Killing by Degradation: Regulation of Apoptosis by the Ubiquitin-Proteasome-System
Abstract
:1. Apoptosis (Programmed Cell Death)
2. The Ubiquitin Proteasome System
E3 Ligases
3. The Regulation of Pro- and Anti-Apoptotic Protein by the UPS
3.1. p53
E3 Ligase | Type | Effect on p53 | Model | Ubiquitination Observed In Vivo/In Vitro | References |
---|---|---|---|---|---|
MDM2 | RING | Degradation | mouse | In vivo and in vitro | [141,142] |
ARF-BP1 | HECT | Degradation | mouse | In vivo and in vitro | [175] |
CHIP | U-box | Degradation | mouse | In vivo and in vitro | [182] |
Cop1 | RING | Degradation | - | In vivo and in vitro | [173] |
Cul1 | RING | Degradation | mouse | In vivo and in vitro | [194] |
Cul4a | RING | Degradation | mouse | In vivo | [195,196,197] |
Cul5 | RING | Degradation | - | In vitro | [198] |
Synoviolin | RING | Degradation | drosophila | In vivo and in vitro | [199] |
TOPORS | RING | Degradation | - | In vivo and in vitro | [200] |
Trim24 | RING | Degradation | drosophila | In vivo and in vitro | [201] |
TRIM28 | RING | Degradation | - | In vitro | [202] |
TRIM39 | RING | Degradation | - | In vitro | [203] |
TRIM65 | RING | Degradation | - | In vivo | [204] |
Carpi | RING | Degradation | - | In vivo | [205] |
Carp2 | RING | Degradation | - | In vivo | [205] |
Pirh2 | RING | Degradation | mouse | In vivo and in vitro | [169] |
TRAF6 | RING | Degradation | mouse | In vivo and in vitro | [206] |
TRAF7 | RING | Degradation | - | In vitro | [207] |
RNF2 | RING | Degradation | mouse | In vivo and in vitro | [208] |
RING1 | RING | Degradation | - | In vivo and in vitro | [209] |
3.2. BCl-2 Family
3.3. Inhibitor of Apoptosis (IAPs) Proteins and IAPs-Antagonists
4. Targeting the UPS for Apoptosis-Induced Cancer Therapy
4.1. Proteasome Inhibitors
4.2. Cancer Therapies Targeting p53 and MDM2 for Degradation
4.3. Cancer Therapies Targeting Bcl-2 Family Proteins for Degradation
4.4. Cancer Therapies Targeting IAPs for Degradation
5. Future Directions and Challenges
Author Contributions
Funding
Conflicts of Interest
References
- Kerr, J.F.; Wyllie, A.H.; Currie, A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meier, P.; Finch, A.; Evan, G. Apoptosis in development. Nature 2000, 407, 796–801. [Google Scholar] [CrossRef] [PubMed]
- Fuchs, Y.; Steller, H. Programmed cell death in animal development and disease. Cell 2011, 147, 742–758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thompson, C.B. Apoptosis in the pathogenesis and treatment of disease. Science 1995, 267, 1456–1462. [Google Scholar] [CrossRef] [PubMed]
- Donepudi, M.; Grutter, M.G. Structure and zymogen activation of caspases. Biophys. Chem. 2002, 101–102, 145–153. [Google Scholar] [CrossRef]
- Aram, L.; Yacobi-Sharon, K.; Arama, E. CDPs: Caspase-dependent non-lethal cellular processes. Cell Death Differ. 2017, 24, 1307–1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arama, E.; Baena-Lopez, L.A.; Fearnhead, H.O. Non-lethal message from the Holy Land: The first international conference on nonapoptotic roles of apoptotic proteins. FEBS J. 2021, 288, 2166–2183. [Google Scholar] [CrossRef] [PubMed]
- Feinstein-Rotkopf, Y.; Arama, E. Can’t live without them, can live with them: Roles of caspases during vital cellular processes. Apoptosis 2009, 14, 980–995. [Google Scholar] [CrossRef]
- Fogarty, C.E.; Bergmann, A. Killers creating new life: Caspases drive apoptosis-induced proliferation in tissue repair and disease. Cell Death Differ. 2017, 24, 1390–1400. [Google Scholar] [CrossRef] [Green Version]
- Baena-Lopez, L.A.; Arthurton, L.; Xu, D.C.; Galasso, A. Non-apoptotic Caspase regulation of stem cell properties. Semin. Cell Dev. Biol. 2018, 82, 118–126. [Google Scholar] [CrossRef]
- Kuranaga, E.; Miura, M. Nonapoptotic functions of caspases: Caspases as regulatory molecules for immunity and cell-fate determination. Trends Cell Biol. 2007, 17, 135–144. [Google Scholar] [CrossRef]
- Buttke, T.M.; Sandstrom, P.A. Oxidative stress as a mediator of apoptosis. Immunol. Today 1994, 15, 7–10. [Google Scholar] [CrossRef]
- Lavrik, I.; Golks, A.; Krammer, P.H. Death receptor signaling. J. Cell Sci. 2005, 118, 265–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kristiansen, M.; Ham, J. Programmed cell death during neuronal development: The sympathetic neuron model. Cell Death Differ. 2014, 21, 1025–1035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, X.; Budihardjo, I.; Zou, H.; Slaughter, C.; Wang, X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 1998, 94, 481–490. [Google Scholar] [CrossRef] [Green Version]
- Li, H.; Zhu, H.; Xu, C.J.; Yuan, J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 1998, 94, 491–501. [Google Scholar] [CrossRef] [Green Version]
- Thornberry, N.A.; Lazebnik, Y. Caspases: Enemies within. Science 1998, 281, 1312–1316. [Google Scholar] [CrossRef] [PubMed]
- McComb, S.; Chan, P.K.; Guinot, A.; Hartmannsdottir, H.; Jenni, S.; Dobay, M.P.; Bourquin, J.P.; Bornhauser, B.C. Efficient apoptosis requires feedback amplification of upstream apoptotic signals by effector caspase-3 or -7. Sci. Adv. 2019, 5, eaau9433. [Google Scholar] [CrossRef] [Green Version]
- Sharma, A.; Trivedi, A.K. Regulation of apoptosis by E3 ubiquitin ligases in ubiquitin proteasome system. Cell Biol. Int. 2020, 44, 721–734. [Google Scholar] [CrossRef]
- Salvesen, G.S.; Duckett, C.S. IAP proteins: Blocking the road to death’s door. Nat. Rev. Mol. Cell Biol. 2002, 3, 401–410. [Google Scholar] [CrossRef] [PubMed]
- Deveraux, Q.L.; Reed, J.C. IAP family proteins-suppressors of apoptosis. Genes Dev. 1999, 13, 239–252. [Google Scholar] [CrossRef]
- Deveraux, Q.L.; Leo, E.; Stennicke, H.R.; Welsh, K.; Salvesen, G.S.; Reed, J.C. Cleavage of human inhibitor of apoptosis protein XIAP results in fragments with distinct specificities for caspases. EMBO J. 1999, 18, 5242–5251. [Google Scholar] [CrossRef] [Green Version]
- Suzuki, Y.; Nakabayashi, Y.; Nakata, K.; Reed, J.C.; Takahashi, R. X-linked inhibitor of apoptosis protein (XIAP) inhibits caspase-3 and -7 in distinct modes. J. Biol. Chem. 2001, 276, 27058–27063. [Google Scholar] [CrossRef] [Green Version]
- Eckelman, B.P.; Salvesen, G.S. The human anti-apoptotic proteins cIAP1 and cIAP2 bind but do not inhibit caspases. J. Biol. Chem. 2006, 281, 3254–3260. [Google Scholar] [CrossRef] [Green Version]
- Eckelman, B.P.; Salvesen, G.S.; Scott, F.L. Human inhibitor of apoptosis proteins: Why XIAP is the black sheep of the family. EMBO Rep. 2006, 7, 988–994. [Google Scholar] [CrossRef] [Green Version]
- Gyrd-Hansen, M.; Darding, M.; Miasari, M.; Santoro, M.M.; Zender, L.; Xue, W.; Tenev, T.; da Fonseca, P.C.; Zvelebil, M.; Bujnicki, J.M.; et al. IAPs contain an evolutionarily conserved ubiquitin-binding domain that regulates NF-kappaB as well as cell survival and oncogenesis. Nat. Cell Biol. 2008, 10, 1309–1317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schile, A.J.; Garcia-Fernandez, M.; Steller, H. Regulation of apoptosis by XIAP ubiquitin-ligase activity. Genes Dev. 2008, 22, 2256–2266. [Google Scholar] [CrossRef] [Green Version]
- Rajalingam, K.; Dikic, I. Inhibitors of apoptosis catch ubiquitin. Biochem. J. 2009, 417, e1–e3. [Google Scholar] [CrossRef] [PubMed]
- Vucic, D.; Stennicke, H.R.; Pisabarro, M.T.; Salvesen, G.S.; Dixit, V.M. ML-IAP, a novel inhibitor of apoptosis that is preferentially expressed in human melanomas. Curr. Biol. 2000, 10, 1359–1366. [Google Scholar] [CrossRef] [Green Version]
- Danial, N.N.; Korsmeyer, S.J. Cell death: Critical control points. Cell 2004, 116, 205–219. [Google Scholar] [CrossRef] [Green Version]
- Lowe, S.W.; Lin, A.W. Apoptosis in cancer. Carcinogenesis 2000, 21, 485–495. [Google Scholar] [CrossRef] [Green Version]
- Cotter, T.G. Apoptosis and cancer: The genesis of a research field. Nat. Rev. Cancer 2009, 9, 501–507. [Google Scholar] [CrossRef] [PubMed]
- Abbas, R.; Larisch, S. Targeting XIAP for Promoting Cancer Cell Death-The Story of ARTS and SMAC. Cells 2020, 9, 663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bornstein, B.; Edison, N.; Gottfried, Y.; Lev, T.; Shekhtman, A.; Gonen, H.; Rajalingam, K.; Larisch, S. X-linked Inhibitor of Apoptosis Protein promotes the degradation of its antagonist, the pro-apoptotic ARTS protein. Int. J. Biochem. Cell Biol. 2012, 44, 489–495. [Google Scholar] [CrossRef]
- Bornstein, B.; Gottfried, Y.; Edison, N.; Shekhtman, A.; Lev, T.; Glaser, F.; Larisch, S. ARTS binds to a distinct domain in XIAP-BIR3 and promotes apoptosis by a mechanism that is different from other IAP-antagonists. Apoptosis 2011, 16, 869–881. [Google Scholar] [CrossRef]
- Edison, N.; Curtz, Y.; Paland, N.; Mamriev, D.; Chorubczyk, N.; Haviv-Reingewertz, T.; Kfir, N.; Morgenstern, D.; Kupervaser, M.; Kagan, J.; et al. Degradation of Bcl-2 by XIAP and ARTS Promotes Apoptosis. Cell Rep. 2017, 21, 442–454. [Google Scholar] [CrossRef] [Green Version]
- Gottfried, Y.; Rotem, A.; Lotan, R.; Steller, H.; Larisch, S. The mitochondrial ARTS protein promotes apoptosis through targeting XIAP. EMBO J. 2004, 23, 1627–1635. [Google Scholar] [CrossRef]
- Lotan, R.; Rotem, A.; Gonen, H.; Finberg, J.P.; Kemeny, S.; Steller, H.; Ciechanover, A.; Larisch, S. Regulation of the proapoptotic ARTS protein by ubiquitin-mediated degradation. J. Biol. Chem. 2005, 280, 25802–25810. [Google Scholar] [CrossRef] [Green Version]
- Bader, M.; Steller, H. Regulation of cell death by the ubiquitin-proteasome system. Curr. Opin. Cell Biol. 2009, 21, 878–884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vaux, D.L.; Silke, J. IAPs—The ubiquitin connection. Cell Death Differ. 2005, 12, 1205–1207. [Google Scholar] [CrossRef]
- Vaux, D.L.; Silke, J. IAPs, RINGs and ubiquitylation. Nat. Rev. Mol. Cell Biol. 2005, 6, 287–297. [Google Scholar] [CrossRef] [PubMed]
- Abu Ahmad, Y.; Oknin-Vaisman, A.; Bitman-Lotan, E.; Orian, A. From the Evasion of Degradation to Ubiquitin-Dependent Protein Stabilization. Cells 2021, 10, 2374. [Google Scholar] [CrossRef] [PubMed]
- Ciechanover, A. Intracellular protein degradation: From a vague idea thru the lysosome and the ubiquitin-proteasome system and onto human diseases and drug targeting. Best Pract. Res. Clin. Haematol. 2017, 30, 341–355. [Google Scholar] [CrossRef] [PubMed]
- Collins, G.A.; Goldberg, A.L. The Logic of the 26S Proteasome. Cell 2017, 169, 792–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciechanover, A.; Orian, A.; Schwartz, A.L. Ubiquitin-mediated proteolysis: Biological regulation via destruction. Bioessays 2000, 22, 442–451. [Google Scholar] [CrossRef]
- Hershko, A.; Ciechanover, A.; Rose, I.A. Resolution of the ATP-dependent proteolytic system from reticulocytes: A component that interacts with ATP. Proc. Natl. Acad. Sci. USA 1979, 76, 3107–3110. [Google Scholar] [CrossRef] [Green Version]
- Ciechanover, A.; Heller, H.; Elias, S.; Haas, A.L.; Hershko, A. ATP-dependent conjugation of reticulocyte proteins with the polypeptide required for protein degradation. Proc. Natl. Acad. Sci. USA 1980, 77, 1365–1368. [Google Scholar] [CrossRef] [Green Version]
- Ciechanover, A.; Hod, Y.; Hershko, A. A heat-stable polypeptide component of an ATP-dependent proteolytic system from reticulocytes. 1978. Biochem. Biophys. Res. Commun. 2012, 425, 565–570. [Google Scholar] [CrossRef] [PubMed]
- Mayor, T.; Sharon, M.; Glickman, M.H. Tuning the proteasome to brighten the end of the journey. Am. J. Physiol Cell Physiol 2016, 311, C793–C804. [Google Scholar] [CrossRef] [Green Version]
- Hershko, A.; Ciechanover, A.; Heller, H.; Haas, A.L.; Rose, I.A. Proposed role of ATP in protein breakdown: Conjugation of protein with multiple chains of the polypeptide of ATP-dependent proteolysis. Proc. Natl. Acad. Sci. USA 1980, 77, 1783–1786. [Google Scholar] [CrossRef] [Green Version]
- Broemer, M.; Meier, P. Ubiquitin-mediated regulation of apoptosis. Trends Cell Biol. 2009, 19, 130–140. [Google Scholar] [CrossRef] [PubMed]
- Thompson, S.J.; Loftus, L.T.; Ashley, M.D.; Meller, R. Ubiquitin-proteasome system as a modulator of cell fate. Curr. Opin. Pharmacol. 2008, 8, 90–95. [Google Scholar] [CrossRef] [Green Version]
- Argentini, M.; Barboule, N.; Wasylyk, B. The contribution of the RING finger domain of MDM2 to cell cycle progression. Oncogene 2000, 19, 3849–3857. [Google Scholar] [CrossRef]
- Huang, H.; Joazeiro, C.A.; Bonfoco, E.; Kamada, S.; Leverson, J.D.; Hunter, T. The inhibitor of apoptosis, cIAP2, functions as a ubiquitin-protein ligase and promotes in vitro monoubiquitination of caspases 3 and 7. J. Biol. Chem. 2000, 275, 26661–26664. [Google Scholar] [CrossRef]
- Suzuki, Y.; Nakabayashi, Y.; Takahashi, R. Ubiquitin-protein ligase activity of X-linked inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3 and enhances its anti-apoptotic effect in Fas-induced cell death. Proc. Natl. Acad. Sci. USA 2001, 98, 8662–8667. [Google Scholar] [CrossRef] [Green Version]
- Wilson, R.; Goyal, L.; Ditzel, M.; Zachariou, A.; Baker, D.A.; Agapite, J.; Steller, H.; Meier, P. The DIAP1 RING finger mediates ubiquitination of Dronc and is indispensable for regulating apoptosis. Nat. Cell Biol. 2002, 4, 445–450. [Google Scholar] [CrossRef]
- Haglund, K.; Dikic, I. Ubiquitylation and cell signaling. EMBO J. 2005, 24, 3353–3359. [Google Scholar] [CrossRef] [Green Version]
- Nakayama, K.I.; Nakayama, K. Ubiquitin ligases: Cell-cycle control and cancer. Nat. Rev. Cancer 2006, 6, 369–381. [Google Scholar] [CrossRef] [PubMed]
- Mukhopadhyay, D.; Riezman, H. Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science 2007, 315, 201–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ikeda, F.; Dikic, I. Atypical ubiquitin chains: New molecular signals. ‘Protein Modifications: Beyond the Usual Suspects’ review series. EMBO Rep. 2008, 9, 536–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swatek, K.N.; Komander, D. Ubiquitin modifications. Cell Res. 2016, 26, 399–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miranda, M.; Sorkin, A. Regulation of receptors and transporters by ubiquitination: New insights into surprisingly similar mechanisms. Mol. Interv. 2007, 7, 157–167. [Google Scholar] [CrossRef]
- Livneh, I.; Kravtsova-Ivantsiv, Y.; Braten, O.; Kwon, Y.T.; Ciechanover, A. Monoubiquitination joins polyubiquitination as an esteemed proteasomal targeting signal. Bioessays 2017, 39, 1700027. [Google Scholar] [CrossRef]
- Nakagawa, T.; Nakayama, K. Protein monoubiquitylation: Targets and diverse functions. Genes Cells 2015, 20, 543–562. [Google Scholar] [CrossRef] [Green Version]
- Shabek, N.; Herman-Bachinsky, Y.; Buchsbaum, S.; Lewinson, O.; Haj-Yahya, M.; Hejjaoui, M.; Lashuel, H.A.; Sommer, T.; Brik, A.; Ciechanover, A. The size of the proteasomal substrate determines whether its degradation will be mediated by mono- or polyubiquitylation. Mol. Cell 2012, 48, 87–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shabek, N.; Herman-Bachinsky, Y.; Ciechanover, A. Ubiquitin degradation with its substrate, or as a monomer in a ubiquitination-independent mode, provides clues to proteasome regulation. Proc. Natl. Acad. Sci. USA 2009, 106, 11907–11912. [Google Scholar] [CrossRef] [Green Version]
- Shabek, N.; Iwai, K.; Ciechanover, A. Ubiquitin is degraded by the ubiquitin system as a monomer and as part of its conjugated target. Biochem. Biophys. Res. Commun. 2007, 363, 425–431. [Google Scholar] [CrossRef]
- Ohtake, F.; Saeki, Y.; Ishido, S.; Kanno, J.; Tanaka, K. The K48-K63 Branched Ubiquitin Chain Regulates NF-kappaB Signaling. Mol. Cell 2016, 64, 251–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yau, R.; Rape, M. The increasing complexity of the ubiquitin code. Nat. Cell Biol. 2016, 18, 579–586. [Google Scholar] [CrossRef]
- Jacobson, A.D.; Zhang, N.Y.; Xu, P.; Han, K.J.; Noone, S.; Peng, J.; Liu, C.W. The lysine 48 and lysine 63 ubiquitin conjugates are processed differently by the 26 s proteasome. J. Biol. Chem. 2009, 284, 35485–35494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, W.; Bennett, E.J.; Huttlin, E.L.; Guo, A.; Li, J.; Possemato, A.; Sowa, M.E.; Rad, R.; Rush, J.; Comb, M.J.; et al. Systematic and quantitative assessment of the ubiquitin-modified proteome. Mol. Cell 2011, 44, 325–340. [Google Scholar] [CrossRef] [Green Version]
- Xu, P.; Duong, D.M.; Seyfried, N.T.; Cheng, D.; Xie, Y.; Robert, J.; Rush, J.; Hochstrasser, M.; Finley, D.; Peng, J. Quantitative proteomics reveals the function of unconventional ubiquitin chains in proteasomal degradation. Cell 2009, 137, 133–145. [Google Scholar] [CrossRef] [Green Version]
- Gendron, J.M.; Webb, K.; Yang, B.; Rising, L.; Zuzow, N.; Bennett, E.J. Using the Ubiquitin-modified Proteome to Monitor Distinct and Spatially Restricted Protein Homeostasis Dysfunction. Mol. Cell Proteom. 2016, 15, 2576–2593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nathan, J.A.; Kim, H.T.; Ting, L.; Gygi, S.P.; Goldberg, A.L. Why do cellular proteins linked to K63-polyubiquitin chains not associate with proteasomes? EMBO J. 2013, 32, 552–565. [Google Scholar] [CrossRef]
- Ashwell, J.D. TWEAKing death. J. Cell Biol. 2008, 182, 15–17. [Google Scholar] [CrossRef] [PubMed]
- Komander, D.; Rape, M. The ubiquitin code. Annu. Rev. Biochem. 2012, 81, 203–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Husnjak, K.; Dikic, I. Ubiquitin-binding proteins: Decoders of ubiquitin-mediated cellular functions. Annu. Rev. Biochem. 2012, 81, 291–322. [Google Scholar] [CrossRef]
- Wolf, D.H.; Hilt, W. The proteasome: A proteolytic nanomachine of cell regulation and waste disposal. Biochim. Biophys. Acta 2004, 1695, 19–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pickart, C.M. Mechanisms underlying ubiquitination. Annu. Rev. Biochem. 2001, 70, 503–533. [Google Scholar] [CrossRef] [PubMed]
- Kleiger, G.; Mayor, T. Perilous journey: A tour of the ubiquitin-proteasome system. Trends Cell Biol. 2014, 24, 352–359. [Google Scholar] [CrossRef] [Green Version]
- Zheng, N.; Shabek, N. Ubiquitin Ligases: Structure, Function, and Regulation. Annu. Rev. Biochem. 2017, 86, 129–157. [Google Scholar] [CrossRef] [PubMed]
- David, Y.; Ternette, N.; Edelmann, M.J.; Ziv, T.; Gayer, B.; Sertchook, R.; Dadon, Y.; Kessler, B.M.; Navon, A. E3 ligases determine ubiquitination site and conjugate type by enforcing specificity on E2 enzymes. J. Biol. Chem. 2011, 286, 44104–44115. [Google Scholar] [CrossRef] [Green Version]
- Ardley, H.C.; Robinson, P.A. E3 ubiquitin ligases. Essays Biochem. 2005, 41, 15–30. [Google Scholar] [CrossRef]
- Berndsen, C.E.; Wolberger, C. New insights into ubiquitin E3 ligase mechanism. Nat. Struct. Mol. Biol. 2014, 21, 301–307. [Google Scholar] [CrossRef]
- Morreale, F.E.; Walden, H. Types of Ubiquitin Ligases. Cell 2016, 165, 248-248.e1. [Google Scholar] [CrossRef]
- Sluimer, J.; Distel, B. Regulating the human HECT E3 ligases. Cell Mol. Life Sci. 2018, 75, 3121–3141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weber, J.; Polo, S.; Maspero, E. HECT E3 Ligases: A Tale With Multiple Facets. Front. Physiol. 2019, 10, 370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Wilt, L.H.; Jansen, G.; Assaraf, Y.G.; van Meerloo, J.; Cloos, J.; Schimmer, A.D.; Chan, E.T.; Kirk, C.J.; Peters, G.J.; Kruyt, F.A. Proteasome-based mechanisms of intrinsic and acquired bortezomib resistance in non-small cell lung cancer. Biochem. Pharmacol. 2012, 83, 207–217. [Google Scholar] [CrossRef]
- Franke, N.E.; Kaspers, G.L.; Assaraf, Y.G.; van Meerloo, J.; Niewerth, D.; Kessler, F.L.; Poddighe, P.J.; Kole, J.; Smeets, S.J.; Ylstra, B.; et al. Exocytosis of polyubiquitinated proteins in bortezomib-resistant leukemia cells: A role for MARCKS in acquired resistance to proteasome inhibitors. Oncotarget 2016, 7, 74779–74796. [Google Scholar] [CrossRef] [Green Version]
- Franke, N.E.; Niewerth, D.; Assaraf, Y.G.; van Meerloo, J.; Vojtekova, K.; van Zantwijk, C.H.; Zweegman, S.; Chan, E.T.; Kirk, C.J.; Geerke, D.P.; et al. Impaired bortezomib binding to mutant beta5 subunit of the proteasome is the underlying basis for bortezomib resistance in leukemia cells. Leukemia 2012, 26, 757–768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeon, Y.K.; Kim, C.K.; Koh, J.; Chung, D.H.; Ha, G.H. Pellino-1 confers chemoresistance in lung cancer cells by upregulating cIAP2 through Lys63-mediated polyubiquitination. Oncotarget 2016, 7, 41811–41824. [Google Scholar] [CrossRef] [Green Version]
- Nelson, J.K.; Cook, E.C.; Loregger, A.; Hoeksema, M.A.; Scheij, S.; Kovacevic, I.; Hordijk, P.L.; Ovaa, H.; Zelcer, N. Deubiquitylase Inhibition Reveals Liver X Receptor-independent Transcriptional Regulation of the E3 Ubiquitin Ligase IDOL and Lipoprotein Uptake. J. Biol. Chem. 2016, 291, 4813–4825. [Google Scholar] [CrossRef] [Green Version]
- Niewerth, D.; Jansen, G.; Assaraf, Y.G.; Zweegman, S.; Kaspers, G.J.; Cloos, J. Molecular basis of resistance to proteasome inhibitors in hematological malignancies. Drug Resist. Updates 2015, 18, 18–35. [Google Scholar] [CrossRef]
- Niewerth, D.; Jansen, G.; Riethoff, L.F.; van Meerloo, J.; Kale, A.J.; Moore, B.S.; Assaraf, Y.G.; Anderl, J.L.; Zweegman, S.; Kaspers, G.J.; et al. Antileukemic activity and mechanism of drug resistance to the marine Salinispora tropica proteasome inhibitor salinosporamide A (Marizomib). Mol. Pharmacol. 2014, 86, 12–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Niewerth, D.; van Meerloo, J.; Jansen, G.; Assaraf, Y.G.; Hendrickx, T.C.; Kirk, C.J.; Anderl, J.L.; Zweegman, S.; Kaspers, G.J.; Cloos, J. Anti-leukemic activity and mechanisms underlying resistance to the novel immunoproteasome inhibitor PR-924. Biochem. Pharmacol. 2014, 89, 43–51. [Google Scholar] [CrossRef]
- Oerlemans, R.; Franke, N.E.; Assaraf, Y.G.; Cloos, J.; van Zantwijk, I.; Berkers, C.R.; Scheffer, G.L.; Debipersad, K.; Vojtekova, K.; Lemos, C.; et al. Molecular basis of bortezomib resistance: Proteasome subunit beta5 (PSMB5) gene mutation and overexpression of PSMB5 protein. Blood 2008, 112, 2489–2499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petzold, G.; Fischer, E.S.; Thoma, N.H. Structural basis of lenalidomide-induced CK1alpha degradation by the CRL4(CRBN) ubiquitin ligase. Nature 2016, 532, 127–130. [Google Scholar] [CrossRef]
- Tanaka, N.; Kosaka, T.; Miyazaki, Y.; Mikami, S.; Niwa, N.; Otsuka, Y.; Minamishima, Y.A.; Mizuno, R.; Kikuchi, E.; Miyajima, A.; et al. Acquired platinum resistance involves epithelial to mesenchymal transition through ubiquitin ligase FBXO32 dysregulation. JCI Insight 2016, 1, e83654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Q.; Hou, Y.X.; Langlais, P.; Erickson, P.; Zhu, J.; Shi, C.X.; Luo, M.; Zhu, Y.; Xu, Y.; Mandarino, L.J.; et al. Expression of the cereblon binding protein argonaute 2 plays an important role for multiple myeloma cell growth and survival. BMC Cancer 2016, 16, 297. [Google Scholar] [CrossRef] [Green Version]
- Yoshino, S.; Hara, T.; Nakaoka, H.J.; Kanamori, A.; Murakami, Y.; Seiki, M.; Sakamoto, T. The ERK signaling target RNF126 regulates anoikis resistance in cancer cells by changing the mitochondrial metabolic flux. Cell Discov. 2016, 2, 16019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.D.; Baladandayuthapani, V.; Lin, H.; Mulligan, G.; Li, B.; Esseltine, D.W.; Qi, L.; Xu, J.; Hunziker, W.; Barlogie, B.; et al. Tight Junction Protein 1 Modulates Proteasome Capacity and Proteasome Inhibitor Sensitivity in Multiple Myeloma via EGFR/JAK1/STAT3 Signaling. Cancer Cell 2016, 29, 639–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, B.; Mao, X. The ubiquitin-proteasomal system is critical for multiple myeloma: Implications in drug discovery. Am. J. Blood Res. 2011, 1, 46–56. [Google Scholar] [PubMed]
- Gandhi, A.K.; Kang, J.; Havens, C.G.; Conklin, T.; Ning, Y.; Wu, L.; Ito, T.; Ando, H.; Waldman, M.F.; Thakurta, A.; et al. Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.). Br. J. Haematol. 2014, 164, 811–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, H.; Weng, H.; Dong, B.; Zhao, P.; Zhou, H.; Qu, L. Oridonin Triggers Chaperon-mediated Proteasomal Degradation of BCR-ABL in Leukemia. Sci. Rep. 2017, 7, 41525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, G.; Middleton, R.E.; Sun, H.; Naniong, M.; Ott, C.J.; Mitsiades, C.S.; Wong, K.K.; Bradner, J.E.; Kaelin, W.G., Jr. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science 2014, 343, 305–309. [Google Scholar] [CrossRef] [Green Version]
- Micel, L.N.; Tentler, J.J.; Smith, P.G.; Eckhardt, G.S. Role of ubiquitin ligases and the proteasome in oncogenesis: Novel targets for anticancer therapies. J. Clin. Oncol. 2013, 31, 1231–1238. [Google Scholar] [CrossRef] [Green Version]
- Wu, B.; Chu, X.; Feng, C.; Hou, J.; Fan, H.; Liu, N.; Li, C.; Kong, X.; Ye, X.; Meng, S. Heat shock protein gp96 decreases p53 stability by regulating Mdm2 E3 ligase activity in liver cancer. Cancer Lett. 2015, 359, 325–334. [Google Scholar] [CrossRef]
- Yerlikaya, A.; Yontem, M. The significance of ubiquitin proteasome pathway in cancer development. Recent Pat. Anticancer Drug Discov. 2013, 8, 298–309. [Google Scholar] [CrossRef] [PubMed]
- Lipkowitz, S.; Weissman, A.M. RINGs of good and evil: RING finger ubiquitin ligases at the crossroads of tumour suppression and oncogenesis. Nat. Rev. Cancer 2011, 11, 629–643. [Google Scholar] [CrossRef] [Green Version]
- Ohi, M.D.; Vander Kooi, C.W.; Rosenberg, J.A.; Chazin, W.J.; Gould, K.L. Structural insights into the U-box, a domain associated with multi-ubiquitination. Nat. Struct. Biol. 2003, 10, 250–255. [Google Scholar] [CrossRef]
- Metzger, M.B.; Pruneda, J.N.; Klevit, R.E.; Weissman, A.M. RING-type E3 ligases: Master manipulators of E2 ubiquitin-conjugating enzymes and ubiquitination. Biochim. Biophys. Acta 2014, 1843, 47–60. [Google Scholar] [CrossRef] [Green Version]
- Eldridge, A.G.; O’Brien, T. Therapeutic strategies within the ubiquitin proteasome system. Cell Death Differ. 2010, 17, 4–13. [Google Scholar] [CrossRef] [Green Version]
- Sarikas, A.; Hartmann, T.; Pan, Z.Q. The cullin protein family. Genome Biol. 2011, 12, 220. [Google Scholar] [CrossRef] [Green Version]
- Spratt, D.E.; Walden, H.; Shaw, G.S. RBR E3 ubiquitin ligases: New structures, new insights, new questions. Biochem. J. 2014, 458, 421–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walden, H.; Rittinger, K. RBR ligase-mediated ubiquitin transfer: A tale with many twists and turns. Nat. Struct. Mol. Biol. 2018, 25, 440–445. [Google Scholar] [CrossRef] [PubMed]
- Vasudevan, D.; Ryoo, H.D. Regulation of Cell Death by IAPs and Their Antagonists. Curr. Top. Dev. Biol. 2015, 114, 185–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bergmann, A. The role of ubiquitylation for the control of cell death in Drosophila. Cell Death Differ. 2010, 17, 61–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schwartz, L.M.; Myer, A.; Kosz, L.; Engelstein, M.; Maier, C. Activation of polyubiquitin gene expression during developmentally programmed cell death. Neuron 1990, 5, 411–419. [Google Scholar] [CrossRef]
- Xu, T.; Jiang, X.; Denton, D.; Kumar, S. Ecdysone controlled cell and tissue deletion. Cell Death Differ. 2020, 27, 1–14. [Google Scholar] [CrossRef]
- Lockshin, R.A.; Williams, C.M. Programmed Cell Death--I. Cytology of Degeneration in the Intersegmental Muscles of the Pernyi Silkmoth. J. Insect Physiol. 1965, 11, 123–133. [Google Scholar] [CrossRef]
- Kaelin, W.G., Jr. The p53 gene family. Oncogene 1999, 18, 7701–7705. [Google Scholar] [CrossRef] [Green Version]
- Oren, M. Regulation of the p53 tumor suppressor protein. J. Biol. Chem. 1999, 274, 36031–36034. [Google Scholar] [CrossRef] [Green Version]
- Labrecque, S.; Naor, N.; Thomson, D.; Matlashewski, G. Analysis of the Anti-p53 Antibody Response in Cancer Patients. Cancer Res. 1993, 53, 3468. [Google Scholar]
- Tokino, T.; Nakamura, Y. The role of p53-target genes in human cancer. Crit. Rev. Oncol. Hematol 2000, 33, 1–6. [Google Scholar] [CrossRef]
- Yang, L.; Song, T.; Cheng, Q.; Chen, L.; Chen, J. Mutant p53 Sequestration of the MDM2 Acidic Domain Inhibits E3 Ligase Activity. Mol. Cell Biol. 2019, 39, e00375-18. [Google Scholar] [CrossRef] [Green Version]
- Khoo, K.H.; Verma, C.S.; Lane, D.P. Drugging the p53 pathway: Understanding the route to clinical efficacy. Nat. Rev. Drug Discov. 2014, 13, 217–236. [Google Scholar] [CrossRef]
- Powell, E.; Piwnica-Worms, D.; Piwnica-Worms, H. Contribution of p53 to metastasis. Cancer Discov. 2014, 4, 405–414. [Google Scholar] [CrossRef] [Green Version]
- Joerger, A.C.; Fersht, A.R. The p53 Pathway: Origins, Inactivation in Cancer, and Emerging Therapeutic Approaches. Annu. Rev. Biochem. 2016, 85, 375–404. [Google Scholar] [CrossRef] [PubMed]
- Klimovich, B.; Mutlu, S.; Schneikert, J.; Elmshauser, S.; Klimovich, M.; Nist, A.; Mernberger, M.; Timofeev, O.; Stiewe, T. Loss of p53 function at late stages of tumorigenesis confers ARF-dependent vulnerability to p53 reactivation therapy. Proc. Natl. Acad. Sci. USA 2019, 116, 22288–22293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brooks, C.L.; Gu, W. p53 regulation by ubiquitin. FEBS Lett. 2011, 585, 2803–2809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hock, A.K.; Vousden, K.H. The role of ubiquitin modification in the regulation of p53. Biochim. Biophys. Acta 2014, 1843, 137–149. [Google Scholar] [CrossRef] [Green Version]
- Levine, A.J.; Oren, M. The first 30 years of p53: Growing ever more complex. Nat. Rev. Cancer 2009, 9, 749–758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aylon, Y.; Oren, M. Living with p53, dying of p53. Cell 2007, 130, 597–600. [Google Scholar] [CrossRef] [Green Version]
- Baresova, P.; Musilova, J.; Pitha, P.M.; Lubyova, B. p53 tumor suppressor protein stability and transcriptional activity are targeted by Kaposi’s sarcoma-associated herpesvirus-encoded viral interferon regulatory factor 3. Mol. Cell Biol. 2014, 34, 386–399. [Google Scholar] [CrossRef] [Green Version]
- Giaccia, A.J.; Kastan, M.B. The complexity of p53 modulation: Emerging patterns from divergent signals. Genes Dev. 1998, 12, 2973–2983. [Google Scholar] [CrossRef] [Green Version]
- Maki, C.G.; Howley, P.M. Ubiquitination of p53 and p21 is differentially affected by ionizing and UV radiation. Mol. Cell Biol. 1997, 17, 355–363. [Google Scholar] [CrossRef] [Green Version]
- Price, B.D.; Calderwood, S.K. Increased sequence-specific p53-DNA binding activity after DNA damage is attenuated by phorbol esters. Oncogene 1993, 8, 3055–3062. [Google Scholar]
- Maltzman, W.; Czyzyk, L. UV irradiation stimulates levels of p53 cellular tumor antigen in nontransformed mouse cells. Mol. Cell Biol. 1984, 4, 1689–1694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vogelstein, B.; Lane, D.; Levine, A.J. Surfing the p53 network. Nature 2000, 408, 307–310. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Simpson, E.R.; Brown, K.A. p53: Protection against Tumor Growth beyond Effects on Cell Cycle and Apoptosis. Cancer Res. 2015, 75, 5001–5007. [Google Scholar] [CrossRef] [Green Version]
- Haupt, Y.; Maya, R.; Kazaz, A.; Oren, M. Mdm2 promotes the rapid degradation of p53. Nature 1997, 387, 296–299. [Google Scholar] [CrossRef] [PubMed]
- Kubbutat, M.H.; Jones, S.N.; Vousden, K.H. Regulation of p53 stability by Mdm2. Nature 1997, 387, 299–303. [Google Scholar] [CrossRef] [PubMed]
- Tao, W.; Levine, A.J. Nucleocytoplasmic shuttling of oncoprotein Hdm2 is required for Hdm2-mediated degradation of p53. Proc. Natl. Acad. Sci. USA 1999, 96, 3077–3080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nag, S.; Zhang, X.; Srivenugopal, K.S.; Wang, M.H.; Wang, W.; Zhang, R. Targeting MDM2-p53 interaction for cancer therapy: Are we there yet? Curr. Med. Chem. 2014, 21, 553–574. [Google Scholar] [CrossRef]
- Poyurovsky, M.V.; Katz, C.; Laptenko, O.; Beckerman, R.; Lokshin, M.; Ahn, J.; Byeon, I.J.; Gabizon, R.; Mattia, M.; Zupnick, A.; et al. The C terminus of p53 binds the N-terminal domain of MDM2. Nat. Struct. Mol. Biol. 2010, 17, 982–989. [Google Scholar] [CrossRef]
- Chi, S.W.; Lee, S.H.; Kim, D.H.; Ahn, M.J.; Kim, J.S.; Woo, J.Y.; Torizawa, T.; Kainosho, M.; Han, K.H. Structural details on mdm2-p53 interaction. J. Biol. Chem. 2005, 280, 38795–38802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Montes de Oca Luna, R.; Wagner, D.S.; Lozano, G. Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature 1995, 378, 203–206. [Google Scholar] [CrossRef] [PubMed]
- Barak, Y.; Juven, T.; Haffner, R.; Oren, M. mdm2 expression is induced by wild type p53 activity. EMBO J. 1993, 12, 461–468. [Google Scholar] [CrossRef]
- Perry, M.E.; Piette, J.; Zawadzki, J.A.; Harvey, D.; Levine, A.J. The mdm-2 gene is induced in response to UV light in a p53-dependent manner. Proc. Natl. Acad. Sci. USA 1993, 90, 11623–11627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, X.; Bayle, J.H.; Olson, D.; Levine, A.J. The p53-mdm-2 autoregulatory feedback loop. Genes Dev. 1993, 7, 1126–1132. [Google Scholar] [CrossRef] [Green Version]
- Fang, S.; Jensen, J.P.; Ludwig, R.L.; Vousden, K.H.; Weissman, A.M. Mdm2 is a RING finger-dependent ubiquitin protein ligase for itself and p53. J. Biol. Chem. 2000, 275, 8945–8951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Honda, R.; Yasuda, H. Activity of MDM2, a ubiquitin ligase, toward p53 or itself is dependent on the RING finger domain of the ligase. Oncogene 2000, 19, 1473–1476. [Google Scholar] [CrossRef] [Green Version]
- Poyurovsky, M.V.; Priest, C.; Kentsis, A.; Borden, K.L.; Pan, Z.Q.; Pavletich, N.; Prives, C. The Mdm2 RING domain C-terminus is required for supramolecular assembly and ubiquitin ligase activity. EMBO J. 2007, 26, 90–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Linke, K.; Mace, P.D.; Smith, C.A.; Vaux, D.L.; Silke, J.; Day, C.L. Structure of the MDM2/MDMX RING domain heterodimer reveals dimerization is required for their ubiquitylation in trans. Cell Death Differ. 2008, 15, 841–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharp, D.A.; Kratowicz, S.A.; Sank, M.J.; George, D.L. Stabilization of the MDM2 oncoprotein by interaction with the structurally related MDMX protein. J. Biol. Chem. 1999, 274, 38189–38196. [Google Scholar] [CrossRef] [Green Version]
- Liu, T.; Zhang, H.; Xiong, J.; Yi, S.; Gu, L.; Zhou, M. Inhibition of MDM2 homodimerization by XIAP IRES stabilizes MDM2, influencing cancer cell survival. Mol. Cancer 2015, 14, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stad, R.; Little, N.A.; Xirodimas, D.P.; Frenk, R.; van der Eb, A.J.; Lane, D.P.; Saville, M.K.; Jochemsen, A.G. Mdmx stabilizes p53 and Mdm2 via two distinct mechanisms. EMBO Rep. 2001, 2, 1029–1034. [Google Scholar] [CrossRef] [Green Version]
- de Polo, A.; Luo, Z.; Gerarduzzi, C.; Chen, X.; Little, J.B.; Yuan, Z.M. AXL receptor signalling suppresses p53 in melanoma through stabilization of the MDMX-MDM2 complex. J. Mol. Cell Biol. 2017, 9, 154–165. [Google Scholar] [CrossRef] [Green Version]
- Shvarts, A.; Steegenga, W.T.; Riteco, N.; van Laar, T.; Dekker, P.; Bazuine, M.; van Ham, R.C.; van der Houven van Oordt, W.; Hateboer, G.; van der Eb, A.J.; et al. MDMX: A novel p53-binding protein with some functional properties of MDM2. EMBO J. 1996, 15, 5349–5357. [Google Scholar] [CrossRef]
- Singh, R.K.; Iyappan, S.; Scheffner, M. Hetero-oligomerization with MdmX rescues the ubiquitin/Nedd8 ligase activity of RING finger mutants of Mdm2. J. Biol. Chem. 2007, 282, 10901–10907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Finch, R.A.; Donoviel, D.B.; Potter, D.; Shi, M.; Fan, A.; Freed, D.D.; Wang, C.Y.; Zambrowicz, B.P.; Ramirez-Solis, R.; Sands, A.T.; et al. mdmx is a negative regulator of p53 activity in vivo. Cancer Res. 2002, 62, 3221–3225. [Google Scholar]
- Parant, J.; Chavez-Reyes, A.; Little, N.A.; Yan, W.; Reinke, V.; Jochemsen, A.G.; Lozano, G. Rescue of embryonic lethality in Mdm4-null mice by loss of Trp53 suggests a nonoverlapping pathway with MDM2 to regulate p53. Nat. Genet. 2001, 29, 92–95. [Google Scholar] [CrossRef]
- Migliorini, D.; Lazzerini Denchi, E.; Danovi, D.; Jochemsen, A.; Capillo, M.; Gobbi, A.; Helin, K.; Pelicci, P.G.; Marine, J.C. Mdm4 (Mdmx) regulates p53-induced growth arrest and neuronal cell death during early embryonic mouse development. Mol. Cell Biol. 2002, 22, 5527–5538. [Google Scholar] [CrossRef] [Green Version]
- Gu, L.; Zhang, H.; Liu, T.; Zhou, S.; Du, Y.; Xiong, J.; Yi, S.; Qu, C.K.; Fu, H.; Zhou, M. Discovery of Dual Inhibitors of MDM2 and XIAP for Cancer Treatment. Cancer Cell 2016, 30, 623–636. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.W.; Wang, X.F.; Ni, S.J.; Qin, W.; Zhao, L.Q.; Hua, R.X.; Lu, Y.W.; Li, J.; Dimri, G.P.; Guo, W.J. UBTD1 induces cellular senescence through an UBTD1-Mdm2/p53 positive feedback loop. J. Pathol. 2015, 235, 656–667. [Google Scholar] [CrossRef]
- Leng, R.P.; Lin, Y.; Ma, W.; Wu, H.; Lemmers, B.; Chung, S.; Parant, J.M.; Lozano, G.; Hakem, R.; Benchimol, S. Pirh2, a p53-induced ubiquitin-protein ligase, promotes p53 degradation. Cell 2003, 112, 779–791. [Google Scholar] [CrossRef] [Green Version]
- Daks, A.; Petukhov, A.; Fedorova, O.; Shuvalov, O.; Merkulov, V.; Vasileva, E.; Antonov, A.; Barlev, N.A. E3 ubiquitin ligase Pirh2 enhances tumorigenic properties of human non-small cell lung carcinoma cells. Genes Cancer 2016, 7, 383–393. [Google Scholar] [CrossRef]
- Hakem, A.; Bohgaki, M.; Lemmers, B.; Tai, E.; Salmena, L.; Matysiak-Zablocki, E.; Jung, Y.S.; Karaskova, J.; Kaustov, L.; Duan, S.; et al. Role of Pirh2 in mediating the regulation of p53 and c-Myc. PLoS Genet. 2011, 7, e1002360. [Google Scholar] [CrossRef]
- Bao, Y.; Wu, X.; Yuan, D.; Shi, W.; Shi, J. High Expression of Pirh2 is Associated with Poor Prognosis in Glioma. Cell Mol. NeuroBiol. 2017, 37, 1501–1509. [Google Scholar] [CrossRef] [PubMed]
- Yang, S.; Chen, Y.; Sun, F.; Ni, Q.; Wang, H.; Huang, Y.; Zhang, C.; Liu, K.; Wang, S.; Qiu, J.; et al. Downregulated PIRH2 Can Decrease the Proliferation of Breast Cancer Cells. Arch. Med. Res. 2016, 47, 186–195. [Google Scholar] [CrossRef]
- Hu, L.; Liu, M.; Chen, L.; Chan, T.H.; Wang, J.; Huo, K.K.; Zheng, B.J.; Xie, D.; Guan, X.Y. SCYL1 binding protein 1 promotes the ubiquitin-dependent degradation of Pirh2 and has tumor-suppressive function in the development of hepatocellular carcinoma. Carcinogenesis 2012, 33, 1581–1588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dornan, D.; Wertz, I.; Shimizu, H.; Arnott, D.; Frantz, G.D.; Dowd, P.; O’Rourke, K.; Koeppen, H.; Dixit, V.M. The ubiquitin ligase COP1 is a critical negative regulator of p53. Nature 2004, 429, 86–92. [Google Scholar] [CrossRef]
- Migliorini, D.; Bogaerts, S.; Defever, D.; Vyas, R.; Denecker, G.; Radaelli, E.; Zwolinska, A.; Depaepe, V.; Hochepied, T.; Skarnes, W.C.; et al. Cop1 constitutively regulates c-Jun protein stability and functions as a tumor suppressor in mice. J. Clin. Investig. 2011, 121, 1329–1343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, D.; Kon, N.; Li, M.; Zhang, W.; Qin, J.; Gu, W. ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor. Cell 2005, 121, 1071–1083. [Google Scholar] [CrossRef] [Green Version]
- Cambiaghi, V.; Giuliani, V.; Lombardi, S.; Marinelli, C.; Toffalorio, F.; Pelicci, P.G. TRIM proteins in cancer. Adv. Exp. Med. Biol. 2012, 770, 77–91. [Google Scholar] [CrossRef]
- Wang, C.; Ivanov, A.; Chen, L.; Fredericks, W.J.; Seto, E.; Rauscher, F.J., 3rd; Chen, J. MDM2 interaction with nuclear corepressor KAP1 contributes to p53 inactivation. EMBO J. 2005, 24, 3279–3290. [Google Scholar] [CrossRef] [Green Version]
- Okamoto, K.; Kitabayashi, I.; Taya, Y. KAP1 dictates p53 response induced by chemotherapeutic agents via Mdm2 interaction. Biochem. Biophys. Res. Commun. 2006, 351, 216–222. [Google Scholar] [CrossRef]
- Jiang, J.; Ballinger, C.A.; Wu, Y.; Dai, Q.; Cyr, D.M.; Hohfeld, J.; Patterson, C. CHIP is a U-box-dependent E3 ubiquitin ligase: Identification of Hsc70 as a target for ubiquitylation. J. Biol. Chem. 2001, 276, 42938–42944. [Google Scholar] [CrossRef] [Green Version]
- Xu, Z.; Devlin, K.I.; Ford, M.G.; Nix, J.C.; Qin, J.; Misra, S. Structure and interactions of the helical and U-box domains of CHIP, the C terminus of HSP70 interacting protein. Biochemistry 2006, 45, 4749–4759. [Google Scholar] [CrossRef] [PubMed]
- Maan, M.; Pati, U. CHIP promotes autophagy-mediated degradation of aggregating mutant p53 in hypoxic conditions. FEBS J. 2018, 285, 3197–3214. [Google Scholar] [CrossRef] [Green Version]
- Esser, C.; Scheffner, M.; Hohfeld, J. The chaperone-associated ubiquitin ligase CHIP is able to target p53 for proteasomal degradation. J. Biol. Chem. 2005, 280, 27443–27448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, A.K.; Barton, M.C. Making sense of ubiquitin ligases that regulate p53. Cancer Biol. Ther. 2010, 10, 665–672. [Google Scholar] [CrossRef] [Green Version]
- O’Connor, L.; Harris, A.W.; Strasser, A. CD95 (Fas/APO-1) and p53 signal apoptosis independently in diverse cell types. Cancer Res. 2000, 60, 1217–1220. [Google Scholar] [PubMed]
- Liu, X.; Yue, P.; Khuri, F.R.; Sun, S.Y. p53 upregulates death receptor 4 expression through an intronic p53 binding site. Cancer Res. 2004, 64, 5078–5083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oda, E.; Ohki, R.; Murasawa, H.; Nemoto, J.; Shibue, T.; Yamashita, T.; Tokino, T.; Taniguchi, T.; Tanaka, N. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 2000, 288, 1053–1058. [Google Scholar] [CrossRef]
- Nakano, K.; Vousden, K.H. PUMA, a novel proapoptotic gene, is induced by p53. Mol. Cell 2001, 7, 683–694. [Google Scholar] [CrossRef]
- Fridman, J.S.; Lowe, S.W. Control of apoptosis by p53. Oncogene 2003, 22, 9030–9040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meulmeester, E.; Jochemsen, A.G. p53: A guide to apoptosis. Curr. Cancer Drug Targets 2008, 8, 87–97. [Google Scholar] [CrossRef]
- Hao, Q.; Chen, J.; Liao, J.; Huang, Y.; Larisch, S.; Zeng, S.X.; Lu, H.; Zhou, X. p53 induces ARTS to promote mitochondrial apoptosis. bioRxiv 2020. [Google Scholar] [CrossRef]
- Tait, S.W.; Green, D.R. Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat. Rev. Mol. Cell Biol. 2010, 11, 621–632. [Google Scholar] [CrossRef] [PubMed]
- Chen, J. The Cell-Cycle Arrest and Apoptotic Functions of p53 in Tumor Initiation and Progression. Cold Spring Harb. Perspect. Med. 2016, 6, a026104. [Google Scholar] [CrossRef]
- Edison, N.; Zuri, D.; Maniv, I.; Bornstein, B.; Lev, T.; Gottfried, Y.; Kemeny, S.; Garcia-Fernandez, M.; Kagan, J.; Larisch, S. The IAP-antagonist ARTS initiates caspase activation upstream of cytochrome C and SMAC/Diablo. Cell Death Differ. 2012, 19, 356–368. [Google Scholar] [CrossRef] [Green Version]
- Sun, L.; Shi, L.; Li, W.; Yu, W.; Liang, J.; Zhang, H.; Yang, X.; Wang, Y.; Li, R.; Yao, X.; et al. JFK, a Kelch domain-containing F-box protein, links the SCF complex to p53 regulation. Proc. Natl. Acad. Sci. USA 2009, 106, 10195–10200. [Google Scholar] [CrossRef] [Green Version]
- Nag, A.; Bagchi, S.; Raychaudhuri, P. Cul4A physically associates with MDM2 and participates in the proteolysis of p53. Cancer Res. 2004, 64, 8152–8155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kopanja, D.; Stoyanova, T.; Okur, M.N.; Huang, E.; Bagchi, S.; Raychaudhuri, P. Proliferation defects and genome instability in cells lacking Cul4A. Oncogene 2009, 28, 2456–2465. [Google Scholar] [CrossRef] [Green Version]
- Banks, D.; Wu, M.; Higa, L.A.; Gavrilova, N.; Quan, J.; Ye, T.; Kobayashi, R.; Sun, H.; Zhang, H. L2DTL/CDT2 and PCNA interact with p53 and regulate p53 polyubiquitination and protein stability through MDM2 and CUL4A/DDB1 complexes. Cell Cycle 2006, 5, 1719–1729. [Google Scholar] [CrossRef] [Green Version]
- Querido, E.; Blanchette, P.; Yan, Q.; Kamura, T.; Morrison, M.; Boivin, D.; Kaelin, W.G.; Conaway, R.C.; Conaway, J.W.; Branton, P.E. Degradation of p53 by adenovirus E4orf6 and E1B55K proteins occurs via a novel mechanism involving a Cullin-containing complex. Genes Dev. 2001, 15, 3104–3117. [Google Scholar] [CrossRef] [Green Version]
- Yamasaki, S.; Yagishita, N.; Sasaki, T.; Nakazawa, M.; Kato, Y.; Yamadera, T.; Bae, E.; Toriyama, S.; Ikeda, R.; Zhang, L.; et al. Cytoplasmic destruction of p53 by the endoplasmic reticulum-resident ubiquitin ligase ‘Synoviolin’. EMBO J. 2007, 26, 113–122. [Google Scholar] [CrossRef] [PubMed]
- Rajendra, R.; Malegaonkar, D.; Pungaliya, P.; Marshall, H.; Rasheed, Z.; Brownell, J.; Liu, L.F.; Lutzker, S.; Saleem, A.; Rubin, E.H. Topors functions as an E3 ubiquitin ligase with specific E2 enzymes and ubiquitinates p53. J. Biol. Chem. 2004, 279, 36440–36444. [Google Scholar] [CrossRef] [Green Version]
- Allton, K.; Jain, A.K.; Herz, H.M.; Tsai, W.W.; Jung, S.Y.; Qin, J.; Bergmann, A.; Johnson, R.L.; Barton, M.C. Trim24 targets endogenous p53 for degradation. Proc. Natl. Acad. Sci. USA 2009, 106, 11612–11616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doyle, J.M.; Gao, J.; Wang, J.; Yang, M.; Potts, P.R. MAGE-RING protein complexes comprise a family of E3 ubiquitin ligases. Mol. Cell 2010, 39, 963–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, L.; Huang, N.J.; Chen, C.; Tang, W.; Kornbluth, S. Ubiquitylation of p53 by the APC/C inhibitor Trim39. Proc. Natl. Acad. Sci. USA 2012, 109, 20931–20936. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Ma, C.; Zhou, T.; Liu, Y.; Sun, L.; Yu, Z. TRIM65 negatively regulates p53 through ubiquitination. Biochem. Biophys. Res. Commun. 2016, 473, 278–282. [Google Scholar] [CrossRef]
- Yang, W.; Rozan, L.M.; McDonald, E.R., 3rd; Navaraj, A.; Liu, J.J.; Matthew, E.M.; Wang, W.; Dicker, D.T.; El-Deiry, W.S. CARPs are ubiquitin ligases that promote MDM2-independent p53 and phospho-p53ser20 degradation. J. Biol. Chem. 2007, 282, 3273–3281. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Li, C.F.; Zhang, L.; Wu, C.Y.; Han, L.; Jin, G.; Rezaeian, A.H.; Han, F.; Liu, C.; Xu, C.; et al. TRAF6 Restricts p53 Mitochondrial Translocation, Apoptosis, and Tumor Suppression. Mol. Cell 2016, 64, 803–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Wang, L.; Zhang, S.; Qu, G.; Zhang, D.; Li, S.; Liu, S. Downregulation of ubiquitin E3 ligase TNF receptor-associated factor 7 leads to stabilization of p53 in breast cancer. Oncol. Rep. 2013, 29, 283–287. [Google Scholar] [CrossRef]
- Su, W.J.; Fang, J.S.; Cheng, F.; Liu, C.; Zhou, F.; Zhang, J. RNF2/Ring1b negatively regulates p53 expression in selective cancer cell types to promote tumor development. Proc. Natl. Acad. Sci. USA 2013, 110, 1720–1725. [Google Scholar] [CrossRef] [Green Version]
- Shen, J.; Li, P.; Shao, X.; Yang, Y.; Liu, X.; Feng, M.; Yu, Q.; Hu, R.; Wang, Z. The E3 Ligase RING1 Targets p53 for Degradation and Promotes Cancer Cell Proliferation and Survival. Cancer Res. 2018, 78, 359–371. [Google Scholar] [CrossRef] [Green Version]
- Cory, S.; Huang, D.C.; Adams, J.M. The Bcl-2 family: Roles in cell survival and oncogenesis. Oncogene 2003, 22, 8590–8607. [Google Scholar] [CrossRef] [Green Version]
- Gross, A.; McDonnell, J.M.; Korsmeyer, S.J. BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 1999, 13, 1899–1911. [Google Scholar] [CrossRef] [Green Version]
- Adams, J.M.; Cory, S. The Bcl-2 protein family: Arbiters of cell survival. Science 1998, 281, 1322–1326. [Google Scholar] [CrossRef] [PubMed]
- Youle, R.J.; Strasser, A. The BCL-2 protein family: Opposing activities that mediate cell death. Nat. Rev. Mol. Cell Biol. 2008, 9, 47–59. [Google Scholar] [CrossRef] [PubMed]
- Siddiqui, W.A.; Ahad, A.; Ahsan, H. The mystery of BCL2 family: Bcl-2 proteins and apoptosis: An update. Arch. Toxicol. 2015, 89, 289–317. [Google Scholar] [CrossRef]
- Vaux, D.L.; Cory, S.; Adams, J.M. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature 1988, 335, 440–442. [Google Scholar] [CrossRef]
- Castle, V.P.; Heidelberger, K.P.; Bromberg, J.; Ou, X.; Dole, M.; Nunez, G. Expression of the apoptosis-suppressing protein bcl-2, in neuroblastoma is associated with unfavorable histology and N-myc amplification. Am. J. Pathol. 1993, 143, 1543–1550. [Google Scholar] [PubMed]
- Krajewska, M.; Krajewski, S.; Epstein, J.I.; Shabaik, A.; Sauvageot, J.; Song, K.; Kitada, S.; Reed, J.C. Immunohistochemical analysis of bcl-2, bax, bcl-X, and mcl-1 expression in prostate cancers. Am. J. Pathol. 1996, 148, 1567–1576. [Google Scholar]
- Robertson, L.E.; Plunkett, W.; McConnell, K.; Keating, M.J.; McDonnell, T.J. Bcl-2 expression in chronic lymphocytic leukemia and its correlation with the induction of apoptosis and clinical outcome. Leukemia 1996, 10, 456–459. [Google Scholar] [PubMed]
- Dimmeler, S.; Breitschopf, K.; Haendeler, J.; Zeiher, A.M. Dephosphorylation targets Bcl-2 for ubiquitin-dependent degradation: A link between the apoptosome and the proteasome pathway. J. Exp. Med. 1999, 189, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
- Kassi, E.; Sourlingas, T.G.; Spiliotaki, M.; Papoutsi, Z.; Pratsinis, H.; Aligiannis, N.; Moutsatsou, P. Ursolic acid triggers apoptosis and Bcl-2 downregulation in MCF-7 breast cancer cells. Cancer Investig. 2009, 27, 723–733. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Chanvorachote, P.; Toledo, D.; Stehlik, C.; Mercer, R.R.; Castranova, V.; Rojanasakul, Y. Peroxide is a key mediator of Bcl-2 down-regulation and apoptosis induction by cisplatin in human lung cancer cells. Mol. Pharmacol. 2008, 73, 119–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhong, Q.; Gao, W.; Du, F.; Wang, X. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell 2005, 121, 1085–1095. [Google Scholar] [CrossRef] [Green Version]
- Mojsa, B.; Lassot, I.; Desagher, S. Mcl-1 ubiquitination: Unique regulation of an essential survival protein. Cells 2014, 3, 418–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Willis, S.N.; Chen, L.; Dewson, G.; Wei, A.; Naik, E.; Fletcher, J.I.; Adams, J.M.; Huang, D.C. Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins. Genes Dev. 2005, 19, 1294–1305. [Google Scholar] [CrossRef] [Green Version]
- Warr, M.R.; Acoca, S.; Liu, Z.; Germain, M.; Watson, M.; Blanchette, M.; Wing, S.S.; Shore, G.C. BH3-ligand regulates access of MCL-1 to its E3 ligase. FEBS Lett. 2005, 579, 5603–5608. [Google Scholar] [CrossRef] [Green Version]
- Magiera, M.M.; Mora, S.; Mojsa, B.; Robbins, I.; Lassot, I.; Desagher, S. Trim17-mediated ubiquitination and degradation of Mcl-1 initiate apoptosis in neurons. Cell Death Differ. 2013, 20, 281–292. [Google Scholar] [CrossRef] [Green Version]
- Carroll, R.G.; Hollville, E.; Martin, S.J. Parkin sensitizes toward apoptosis induced by mitochondrial depolarization through promoting degradation of Mcl-1. Cell Rep. 2014, 9, 1538–1553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Inuzuka, H.; Shaik, S.; Onoyama, I.; Gao, D.; Tseng, A.; Maser, R.S.; Zhai, B.; Wan, L.; Gutierrez, A.; Lau, A.W.; et al. SCF(FBW7) regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruction. Nature 2011, 471, 104–109. [Google Scholar] [CrossRef] [PubMed]
- Wertz, I.E.; Kusam, S.; Lam, C.; Okamoto, T.; Sandoval, W.; Anderson, D.J.; Helgason, E.; Ernst, J.A.; Eby, M.; Liu, J.; et al. Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature 2011, 471, 110–114. [Google Scholar] [CrossRef]
- Ding, Q.; He, X.; Hsu, J.M.; Xia, W.; Chen, C.T.; Li, L.Y.; Lee, D.F.; Liu, J.C.; Zhong, Q.; Wang, X.; et al. Degradation of Mcl-1 by beta-TrCP mediates glycogen synthase kinase 3-induced tumor suppression and chemosensitization. Mol. Cell Biol. 2007, 27, 4006–4017. [Google Scholar] [CrossRef] [Green Version]
- Allan, L.A.; Skowyra, A.; Rogers, K.I.; Zeller, D.; Clarke, P.R. Atypical APC/C-dependent degradation of Mcl-1 provides an apoptotic timer during mitotic arrest. EMBO J. 2018, 37, e96831. [Google Scholar] [CrossRef]
- Harley, M.E.; Allan, L.A.; Sanderson, H.S.; Clarke, P.R. Phosphorylation of Mcl-1 by CDK1-cyclin B1 initiates its Cdc20-dependent destruction during mitotic arrest. EMBO J. 2010, 29, 2407–2420. [Google Scholar] [CrossRef] [Green Version]
- Azakir, B.A.; Desrochers, G.; Angers, A. The ubiquitin ligase Itch mediates the antiapoptotic activity of epidermal growth factor by promoting the ubiquitylation and degradation of the truncated C-terminal portion of Bid. FEBS J. 2010, 277, 1319–1330. [Google Scholar] [CrossRef]
- Ghibelli, L.; Diederich, M. multistep and multitask Bax activation. Mitochondria Res. Soc. 2010, 10, 604–613. [Google Scholar] [CrossRef] [Green Version]
- Hsu, Y.T.; Wolter, K.G.; Youle, R.J. Cytosol-to-membrane redistribution of Bax and Bcl-X(L) during apoptosis. Proc. Natl. Acad. Sci. USA 1997, 94, 3668–3672. [Google Scholar] [CrossRef] [Green Version]
- Wolter, K.G.; Hsu, Y.T.; Smith, C.L.; Nechushtan, A.; Xi, X.G.; Youle, R.J. Movement of Bax from the cytosol to mitochondria during apoptosis. J. Cell Biol. 1997, 139, 1281–1292. [Google Scholar] [CrossRef]
- Kuwana, T.; Newmeyer, D.D. Bcl-2-family proteins and the role of mitochondria in apoptosis. Curr. Opin. Cell Biol. 2003, 15, 691–699. [Google Scholar] [CrossRef]
- Li, B.; Dou, Q.P. Bax degradation by the ubiquitin/proteasome-dependent pathway: Involvement in tumor survival and progression. Proc. Natl. Acad. Sci. USA 2000, 97, 3850–3855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, F.T.; Agrawal, S.G.; Gribben, J.G.; Ye, H.; Du, M.Q.; Newland, A.C.; Jia, L. Bortezomib blocks Bax degradation in malignant B cells during treatment with TRAIL. Blood 2008, 111, 2797–2805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, Y.C.; Lee, Y.S.; Tejima, T.; Tanaka, K.; Omura, S.; Heintz, N.H.; Mitsui, Y.; Magae, J. mdm2 and bax, downstream mediators of the p53 response, are degraded by the ubiquitin-proteasome pathway. Cell Growth Differ. 1998, 9, 79–84. [Google Scholar] [PubMed]
- Benard, G.; Neutzner, A.; Peng, G.; Wang, C.; Livak, F.; Youle, R.J.; Karbowski, M. IBRDC2, an IBR-type E3 ubiquitin ligase, is a regulatory factor for Bax and apoptosis activation. EMBO J. 2010, 29, 1458–1471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnson, B.N.; Berger, A.K.; Cortese, G.P.; Lavoie, M.J. The ubiquitin E3 ligase parkin regulates the proapoptotic function of Bax. Proc. Natl. Acad. Sci. USA 2012, 109, 6283–6288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Srinivasula, S.M.; Ashwell, J.D. IAPs: What’s in a name? Mol. Cell 2008, 30, 123–135. [Google Scholar] [CrossRef]
- Schimmer, A.D. Inhibitor of apoptosis proteins: Translating basic knowledge into clinical practice. Cancer Res. 2004, 64, 7183–7190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rothe, M.; Pan, M.G.; Henzel, W.J.; Ayres, T.M.; Goeddel, D.V. The TNFR2-TRAF signaling complex contains two novel proteins related to baculoviral inhibitor of apoptosis proteins. Cell 1995, 83, 1243–1252. [Google Scholar] [CrossRef] [Green Version]
- Shu, H.B.; Takeuchi, M.; Goeddel, D.V. The tumor necrosis factor receptor 2 signal transducers TRAF2 and c-IAP1 are components of the tumor necrosis factor receptor 1 signaling complex. Proc. Natl. Acad. Sci. USA 1996, 93, 13973–13978. [Google Scholar] [CrossRef] [Green Version]
- Varfolomeev, E.; Blankenship, J.W.; Wayson, S.M.; Fedorova, A.V.; Kayagaki, N.; Garg, P.; Zobel, K.; Dynek, J.N.; Elliott, L.O.; Wallweber, H.J.; et al. IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB activation, and TNFalpha-dependent apoptosis. Cell 2007, 131, 669–681. [Google Scholar] [CrossRef] [Green Version]
- Gyrd-Hansen, M.; Meier, P. IAPs: From caspase inhibitors to modulators of NF-kappaB, inflammation and cancer. Nat. Rev. Cancer 2010, 10, 561–574. [Google Scholar] [CrossRef]
- Vallabhapurapu, S.; Matsuzawa, A.; Zhang, W.; Tseng, P.H.; Keats, J.J.; Wang, H.; Vignali, D.A.; Bergsagel, P.L.; Karin, M. Nonredundant and complementary functions of TRAF2 and TRAF3 in a ubiquitination cascade that activates NIK-dependent alternative NF-kappaB signaling. Nat. Immunol. 2008, 9, 1364–1370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bertrand, M.J.; Milutinovic, S.; Dickson, K.M.; Ho, W.C.; Boudreault, A.; Durkin, J.; Gillard, J.W.; Jaquith, J.B.; Morris, S.J.; Barker, P.A. cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol. Cell 2008, 30, 689–700. [Google Scholar] [CrossRef] [PubMed]
- Varfolomeev, E.; Goncharov, T.; Fedorova, A.V.; Dynek, J.N.; Zobel, K.; Deshayes, K.; Fairbrother, W.J.; Vucic, D. c-IAP1 and c-IAP2 are critical mediators of tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB activation. J. Biol. Chem. 2008, 283, 24295–24299. [Google Scholar] [CrossRef] [Green Version]
- Vince, J.E.; Wong, W.W.; Khan, N.; Feltham, R.; Chau, D.; Ahmed, A.U.; Benetatos, C.A.; Chunduru, S.K.; Condon, S.M.; McKinlay, M.; et al. IAP antagonists target cIAP1 to induce TNFalpha-dependent apoptosis. Cell 2007, 131, 682–693. [Google Scholar] [CrossRef] [Green Version]
- Gaither, A.; Porter, D.; Yao, Y.; Borawski, J.; Yang, G.; Donovan, J.; Sage, D.; Slisz, J.; Tran, M.; Straub, C.; et al. A Smac mimetic rescue screen reveals roles for inhibitor of apoptosis proteins in tumor necrosis factor-alpha signaling. Cancer Res. 2007, 67, 11493–11498. [Google Scholar] [CrossRef] [Green Version]
- Bonizzi, G.; Karin, M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004, 25, 280–288. [Google Scholar] [CrossRef] [PubMed]
- Dubrez, L.; Berthelet, J.; Glorian, V. IAP proteins as targets for drug development in oncology. Onco Targets Ther. 2013, 9, 1285–1304. [Google Scholar] [CrossRef] [Green Version]
- Danson, S.; Dean, E.; Dive, C.; Ranson, M. IAPs as a target for anticancer therapy. Curr. Cancer Drug Targets 2007, 7, 785–794. [Google Scholar] [CrossRef]
- Hegde, R.; Srinivasula, S.M.; Zhang, Z.; Wassell, R.; Mukattash, R.; Cilenti, L.; DuBois, G.; Lazebnik, Y.; Zervos, A.S.; Fernandes-Alnemri, T.; et al. Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that disrupts inhibitor of apoptosis protein-caspase interaction. J. Biol. Chem. 2002, 277, 432–438. [Google Scholar] [CrossRef] [Green Version]
- Elhasid, R.; Sahar, D.; Merling, A.; Zivony, Y.; Rotem, A.; Ben-Arush, M.; Izraeli, S.; Bercovich, D.; Larisch, S. Mitochondrial pro-apoptotic ARTS protein is lost in the majority of acute lymphoblastic leukemia patients. Oncogene 2004, 23, 5468–5475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wing, J.P.; Schwartz, L.M.; Nambu, J.R. The RHG motifs of Drosophila Reaper and Grim are important for their distinct cell death-inducing abilities. Mech. Dev. 2001, 102, 193–203. [Google Scholar] [CrossRef]
- Jost, P.J.; Vucic, D. Regulation of Cell Death and Immunity by XIAP. Cold Spring Harb. Perspect. Biol. 2019. [Google Scholar] [CrossRef]
- Steller, H. Regulation of apoptosis in Drosophila. Cell Death Differ. 2008, 15, 1132–1138. [Google Scholar] [CrossRef] [Green Version]
- Kornbluth, S.; White, K. Apoptosis in Drosophila: Neither fish nor fowl (nor man, nor worm). J. Cell Sci. 2005, 118, 1779–1787. [Google Scholar] [CrossRef] [Green Version]
- Du, C.; Fang, M.; Li, Y.; Li, L.; Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000, 102, 33–42. [Google Scholar] [CrossRef] [Green Version]
- Verhagen, A.M.; Ekert, P.G.; Pakusch, M.; Silke, J.; Connolly, L.M.; Reid, G.E.; Moritz, R.L.; Simpson, R.J.; Vaux, D.L. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000, 102, 43–53. [Google Scholar] [CrossRef] [Green Version]
- Yin, W.; Cheepala, S.; Clifford, J.L. Identification of a novel splice variant of X-linked inhibitor of apoptosis-associated factor 1. Biochem. Biophys. Res. Commun. 2006, 339, 1148–1154. [Google Scholar] [CrossRef]
- Larisch, S.; Yi, Y.; Lotan, R.; Kerner, H.; Eimerl, S.; Tony Parks, W.; Gottfried, Y.; Birkey Reffey, S.; de Caestecker, M.P.; Danielpour, D.; et al. A novel mitochondrial septin-like protein, ARTS, mediates apoptosis dependent on its P-loop motif. Nat. Cell Biol. 2000, 2, 915–921. [Google Scholar] [CrossRef] [PubMed]
- van Loo, G.; van Gurp, M.; Depuydt, B.; Srinivasula, S.M.; Rodriguez, I.; Alnemri, E.S.; Gevaert, K.; Vandekerckhove, J.; Declercq, W.; Vandenabeele, P. The serine protease Omi/HtrA2 is released from mitochondria during apoptosis. Omi interacts with caspase-inhibitor XIAP and induces enhanced caspase activity. Cell Death Differ. 2002, 9, 20–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leaman, D.W.; Chawla-Sarkar, M.; Vyas, K.; Reheman, M.; Tamai, K.; Toji, S.; Borden, E.C. Identification of X-linked inhibitor of apoptosis-associated factor-1 as an interferon-stimulated gene that augments TRAIL Apo2L-induced apoptosis. J. Biol. Chem. 2002, 277, 28504–28511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Srinivasula, S.M.; Hegde, R.; Saleh, A.; Datta, P.; Shiozaki, E.; Chai, J.; Lee, R.A.; Robbins, P.D.; Fernandes-Alnemri, T.; Shi, Y.; et al. A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature 2001, 410, 112–116. [Google Scholar] [CrossRef]
- Shi, Y. A conserved tetrapeptide motif: Potentiating apoptosis through IAP-binding. Cell Death Differ. 2002, 9, 93–95. [Google Scholar] [CrossRef]
- Goyal, L.; McCall, K.; Agapite, J.; Hartwieg, E.; Steller, H. Induction of apoptosis by Drosophila reaper, hid and grim through inhibition of IAP function. EMBO J. 2000, 19, 589–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, G.; Chai, J.; Suber, T.L.; Wu, J.W.; Du, C.; Wang, X.; Shi, Y. Structural basis of IAP recognition by Smac/DIABLO. Nature 2000, 408, 1008–1012. [Google Scholar] [CrossRef] [PubMed]
- Grether, M.E.; Abrams, J.M.; Agapite, J.; White, K.; Steller, H. The head involution defective gene of Drosophila melanogaster functions in programmed cell death. Genes Dev. 1995, 9, 1694–1708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, K.; Grether, M.E.; Abrams, J.M.; Young, L.; Farrell, K.; Steller, H. Genetic control of programmed cell death in Drosophila. Science 1994, 264, 677–683. [Google Scholar] [CrossRef]
- Hay, B.A.; Wassarman, D.A.; Rubin, G.M. Drosophila homologs of baculovirus inhibitor of apoptosis proteins function to block cell death. Cell 1995, 83, 1253–1262. [Google Scholar] [CrossRef] [Green Version]
- Wang, S.L.; Hawkins, C.J.; Yoo, S.J.; Muller, H.A.; Hay, B.A. The Drosophila caspase inhibitor DIAP1 is essential for cell survival and is negatively regulated by HID. Cell 1999, 98, 453–463. [Google Scholar] [CrossRef] [Green Version]
- Burri, L.; Strahm, Y.; Hawkins, C.J.; Gentle, I.E.; Puryer, M.A.; Verhagen, A.; Callus, B.; Vaux, D.; Lithgow, T. Mature DIABLO/Smac is produced by the IMP protease complex on the mitochondrial inner membrane. Mol. Biol. Cell 2005, 16, 2926–2933. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez, J.; Lazebnik, Y. Caspase-9 and APAF-1 form an active holoenzyme. Genes Dev. 1999, 13, 3179–3184. [Google Scholar] [CrossRef] [Green Version]
- Adrain, C.; Creagh, E.M.; Martin, S.J. Apoptosis-associated release of Smac/DIABLO from mitochondria requires active caspases and is blocked by Bcl-2. EMBO J. 2001, 20, 6627–6636. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Thomas, R.M.; Suzuki, H.; De Brabander, J.K.; Wang, X.; Harran, P.G. A small molecule Smac mimic potentiates TRAIL- and TNFalpha-mediated cell death. Science 2004, 305, 1471–1474. [Google Scholar] [CrossRef] [Green Version]
- Chai, J.; Du, C.; Wu, J.W.; Kyin, S.; Wang, X.; Shi, Y. Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature 2000, 406, 855–862. [Google Scholar] [CrossRef]
- Yang, Q.H.; Du, C. Smac/DIABLO selectively reduces the levels of c-IAP1 and c-IAP2 but not that of XIAP and livin in HeLa cells. J. Biol. Chem. 2004, 279, 16963–16970. [Google Scholar] [CrossRef] [Green Version]
- Creagh, E.M.; Murphy, B.M.; Duriez, P.J.; Duckett, C.S.; Martin, S.J. Smac/Diablo antagonizes ubiquitin ligase activity of inhibitor of apoptosis proteins. J. Biol. Chem. 2004, 279, 26906–26914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mandel-Gutfreund, Y.; Kosti, I.; Larisch, S. ARTS, the unusual septin: Structural and functional aspects. Biol. Chem. 2011, 392, 783–790. [Google Scholar] [CrossRef]
- Lakhani, S.A.; Masud, A.; Kuida, K.; Porter, G.A., Jr.; Booth, C.J.; Mehal, W.Z.; Inayat, I.; Flavell, R.A. Caspases 3 and 7: Key mediators of mitochondrial events of apoptosis. Science 2006, 311, 847–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Braun, T.; Dar, S.; Vorobiov, D.; Lindenboim, L.; Dascal, N.; Stein, R. Expression of Bcl-x(S) in Xenopus oocytes induces BH3-dependent and caspase-dependent cytochrome c release and apoptosis. Mol. Cancer Res. 2003, 1, 186–194. [Google Scholar] [PubMed]
- Gao, C.F.; Ren, S.; Zhang, L.; Nakajima, T.; Ichinose, S.; Hara, T.; Koike, K.; Tsuchida, N. Caspase-dependent cytosolic release of cytochrome c and membrane translocation of Bax in p53-induced apoptosis. Exp. Cell Res. 2001, 265, 145–151. [Google Scholar] [CrossRef] [PubMed]
- Ho, A.T.; Zacksenhaus, E. Splitting the apoptosome. Cell Cycle 2004, 3, 446–448. [Google Scholar] [CrossRef] [Green Version]
- Shawgo, M.E.; Shelton, S.N.; Robertson, J.D. Caspase-mediated Bak activation and cytochrome c release during intrinsic apoptotic cell death in Jurkat cells. J. Biol. Chem. 2008, 283, 35532–35538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manns, J.; Daubrawa, M.; Driessen, S.; Paasch, F.; Hoffmann, N.; Loffler, A.; Lauber, K.; Dieterle, A.; Alers, S.; Iftner, T.; et al. Triggering of a novel intrinsic apoptosis pathway by the kinase inhibitor staurosporine: Activation of caspase-9 in the absence of Apaf-1. FASEB J. 2011, 25, 3250–3261. [Google Scholar] [CrossRef] [PubMed]
- Morizane, Y.; Honda, R.; Fukami, K.; Yasuda, H. X-linked inhibitor of apoptosis functions as ubiquitin ligase toward mature caspase-9 and cytosolic Smac/DIABLO. J. Biochem. 2005, 137, 125–132. [Google Scholar] [CrossRef]
- Wilkinson, J.C.; Wilkinson, A.S.; Galban, S.; Csomos, R.A.; Duckett, C.S. Apoptosis-inducing factor is a target for ubiquitination through interaction with XIAP. Mol. Cell Biol. 2008, 28, 237–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dumetier, B.; Zadoroznyj, A.; Dubrez, L. IAP-Mediated Protein Ubiquitination in Regulating Cell Signaling. Cells 2020, 9, 1118. [Google Scholar] [CrossRef] [PubMed]
- MacFarlane, M.; Merrison, W.; Bratton, S.B.; Cohen, G.M. Proteasome-mediated degradation of Smac during apoptosis: XIAP promotes Smac ubiquitination in vitro. J. Biol. Chem. 2002, 277, 36611–36616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qin, S.; Yang, C.; Zhang, B.; Li, X.; Sun, X.; Li, G.; Zhang, J.; Xiao, G.; Gao, X.; Huang, G.; et al. XIAP inhibits mature Smac-induced apoptosis by degrading it through ubiquitination in NSCLC. Int. J. Oncol. 2016, 49, 1289–1296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kemeny, S.; Dery, D.; Loboda, Y.; Rovner, M.; Lev, T.; Zuri, D.; Finberg, J.P.M.; Larisch, S. Parkin promotes degradation of the mitochondrial pro-apoptotic ARTS protein. PLoS ONE 2012, 7, e38837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Edison, N.; Reingewertz, T.H.; Gottfried, Y.; Lev, T.; Zuri, D.; Maniv, I.; Carp, M.J.; Shalev, G.; Friedler, A.; Larisch, S. Peptides Mimicking the Unique ARTS-XIAP Binding Site Promote Apoptotic Cell Death in Cultured Cancer Cells. Clin. Cancer Res. 2012, 18, 2569–2578. [Google Scholar] [CrossRef] [Green Version]
- Garrison, J.B.; Correa, R.G.; Gerlic, M.; Yip, K.W.; Krieg, A.; Tamble, C.M.; Shi, R.; Welsh, K.; Duggineni, S.; Huang, Z.; et al. ARTS and Siah collaborate in a pathway for XIAP degradation. Mol. Cell 2011, 41, 107–116. [Google Scholar] [CrossRef] [Green Version]
- Garcia-Fernandez, M.; Kissel, H.; Brown, S.; Gorenc, T.; Schile, A.J.; Rafii, S.; Larisch, S.; Steller, H. Sept4/ARTS is required for stem cell apoptosis and tumor suppression. Genes Dev. 2010, 24, 2282–2293. [Google Scholar] [CrossRef] [Green Version]
- Samuel, T.; Welsh, K.; Lober, T.; Togo, S.H.; Zapata, J.M.; Reed, J.C. Distinct BIR domains of cIAP1 mediate binding to and ubiquitination of tumor necrosis factor receptor-associated factor 2 and second mitochondrial activator of caspases. J. Biol. Chem. 2006, 281, 1080–1090. [Google Scholar] [CrossRef] [Green Version]
- Tenev, T.; Bianchi, K.; Darding, M.; Broemer, M.; Langlais, C.; Wallberg, F.; Zachariou, A.; Lopez, J.; MacFarlane, M.; Cain, K.; et al. The Ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Mol. Cell 2011, 43, 432–448. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Yang, Y.; Ashwell, J.D. TNF-RII and c-IAP1 mediate ubiquitination and degradation of TRAF2. Nature 2002, 416, 345–347. [Google Scholar] [CrossRef] [Green Version]
- Matsuzawa, A.; Tseng, P.H.; Vallabhapurapu, S.; Luo, J.L.; Zhang, W.; Wang, H.; Vignali, D.A.; Gallagher, E.; Karin, M. Essential cytoplasmic translocation of a cytokine receptor-assembled signaling complex. Science 2008, 321, 663–668. [Google Scholar] [CrossRef] [Green Version]
- Tseng, P.H.; Matsuzawa, A.; Zhang, W.; Mino, T.; Vignali, D.A.; Karin, M. Different modes of ubiquitination of the adaptor TRAF3 selectively activate the expression of type I interferons and proinflammatory cytokines. Nat. Immunol. 2010, 11, 70–75. [Google Scholar] [CrossRef] [Green Version]
- Ma, L.; Huang, Y.; Song, Z.; Feng, S.; Tian, X.; Du, W.; Qiu, X.; Heese, K.; Wu, M. Livin promotes Smac/DIABLO degradation by ubiquitin-proteasome pathway. Cell Death Differ. 2006, 13, 2079–2088. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.B.; Kim, S.Y.; Kim, B.M.; Lee, H.; Kim, I.; Yun, J.; Jo, Y.; Oh, T.; Jo, Y.; Chae, H.D.; et al. Identification of a novel anti-apoptotic E3 ubiquitin ligase that ubiquitinates antagonists of inhibitor of apoptosis proteins SMAC, HtrA2, and ARTS. J. Biol. Chem. 2013, 288, 12014–12021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciechanover, A. The ubiquitin-proteasome proteolytic pathway. Cell 1994, 79, 13–21. [Google Scholar] [CrossRef]
- Jana, N.R. Protein homeostasis and aging: Role of ubiquitin protein ligases. Neurochem. Int. 2012, 60, 443–447. [Google Scholar] [CrossRef] [PubMed]
- Jarpe, M.B.; Widmann, C.; Knall, C.; Schlesinger, T.K.; Gibson, S.; Yujiri, T.; Fanger, G.R.; Gelfand, E.W.; Johnson, G.L. Anti-apoptotic versus pro-apoptotic signal transduction: Checkpoints and stop signs along the road to death. Oncogene 1998, 17, 1475–1482. [Google Scholar] [CrossRef] [Green Version]
- Opferman, J.T.; Kothari, A. Anti-apoptotic BCL-2 family members in development. Cell Death Differ. 2018, 25, 37–45. [Google Scholar] [CrossRef]
- Yang, E.; Korsmeyer, S.J. Molecular thanatopsis: A discourse on the BCL2 family and cell death. Blood 1996, 88, 386–401. [Google Scholar] [CrossRef] [Green Version]
- Prehn, J.H.; Bindokas, V.P.; Marcuccilli, C.J.; Krajewski, S.; Reed, J.C.; Miller, R.J. Regulation of neuronal Bcl2 protein expression and calcium homeostasis by transforming growth factor type beta confers wide-ranging protection on rat hippocampal neurons. Proc. Natl. Acad. Sci. USA 1994, 91, 12599–12603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fricker, L.D. Proteasome Inhibitor Drugs. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 457–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gandolfi, S.; Laubach, J.P.; Hideshima, T.; Chauhan, D.; Anderson, K.C.; Richardson, P.G. The proteasome and proteasome inhibitors in multiple myeloma. Cancer Metastasis Rev. 2017, 36, 561–584. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Yang, P.L.; Gray, N.S. Targeting cancer with small molecule kinase inhibitors. Nat. Rev. Cancer 2009, 9, 28–39. [Google Scholar] [CrossRef]
- Huang, X.; Dixit, V.M. Drugging the undruggables: Exploring the ubiquitin system for drug development. Cell Res. 2016, 26, 484–498. [Google Scholar] [CrossRef]
- Narayanan, S.; Cai, C.Y.; Assaraf, Y.G.; Guo, H.Q.; Cui, Q.; Wei, L.; Huang, J.J.; Ashby, C.R., Jr.; Chen, Z.S. Targeting the ubiquitin-proteasome pathway to overcome anti-cancer drug resistance. Drug Resist. Updates 2020, 48, 100663. [Google Scholar] [CrossRef]
- Crew, A.P.; Raina, K.; Dong, H.; Qian, Y.; Wang, J.; Vigil, D.; Serebrenik, Y.V.; Hamman, B.D.; Morgan, A.; Ferraro, C.; et al. Identification and Characterization of Von Hippel-Lindau-Recruiting Proteolysis Targeting Chimeras (PROTACs) of TANK-Binding Kinase 1. J. Med. Chem. 2018, 61, 583–598. [Google Scholar] [CrossRef]
- Kane, R.C.; Bross, P.F.; Farrell, A.T.; Pazdur, R. Velcade: U.S. FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist 2003, 8, 508–513. [Google Scholar] [CrossRef] [PubMed]
- Richardson, P.G.; Mitsiades, C.; Hideshima, T.; Anderson, K.C. Bortezomib: Proteasome inhibition as an effective anticancer therapy. Annu. Rev. Med. 2006, 57, 33–47. [Google Scholar] [CrossRef] [PubMed]
- Goldberg, A.L. Development of proteasome inhibitors as research tools and cancer drugs. J. Cell Biol. 2012, 199, 583–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goldberg, A.L. Bortezomib’s Scientific Origins and Its Tortuous Path to the Clinic. In Bortezomib in the Treatment of Multiple Myeloma; Ghobrial, I.M., Richardson, P.G., Anderson, K.C., Eds.; Springer: Basel, Switzerland, 2011; pp. 1–27. [Google Scholar]
- Reinstein, E.; Ciechanover, A. Narrative review: Protein degradation and human diseases: The ubiquitin connection. Ann. Intern. Med. 2006, 145, 676–684. [Google Scholar] [CrossRef]
- Qin, J.Z.; Ziffra, J.; Stennett, L.; Bodner, B.; Bonish, B.K.; Chaturvedi, V.; Bennett, F.; Pollock, P.M.; Trent, J.M.; Hendrix, M.J.; et al. Proteasome inhibitors trigger NOXA-mediated apoptosis in melanoma and myeloma cells. Cancer Res. 2005, 65, 6282–6293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kortuem, K.M.; Stewart, A.K. Carfilzomib. Blood 2013, 121, 893–897. [Google Scholar] [CrossRef]
- Kuhn, D.J.; Chen, Q.; Voorhees, P.M.; Strader, J.S.; Shenk, K.D.; Sun, C.M.; Demo, S.D.; Bennett, M.K.; van Leeuwen, F.W.; Chanan-Khan, A.A.; et al. Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. Blood 2007, 110, 3281–3290. [Google Scholar] [CrossRef] [Green Version]
- Parlati, F.; Lee, S.J.; Aujay, M.; Suzuki, E.; Levitsky, K.; Lorens, J.B.; Micklem, D.R.; Ruurs, P.; Sylvain, C.; Lu, Y.; et al. Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome. Blood 2009, 114, 3439–3447. [Google Scholar] [CrossRef] [Green Version]
- Al-Salama, Z.T.; Garnock-Jones, K.P.; Scott, L.J. Ixazomib: A Review in Relapsed and/or Refractory Multiple Myeloma. Target. Oncol. 2017, 12, 535–542. [Google Scholar] [CrossRef]
- Manasanch, E.E.; Orlowski, R.Z. Proteasome inhibitors in cancer therapy. Nat. Rev. Clin. Oncol. 2017, 14, 417–433. [Google Scholar] [CrossRef]
- Chauhan, D.; Tian, Z.; Zhou, B.; Kuhn, D.; Orlowski, R.; Raje, N.; Richardson, P.; Anderson, K.C. In vitro and in vivo selective antitumor activity of a novel orally bioavailable proteasome inhibitor MLN9708 against multiple myeloma cells. Clin. Cancer Res 2011, 17, 5311–5321. [Google Scholar] [CrossRef] [Green Version]
- Piva, R.; Ruggeri, B.; Williams, M.; Costa, G.; Tamagno, I.; Ferrero, D.; Giai, V.; Coscia, M.; Peola, S.; Massaia, M.; et al. CEP-18770: A novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib. Blood 2008, 111, 2765–2775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vogl, D.T.; Martin, T.G.; Vij, R.; Hari, P.; Mikhael, J.R.; Siegel, D.; Wu, K.L.; Delforge, M.; Gasparetto, C. Phase I/II study of the novel proteasome inhibitor delanzomib (CEP-18770) for relapsed and refractory multiple myeloma. Leuk Lymphoma 2017, 58, 1872–1879. [Google Scholar] [CrossRef] [PubMed]
- Chauhan, D.; Catley, L.; Li, G.; Podar, K.; Hideshima, T.; Velankar, M.; Mitsiades, C.; Mitsiades, N.; Yasui, H.; Letai, A.; et al. A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib. Cancer Cell 2005, 8, 407–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shah, J.; Usmani, S.; Stadtmauer, E.A.; Rifkin, R.M.; Berenson, J.R.; Berdeja, J.G.; Lyons, R.M.; Klippel, Z.; Chang, Y.L.; Niesvizky, R. Oprozomib, pomalidomide, and Dexamethasone in Patients With Relapsed and/or Refractory Multiple Myeloma. Clin. Lymphoma Myeloma Leuk 2019, 19, 570-578.e1. [Google Scholar] [CrossRef] [PubMed]
- Zhu, H.; Wang, T.; Xin, Z.; Zhan, Y.; Gu, G.; Li, X.; Wang, X.; Yang, S.; Liu, C. An oral second-generation proteasome inhibitor oprozomib significantly inhibits lung cancer in a p53 independent manner in vitro. Acta Biochim. Biophys. Sin. 2019, 51, 1034–1040. [Google Scholar] [CrossRef] [PubMed]
- Potts, B.C.; Albitar, M.X.; Anderson, K.C.; Baritaki, S.; Berkers, C.; Bonavida, B.; Chandra, J.; Chauhan, D.; Cusack, J.C., Jr.; Fenical, W.; et al. Marizomib, a proteasome inhibitor for all seasons: Preclinical profile and a framework for clinical trials. Curr. Cancer Drug Targets 2011, 11, 254–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, Z.; Liu, T.; Zheng, J.; Hu, J. Clarifying the molecular mechanism associated with carfilzomib resistance in human multiple myeloma using microarray gene expression profile and genetic interaction network. Onco Targets Ther. 2017, 10, 1327–1334. [Google Scholar] [CrossRef] [Green Version]
- Chene, P. Inhibiting the p53-MDM2 interaction: An important target for cancer therapy. Nat. Rev. Cancer 2003, 3, 102–109. [Google Scholar] [CrossRef]
- Quesnel, B.; Preudhomme, C.; Oscier, D.; Lepelley, P.; Collyn-d’Hooghe, M.; Facon, T.; Zandecki, M.; Fenaux, P. Over-expression of the MDM2 gene is found in some cases of haematological malignancies. Br. J. Haematol. 1994, 88, 415–418. [Google Scholar] [CrossRef] [PubMed]
- Vassilev, L.T.; Vu, B.T.; Graves, B.; Carvajal, D.; Podlaski, F.; Filipovic, Z.; Kong, N.; Kammlott, U.; Lukacs, C.; Klein, C.; et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004, 303, 844–848. [Google Scholar] [CrossRef] [Green Version]
- Ding, Q.; Zhang, Z.; Liu, J.J.; Jiang, N.; Zhang, J.; Ross, T.M.; Chu, X.J.; Bartkovitz, D.; Podlaski, F.; Janson, C.; et al. Discovery of RG7388, a potent and selective p53-MDM2 inhibitor in clinical development. J. Med. Chem. 2013, 56, 5979–5983. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Chu, X.J.; Liu, J.J.; Ding, Q.; Zhang, J.; Bartkovitz, D.; Jiang, N.; Karnachi, P.; So, S.S.; Tovar, C.; et al. Discovery of Potent and Orally Active p53-MDM2 Inhibitors RO5353 and RO2468 for Potential Clinical Development. ACS Med. Chem. Lett. 2014, 5, 124–127. [Google Scholar] [CrossRef] [Green Version]
- Vu, B.; Wovkulich, P.; Pizzolato, G.; Lovey, A.; Ding, Q.; Jiang, N.; Liu, J.J.; Zhao, C.; Glenn, K.; Wen, Y.; et al. Discovery of RG7112: A Small-Molecule MDM2 Inhibitor in Clinical Development. ACS Med. Chem. Lett. 2013, 4, 466–469. [Google Scholar] [CrossRef] [Green Version]
- Ray-Coquard, I.; Blay, J.Y.; Italiano, A.; Le Cesne, A.; Penel, N.; Zhi, J.; Heil, F.; Rueger, R.; Graves, B.; Ding, M.; et al. Effect of the MDM2 antagonist RG7112 on the P53 pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: An exploratory proof-of-mechanism study. Lancet Oncol. 2012, 13, 1133–1140. [Google Scholar] [CrossRef]
- Fang, Y.; Liao, G.; Yu, B. Small-molecule MDM2/X inhibitors and PROTAC degraders for cancer therapy: Advances and perspectives. Acta Pharm. Sin. B 2020, 10, 1253–1278. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, D.; Liao, W.; Zeng, S.X.; Lu, H. Reviving the guardian of the genome: Small molecule activators of p53. Pharmacol. Ther. 2017, 178, 92–108. [Google Scholar] [CrossRef]
- Ladds, M.; Lain, S. Small molecule activators of the p53 response. J. Mol. Cell Biol. 2019, 11, 245–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Konopleva, M.; Martinelli, G.; Daver, N.; Papayannidis, C.; Wei, A.; Higgins, B.; Ott, M.; Mascarenhas, J.; Andreeff, M. MDM2 inhibition: An important step forward in cancer therapy. Leukemia 2020, 34, 2858–2874. [Google Scholar] [CrossRef]
- Fischer, K.; Al-Sawaf, O.; Fink, A.M.; Dixon, M.; Bahlo, J.; Warburton, S.; Kipps, T.J.; Weinkove, R.; Robinson, S.; Seiler, T.; et al. Venetoclax and obinutuzumab in chronic lymphocytic leukemia. Blood 2017, 129, 2702–2705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mihalyova, J.; Jelinek, T.; Growkova, K.; Hrdinka, M.; Simicek, M.; Hajek, R. Venetoclax: A new wave in hematooncology. Exp. Hematol 2018, 61, 10–25. [Google Scholar] [CrossRef] [PubMed]
- Pollyea, D.A.; Stevens, B.M.; Jones, C.L.; Winters, A.; Pei, S.; Minhajuddin, M.; D’Alessandro, A.; Culp-Hill, R.; Riemondy, K.A.; Gillen, A.E.; et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat. Med. 2018, 24, 1859–1866. [Google Scholar] [CrossRef] [PubMed]
- Rogers, K.A.; Huang, Y.; Ruppert, A.S.; Awan, F.T.; Heerema, N.A.; Hoffman, C.; Lozanski, G.; Maddocks, K.J.; Moran, M.E.; Reid, M.A.; et al. Phase 1b study of obinutuzumab, ibrutinib, and venetoclax in relapsed and refractory chronic lymphocytic leukemia. Blood 2018, 132, 1568–1572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stilgenbauer, S.; Eichhorst, B.; Schetelig, J.; Coutre, S.; Seymour, J.F.; Munir, T.; Puvvada, S.D.; Wendtner, C.-M.; Roberts, A.W.; Jurczak, W.; et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: A multicentre, open-label, phase 2 study. Lancet Oncol. 2016, 17, 768–778. [Google Scholar] [CrossRef]
- Shahar, N.; Larisch, S. Inhibiting the inhibitors: Targeting anti-apoptotic proteins in cancer and therapy resistance. Drug Resist. Updates 2020, 52, 100712. [Google Scholar] [CrossRef] [PubMed]
- Crews, C.M. Targeting the undruggable proteome: The small molecules of my dreams. Chem. Biol. 2010, 17, 551–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lai, A.C.; Crews, C.M. Induced protein degradation: An emerging drug discovery paradigm. Nat. Rev. Drug Discov. 2017, 16, 101–114. [Google Scholar] [CrossRef] [Green Version]
- Overington, J.P.; Al-Lazikani, B.; Hopkins, A.L. How many drug targets are there? Nat. Rev. Drug Discov. 2006, 5, 993–996. [Google Scholar] [CrossRef]
- Sakamoto, K.M.; Kim, K.B.; Kumagai, A.; Mercurio, F.; Crews, C.M.; Deshaies, R.J. Protacs: Chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. USA 2001, 98, 8554–8559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, X.; Gao, H.; Yang, Y.; He, M.; Wu, Y.; Song, Y.; Tong, Y.; Rao, Y. PROTACs: Great opportunities for academia and industry. Signal. Transduct. Target. Ther. 2019, 4, 64. [Google Scholar] [CrossRef] [Green Version]
- Sun, B.; Fiskus, W.; Qian, Y.; Rajapakshe, K.; Raina, K.; Coleman, K.G.; Crew, A.P.; Shen, A.; Saenz, D.T.; Mill, C.P.; et al. BET protein proteolysis targeting chimera (PROTAC) exerts potent lethal activity against mantle cell lymphoma cells. Leukemia 2018, 32, 343–352. [Google Scholar] [CrossRef]
- He, Y.; Koch, R.; Budamagunta, V.; Zhang, P.; Zhang, X.; Khan, S.; Thummuri, D.; Ortiz, Y.T.; Zhang, X.; Lv, D.; et al. DT2216-a Bcl-xL-specific degrader is highly active against Bcl-xL-dependent T cell lymphomas. J. Hematol. Oncol. 2020, 13, 95. [Google Scholar] [CrossRef]
- Khan, S.; Zhang, X.; Lv, D.; Zhang, Q.; He, Y.; Zhang, P.; Liu, X.; Thummuri, D.; Yuan, Y.; Wiegand, J.S.; et al. A selective BCL-XL PROTAC degrader achieves safe and potent antitumor activity. Nat. Med. 2019, 25, 1938–1947. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Liu, X.; Zhou, D.; Zheng, G. Targeting anti-apoptotic BCL-2 family proteins for cancer treatment. Future Med. Chem. 2020, 12, 563–565. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; He, N.; Guo, Z.; Niu, C.; Song, T.; Guo, Y.; Cao, K.; Wang, A.; Zhu, J.; Zhang, X.; et al. Proteolysis Targeting Chimeras for the Selective Degradation of Mcl-1/Bcl-2 Derived from Nonselective Target Binding Ligands. J. Med. Chem. 2019, 62, 8152–8163. [Google Scholar] [CrossRef] [PubMed]
- Papatzimas, J.W.; Gorobets, E.; Maity, R.; Muniyat, M.I.; MacCallum, J.L.; Neri, P.; Bahlis, N.J.; Derksen, D.J. From Inhibition to Degradation: Targeting the Antiapoptotic Protein Myeloid Cell Leukemia 1 (MCL1). J. Med. Chem. 2019, 62, 5522–5540. [Google Scholar] [CrossRef]
- Yang, Y.; Fang, S.; Jensen, J.P.; Weissman, A.M.; Ashwell, J.D. Ubiquitin protein ligase activity of IAPs and their degradation in proteasomes in response to apoptotic stimuli. Science 2000, 288, 874–877. [Google Scholar] [CrossRef] [PubMed]
- Esposito, I.; Kleeff, J.; Abiatari, I.; Shi, X.; Giese, N.; Bergmann, F.; Roth, W.; Friess, H.; Schirmacher, P. Overexpression of cellular inhibitor of apoptosis protein 2 is an early event in the progression of pancreatic cancer. J. Clin. Pathol. 2007, 60, 885–895. [Google Scholar] [CrossRef] [Green Version]
- Che, X.; Yang, D.; Zong, H.; Wang, J.; Li, X.; Chen, F.; Chen, X.; Song, X. Nuclear cIAP1 overexpression is a tumor stage- and grade-independent predictor of poor prognosis in human bladder cancer patients. Urol Oncol. 2012, 30, 450–456. [Google Scholar] [CrossRef] [PubMed]
- Oost, T.K.; Sun, C.; Armstrong, R.C.; Al-Assaad, A.S.; Betz, S.F.; Deckwerth, T.L.; Ding, H.; Elmore, S.W.; Meadows, R.P.; Olejniczak, E.T.; et al. Discovery of potent antagonists of the antiapoptotic protein XIAP for the treatment of cancer. J. Med. Chem. 2004, 47, 4417–4426. [Google Scholar] [CrossRef] [PubMed]
- Bergmann, A.; Yang, A.Y.; Srivastava, M. Regulators of IAP function: Coming to grips with the grim reaper. Curr. Opin. Cell Biol. 2003, 15, 717–724. [Google Scholar] [CrossRef]
- Sun, H.; Nikolovska-Coleska, Z.; Yang, C.Y.; Qian, D.; Lu, J.; Qiu, S.; Bai, L.; Peng, Y.; Cai, Q.; Wang, S. Design of small-molecule peptidic and nonpeptidic Smac mimetics. Acc. Chem. Res. 2008, 41, 1264–1277. [Google Scholar] [CrossRef] [Green Version]
- Sun, H.; Nikolovska-Coleska, Z.; Yang, C.Y.; Xu, L.; Liu, M.; Tomita, Y.; Pan, H.; Yoshioka, Y.; Krajewski, K.; Roller, P.P.; et al. Structure-based design of potent, conformationally constrained Smac mimetics. J. Am. Chem. Soc. 2004, 126, 16686–16687. [Google Scholar] [CrossRef] [PubMed]
- Corti, A.; Milani, M.; Lecis, D.; Seneci, P.; de Rosa, M.; Mastrangelo, E.; Cossu, F. Structure-based design and molecular profiling of Smac-mimetics selective for cellular IAPs. FEBS J. 2018, 285, 3286–3298. [Google Scholar] [CrossRef] [Green Version]
- Welsh, K.; Milutinovic, S.; Ardecky, R.J.; Gonzalez-Lopez, M.; Ganji, S.R.; Teriete, P.; Finlay, D.; Riedl, S.; Matsuzawa, S.; Pinilla, C.; et al. Characterization of Potent SMAC Mimetics that Sensitize Cancer Cells to TNF Family-Induced Apoptosis. PLoS ONE 2016, 11, e0161952. [Google Scholar] [CrossRef]
- Liu, Z.; Sun, C.; Olejniczak, E.T.; Meadows, R.P.; Betz, S.F.; Oost, T.; Herrmann, J.; Wu, J.C.; Fesik, S.W. Structural basis for binding of Smac/DIABLO to the XIAP BIR3 domain. Nature 2000, 408, 1004–1008. [Google Scholar] [CrossRef] [PubMed]
- Sharma, S.K.; Straub, C.; Zawel, L. Development of Peptidomimetics Targeting IAPs. Int. J. Pept. Res. Ther. 2006, 12, 21–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zobel, K.; Wang, L.; Varfolomeev, E.; Franklin, M.C.; Elliott, L.O.; Wallweber, H.J.; Okawa, D.C.; Flygare, J.A.; Vucic, D.; Fairbrother, W.J.; et al. Design, synthesis, and biological activity of a potent Smac mimetic that sensitizes cancer cells to apoptosis by antagonizing IAPs. ACS Chem. Biol. 2006, 1, 525–533. [Google Scholar] [CrossRef]
- Darding, M.; Feltham, R.; Tenev, T.; Bianchi, K.; Benetatos, C.; Silke, J.; Meier, P. Molecular determinants of Smac mimetic induced degradation of cIAP1 and cIAP2. Cell Death Differ. 2011, 18, 1376–1386. [Google Scholar] [CrossRef] [PubMed]
- He, S.; Wang, L.; Miao, L.; Wang, T.; Du, F.; Zhao, L.; Wang, X. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 2009, 137, 1100–1111. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Du, F.; Wang, X. TNF-alpha induces two distinct caspase-8 activation pathways. Cell 2008, 133, 693–703. [Google Scholar] [CrossRef] [Green Version]
- Barnhart, B.C.; Peter, M.E. The TNF receptor 1: A split personality complex. Cell 2003, 114, 148–150. [Google Scholar] [CrossRef] [Green Version]
- Schutze, S.; Tchikov, V.; Schneider-Brachert, W. Regulation of TNFR1 and CD95 signalling by receptor compartmentalization. Nat. Rev. Mol. Cell Biol. 2008, 9, 655–662. [Google Scholar] [CrossRef] [PubMed]
- Petersen, S.L.; Wang, L.; Yalcin-Chin, A.; Li, L.; Peyton, M.; Minna, J.; Harran, P.; Wang, X. Autocrine TNFalpha signaling renders human cancer cells susceptible to Smac-mimetic-induced apoptosis. Cancer Cell 2007, 12, 445–456. [Google Scholar] [CrossRef] [Green Version]
- Darding, M.; Meier, P. IAPs: Guardians of RIPK1. Cell Death Differ. 2012, 19, 58–66. [Google Scholar] [CrossRef] [Green Version]
- Derakhshan, A.; Chen, Z.; Van Waes, C. Therapeutic Small Molecules Target Inhibitor of Apoptosis Proteins in Cancers with Deregulation of Extrinsic and Intrinsic Cell Death Pathways. Clin. Cancer Res. 2017, 23, 1379–1387. [Google Scholar] [CrossRef] [Green Version]
- Collart, M.A.; Baeuerle, P.; Vassalli, P. Regulation of tumor necrosis factor alpha transcription in macrophages: Involvement of four kappa B-like motifs and of constitutive and inducible forms of NF-kappa B. Mol. Cell Biol. 1990, 10, 1498–1506. [Google Scholar] [CrossRef] [Green Version]
- Kondylis, V.; Kumari, S.; Vlantis, K.; Pasparakis, M. The interplay of IKK, NF-kappaB and RIPK1 signaling in the regulation of cell death, tissue homeostasis and inflammation. Immunol. Rev. 2017, 277, 113–127. [Google Scholar] [CrossRef]
- Condon, S.M.; Mitsuuchi, Y.; Deng, Y.; LaPorte, M.G.; Rippin, S.R.; Haimowitz, T.; Alexander, M.D.; Kumar, P.T.; Hendi, M.S.; Lee, Y.H.; et al. Birinapant, a smac-mimetic with improved tolerability for the treatment of solid tumors and hematological malignancies. J. Med. Chem. 2014, 57, 3666–3677. [Google Scholar] [CrossRef]
- Amaravadi, R.K.; Schilder, R.J.; Martin, L.P.; Levin, M.; Graham, M.A.; Weng, D.E.; Adjei, A.A. A Phase I Study of the SMAC-Mimetic Birinapant in Adults with Refractory Solid Tumors or Lymphoma. Mol. Cancer Ther. 2015, 14, 2569–2575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benetatos, C.A.; Mitsuuchi, Y.; Burns, J.M.; Neiman, E.M.; Condon, S.M.; Yu, G.; Seipel, M.E.; Kapoor, G.S.; Laporte, M.G.; Rippin, S.R.; et al. Birinapant (TL32711), a bivalent SMAC mimetic, targets TRAF2-associated cIAPs, abrogates TNF-induced NF-kappaB activation, and is active in patient-derived xenograft models. Mol. Cancer Ther. 2014, 13, 867–879. [Google Scholar] [CrossRef] [Green Version]
- Finlay, D.; Teriete, P.; Vamos, M.; Cosford, N.D.P.; Vuori, K. Inducing death in tumor cells: Roles of the inhibitor of apoptosis proteins. F1000Research 2017, 6, 587. [Google Scholar] [CrossRef]
- Maas, C.; Tromp, J.M.; van Laar, J.; Thijssen, R.; Elias, J.A.; Malara, A.; Krippner-Heidenreich, A.; Silke, J.; van Oers, M.H.; Eldering, E. CLL cells are resistant to smac mimetics because of an inability to form a ripoptosome complex. Cell Death Dis. 2013, 4, e782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petersen, S.L.; Peyton, M.; Minna, J.D.; Wang, X. Overcoming cancer cell resistance to Smac mimetic induced apoptosis by modulating cIAP-2 expression. Proc. Natl. Acad. Sci. USA 2010, 107, 11936–11941. [Google Scholar] [CrossRef] [Green Version]
- Shekhar, T.M.; Burvenich, I.J.G.; Harris, M.A.; Rigopoulos, A.; Zanker, D.; Spurling, A.; Parker, B.S.; Walkley, C.R.; Scott, A.M.; Hawkins, C.J. Smac mimetics LCL161 and GDC-0152 inhibit osteosarcoma growth and metastasis in mice. BMC Cancer 2019, 19, 924. [Google Scholar] [CrossRef]
- Chen, Z.; Chen, J.; Liu, H.; Dong, W.; Huang, X.; Yang, D.; Hou, J.; Zhang, X. The SMAC Mimetic APG-1387 Sensitizes Immune-Mediated Cell Apoptosis in Hepatocellular Carcinoma. Front. Pharmacol. 2018, 9, 1298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scheurer, M.J.J.; Seher, A.; Steinacker, V.; Linz, C.; Hartmann, S.; Kubler, A.C.; Muller-Richter, U.D.A.; Brands, R.C. Targeting inhibitors of apoptosis in oral squamous cell carcinoma in vitro. J. Craniomaxillofac. Surg. 2019, 47, 1589–1599. [Google Scholar] [CrossRef] [PubMed]
- Mamriev, D.; Abbas, R.; Klingler, F.M.; Kagan, J.; Kfir, N.; Donald, A.; Weidenfeld, K.; Sheppard, D.W.; Barkan, D.; Larisch, S. A small-molecule ARTS mimetic promotes apoptosis through degradation of both XIAP and Bcl-2. Cell Death Dis. 2020, 11, 483. [Google Scholar] [CrossRef]
- Naito, M.; Ohoka, N.; Shibata, N. SNIPERs-Hijacking IAP activity to induce protein degradation. Drug Discov. Today Technol 2019, 31, 35–42. [Google Scholar] [CrossRef] [PubMed]
- Ohoka, N.; Ujikawa, O.; Shimokawa, K.; Sameshima, T.; Shibata, N.; Hattori, T.; Nara, H.; Cho, N.; Naito, M. Different Degradation Mechanisms of Inhibitor of Apoptosis Proteins (IAPs) by the Specific and Nongenetic IAP-Dependent Protein Eraser (SNIPER). Chem. Pharm. Bull. 2019, 67, 203–209. [Google Scholar] [CrossRef] [Green Version]
- Ishikawa, M.; Tomoshige, S.; Demizu, Y.; Naito, M. Selective Degradation of Target Proteins by Chimeric Small-Molecular Drugs, PROTACs and SNIPERs. Pharmaceuticals 2020, 13, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, Z.; Ji, Y.; Yu, Y.; Liang, D. Specific non-genetic IAP-based protein erasers (SNIPERs) as a potential therapeutic strategy. Eur. J. Med. Chem. 2021, 216, 113247. [Google Scholar] [CrossRef] [PubMed]
- Itoh, Y.; Ishikawa, M.; Naito, M.; Hashimoto, Y. Protein knockdown using methyl bestatin-ligand hybrid molecules: Design and synthesis of inducers of ubiquitination-mediated degradation of cellular retinoic acid-binding proteins. J. Am. Chem. Soc. 2010, 132, 5820–5826. [Google Scholar] [CrossRef]
- Ohoka, N.; Okuhira, K.; Ito, M.; Nagai, K.; Shibata, N.; Hattori, T.; Ujikawa, O.; Shimokawa, K.; Sano, O.; Koyama, R.; et al. In Vivo Knockdown of Pathogenic Proteins via Specific and Nongenetic Inhibitor of Apoptosis Protein (IAP)-dependent Protein Erasers (SNIPERs). J Biol. Chem. 2017, 292, 4556–4570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dueber, E.C.; Schoeffler, A.J.; Lingel, A.; Elliott, J.M.; Fedorova, A.V.; Giannetti, A.M.; Zobel, K.; Maurer, B.; Varfolomeev, E.; Wu, P.; et al. Antagonists induce a conformational change in cIAP1 that promotes autoubiquitination. Science 2011, 334, 376–380. [Google Scholar] [CrossRef] [PubMed]
- Edmondson, S.D.; Yang, B.; Fallan, C. Proteolysis targeting chimeras (PROTACs) in ‘beyond rule-of-five’ chemical space: Recent progress and future challenges. Bioorg. Med. Chem. Lett. 2019, 29, 1555–1564. [Google Scholar] [CrossRef]
- Pop, C.; Timmer, J.; Sperandio, S.; Salvesen, G.S. The apoptosome activates caspase-9 by dimerization. Mol. Cell 2006, 22, 269–275. [Google Scholar] [CrossRef] [PubMed]
- Hideshima, T.; Bradner, J.E.; Wong, J.; Chauhan, D.; Richardson, P.; Schreiber, S.L.; Anderson, K.C. Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proc. Natl. Acad. Sci. USA 2005, 102, 8567–8572. [Google Scholar] [CrossRef] [Green Version]
- Cenci, S.; Oliva, L.; Cerruti, F.; Milan, E.; Bianchi, G.; Raule, M.; Mezghrani, A.; Pasqualetto, E.; Sitia, R.; Cascio, P. Pivotal Advance: Protein synthesis modulates responsiveness of differentiating and malignant plasma cells to proteasome inhibitors. J. Leukoc Biol. 2012, 92, 921–931. [Google Scholar] [CrossRef]
- Fuchs, D.; Berges, C.; Opelz, G.; Daniel, V.; Naujokat, C. Increased expression and altered subunit composition of proteasomes induced by continuous proteasome inhibition establish apoptosis resistance and hyperproliferation of Burkitt lymphoma cells. J. Cell Biochem. 2008, 103, 270–283. [Google Scholar] [CrossRef]
- Leung-Hagesteijn, C.; Erdmann, N.; Cheung, G.; Keats, J.J.; Stewart, A.K.; Reece, D.E.; Chung, K.C.; Tiedemann, R.E. Xbp1s-Negative Tumor B Cells and Pre-Plasmablasts Mediate Therapeutic Proteasome Inhibitor Resistance in Multiple Myeloma. Cancer Cell 2015, 28, 541–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reimold, A.M.; Iwakoshi, N.N.; Manis, J.; Vallabhajosyula, P.; Szomolanyi-Tsuda, E.; Gravallese, E.M.; Friend, D.; Grusby, M.J.; Alt, F.; Glimcher, L.H. Plasma cell differentiation requires the transcription factor XBP-1. Nature 2001, 412, 300–307. [Google Scholar] [CrossRef]
- Perez-Galan, P.; Mora-Jensen, H.; Weniger, M.A.; Shaffer, A.L., 3rd; Rizzatti, E.G.; Chapman, C.M.; Mo, C.C.; Stennett, L.S.; Rader, C.; Liu, P.; et al. Bortezomib resistance in mantle cell lymphoma is associated with plasmacytic differentiation. Blood 2011, 117, 542–552. [Google Scholar] [CrossRef] [Green Version]
- Mani, A.; Gelmann, E.P. The ubiquitin-proteasome pathway and its role in cancer. J. Clin. Oncol. 2005, 23, 4776–4789. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Duan, C.; Zhang, C. E3 Ubiquitin Ligase in Anticancer Drugdsla Resistance: Recent Advances and Future Potential. Front. Pharmacol. 2021, 12, 645864. [Google Scholar] [CrossRef] [PubMed]
- Um, H.D. Bcl-2 family proteins as regulators of cancer cell invasion and metastasis: A review focusing on mitochondrial respiration and reactive oxygen species. Oncotarget 2016, 7, 5193–5203. [Google Scholar] [CrossRef] [Green Version]
- Tu, H.; Costa, M. XIAP’s Profile in Human Cancer. Biomolecules 2020, 10, 1493. [Google Scholar] [CrossRef]
- Ku, B.; Liang, C.; Jung, J.U.; Oh, B.H. Evidence that inhibition of BAX activation by BCL-2 involves its tight and preferential interaction with the BH3 domain of BAX. Cell Res. 2011, 21, 627–641. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Abbas, R.; Larisch, S. Killing by Degradation: Regulation of Apoptosis by the Ubiquitin-Proteasome-System. Cells 2021, 10, 3465. https://doi.org/10.3390/cells10123465
Abbas R, Larisch S. Killing by Degradation: Regulation of Apoptosis by the Ubiquitin-Proteasome-System. Cells. 2021; 10(12):3465. https://doi.org/10.3390/cells10123465
Chicago/Turabian StyleAbbas, Ruqaia, and Sarit Larisch. 2021. "Killing by Degradation: Regulation of Apoptosis by the Ubiquitin-Proteasome-System" Cells 10, no. 12: 3465. https://doi.org/10.3390/cells10123465
APA StyleAbbas, R., & Larisch, S. (2021). Killing by Degradation: Regulation of Apoptosis by the Ubiquitin-Proteasome-System. Cells, 10(12), 3465. https://doi.org/10.3390/cells10123465