Kim, S.H.; Cho, Y.Y.; Nadas, J.; Jeong, C.H.; Yao, K.; Kim, D.J.; Yu, D.H.; Keum, Y.S.; Lee, K.Y.; et al. ERK1 and ERK2 regulate embryonic stem cell self-renewal through phosphorylation of Klf4. Nat. Struct. Mol. Biol. 2012, 19, 283–290. [Google Scholar] [CrossRef]
- de Dieuleveult, M.; Leduc, M.; Salataj, E.; Ransy, C.; Dairou, J.; Homma, K.; Le Gall, M.; Bossard, P.; Lombès, A.; Bouillaud, F.; et al. USP9X deubiquitinase couples the pluripotency network and cell metabolism to regulate ESC differentiation potential. bioRxiv 2020. [Google Scholar] [CrossRef]
- Wang, X.; Xia, S.; Li, H.; Wang, X.; Li, C.; Chao, Y.; Zhang, L.; Han, C. The deubiquitinase USP10 regulates KLF4 stability and suppresses lung tumorigenesis. Cell Death Differ. 2019, 27, 1747–1764. [Google Scholar] [CrossRef]
- Kapoor, N.; Niu, J.; Saad, Y.; Kumar, S.; Sirakova, T.; Becerra, E.; Li, X.; Kolattukudy, P.E. Transcription factors STAT6 and KLF4 implement macrophage polarization via the dual catalytic powers of MCPIP. J. Immunol. 2015, 194, 6011–6023. [Google Scholar] [CrossRef] [Green Version]
- Ludwig, S.R.; Habera, L.F.; Dellaporta, S.L.; Wessler, S.R. Lc, a member of the maize R gene family responsible for tissue-specific anthocyanin production, encodes a protein similar to transcriptional activators and contains the myc-homology region. Proc. Natl. Acad. Sci. USA 1989, 86, 7092–7096. [Google Scholar] [CrossRef] [Green Version]
- Chappell, J.; Dalton, S. Roles for MYC in the establishment and maintenance of pluripotency. Cold Spring Harb. Perspect. Med. 2013, 3, a014381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, K.; Yang, L.; Wang, J.; Sun, T.; Guo, Y.; Nelson, R.; Tong, T.R.; Pangeni, R.; Salgia, R.; Raz, D.J. Ubiquitin-specific protease 22 is critical to in vivo angiogenesis, growth and metastasis of non-small cell lung cancer. Cell Commun. Signal. 2019, 17, 167. [Google Scholar] [CrossRef] [Green Version]
- Popov, N.; Wanzel, M.; Madiredjo, M.; Zhang, D.; Beijersbergen, R.; Bernards, R.; Moll, R.; Elledge, S.J.; Eilers, M. The ubiquitin-specific protease USP28 is required for MYC stability. Nat. Cell Biol. 2007, 9, 765–774. [Google Scholar] [CrossRef]
- Pan, J.; Deng, Q.; Jiang, C.; Wang, X.; Niu, T.; Li, H.; Chen, T.; Jin, J.; Pan, W.; Cai, X.; et al. USP37 directly deubiquitinates and stabilizes c-Myc in lung cancer. Oncogene 2015, 34, 3957–3967. [Google Scholar] [CrossRef]
- Sun, X.-X.; He, X.; Yin, L.; Komada, M.; Sears, R.C.; Dai, M.-S. The nucleolar ubiquitin-specific protease USP36 deubiquitinates and stabilizes c-Myc. Proc. Natl. Acad. Sci. USA 2015, 112, 3734. [Google Scholar] [CrossRef] [Green Version]
- Chambers, I.; Colby, D.; Robertson, M.; Nichols, J.; Lee, S.; Tweedie, S.; Smith, A. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell 2003, 113, 643–655. [Google Scholar] [CrossRef] [Green Version]
- Nagata, T.; Shimada, Y.; Sekine, S.; Hori, R.; Matsui, K.; Okumura, T.; Sawada, S.; Fukuoka, J.; Tsukada, K. Prognostic significance of NANOG and KLF4 for breast cancer. Breast Cancer 2014, 21, 96–101. [Google Scholar] [CrossRef]
- Lin, T.; Ding, Y.Q.; Li, J.M. Overexpression of Nanog protein is associated with poor prognosis in gastric adenocarcinoma. Med. Oncol. 2012, 29, 878–885. [Google Scholar] [CrossRef]
- Yu, C.C.; Chen, Y.W.; Chiou, G.Y.; Tsai, L.L.; Huang, P.I.; Chang, C.Y.; Tseng, L.M.; Chiou, S.H.; Yen, S.H.; Chou, M.Y.; et al. MicroRNA let-7a represses chemoresistance and tumourigenicity in head and neck cancer via stem-like properties ablation. Oral Oncol. 2011, 47, 202–210. [Google Scholar] [CrossRef]
- Ibrahim, E.E.; Babaei-Jadidi, R.; Saadeddin, A.; Spencer-Dene, B.; Hossaini, S.; Abuzinadah, M.; Li, N.; Fadhil, W.; Ilyas, M.; Bonnet, D.; et al. Embryonic NANOG activity defines colorectal cancer stem cells and modulates through AP1- and TCF-dependent mechanisms. Stem Cells 2012, 30, 2076–2087. [Google Scholar] [CrossRef]
- Wang, X.Q.; Ng, R.K.; Ming, X.; Zhang, W.; Chen, L.; Chu, A.C.; Pang, R.; Lo, C.M.; Tsao, S.W.; Liu, X.; et al. Epigenetic regulation of pluripotent genes mediates stem cell features in human hepatocellular carcinoma and cancer cell lines. PLoS ONE 2013, 8, e72435. [Google Scholar] [CrossRef] [Green Version]
- Wei, X.; Guo, J.; Li, Q.; Jia, Q.; Jing, Q.; Li, Y.; Zhou, B.; Chen, J.; Gao, S.; Zhang, X.; et al. Bach1 regulates self-renewal and impedes mesendodermal differentiation of human embryonic stem cells. Sci. Adv. 2019, 5, eaau7887. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Yao, Y.; Ding, H.; Han, C.; Chen, Y.; Zhang, Y.; Wang, C.; Zhang, X.; Zhang, Y.; Zhai, Y.; et al. USP21 deubiquitylates Nanog to regulate protein stability and stem cell pluripotency. Signal Transduct. Target. Ther. 2016, 1, 16024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pei, D. Deubiquitylating Nanog: Novel role of USP21 in embryonic stem cell maintenance. Signal Transduct. Target. Ther. 2017, 2, 17014. [Google Scholar] [CrossRef] [Green Version]
- Kwon, S.K.; Lee, D.H.; Kim, S.Y.; Park, J.H.; Choi, J.; Baek, K.H. Ubiquitin-specific protease 21 regulating the K48-linked polyubiquitination of NANOG. Biochem. Biophys. Res. Commun. 2017, 482, 1443–1448. [Google Scholar] [CrossRef]
- Jin, J.; Liu, J.; Chen, C.; Liu, Z.; Jiang, C.; Chu, H.; Pan, W.; Wang, X.; Zhang, L.; Li, B.; et al. The deubiquitinase USP21 maintains the stemness of mouse embryonic stem cells via stabilization of Nanog. Nat. Commun. 2016, 7, 13594. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.H.; Kim, M.O.; Cho, Y.Y.; Yao, K.; Kim, D.J.; Jeong, C.H.; Yu, D.H.; Bae, K.B.; Cho, E.J.; Jung, S.K.; et al. ERK1 phosphorylates Nanog to regulate protein stability and stem cell self-renewal. Stem Cell Res. 2014, 13, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Fong, S.; Itahana, Y.; Sumida, T.; Singh, J.; Coppe, J.P.; Liu, Y.; Richards, P.C.; Bennington, J.L.; Lee, N.M.; Debs, R.J.; et al. Id-1 as a molecular target in therapy for breast cancer cell invasion and metastasis. Proc. Natl. Acad. Sci. USA 2003, 100, 13543–13548. [Google Scholar] [CrossRef] [Green Version]
- Iavarone, A.; Lasorella, A. ID proteins as targets in cancer and tools in neurobiology. Trends Mol. Med. 2006, 12, 588–594. [Google Scholar] [CrossRef]
- Williams, S.A.; Maecker, H.L.; French, D.M.; Liu, J.; Gregg, A.; Silverstein, L.B.; Cao, T.C.; Carano, R.A.; Dixit, V.M. USP1 deubiquitinates ID proteins to preserve a mesenchymal stem cell program in osteosarcoma. Cell 2011, 146, 918–930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goncalves, J.M.; Cordeiro, M.M.R.; Rivero, E.R.C. The Role of the Complex USP1/WDR48 in Differentiation and Proliferation Processes in Cancer Stem Cells. Curr. Stem Cell Res. Ther. 2017, 12, 416–422. [Google Scholar] [CrossRef]
- Sari, I.N.; Phi, L.T.H.; Jun, N.; Wijaya, Y.T.; Lee, S.; Kwon, H.Y. Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells. Cells 2018, 7, 208. [Google Scholar] [CrossRef] [Green Version]
- Clement, V.; Sanchez, P.; de Tribolet, N.; Radovanovic, I.; Ruiz i Altaba, A. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr. Biol. 2007, 17, 165–172. [Google Scholar] [CrossRef]
- Batsaikhan, B.E.; Yoshikawa, K.; Kurita, N.; Iwata, T.; Takasu, C.; Kashihara, H.; Shimada, M. Cyclopamine decreased the expression of Sonic Hedgehog and its downstream genes in colon cancer stem cells. Anticancer Res. 2014, 34, 6339–6344. [Google Scholar]
- Justilien, V.; Walsh, M.P.; Ali, S.A.; Thompson, E.A.; Murray, N.R.; Fields, A.P. The PRKCI and SOX2 oncogenes are coamplified and cooperate to activate Hedgehog signaling in lung squamous cell carcinoma. Cancer Cell 2014, 25, 139–151. [Google Scholar] [CrossRef] [Green Version]
- Najafi, M.; Abbaszadegan, M.R.; Rad, A.; Dastpak, M.; Boroumand-Noughabi, S.; Forghanifard, M.M. Crosstalk between SHH and stemness state signaling pathways in esophageal squamous cell carcinoma. J. Cell Commun. Signal 2017, 11, 147–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Po, A.; Ferretti, E.; Miele, E.; De Smaele, E.; Paganelli, A.; Canettieri, G.; Coni, S.; Di Marcotullio, L.; Biffoni, M.; Massimi, L.; et al. Hedgehog controls neural stem cells through p53-independent regulation of Nanog. EMBO J. 2010, 29, 2646–2658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Z.; Yao, X.; Li, S.; Xiong, Y.; Dong, X.; Zhao, Y.; Jiang, J.; Zhang, Q. Deubiquitination of Ci/Gli by Usp7/HAUSP Regulates Hedgehog Signaling. Dev. Cell 2015, 34, 58–72. [Google Scholar] [CrossRef] [Green Version]
- Heride, C.; Rigden, D.J.; Bertsoulaki, E.; Cucchi, D.; De Smaele, E.; Clague, M.J.; Urbe, S. The centrosomal deubiquitylase USP21 regulates Gli1 transcriptional activity and stability. J. Cell Sci. 2016, 129, 4001–4013. [Google Scholar] [CrossRef] [Green Version]
- Zhou, A.; Lin, K.; Zhang, S.; Ma, L.; Xue, J.; Morris, S.A.; Aldape, K.D.; Huang, S. Gli1-induced deubiquitinase USP48 aids glioblastoma tumorigenesis by stabilizing Gli1. EMBO Rep. 2017, 18, 1318–1330. [Google Scholar] [CrossRef]
- Qin, T.; Li, B.; Feng, X.; Fan, S.; Liu, L.; Liu, D.; Mao, J.; Lu, Y.; Yang, J.; Yu, X.; et al. Abnormally elevated USP37 expression in breast cancer stem cells regulates stemness, epithelial-mesenchymal transition and cisplatin sensitivity. J. Exp. Clin. Cancer Res. 2018, 37, 287. [Google Scholar] [CrossRef] [Green Version]
- Li, X.Y.; Mao, X.F.; Tang, X.Q.; Han, Q.Q.; Jiang, L.X.; Qiu, Y.M.; Dai, J.; Wang, Y.X. Regulation of Gli2 stability by deubiquitinase OTUB2. Biochem. Biophys. Res. Commun. 2018, 505, 113–118. [Google Scholar] [CrossRef] [PubMed]
- Xia, R.; Jia, H.; Fan, J.; Liu, Y.; Jia, J. USP8 promotes smoothened signaling by preventing its ubiquitination and changing its subcellular localization. PLoS Biol. 2012, 10, e1001238. [Google Scholar] [CrossRef] [Green Version]
- Massa, F.; Tammaro, R.; Prado, M.A.; Cesana, M.; Lee, B.H.; Finley, D.; Franco, B.; Morleo, M. The deubiquitinating enzyme Usp14 controls ciliogenesis and Hedgehog signaling. Hum. Mol. Genet. 2019, 28, 764–777. [Google Scholar] [CrossRef]
- Gedaly, R.; Galuppo, R.; Daily, M.F.; Shah, M.; Maynard, E.; Chen, C.; Zhang, X.; Esser, K.A.; Cohen, D.A.; Evers, B.M.; et al. Targeting the Wnt/beta-catenin signaling pathway in liver cancer stem cells and hepatocellular carcinoma cell lines with FH535. PLoS ONE 2014, 9, e99272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, Y.M.; Kahn, M. The role of the Wnt signaling pathway in cancer stem cells: Prospects for drug development. Res. Rep. Biochem. 2014, 4, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katoh, M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review). Int. J. Oncol. 2017, 51, 1357–1369. [Google Scholar] [CrossRef] [Green Version]
- Zhan, T.; Rindtorff, N.; Boutros, M. Wnt signaling in cancer. Oncogene 2017, 36, 1461–1473. [Google Scholar] [CrossRef]
- Morin, P.J. beta-catenin signaling and cancer. Bioessays 1999, 21, 1021–1030. [Google Scholar] [CrossRef]
- Kim, J.; Alavi Naini, F.; Sun, Y.; Ma, L. Ubiquitin-specific peptidase 2a (USP2a) deubiquitinates and stabilizes β-catenin. Am. J. Cancer Res. 2018, 8, 1823–1836. [Google Scholar] [PubMed]
- Greenblatt, M.B.; Shin, D.Y.; Oh, H.; Lee, K.Y.; Zhai, B.; Gygi, S.P.; Lotinun, S.; Baron, R.; Liu, D.; Su, B.; et al. MEKK2 mediates an alternative β-catenin pathway that promotes bone formation. Proc. Natl. Acad. Sci. USA 2016, 113, E1226–E1235. [Google Scholar] [CrossRef] [Green Version]
- Huang, X.; Langelotz, C.; Hetfeld-Pechoc, B.K.; Schwenk, W.; Dubiel, W. The COP9 signalosome mediates beta-catenin degradation by deneddylation and blocks adenomatous polyposis coli destruction via USP15. J. Mol. Biol. 2009, 391, 691–702. [Google Scholar] [CrossRef]
- Yun, S.I.; Kim, H.H.; Yoon, J.H.; Park, W.S.; Hahn, M.J.; Kim, H.C.; Chung, C.H.; Kim, K.K. Ubiquitin specific protease 4 positively regulates the WNT/β-catenin signaling in colorectal cancer. Mol. Oncol. 2015, 9, 1834–1851. [Google Scholar] [CrossRef]
- Madan, B.; Walker, M.P.; Young, R.; Quick, L.; Orgel, K.A.; Ryan, M.; Gupta, P.; Henrich, I.C.; Ferrer, M.; Marine, S.; et al. USP6 oncogene promotes Wnt signaling by deubiquitylating Frizzleds. Proc. Natl. Acad. Sci. USA 2016, 113, E2945–E2954. [Google Scholar] [CrossRef] [Green Version]
- Sun, K.; He, S.B.; Yao, Y.Z.; Qu, J.G.; Xie, R.; Ma, Y.Q.; Zong, M.H.; Chen, J.X. Tre2 (USP6NL) promotes colorectal cancer cell proliferation via Wnt/β-catenin pathway. Cancer Cell Int. 2019, 19, 102. [Google Scholar] [CrossRef]
- Mukai, A.; Yamamoto-Hino, M.; Awano, W.; Watanabe, W.; Komada, M.; Goto, S. Balanced ubiquitylation and deubiquitylation of Frizzled regulate cellular responsiveness to Wg/Wnt. EMBO J. 2010, 29, 2114–2125. [Google Scholar] [CrossRef] [Green Version]
- Yang, B.; Zhang, S.; Wang, Z.; Yang, C.; Ouyang, W.; Zhou, F.; Zhou, Y.; Xie, C. Deubiquitinase USP9X deubiquitinates β-catenin and promotes high grade glioma cell growth. Oncotarget 2016, 7, 79515–79525. [Google Scholar] [CrossRef] [Green Version]
- Shang, Z.; Zhao, J.; Zhang, Q.; Cao, C.; Tian, S.; Zhang, K.; Liu, L.; Shi, L.; Yu, N.; Yang, S. USP9X-mediated deubiquitination of B-cell CLL/lymphoma 9 potentiates Wnt signaling and promotes breast carcinogenesis. J. Biol. Chem. 2019, 294, 9844–9857. [Google Scholar] [CrossRef]
- Jung, H.; Kim, B.G.; Han, W.H.; Lee, J.H.; Cho, J.Y.; Park, W.S.; Maurice, M.M.; Han, J.K.; Lee, M.J.; Finley, D.; et al. Deubiquitination of Dishevelled by Usp14 is required for Wnt signaling. Oncogenesis 2013, 2, e64. [Google Scholar] [CrossRef]
- Xia, X.; Huang, C.; Liao, Y.; Liu, Y.; He, J.; Guo, Z.; Jiang, L.; Wang, X.; Liu, J.; Huang, H. Inhibition of USP14 enhances the sensitivity of breast cancer to enzalutamide. J. Exp. Clin. Cancer Res. 2019, 38, 220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tauriello, D.V.; Haegebarth, A.; Kuper, I.; Edelmann, M.J.; Henraat, M.; Canninga-van Dijk, M.R.; Kessler, B.M.; Clevers, H.; Maurice, M.M. Loss of the tumor suppressor CYLD enhances Wnt/beta-catenin signaling through K63-linked ubiquitination of Dvl. Mol. Cell 2010, 37, 607–619. [Google Scholar] [CrossRef] [PubMed]
- Xu, D.; Liu, J.; Fu, T.; Shan, B.; Qian, L.; Pan, L.; Yuan, J. USP25 regulates Wnt signaling by controlling the stability of tankyrases. Genes Dev. 2017, 31, 1024–1035. [Google Scholar] [CrossRef] [Green Version]
- Shi, J.; Liu, Y.; Xu, X.; Zhang, W.; Yu, T.; Jia, J.; Liu, C. Deubiquitinase USP47/UBP64E Regulates β-Catenin Ubiquitination and Degradation and Plays a Positive Role in Wnt Signaling. Mol. Cell Biol. 2015, 35, 3301–3311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tran, H.; Hamada, F.; Schwarz-Romond, T.; Bienz, M. Trabid, a new positive regulator of Wnt-induced transcription with preference for binding and cleaving K63-linked ubiquitin chains. Genes Dev. 2008, 22, 528–542. [Google Scholar] [CrossRef] [Green Version]
- Ji, L.; Lu, B.; Zamponi, R.; Charlat, O.; Aversa, R.; Yang, Z.; Sigoillot, F.; Zhu, X.; Hu, T.; Reece-Hoyes, J.S.; et al. USP7 inhibits Wnt/β-catenin signaling through promoting stabilization of Axin. Nat. Commun. 2019, 10, 4184. [Google Scholar] [CrossRef]
- Lui, T.T.; Lacroix, C.; Ahmed, S.M.; Goldenberg, S.J.; Leach, C.A.; Daulat, A.M.; Angers, S. The ubiquitin-specific protease USP34 regulates axin stability and Wnt/β-catenin signaling. Mol. Cell Biol. 2011, 31, 2053–2065. [Google Scholar] [CrossRef] [Green Version]
- Huang, T.; Zhang, Q.; Ren, W.; Yan, B.; Yi, L.; Tang, T.; Lin, H.; Zhang, Y. USP44 suppresses proliferation and enhances apoptosis in colorectal cancer cells by inactivating the Wnt/β-catenin pathway via Axin1 deubiquitination. Cell Biol. Int. 2020, 44, 1651–1659. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.M. Notch signaling and Notch signaling modifiers. Int. J. Biochem. Cell Biol. 2011, 43, 1550–1562. [Google Scholar] [CrossRef] [Green Version]
- Bray, S.J. Notch signalling: A simple pathway becomes complex. Nat. Rev. Mol. Cell Biol. 2006, 7, 678–689. [Google Scholar] [CrossRef]
- Ranganathan, P.; Weaver, K.L.; Capobianco, A.J. Notch signalling in solid tumours: A little bit of everything but not all the time. Nat. Rev. Cancer 2011, 11, 338–351. [Google Scholar] [CrossRef] [PubMed]
- Venkatesh, V.; Nataraj, R.; Thangaraj, G.S.; Karthikeyan, M.; Gnanasekaran, A.; Kaginelli, S.B.; Kuppanna, G.; Kallappa, C.G.; Basalingappa, K.M. Targeting Notch signalling pathway of cancer stem cells. Stem Cell Investig. 2018, 5, 5. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Masiero, M.; Banham, A.H.; Harris, A.L. The notch ligand JAGGED1 as a target for anti-tumor therapy. Front. Oncol. 2014, 4, 254. [Google Scholar] [CrossRef] [Green Version]
- Androutsellis-Theotokis, A.; McCormack, W.J.; Bradford, H.F.; Stern, G.M.; Pliego-Rivero, F.B. The depolarisation-induced release of [125I]BDNF from brain tissue. Brain Res. 1996, 743, 40–48. [Google Scholar] [CrossRef]
- Zhang, J.; Liu, M.; Su, Y.; Du, J.; Zhu, A.J. A targeted in vivo RNAi screen reveals deubiquitinases as new regulators of Notch signaling. G3 2012, 2, 1563–1575. [Google Scholar] [CrossRef] [Green Version]
- Sarkari, F.; Wheaton, K.; La Delfa, A.; Mohamed, M.; Shaikh, F.; Khatun, R.; Arrowsmith, C.H.; Frappier, L.; Saridakis, V.; Sheng, Y. Ubiquitin-specific protease 7 is a regulator of ubiquitin-conjugating enzyme UbE2E1. J. Biol. Chem. 2013, 288, 16975–16985. [Google Scholar] [CrossRef] [Green Version]
- Shan, H.; Li, X.; Xiao, X.; Dai, Y.; Huang, J.; Song, J.; Liu, M.; Yang, L.; Lei, H.; Tong, Y.; et al. USP7 deubiquitinates and stabilizes NOTCH1 in T-cell acute lymphoblastic leukemia. Signal Transduct. Target. Ther. 2018, 3, 29. [Google Scholar] [CrossRef] [Green Version]
- Mouchantaf, R.; Azakir, B.A.; McPherson, P.S.; Millard, S.M.; Wood, S.A.; Angers, A. The ubiquitin ligase itch is auto-ubiquitylated in vivo and in vitro but is protected from degradation by interacting with the deubiquitylating enzyme FAM/USP9X. J. Biol. Chem. 2006, 281, 38738–38747. [Google Scholar] [CrossRef] [Green Version]
- Moretti, J.; Chastagner, P.; Liang, C.C.; Cohn, M.A.; Israël, A.; Brou, C. The ubiquitin-specific protease 12 (USP12) is a negative regulator of notch signaling acting on notch receptor trafficking toward degradation. J. Biol. Chem. 2012, 287, 29429–29441. [Google Scholar] [CrossRef] [Green Version]
- Izrailit, J.; Jaiswal, A.; Zheng, W.; Moran, M.F.; Reedijk, M. Cellular stress induces TRB3/USP9x-dependent Notch activation in cancer. Oncogene 2017, 36, 1048–1057. [Google Scholar] [CrossRef]
- Rajan, N.; Elliott, R.J.; Smith, A.; Sinclair, N.; Swift, S.; Lord, C.J.; Ashworth, A. The cylindromatosis gene product, CYLD, interacts with MIB2 to regulate notch signalling. Oncotarget 2014, 5, 12126–12140. [Google Scholar] [CrossRef] [Green Version]
- Lim, R.; Sugino, T.; Nolte, H.; Andrade, J.; Zimmermann, B.; Shi, C.; Doddaballapur, A.; Ong, Y.T.; Wilhelm, K.; Fasse, J.W.D.; et al. Deubiquitinase USP10 regulates Notch signaling in the endothelium. Science 2019, 364, 188–193. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.C.; Lin, Y.C.; Liu, C.H.; Chung, H.C.; Wang, Y.T.; Lin, Y.W.; Ma, H.I.; Tu, P.H.; Lawler, S.E.; Chen, R.H. USP11 regulates PML stability to control Notch-induced malignancy in brain tumours. Nat. Commun. 2014, 5, 3214. [Google Scholar] [CrossRef] [Green Version]
- Kusanagi, K.; Inoue, H.; Ishidou, Y.; Mishima, H.K.; Kawabata, M.; Miyazono, K. Characterization of a bone morphogenetic protein-responsive Smad-binding element. Mol. Biol. Cell 2000, 11, 555–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moustakas, A.; Souchelnytskyi, S.; Heldin, C.H. Smad regulation in TGF-beta signal transduction. J. Cell Sci. 2001, 114, 4359–4369. [Google Scholar] [PubMed]
- Shen, Z.; Seppänen, H.; Kauttu, T.; Vainionpää, S.; Ye, Y.; Wang, S.; Mustonen, H.; Puolakkainen, P. Vasohibin-1 expression is regulated by transforming growth factor-β/bone morphogenic protein signaling pathway between tumor-associated macrophages and pancreatic cancer cells. J. Interferon Cytokine Res. 2013, 33, 428–433. [Google Scholar] [CrossRef] [Green Version]
- Colak, S.; Ten Dijke, P. Targeting TGF-β Signaling in Cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef]
- Zhao, Y.; Wang, X.; Wang, Q.; Deng, Y.; Li, K.; Zhang, M.; Zhang, Q.; Zhou, J.; Wang, H.Y.; Bai, P.; et al. USP2a Supports Metastasis by Tuning TGF-β Signaling. Cell Rep. 2018, 22, 2442–2454. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Zhou, F.; Drabsch, Y.; Gao, R.; Snaar-Jagalska, B.E.; Mickanin, C.; Huang, H.; Sheppard, K.A.; Porter, J.A.; Lu, C.X.; et al. USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-β type I receptor. Nat. Cell Biol. 2012, 14, 717–726. [Google Scholar] [CrossRef]
- Dupont, S.; Mamidi, A.; Cordenonsi, M.; Montagner, M.; Zacchigna, L.; Adorno, M.; Martello, G.; Stinchfield, M.J.; Soligo, S.; Morsut, L.; et al. FAM/USP9x, a deubiquitinating enzyme essential for TGFbeta signaling, controls Smad4 monoubiquitination. Cell 2009, 136, 123–135. [Google Scholar] [CrossRef]
- Stegeman, S.; Jolly, L.A.; Premarathne, S.; Gecz, J.; Richards, L.J.; Mackay-Sim, A.; Wood, S.A. Loss of Usp9x disrupts cortical architecture, hippocampal development and TGFβ-mediated axonogenesis. PLoS ONE 2013, 8, e68287. [Google Scholar] [CrossRef] [Green Version]
- Al-Salihi, M.A.; Herhaus, L.; Macartney, T.; Sapkota, G.P. USP11 augments TGFβ signalling by deubiquitylating ALK5. Open Biol. 2012, 2, 120063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacko, A.M.; Nan, L.; Li, S.; Tan, J.; Zhao, J.; Kass, D.J.; Zhao, Y. De-ubiquitinating enzyme, USP11, promotes transforming growth factor β-1 signaling through stabilization of transforming growth factor β receptor II. Cell Death Dis. 2016, 7, e2474. [Google Scholar] [CrossRef] [Green Version]
- Garcia, D.A.; Baek, C.; Estrada, M.V.; Tysl, T.; Bennett, E.J.; Yang, J.; Chang, J.T. USP11 Enhances TGFβ-Induced Epithelial-Mesenchymal Plasticity and Human Breast Cancer Metastasis. Mol. Cancer Res. 2018, 16, 1172–1184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eichhorn, P.J.; Rodón, L.; Gonzàlez-Juncà, A.; Dirac, A.; Gili, M.; Martínez-Sáez, E.; Aura, C.; Barba, I.; Peg, V.; Prat, A.; et al. USP15 stabilizes TGF-β receptor I and promotes oncogenesis through the activation of TGF-β signaling in glioblastoma. Nat. Med. 2012, 18, 429–435. [Google Scholar] [CrossRef]
- Wicks, S.J.; Haros, K.; Maillard, M.; Song, L.; Cohen, R.E.; Dijke, P.T.; Chantry, A. The deubiquitinating enzyme UCH37 interacts with Smads and regulates TGF-beta signalling. Oncogene 2005, 24, 8080–8084. [Google Scholar] [CrossRef] [Green Version]
- Kit Leng Lui, S.; Iyengar, P.V.; Jaynes, P.; Isa, Z.; Pang, B.; Tan, T.Z.; Eichhorn, P.J.A. USP26 regulates TGF-β signaling by deubiquitinating and stabilizing SMAD7. EMBO Rep. 2017, 18, 797–808. [Google Scholar] [CrossRef]
- Herhaus, L.; Al-Salihi, M.; Macartney, T.; Weidlich, S.; Sapkota, G.P. OTUB1 enhances TGFβ signalling by inhibiting the ubiquitylation and degradation of active SMAD2/3. Nat. Commun. 2013, 4, 2519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fan, Y.H.; Yu, Y.; Mao, R.F.; Tan, X.J.; Xu, G.F.; Zhang, H.; Lu, X.B.; Fu, S.B.; Yang, J. USP4 targets TAK1 to downregulate TNFα-induced NF-κB activation. Cell Death Differ. 2011, 18, 1547–1560. [Google Scholar] [CrossRef]
- Lim, J.H.; Jono, H.; Komatsu, K.; Woo, C.H.; Lee, J.; Miyata, M.; Matsuno, T.; Xu, X.; Huang, Y.; Zhang, W.; et al. CYLD negatively regulates transforming growth factor-β-signalling via deubiquitinating Akt. Nat. Commun. 2012, 3, 771. [Google Scholar] [CrossRef] [Green Version]
- Zanconato, F.; Battilana, G.; Cordenonsi, M.; Piccolo, S. YAP/TAZ as therapeutic targets in cancer. Curr. Opin. Pharmacol. 2016, 29, 26–33. [Google Scholar] [CrossRef]
- Maugeri-Sacca, M.; De Maria, R. Hippo pathway and breast cancer stem cells. Crit. Rev. Oncol. Hematol. 2016, 99, 115–122. [Google Scholar] [CrossRef] [PubMed]
- Cordenonsi, M.; Zanconato, F.; Azzolin, L.; Forcato, M.; Rosato, A.; Frasson, C.; Inui, M.; Montagner, M.; Parenti, A.R.; Poletti, A.; et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell 2011, 147, 759–772. [Google Scholar] [CrossRef]
- Morin-Kensicki, E.M.; Boone, B.N.; Howell, M.; Stonebraker, J.R.; Teed, J.; Alb, J.G.; Magnuson, T.R.; O’Neal, W.; Milgram, S.L. Defects in yolk sac vasculogenesis, chorioallantoic fusion, and embryonic axis elongation in mice with targeted disruption of Yap65. Mol. Cell Biol. 2006, 26, 77–87. [Google Scholar] [CrossRef] [Green Version]
- Basu-Roy, U.; Basilico, C.; Mansukhani, A. Perspectives on cancer stem cells in osteosarcoma. Cancer Lett. 2013, 338, 158–167. [Google Scholar] [CrossRef] [Green Version]
- Snigdha, K.; Gangwani, K.S.; Lapalikar, G.V.; Singh, A.; Kango-Singh, M. Hippo Signaling in Cancer: Lessons From Drosophila Models. Front. Cell Dev. Biol. 2019, 7, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nguyen, T.H.; Kugler, J.M. Ubiquitin-Dependent Regulation of the Mammalian Hippo Pathway: Therapeutic Implications for Cancer. Cancers 2018, 10, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thanh Nguyen, H.; Andrejeva, D.; Gupta, R.; Choudhary, C.; Hong, X.; Eichhorn, P.J.A.; Loya, A.C.; Cohen, S.M. Deubiquitylating enzyme USP9x regulates hippo pathway activity by controlling angiomotin protein turnover. Cell Discov. 2016, 2, 16001. [Google Scholar] [CrossRef]
- Nguyen, H.T.; Kugler, J.M.; Cohen, S.M. DUB3 Deubiquitylating Enzymes Regulate Hippo Pathway Activity by Regulating the Stability of ITCH, LATS and AMOT Proteins. PLoS ONE 2017, 12, e0169587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pan, B.; Yang, Y.; Li, J.; Wang, Y.; Fang, C.; Yu, F.-X.; Xu, Y. USP47-mediated deubiquitination and stabilization of YAP contributes to the progression of colorectal cancer. Protein Cell 2020, 11, 138–143. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.; Gao, Y.; Li, P.; Shi, Z.; Guo, T.; Li, F.; Han, X.; Feng, Y.; Zheng, C.; Wang, Z.; et al. VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex. Cell Res. 2014, 24, 331–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, E.; Shen, B.; Mu, X.; Qin, Y.; Zhang, F.; Liu, Y.; Xiao, J.; Zhang, P.; Wang, C.; Tan, M.; et al. Ubiquitin-specific protease 11 (USP11) functions as a tumor suppressor through deubiquitinating and stabilizing VGLL4 protein. Am. J. Cancer Res. 2016, 6, 2901–2909. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, H.T.; Kugler, J.M.; Loya, A.C.; Cohen, S.M. USP21 regulates Hippo pathway activity by mediating MARK protein turnover. Oncotarget 2017, 8, 64095–64105. [Google Scholar] [CrossRef] [Green Version]
- Kim, Y.; Kim, W.; Song, Y.; Kim, J.R.; Cho, K.; Moon, H.; Ro, S.W.; Seo, E.; Ryu, Y.M.; Myung, S.J.; et al. Deubiquitinase YOD1 potentiates YAP/TAZ activities through enhancing ITCH stability. Proc. Natl. Acad. Sci. USA 2017, 114, 4691–4696. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Du, J.; Wang, S.; Shao, L.; Jin, K.; Li, F.; Wei, B.; Ding, W.; Fu, P.; van Dam, H.; et al. OTUB2 Promotes Cancer Metastasis via Hippo-Independent Activation of YAP and TAZ. Mol. Cell 2019, 73, 7–21.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, F.; Zhou, Z.; Kim, J.; Hang, Q.; Xiao, Z.; Ton, B.N.; Chang, L.; Liu, N.; Zeng, L.; Wang, W.; et al. SKP2- and OTUD1-regulated non-proteolytic ubiquitination of YAP promotes YAP nuclear localization and activity. Nat. Commun. 2018, 9, 2269. [Google Scholar] [CrossRef]
- Zhou, Z.; Zhou, H.; Ponzoni, L.; Luo, A.; Zhu, R.; He, M.; Huang, Y.; Guan, K.L.; Bahar, I.; Liu, Z.; et al. EIF3H Orchestrates Hippo Pathway-Mediated Oncogenesis via Catalytic Control of YAP Stability. Cancer Res. 2020, 80, 2550–2563. [Google Scholar] [CrossRef] [Green Version]
- Chen, S.M.; Li, Y.Y.; Tu, C.H.; Salazar, N.; Tseng, Y.Y.; Huang, S.F.; Hsieh, L.L.; Lui, T.N. Blockade of Inhibitors of Apoptosis Proteins in Combination with Conventional Chemotherapy Leads to Synergistic Antitumor Activity in Medulloblastoma and Cancer Stem-Like Cells. PLoS ONE 2016, 11, e0161299. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.; Brocker, C.; Koppaka, V.; Chen, Y.; Jackson, B.C.; Matsumoto, A.; Thompson, D.C.; Vasiliou, V. Aldehyde dehydrogenases in cellular responses to oxidative/electrophilic stress. Free Radic Biol. Med. 2013, 56, 89–101. [Google Scholar] [CrossRef] [Green Version]
- Begicevic, R.R.; Falasca, M. ABC Transporters in Cancer Stem Cells: Beyond Chemoresistance. Int. J. Mol. Sci. 2017, 18, 2362. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.; Alexander, C.M. Tumorsphere assay provides more accurate prediction of in vivo responses to chemotherapeutics. Biotechnol. Lett. 2014, 36, 481–488. [Google Scholar] [CrossRef] [Green Version]
- Zhou, J.; Wulfkuhle, J.; Zhang, H.; Gu, P.; Yang, Y.; Deng, J.; Margolick, J.B.; Liotta, L.A.; Petricoin, E., 3rd; Zhang, Y. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc. Natl. Acad. Sci. USA 2007, 104, 16158–16163. [Google Scholar] [CrossRef] [Green Version]
- Zuo, M.; Rashid, A.; Churi, C.; Vauthey, J.N.; Chang, P.; Li, Y.; Hung, M.C.; Li, D.; Javle, M. Novel therapeutic strategy targeting the Hedgehog signalling and mTOR pathways in biliary tract cancer. Br. J. Cancer 2015, 112, 1042–1051. [Google Scholar] [CrossRef] [Green Version]
- Trivigno, D.; Essmann, F.; Huber, S.M.; Rudner, J. Deubiquitinase USP9x confers radioresistance through stabilization of Mcl-1. Neoplasia 2012, 14, 893–904. [Google Scholar] [CrossRef]
- Lee, E.W.; Seong, D.; Seo, J.; Jeong, M.; Lee, H.K.; Song, J. USP11-dependent selective cIAP2 deubiquitylation and stabilization determine sensitivity to Smac mimetics. Cell Death Differ. 2015, 22, 1463–1476. [Google Scholar] [CrossRef] [Green Version]
- Mei, Y.; Hahn, A.A.; Hu, S.; Yang, X. The USP19 deubiquitinase regulates the stability of c-IAP1 and c-IAP2. J. Biol. Chem. 2011, 286, 35380–35387. [Google Scholar] [CrossRef] [Green Version]
- Goncharov, T.; Niessen, K.; de Almagro, M.C.; Izrael-Tomasevic, A.; Fedorova, A.V.; Varfolomeev, E.; Arnott, D.; Deshayes, K.; Kirkpatrick, D.S.; Vucic, D. OTUB1 modulates c-IAP1 stability to regulate signalling pathways. EMBO J. 2013, 32, 1103–1114. [Google Scholar] [CrossRef] [PubMed]
- Faustrup, H.; Bekker-Jensen, S.; Bartek, J.; Lukas, J.; Mailand, N. USP7 counteracts SCFbetaTrCP- but not APCCdh1-mediated proteolysis of Claspin. J. Cell Biol. 2009, 184, 13–19. [Google Scholar] [CrossRef]
- Zhu, M.; Zhao, H.; Liao, J.; Xu, X. HERC2/USP20 coordinates CHK1 activation by modulating CLASPIN stability. Nucleic Acids Res. 2014, 42, 13074–13081. [Google Scholar] [CrossRef] [Green Version]
- Bassermann, F.; Frescas, D.; Guardavaccaro, D.; Busino, L.; Peschiaroli, A.; Pagano, M. The Cdc14B-Cdh1-Plk1 axis controls the G2 DNA-damage-response checkpoint. Cell 2008, 134, 256–267. [Google Scholar] [CrossRef] [Green Version]
- Panner, A.; Crane, C.A.; Weng, C.; Feletti, A.; Fang, S.; Parsa, A.T.; Pieper, R.O. Ubiquitin-specific protease 8 links the PTEN-Akt-AIP4 pathway to the control of FLIPS stability and TRAIL sensitivity in glioblastoma multiforme. Cancer Res. 2010, 70, 5046–5053. [Google Scholar] [CrossRef] [Green Version]
- Guervilly, J.H.; Renaud, E.; Takata, M.; Rosselli, F. USP1 deubiquitinase maintains phosphorylated CHK1 by limiting its DDB1-dependent degradation. Hum. Mol. Genet. 2011, 20, 2171–2181. [Google Scholar] [CrossRef] [Green Version]
- Cheng, Y.C.; Shieh, S.Y. Deubiquitinating enzyme USP3 controls CHK1 chromatin association and activation. Proc. Natl. Acad. Sci. USA 2018, 115, 5546–5551. [Google Scholar] [CrossRef] [Green Version]
- Alonso-de Vega, I.; Martín, Y.; Smits, V.A. USP7 controls Chk1 protein stability by direct deubiquitination. Cell Cycle 2014, 13, 3921–3926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, D.; Zaugg, K.; Mak, T.W.; Elledge, S.J. A role for the deubiquitinating enzyme USP28 in control of the DNA-damage response. Cell 2006, 126, 529–542. [Google Scholar] [CrossRef] [Green Version]
- Lathia, J.D.; Heddleston, J.M.; Venere, M.; Rich, J.N. Deadly teamwork: Neural cancer stem cells and the tumor microenvironment. Cell Stem Cell 2011, 8, 482–485. [Google Scholar] [CrossRef] [Green Version]
- Clara, J.A.; Monge, C.; Yang, Y.; Takebe, N. Targeting signalling pathways and the immune microenvironment of cancer stem cells—A clinical update. Nat. Rev. Clin. Oncol. 2020, 17, 204–232. [Google Scholar] [CrossRef]
- Kise, K.; Kinugasa-Katayama, Y.; Takakura, N. Tumor microenvironment for cancer stem cells. Adv. Drug Deliv. Rev. 2016, 99, 197–205. [Google Scholar] [CrossRef]
- Lau, E.Y.; Ho, N.P.; Lee, T.K. Cancer Stem Cells and Their Microenvironment: Biology and Therapeutic Implications. Stem Cells Int. 2017, 2017, 3714190. [Google Scholar] [CrossRef]
- Sattiraju, A.; Sai, K.K.S.; Mintz, A. Glioblastoma Stem Cells and Their Microenvironment. Adv. Exp. Med. Biol. 2017, 1041, 119–140. [Google Scholar] [CrossRef] [PubMed]
- Vermeulen, L.; De Sousa, E.M.F.; van der Heijden, M.; Cameron, K.; de Jong, J.H.; Borovski, T.; Tuynman, J.B.; Todaro, M.; Merz, C.; Rodermond, H.; et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol. 2010, 12, 468–476. [Google Scholar] [CrossRef] [PubMed]
- Mittal, S.; Brown, N.J.; Holen, I. The breast tumor microenvironment: Role in cancer development, progression and response to therapy. Expert Rev. Mol. Diagn. 2018, 18, 227–243. [Google Scholar] [CrossRef] [PubMed]
- Jain, M.V.; Jangamreddy, J.R.; Grabarek, J.; Schweizer, F.; Klonisch, T.; Cieslar-Pobuda, A.; Los, M.J. Nuclear localized Akt enhances breast cancer stem-like cells through counter-regulation of p21(Waf1/Cip1) and p27(kip1). Cell Cycle 2015, 14, 2109–2120. [Google Scholar] [CrossRef] [Green Version]
- Korkaya, H.; Liu, S.; Wicha, M.S. Breast cancer stem cells, cytokine networks, and the tumor microenvironment. J. Clin. Investig. 2011, 121, 3804–3809. [Google Scholar] [CrossRef]
- Yamamoto, M.; Taguchi, Y.; Ito-Kureha, T.; Semba, K.; Yamaguchi, N.; Inoue, J. NF-kappaB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype. Nat. Commun. 2013, 4, 2299. [Google Scholar] [CrossRef] [Green Version]
- Chefetz, I.; Alvero, A.B.; Holmberg, J.C.; Lebowitz, N.; Craveiro, V.; Yang-Hartwich, Y.; Yin, G.; Squillace, L.; Gurrea Soteras, M.; Aldo, P.; et al. TLR2 enhances ovarian cancer stem cell self-renewal and promotes tumor repair and recurrence. Cell Cycle 2013, 12, 511–521. [Google Scholar] [CrossRef] [Green Version]
- Feng, X.; Yu, Y.; He, S.; Cheng, J.; Gong, Y.; Zhang, Z.; Yang, X.; Xu, B.; Liu, X.; Li, C.Y.; et al. Dying glioma cells establish a proangiogenic microenvironment through a caspase 3 dependent mechanism. Cancer Lett. 2017, 385, 12–20. [Google Scholar] [CrossRef] [Green Version]
- Inukai, M.; Hara, A.; Yasui, Y.; Kumabe, T.; Matsumoto, T.; Saegusa, M. Hypoxia-mediated cancer stem cells in pseudopalisades with activation of hypoxia-inducible factor-1alpha/Akt axis in glioblastoma. Hum. Pathol. 2015, 46, 1496–1505. [Google Scholar] [CrossRef]
- Li, Q.; Yu, L.; Xin, T.; Du, S.; Yi, X.; Zhang, B.; Wu, B.; Sun, H. Increased IL-9 mRNA expression as a biomarker to diagnose childhood tuberculosis in a high burden settings. J. Infect. 2015, 71, 273–276. [Google Scholar] [CrossRef]
- Carnero, A.; Lleonart, M. The hypoxic microenvironment: A determinant of cancer stem cell evolution. Bioessays 2016, 38 (Suppl. 1), S65–S74. [Google Scholar] [CrossRef]
- Zhao, M.; Zhang, Y.; Zhang, H.; Wang, S.; Zhang, M.; Chen, X.; Wang, H.; Zeng, G.; Chen, X.; Liu, G.; et al. Hypoxia-induced cell stemness leads to drug resistance and poor prognosis in lung adenocarcinoma. Lung Cancer 2015, 87, 98–106. [Google Scholar] [CrossRef]
- Maxwell, P.H.; Wiesener, M.S.; Chang, G.W.; Clifford, S.C.; Vaux, E.C.; Cockman, M.E.; Wykoff, C.C.; Pugh, C.W.; Maher, E.R.; Ratcliffe, P.J. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 1999, 399, 271–275. [Google Scholar] [CrossRef]
- Katoh, M. Networking of WNT, FGF, Notch, BMP, and Hedgehog signaling pathways during carcinogenesis. Stem Cell Rev. 2007, 3, 30–38. [Google Scholar] [CrossRef]
- He, J.; Sheng, T.; Stelter, A.A.; Li, C.; Zhang, X.; Sinha, M.; Luxon, B.A.; Xie, J. Suppressing Wnt signaling by the hedgehog pathway through sFRP-1. J. Biol. Chem. 2006, 281, 35598–35602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noubissi, F.K.; Goswami, S.; Sanek, N.A.; Kawakami, K.; Minamoto, T.; Moser, A.; Grinblat, Y.; Spiegelman, V.S. Wnt signaling stimulates transcriptional outcome of the Hedgehog pathway by stabilizing GLI1 mRNA. Cancer Res. 2009, 69, 8572–8578. [Google Scholar] [CrossRef] [Green Version]
- Schreck, K.C.; Taylor, P.; Marchionni, L.; Gopalakrishnan, V.; Bar, E.E.; Gaiano, N.; Eberhart, C.G. The Notch target Hes1 directly modulates Gli1 expression and Hedgehog signaling: A potential mechanism of therapeutic resistance. Clin. Cancer Res. 2010, 16, 6060–6070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nawshad, A.; Medici, D.; Liu, C.C.; Hay, E.D. TGFbeta3 inhibits E-cadherin gene expression in palate medial-edge epithelial cells through a Smad2-Smad4-LEF1 transcription complex. J. Cell Sci. 2007, 120, 1646–1653. [Google Scholar] [CrossRef] [Green Version]
- Galceran, J.; Fariñas, I.; Depew, M.J.; Clevers, H.; Grosschedl, R. Wnt3a-/--like phenotype and limb deficiency in Lef1(-/-)Tcf1(-/-) mice. Genes Dev. 1999, 13, 709–717. [Google Scholar] [CrossRef]
- Taya, Y.; O’Kane, S.; Ferguson, M.W. Pathogenesis of cleft palate in TGF-beta3 knockout mice. Development 1999, 126, 3869–3879. [Google Scholar] [PubMed]
- Wu, W.; Chen, F.; Cui, X.; Yang, L.; Chen, J.; Zhao, J.; Huang, D.; Liu, J.; Yang, L.; Zeng, J.; et al. LncRNA NKILA suppresses TGF-β-induced epithelial-mesenchymal transition by blocking NF-κB signaling in breast cancer. Int. J. Cancer 2018, 143, 2213–2224. [Google Scholar] [CrossRef]
- Serra, R.; Easter, S.L.; Jiang, W.; Baxley, S.E. Wnt5a as an effector of TGFβ in mammary development and cancer. J. Mammary Gland Biol. Neoplasia 2011, 16, 157–167. [Google Scholar] [CrossRef] [Green Version]
- Luu, A.K.; Schott, C.R.; Jones, R.; Poon, A.C.; Golding, B.; Hamed, R.; Deheshi, B.; Mutsaers, A.; Wood, G.A.; Viloria-Petit, A.M. An evaluation of TAZ and YAP crosstalk with TGFβ signalling in canine osteosarcoma suggests involvement of hippo signalling in disease progression. BMC Vet. Res. 2018, 14, 365. [Google Scholar] [CrossRef]
- Ward, D.; Montes Olivas, S.; Fletcher, A.; Homer, M.; Marucci, L. Cross-talk between Hippo and Wnt signalling pathways in intestinal crypts: Insights from an agent-based model. Comput. Struct. Biotechnol. J. 2020, 18, 230–240. [Google Scholar] [CrossRef]
- Munakata, K.; Uemura, M.; Tanaka, S.; Kawai, K.; Kitahara, T.; Miyo, M.; Kano, Y.; Nishikawa, S.; Fukusumi, T.; Takahashi, Y.; et al. Cancer Stem-like Properties in Colorectal Cancer Cells with Low Proteasome Activity. Clin. Cancer Res. 2016, 22, 5277–5286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Diehn, M.; Cho, R.W.; Lobo, N.A.; Kalisky, T.; Dorie, M.J.; Kulp, A.N.; Qian, D.; Lam, J.S.; Ailles, L.E.; Wong, M.; et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009, 458, 780–783. [Google Scholar] [CrossRef]
- Tian, Z.; D’Arcy, P.; Wang, X.; Ray, A.; Tai, Y.-T.; Hu, Y.; Carrasco, R.D.; Richardson, P.; Linder, S.; Chauhan, D.; et al. A novel small molecule inhibitor of deubiquitylating enzyme USP14 and UCHL5 induces apoptosis in multiple myeloma and overcomes bortezomib resistance. Blood 2014, 123, 706–716. [Google Scholar] [CrossRef]
- Lee, J.K.; Chang, N.; Yoon, Y.; Yang, H.; Cho, H.; Kim, E.; Shin, Y.; Kang, W.; Oh, Y.T.; Mun, G.I.; et al. USP1 targeting impedes GBM growth by inhibiting stem cell maintenance and radioresistance. Neuro Oncol. 2016, 18, 37–47. [Google Scholar] [CrossRef] [Green Version]
- Gopinath, P.; Ohayon, S.; Nawatha, M.; Brik, A. Chemical and semisynthetic approaches to study and target deubiquitinases. Chem. Soc. Rev. 2016, 45, 4171–4198. [Google Scholar] [CrossRef] [Green Version]
- Reverdy, C.; Conrath, S.; Lopez, R.; Planquette, C.; Atmanene, C.; Collura, V.; Harpon, J.; Battaglia, V.; Vivat, V.; Sippl, W.; et al. Discovery of specific inhibitors of human USP7/HAUSP deubiquitinating enzyme. Chem. Biol. 2012, 19, 467–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lim, K.H.; Song, M.H.; Baek, K.H. Decision for cell fate: Deubiquitinating enzymes in cell cycle checkpoint. Cell Mol. Life Sci. 2016, 73, 1439–1455. [Google Scholar] [CrossRef]
- Chauhan, D.; Tian, Z.; Nicholson, B.; Kumar, K.G.; Zhou, B.; Carrasco, R.; McDermott, J.L.; Leach, C.A.; Fulcinniti, M.; Kodrasov, M.P.; et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell 2012, 22, 345–358. [Google Scholar] [CrossRef] [Green Version]
- Pal, A.; Young, M.A.; Donato, N.J. Emerging potential of therapeutic targeting of ubiquitin-specific proteases in the treatment of cancer. Cancer Res. 2014, 74, 4955–4966. [Google Scholar] [CrossRef] [Green Version]
- Weisberg, E.L.; Schauer, N.J.; Yang, J.; Lamberto, I.; Doherty, L.; Bhatt, S.; Nonami, A.; Meng, C.; Letai, A.; Wright, R.; et al. Inhibition of USP10 induces degradation of oncogenic FLT3. Nat. Chem. Biol. 2017, 13, 1207–1215. [Google Scholar] [CrossRef]
- Liu, J.; Xia, H.; Kim, M.; Xu, L.; Li, Y.; Zhang, L.; Cai, Y.; Norberg, H.V.; Zhang, T.; Furuya, T.; et al. Beclin1 controls the levels of p53 by regulating the deubiquitination activity of USP10 and USP13. Cell 2011, 147, 223–234. [Google Scholar] [CrossRef] [Green Version]
- Yang, M.; Li, C.; Zhu, S.; Cao, L.; Kroemer, G.; Zeh, H.; Tang, D.; Kang, R. TFAM is a novel mediator of immunogenic cancer cell death. Oncoimmunology 2018, 7, e1431086. [Google Scholar] [CrossRef] [PubMed]
- Shen, G.; Lin, Y.; Yang, X.; Zhang, J.; Xu, Z.; Jia, H. MicroRNA-26b inhibits epithelial-mesenchymal transition in hepatocellular carcinoma by targeting USP9X. BMC Cancer 2014, 14, 393. [Google Scholar] [CrossRef]
- Fu, P.; Du, F.; Liu, Y.; Yao, M.; Zhang, S.; Zheng, X.; Zheng, S. WP1130 increases cisplatin sensitivity through inhibition of usp9x in estrogen receptor-negative breast cancer cells. Am. J. Transl. Res. 2017, 9, 1783–1791. [Google Scholar]
- Jin, W.L.; Mao, X.Y.; Qiu, G.Z. Targeting Deubiquitinating Enzymes in Glioblastoma Multiforme: Expectations and Challenges. Med. Res. Rev. 2017, 37, 627–661. [Google Scholar] [CrossRef]
- Kapuria, V.; Peterson, L.F.; Fang, D.; Bornmann, W.G.; Talpaz, M.; Donato, N.J. Deubiquitinase inhibition by small-molecule WP1130 triggers aggresome formation and tumor cell apoptosis. Cancer Res. 2010, 70, 9265–9276. [Google Scholar] [CrossRef] [Green Version]
- Pham, L.V.; Tamayo, A.T.; Li, C.; Bornmann, W.; Priebe, W.; Ford, R.J. Degrasyn potentiates the antitumor effects of bortezomib in mantle cell lymphoma cells in vitro and in vivo: Therapeutic implications. Mol. Cancer Ther. 2010, 9, 2026–2036. [Google Scholar] [CrossRef] [Green Version]
- D’Arcy, P.; Brnjic, S.; Olofsson, M.H.; Fryknäs, M.; Lindsten, K.; De Cesare, M.; Perego, P.; Sadeghi, B.; Hassan, M.; Larsson, R.; et al. Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat. Med. 2011, 17, 1636–1640. [Google Scholar] [CrossRef]
- Farshi, P.; Deshmukh, R.R.; Nwankwo, J.O.; Arkwright, R.T.; Cvek, B.; Liu, J.; Dou, Q.P. Deubiquitinases (DUBs) and DUB inhibitors: A patent review. Expert Opin. Ther. Pat. 2015, 25, 1191–1208. [Google Scholar] [CrossRef] [Green Version]
- Mistry, H.; Hsieh, G.; Buhrlage, S.J.; Huang, M.; Park, E.; Cuny, G.D.; Galinsky, I.; Stone, R.M.; Gray, N.S.; D’Andrea, A.D.; et al. Small-molecule inhibitors of USP1 target ID1 degradation in leukemic cells. Mol. Cancer Ther. 2013, 12, 2651–2662. [Google Scholar] [CrossRef] [Green Version]
- Harrigan, J.A.; Jacq, X.; Martin, N.M.; Jackson, S.P. Deubiquitylating enzymes and drug discovery: Emerging opportunities. Nat. Rev. Drug Discov. 2018, 17, 57–78. [Google Scholar] [CrossRef]
- D’Arcy, P.; Wang, X.; Linder, S. Deubiquitinase inhibition as a cancer therapeutic strategy. Pharmacol. Ther. 2015, 147, 32–54. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Mazurkiewicz, M.; Hillert, E.K.; Olofsson, M.H.; Pierrou, S.; Hillertz, P.; Gullbo, J.; Selvaraju, K.; Paulus, A.; Akhtar, S.; et al. The proteasome deubiquitinase inhibitor VLX1570 shows selectivity for ubiquitin-specific protease-14 and induces apoptosis of multiple myeloma cells. Sci. Rep. 2016, 6, 26979. [Google Scholar] [CrossRef]
- Advani, A.S.; Cooper, B.; Visconte, V.; Elson, P.; Chan, R.; Carew, J.; Wei, W.; Mukherjee, S.; Gerds, A.; Carraway, H.; et al. A Phase I/II Trial of MEC (Mitoxantrone, Etoposide, Cytarabine) in Combination with Ixazomib for Relapsed Refractory Acute Myeloid Leukemia. Clin. Cancer Res. 2019, 25, 4231–4237. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.; Shi, Y.; Li, C.; Gui, L.; Zhao, X.; Liu, P.; Han, X.; Song, Y.; Li, N.; Du, P.; et al. Phase I clinical trial of pegylated liposomal mitoxantrone plm60-s: Pharmacokinetics, toxicity and preliminary efficacy. Cancer Chemother. Pharmacol. 2014, 74, 637–646. [Google Scholar] [CrossRef] [PubMed]
Factor | Associated DUB | Function | Representative CSC Type |
---|---|---|---|
Sox2 | USP22, USP9X | Regulates transcription and growth of cancer cells | Brain, pancreatic, prostrate, lung |
Nanog | USP21 USP3 USP16 | Protein stabilization | Pancreatic, lung, glioma, ovarian |
Oct 4 | USP7 USP 44 | Transcription factor Promotes differentiation | Melanoma, breast |
c-Myc | USP36 USP37 USP22 USP28 | Protein stabilization | Glioma, liver, lung |
ID proteins | USP1 | Protein stabilization | Glioma, osteosarcoma |
SIRT1 | USP22 | Positive regulator | Colorectal |
Klf4 | MCPIP | Transcription factor | Pancreatic |
Lin28 | USP28 | Promotes translation | Liver, oral |
P53 | USP2a, OTUB1 USP10 Ataxin−3 USP7 OTUD1 OTUD5 USP11 | Protein stabilization | Osteosarcoma, glioma |
PTEN | ATXN3 USP18 USP7 | Transcription Protein stabilization Location Tumor suppression | Prostate, endometrial |
c-met | USP8 | Organ regeneration Promotes cancer | Liver, prostate |
Bmi1 | USP7 USP11 | Embryonic development DNA damage repair Self-renewal | Glioma, lung, head, neck |
LSD1/KDM1A | USP7 USP11 USP28 | Protein stabilization | Breast, glioma |
REST | USP7 USP15 | Transcriptional repression | Glioma |
PRC1 | USP7 USP11 USP26 | Protein stabilization | Prostate, ovarian, uterine |
PRC2 | BAP1 | Regulation of gene expression | Prostate |
Signaling Pathway | CSC Fate | DUBs Involved | Functions |
---|---|---|---|
Hedgehog | Breast glioblastoma adenocarcinoma | USP21 USP 8 | Cell growth, cell specialization, patterning of the body |
Wnt signaling pathway | Glioma | USP4 USP8 | Embryonic development |
Notch | Ovarian | EIF3f USP9X USP12 CYLD USP28 | Cell–cell communication |
Hippo | Osteosarcoma | USP9X USP11 | Modulates cell proliferation |
TGF/BMP | Breast | OTUB1 USP9X | Embryonal development, cellular differentiation, hormone secretion, immune function |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kaushal, K.; Ramakrishna, S. Deubiquitinating Enzyme-Mediated Signaling Networks in Cancer Stem Cells. Cancers 2020, 12, 3253. https://doi.org/10.3390/cancers12113253
Kaushal K, Ramakrishna S. Deubiquitinating Enzyme-Mediated Signaling Networks in Cancer Stem Cells. Cancers. 2020; 12(11):3253. https://doi.org/10.3390/cancers12113253
Chicago/Turabian StyleKaushal, Kamini, and Suresh Ramakrishna. 2020. "Deubiquitinating Enzyme-Mediated Signaling Networks in Cancer Stem Cells" Cancers 12, no. 11: 3253. https://doi.org/10.3390/cancers12113253
APA StyleKaushal, K., & Ramakrishna, S. (2020). Deubiquitinating Enzyme-Mediated Signaling Networks in Cancer Stem Cells. Cancers, 12(11), 3253. https://doi.org/10.3390/cancers12113253