Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-26T18:20:05.662Z Has data issue: false hasContentIssue false

24 - 14-3-3 proteins in cancer

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Alexandra K. Gardino
Affiliation:
David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
Michael B. Yaffe
Affiliation:
David H. Koch Institute for Integrative Cancer Research and Department of Biological Engineering,Massachusetts Institute of Technology, Cambridge, MA, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

14-3-3 proteins constitute a family of acidic α-helical cup-shaped dimers that are ubiquitously expressed in all eukaryotic cells. Well over 200 proteins have been identified as 14-3-3-binding ligands, and many of these proteins have established roles in cell-cycle control, DNA damage responses, growth-factor receptor signaling, regulation of gene expression, metabolism, and apoptosis (1–5; Figure 24.1). Early work that hinted at a connection between protein phosphorylation and 14-3-3 binding came from work on tryptophan hydroxylase (6), an enzyme involved in neurotransmitter biosynthesis, and Raf, the upstream activator of the classical MAP kinase pathway (7). Work from the Shaw lab (8), building on detailed phosphorylation studies of Raf by Debbie Morrison's group, revealed that 14-3-3 proteins specifically bound to phosphoserine-containing sequences. That work, together with oriented peptide library screening on all mammalian 14-3-3 proteins by Yaffe and colleagues (9), led to the identification of two optimal phosphoserine/threonine-containing motifs – RSx[pS/pT]xP (mode-1) and Rxxx[pS/pT]xP (mode-2) – that are recognized by all 14-3-3 isotypes (9). Pro in the pS/pT+2 position is not absolutely required (9–11), and an additional motif (mode-3) has been found in proteins where the pS/pT residue is the final or next to the last residue at the C-terminus (12). Although there are clear examples of proteins and peptides that differ from these motifs, or that do not require phosphorylation at all for binding, most 14-3-3 ligands use phosphorylated sequences that closely resemble the optimal 14-3-3 consensus motifs for binding. This ability of 14-3-3 proteins to bind to large numbers of ligands in a phospho-regulated manner provides a mechanism for cytoplasmic Ser/Thr kinases, including kinases that are misregulated in cancer, such as mTor, AKT, and PKC family members, to broadly exert control over many key signaling events required for tumor cell survival.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 293 - 304
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Fu, H, Subramanian, RR, Masters, SC. 14-3-3 proteins: structure, function, and regulation. Annual Review of Pharmacology and Toxicology 2000;40:617–47.CrossRef
Jin, J, Smith, FD, Stark, C, et al. Proteomic, functional, and domain-based analysis of in vivo 14-3-3 binding proteins involved in cytoskeletal regulation and cellular organization. Current Biology 2004;14:1436–50.CrossRef
Meek, SE, Lane, WS, Piwnica-Worms, H. Comprehensive proteomic analysis of interphase and mitotic 14-3-3-binding proteins. Journal of Biological Chemistry 2004;279:32 046–54.CrossRefGoogle ScholarPubMed
Pozuelo Rubio, M, Geraghty, KM, Wong, BH, et al. 14-3-3-affinity purification of over 200 human phosphoproteins reveals new links to regulation of cellular metabolism, proliferation and trafficking. Biochemical Journal 2004;379:395–408.CrossRef
van Heusden, GP. 14-3-3 proteins: regulators of numerous eukaryotic proteins. IUBMB Life 2005;57:623–9.CrossRef
Furukawa, Y, Ikuta, N, Omata, S, et al. Demonstration of the phosphorylation-dependent interaction of tryptophan hydroxylase with the 14-3-3 protein. Biochemical and Biophysical Research Communications 1993;194:144–9.CrossRef
Michaud, NR, Fabian, JR, Mathes, KD, Morrison, DK. 14-3-3 is not essential for Raf-1 function: identification of Raf-1 proteins that are biologically activated in a 14-3-3- and Ras-independent manner. Molecular and Cellular Biology 1995;15:3390–7.CrossRef
Muslin, AJ, Tanner, JW, Allen, PM, Shaw, AS. Interaction of 14-3-3 with signaling proteins is mediated by the recognition of phosphoserine. Cell 1996;84:889–97.CrossRef
Yaffe, MB, Rittinger, K, Volinia, S, et al. The structural basis for 14–3–3: phosphopeptide binding specificity. Cell 1997;91:961–71.CrossRef
Rittinger, K, Budman, J, Xu, J, et al. Structural analysis of 14-3-3 phosphopeptide complexes identifies a dual role for the nuclear export signal of 14-3-3 in ligand binding. Molecular Cell 1999;4:153–66.CrossRef
Johnson, C, Crowther, S, Stafford, MJ, et al. Bioinformatic and experimental survey of 14–3–3-binding sites. Biochemical Journal; 2010:69–78.
Coblitz, B, Shikano, S, Wu, M, et al. C-terminal recognition by 14-3-3 proteins for surface expression of membrane receptors. Journal of Biological Chemistry 2005;280:36 263–72.CrossRefGoogle ScholarPubMed
Wang, W, Shakes, DC. Molecular evolution of the 14-3-3 protein family. Journal of Molecular Evolution 1996;43:384–98.CrossRefGoogle ScholarPubMed
Rosenquist, M, Alsterfjord, M, Larsson, C, Sommarin, M. Data mining the Arabidopsis genome reveals fifteen 14-3-3 genes: expression is demonstrated for two out of five novel genes. Plant Physiology 2001;127:142–9.CrossRef
Vincenz, C, Dixit, VM. 14-3-3 proteins associate with A20 in an isoform-specific manner and function both as chaperone and adapter molecules. Journal of Biological Chemistry 1996;271:20 029–34.CrossRefGoogle Scholar
Mils, V, Baldin, V, Goubin, F, et al. Specific interaction between 14-3-3 isoforms and the human CDC25B phosphatase. Oncogene 2000;19:1257–65.CrossRef
Dalal, SN, Yaffe, MB, DeCaprio, JA. 14-3-3 family members act coordinately to regulate mitotic progression. Cell Cycle 2004;3:672–7.
Jagemann, LR, Perez-Rivas, LG, Ruiz, EJ, et al. The functional interaction of 14-3-3 proteins with the ERK1/2 scaffold KSR1 occurs in an isoform-specific manner. Journal of Biological Chemistry 2008;283:17 450–62.CrossRefGoogle Scholar
Hermeking, H. The 14-3-3 cancer connection. Nature Reviews Cancer 2003;3:931–43.CrossRef
Aitken, A. Functional specificity in 14-3-3 isoform interactions through dimer formation and phosphorylation: chromosome location of mammalian isoforms and variants. Plant Molecular Biology 2002;50:993–1010.CrossRef
Wilker, EW, Grant, RA, Artim, SC, Yaffe, MB. A structural basis for 14–3–3sigma functional specificity. Journal of Biological Chemistry 2005;280:18 891–8.CrossRefGoogle ScholarPubMed
Chaudhri, M, Scarabel, M, Aitken, A. Mammalian and yeast 14-3-3 isoforms form distinct patterns of dimers in vivo. Biochemical and Biophysical Research Communications 2003;300:679–85.CrossRef
Benzinger, A, Muster, N, Koch, HB, Yates, JR, Hermeking, H. Targeted proteomic analysis of 14-3-3 sigma, a p53 effector commonly silenced in cancer. Molecular Cell Proteomics 2005;4:785–95.CrossRef
Gardino, AK, Smerdon, SJ, Yaffe, MB. Structural determinants of 14-3-3 binding specificities and regulation of subcellular localization of 14–3–3-ligand complexes: a comparison of the X-ray crystal structures of all human 14-3-3 isoforms. Seminars in Cancer Biology 2006;16:173–82.CrossRef
Rosenquist, M, Sehnke, P, Ferl, RJ, Sommarin, M, Larsson, C. Evolution of the 14-3-3 protein family: does the large number of isoforms in multicellular organisms reflect functional specificity?Journal of Molecular Evolution 2000;51:446–58.CrossRefGoogle ScholarPubMed
Yang, X, Lee, WH, Sobott, F, et al. Structural basis for protein-protein interactions in the 14-3-3 protein family. Proceedings of the National Academy of Sciences USA 2006;103:17 237–42.
Xiao, B, Smerdon, SJ, Jones, DH, et al. Structure of a 14-3-3 protein and implications for coordination of multiple signalling pathways. Nature 1995;376:188–91.CrossRef
Liu, D, Bienkowska, J, Petosa, C, et al. Crystal structure of the zeta isoform of the 14-3-3 protein. Nature 1995;376:191–4.CrossRef
Braselmann, S, McCormick, F. Bcr and Raf form a complex in vivo via 14-3-3 proteins. EMBO Journal 1995;14:4839–48.
Van Der Hoeven, PC, Van Der Wal, JC, Ruurs, P, Van Dijk, MC, Van Blitterswijk, J. 14-3-3 isotypes facilitate coupling of protein kinase C-zeta to Raf-1: negative regulation by 14-3-3 phosphorylation. Biochemical Journal 2000;345 Pt 2:297–306.
Morrison, DK, Heidecker, G, Rapp, UR, Copeland, TD. Identification of the major phosphorylation sites of the Raf-1 kinase. Journal of Biological Chemistry 1993;268:17 309–16.Google ScholarPubMed
Zha, J, Harada, H, Yang, E, Jockel, J, Korsmeyer, SJ. Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L). Cell 1996;87:619–28.CrossRef
Brunet, A, Bonni, A, Zigmond, MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 1999;96:857–68.CrossRef
Yaffe, MB. How do 14-3-3 proteins work? Gatekeeper phosphorylation and the molecular anvil hypothesis. FEBS Letters 2002;513:53–7.CrossRef
Aitken, A, Howell, S, Jones, D, Madrazo, J, Patel, Y. 14-3-3 alpha and delta are the phosphorylated forms of raf-activating 14-3-3 beta and zeta. In vivo stoichiometric phosphorylation in brain at a Ser-Pro-Glu-Lys MOTIF. Journal of Biological Chemistry 1995;270:5706–9.CrossRefGoogle Scholar
Dubois, T, Rommel, C, Howell, S, et al. 14-3-3 is phosphorylated by casein kinase I on residue 233. Phosphorylation at this site in vivo regulates Raf/14-3-3 interaction. Journal of Biological Chemistry 1997;272:28 882–8.CrossRefGoogle ScholarPubMed
Megidish, T, Cooper, J, Zhang, L, Fu, H, Hakomori, S. A novel sphingosine-dependent protein kinase (SDK1) specifically phosphorylates certain isoforms of 14-3-3 protein. Journal of Biological Chemistry 1998;273:21 834–45.CrossRefGoogle ScholarPubMed
Powell, DW, Rane, MJ, Joughin, BA, et al. Proteomic identification of 14–3–3zeta as a mitogen-activated protein kinase-activated protein kinase 2 substrate: role in dimer formation and ligand binding. Molecular and Cellular Biology 2003;23:5376–87.CrossRef
Sunayama, J, Tsuruta, F, Masuyama, N, Gotoh, Y. JNK antagonizes Akt-mediated survival signals by phosphorylating 14-3-3. Journal of Cell Biology 2005;170:295–304.CrossRefGoogle ScholarPubMed
Tsuruta, F, Sunayama, J, Mori, Y, et al. JNK promotes Bax translocation to mitochondria through phosphorylation of 14-3-3 proteins. EMBO Journal 2004;23:1889–99.CrossRef
Lalle, M, Salzano, AM, Crescenzi, M, Pozio, E. The Giardia duodenalis 14-3-3 protein is post-translationally modified by phosphorylation and polyglycylation of the C-terminal tail. Journal of Biological Chemistry 2006;281:5137–48.CrossRefGoogle ScholarPubMed
Choudhary, C, Kumar, C, Gnad, F, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 2009;325:834–40.CrossRef
Roy, S, McPherson, RA, Apolloni, A, et al. 14-3-3 facilitates Ras-dependent Raf-1 activation in vitro and in vivo. Molecular and Cellular Biology 1998;18:3947–55.CrossRef
Tzivion, G, Luo, Z, Avruch, J. A dimeric 14-3-3 protein is an essential cofactor for Raf kinase activity. Nature 1998;394:88–92.CrossRef
Obsil, T, Ghirlando, R, Klein, DC, Ganguly, S, Dyda, F. Crystal structure of the 14–3–3zeta:serotonin N-acetyltransferase complex. a role for scaffolding in enzyme regulation. Cell 2001;105:257–67.
Dumaz, N, Marais, R. Protein kinase A blocks Raf-1 activity by stimulating 14-3-3 binding and blocking Raf-1 interaction with Ras. Journal of Biological Chemistry 2003;278:29 819–23.CrossRefGoogle ScholarPubMed
Hsu, SY, Kaipia, A, Zhu, L, Hsueh, AJ. Interference of BAD (Bcl-xL/Bcl-2-associated death promoter)-induced apoptosis in mammalian cells by 14-3-3 isoforms and P11. Molecular Endocrinology 1997;11:1858–67.
Zhou, GL, Yamamoto, T, Ozoe, F, et al. Identification of a 14-3-3 protein from Lentinus edodes that interacts with CAP (adenylyl cyclase-associated protein), and conservation of this interaction in fission yeast. Bioscience, Biotechnology and Biochemistry 2000;64:149–59.CrossRef
Tan, Y, Demeter, MR, Ruan, H, Comb, MJ. BAD Ser-155 phosphorylation regulates BAD/Bcl-XL interaction and cell survival. Journal of Biological Chemistry 2000;275:25 865–9.CrossRefGoogle ScholarPubMed
Lizcano, JM, Morrice, N, Cohen, P. Regulation of BAD by cAMP-dependent protein kinase is mediated via phosphorylation of a novel site, Ser155. Biochemical Journal 2000;349:547–57.CrossRef
Datta, SR, Katsov, A, Hu, L, et al. 14-3-3 proteins and survival kinases cooperate to inactivate BAD by BH3 domain phosphorylation. Molecular Cell 2000;6:41–51.CrossRef
Cahill, CM, Tzivion, G, Nasrin, N, et al. Phosphatidylinositol 3-kinase signaling inhibits DAF-16 DNA binding and function via 14–3–3-dependent and 14–3–3-independent pathways. Journal of Biological Chemistry 2001;276:13 402–10.CrossRefGoogle ScholarPubMed
Muslin, AJ, Xing, H. 14-3-3 proteins: regulation of subcellular localization by molecular interference. Cellular Signaling 2000;12:703–9.CrossRef
Brunet, A, Kanai, F, Stehn, J, et al. 14-3-3 transits to the nucleus and participates in dynamic nucleocytoplasmic transport. Journal of Cell Biology 2002;156:817–28.CrossRefGoogle ScholarPubMed
Rena, G, Prescott, AR, Guo, S, Cohen, P, Unterman, TG. Roles of the forkhead in rhabdomyosarcoma (FKHR) phosphorylation sites in regulating 14-3-3 binding, transactivation and nuclear targetting. Biochemical Journal 2001;354:605–12.CrossRef
McKinsey, TA, Zhang, CL, Olson, EN. Identification of a signal-responsive nuclear export sequence in class II histone deacetylases. Molecular and Cellular Biology 2001;21:6312–21.CrossRef
Kumagai, A, Dunphy, WG. Binding of 14-3-3 proteins and nuclear export control the intracellular localization of the mitotic inducer Cdc25. Genes and Development 1999;13:1067–72.CrossRef
Graves, PR, Lovly, CM, Uy, GL, Piwnica-Worms, H. Localization of human Cdc25C is regulated both by nuclear export and 14-3-3 protein binding. Oncogene 2001;20:1839–51.CrossRef
Faul, C, Huttelmaier, S, Oh, J, et al. Promotion of importin alpha-mediated nuclear import by the phosphorylation-dependent binding of cargo protein to 14–3–3. Journal of Cell Biology 2005;169:415–24.CrossRefGoogle ScholarPubMed
LeBron, C, Chen, L, Gilkes, DM, Chen, J. Regulation of MDMX nuclear import and degradation by Chk2 and 14–3–3. EMBO Journal 2006;25:1196–206.CrossRef
Kim, HH, Abdelmohsen, K, Lal, A, et al. Nuclear HuR accumulation through phosphorylation by Cdk1. Genes and Development 2008;22:1804–15.CrossRef
O’Kelly, I, Butler, MH, Zilberberg, N, Goldstein, SA. Forward transport. 14-3-3 binding overcomes retention in endoplasmic reticulum by dibasic signals. Cell 2002;111:577–88.
Shikano, S, Coblitz, B, Wu, M, Li, M. 14-3-3 proteins: regulation of endoplasmic reticulum localization and surface expression of membrane proteins. Trends in Cell Biology 2006;16:370–5.CrossRef
Forrest, A, Gabrielli, B. Cdc25B activity is regulated by 14–3–3. Oncogene 2001;20:4393–401.CrossRef
Kumagai, A, Yakowec, PS, Dunphy, WG. 14-3-3 proteins act as negative regulators of the mitotic inducer Cdc25 in Xenopus egg extracts. Molecular Biology of the Cell 1998;9:345–54.CrossRef
Peng, CY, Graves, PR, Thoma, RS, et al. Mitotic and G2 checkpoint control: regulation of 14-3-3 protein binding by phosphorylation of Cdc25C on serine-216. Science 1997;277:1501–5.CrossRef
Rothblum-Oviatt, CJ, Ryan, CE, Piwnica-Worms, H. 14-3-3 binding regulates catalytic activity of human Wee1 kinase. Cell Growth and Differentiation 2001;12:581–9.
Wang, B, Liu, K, Lin, HY, et al. 14–3–3Tau regulates ubiquitin-independent proteasomal degradation of p21, a novel mechanism of p21 downregulation in breast cancer. Molecular and Cellular Biology 2010;30:1508–27.CrossRef
Hong, HY, Jeon, WK, Bae, EJ, et al. 14-3-3 sigma and 14-3-3 zeta plays an opposite role in cell growth inhibition mediated by transforming growth factor-beta 1. Molecules and Cells 2010;29:305–9.CrossRef
Rajagopalan, S, Jaulent, AM, Wells, M, Veprintsev, DB, Fersht, AR. 14-3-3 activation of DNA binding of p53 by enhancing its association into tetramers. Nucleic Acids Research 2008;36:5983–91.CrossRef
Waterman, MJ, Stavridi, ES, Waterman, JL, Halazonetis, TD. ATM-dependent activation of p53 involves dephosphorylation and association with 14-3-3 proteins. Nature Genetics 1998;19:175–8.CrossRef
Rajagopalan, S, Sade, RS, Townsley, FM, Fersht, AR. Mechanistic differences in the transcriptional activation of p53 by 14-3-3 isoforms. Nucleic Acids Research 2010;38:893–906.CrossRef
Yang, HY, Wen, YY, Chen, CH, Lozano, G, Lee, MH. 14-3-3 sigma positively regulates p53 and suppresses tumor growth. Molecular and Cellular Biology 2003;23:7096–107.CrossRef
Yang, HY, Wen, YY, Lin, YI, et al. Roles for negative cell regulator 14–3–3sigma in control of MDM2 activities. Oncogene 2007;26:7355–62.CrossRef
Morrison, DK. The 14-3-3 proteins: integrators of diverse signaling cues that impact cell fate and cancer development. Trends in Cell Biology 2009;19:16–23.CrossRef
Tzivion, G, Gupta, VS, Kaplun, L, Balan, V. 14-3-3 proteins as potential oncogenes. Seminars in Cancer Biology 2006;16:203–13.CrossRef
Wilker, E, Yaffe, MB. 14-3-3 Proteins–a focus on cancer and human disease. Journal of Molecular and Cellular Cardiology 2004;37:633–42.CrossRefGoogle ScholarPubMed
Chan, TA, Hermeking, H, Lengauer, C, Kinzler, KW, Vogelstein, B. 14–3–3Sigma is required to prevent mitotic catastrophe after DNA damage. Nature 1999;401:616–20.CrossRef
Wilker, EW, van Vugt, MA, Artim, SA, et al. 14–3–3sigma controls mitotic translation to facilitate cytokinesis. Nature 2007;446:329–32.CrossRef
Pines, J. Four-dimensional control of the cell cycle. Nature Cell Biology 1999;1:E73–9.
Boutros, R, Lobjois, V, Ducommun, B. CDC25 phosphatases in cancer cells: key players? Good targets? Nature Reviews Cancer 2007;7:495–507.
Donzelli, M, Draetta, GF. Regulating mammalian checkpoints through Cdc25 inactivation. EMBO Reports 2003;4:671–7.CrossRef
Karlsson, C, Katich, S, Hagting, A, Hoffmann, I, Pines, J. Cdc25B and Cdc25C differ markedly in their properties as initiators of mitosis. Journal of Cell Biology 1999;146:573–84.CrossRefGoogle ScholarPubMed
Gabrielli, BG, De Souza, CP, Tonks, ID, et al. Cytoplasmic accumulation of cdc25B phosphatase in mitosis triggers centrosomal microtubule nucleation in HeLa cells. Journal of Cell Science 1996;109:1081–93.Google ScholarPubMed
Nishijima, H, Nishitani, H, Seki, T, Nishimoto, T. A dual-specificity phosphatase Cdc25B is an unstable protein and triggers p34(cdc2)/cyclin B activation in hamster BHK21 cells arrested with hydroxyurea. Journal of Cell Biology 1997;138:1105–16.CrossRefGoogle ScholarPubMed
Donzelli, M, Squatrito, M, Ganoth, D, et al. Dual mode of degradation of Cdc25 A phosphatase. EMBO Journal 2002;21:4875–84.CrossRef
Molinari, M, Mercurio, C, Dominguez, J, Goubin, F, Draetta, GF. Human Cdc25 A inactivation in response to S phase inhibition and its role in preventing premature mitosis. EMBO Reports 2000;1:71–9.CrossRef
Lee, G, White, LS, Hurov, KE, Stappenbeck, TS, Piwnica-Worms, H. Response of small intestinal epithelial cells to acute disruption of cell division through CDC25 deletion. Proceedings of the National Academy of Sciences USA 2009;106:4701–6.CrossRef
Toyoshima-Morimoto, F, Taniguchi, E, Nishida, E. Plk1 promotes nuclear translocation of human Cdc25C during prophase. EMBO Reports 2002;3:341–8.CrossRef
Wang, R, He, G, Nelman-Gonzalez, M, et al. Regulation of Cdc25C by ERK-MAP kinases during the G2/M transition. Cell 2007;128:1119–32.CrossRef
Hoffmann, I, Clarke, PR, Marcote, MJ, Karsenti, E, Draetta, G. Phosphorylation and activation of human cdc25-C by cdc2–cyclin B and its involvement in the self-amplification of MPF at mitosis. EMBO Journal 1993;12:53–63.
Izumi, T, Maller, JL. Elimination of cdc2 phosphorylation sites in the cdc25 phosphatase blocks initiation of M-phase. Molecular Biology of the Cell 1993;4:1337–50.CrossRef
Elia, EAH, Rellos, P, Haire, LF, et al. The molecular basis for phosphodependent substrate targeting and regulation of Plks by the Polo-box domain. Cell 2003;115:83–95.CrossRef
Reinhardt, HC, Yaffe, MB. Kinases that control the cell cycle in response to DNA damage: Chk1, Chk2, and MK2. Current Opinion in Cell Biology 2009;21:245–55.CrossRef
Chen, MS, Ryan, CE, Piwnica-Worms, H. Chk1 kinase negatively regulates mitotic function of Cdc25A phosphatase through 14-3-3 binding. Molecular and Cellular Biology 2003;23:7488–97.CrossRef
Davezac, N, Baldin, V, Gabrielli, B, et al. Regulation of CDC25B phosphatases subcellular localization. Oncogene 2000;19:2179–85.CrossRef
Ogg, S, Gabrielli, B, Piwnica-Worms, H. Purification of a serine kinase that associates with and phosphorylates human Cdc25C on serine 216. Journal of Biological Chemistry 1994;269:30 461–9.Google ScholarPubMed
Yang, J, Winkler, K, Yoshida, M, Kornbluth, S. Maintenance of G2 arrest in the Xenopus oocyte: a role for 14–3–3-mediated inhibition of Cdc25 nuclear import. EMBO Journal 1999;18:2174–83.CrossRef
Peng, CY, Graves, PR, Ogg, S, et al. C-TAK1 protein kinase phosphorylates human Cdc25C on serine 216 and promotes 14-3-3 protein binding. Cell Growth and Differentiation 1998;9:197–208.
Ouyang, B, Li, W, Pan, H, et al. The physical association and phosphorylation of Cdc25C protein phosphatase by Prk. Oncogene 1999;18:6029–36.CrossRef
Walworth, NC. DNA damage: Chk1 and Cdc25, more than meets the eye. Current Opinion in Genetic Development 2001;11:78–82.CrossRef
Zhou, BB, Elledge, SJ. The DNA damage response: putting checkpoints in perspective. Nature 2000;408:433–9.CrossRef
Wang, JY. Cellular responses to DNA damage. Current Opinion in Cell Biology 1998;10:240–7.CrossRef
Zeng, Y, Forbes, KC, Wu, Z, et al. Replication checkpoint requires phosphorylation of the phosphatase Cdc25 by Cds1 or Chk1. Nature 1998;395:507–10.CrossRef
Manke, IA, Nguyen, A, Lim, D, et al. MAPKAP kinase-2 is a cell cycle checkpoint kinase that regulates the G2/M transition and S phase progression in response to UV irradiation. Molecular Cell 2005;17:37–48.CrossRef
Reinhardt, HC, Aslanian, AS, Lees, JA, Yaffe, MB. p53-deficient cells rely on ATM- and ATR-mediated checkpoint signaling through the p38MAPK/MK2 pathway for survival after DNA damage. Cancer Cell 2007;11:175–89.CrossRef
Jazayeri, A, Falck, J, Lukas, C, et al. ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nature Cell Biology 2006;8:37–45.CrossRef
Lee, J, Kumagai, A, Dunphy, WG. Positive regulation of Wee1 by Chk1 and 14-3-3 proteins. Molecular Biology of the Cell 2001;12:551–63.CrossRef
Laronga, C, Yang, HY, Neal, C, Lee, MH. Association of the cyclin-dependent kinases and 14-3-3 sigma negatively regulates cell cycle progression. Journal of Biological Chemistry 2000;275:23 106–12.CrossRefGoogle ScholarPubMed
Bartkova, J, Horejsi, Z, Koed, K, et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature 2005;434:864–70.CrossRef
Margolis, SS, Walsh, S, Weiser, DC, et al. PP1 control of M phase entry exerted through 14–3–3-regulated Cdc25 dephosphorylation. EMBO Journal 2003;22:5734–45.CrossRef
Margolis, SS, Perry, JA, Forester, CM, et al. Role for the PP2A/B56delta phosphatase in regulating 14-3-3 release from Cdc25 to control mitosis. Cell 2006;127:759–73.CrossRef
Mailand, N, Bekker-Jensen, S, Bartek, J, Lukas, J. Destruction of Claspin by SCFbetaTrCP restrains Chk1 activation and facilitates recovery from genotoxic stress. Molecular Cell 2006;23:307–18.CrossRef
Mamely, I, van Vugt, MA, Smits, VA, et al. Polo-like kinase-1 controls proteasome-dependent degradation of Claspin during checkpoint recovery. Current Biology 2006;16:1950–5.CrossRef
Peschiaroli, A, Dorrello, NV, Guardavaccaro D, et al. SCFbetaTrCP-mediated degradation of Claspin regulates recovery from the DNA replication checkpoint response. Molecular Cell 2006;23:319–29.CrossRef
van Vugt, MA, Gardino, AK, Linding, R, et al. A mitotic phosphorylation feedback network connects Cdk1, Plk1, 53BP1, and Chk2 to inactivate the G(2)/M DNA damage checkpoint. PLoS Biology 2010;8:e1000287.
Lodygin, D, Hermeking, H. Epigenetic silencing of 14–3–3sigma in cancer. Seminars in Cancer Biology 2006;16:214–24.CrossRef
Hu, D, Valentine, M, Kidd, VJ, Lahti, JM. CDK11(p58) is required for the maintenance of sister chromatid cohesion. Journal of Cell Science 2007;120:2424–34.CrossRefGoogle ScholarPubMed
Petretti, C, Savoian, M, Montembault, E, et al. The PITSLRE/CDK11p58 protein kinase promotes centrosome maturation and bipolar spindle formation. EMBO Reports 2006;7:418–24.
Saurin, AT, Durgan, J, Cameron, AJ, et al. The regulated assembly of a PKCepsilon complex controls the completion of cytokinesis. Nature Cell Biology 2008;10:891–901.CrossRef
Cornelis, S, Bruynooghe, Y, Denecker, G, et al. Identification and characterization of a novel cell cycle-regulated internal ribosome entry site. Molecular Cell 2000;5:597–605.CrossRef
Gorin, MA, Pan, Q. Protein kinase C epsilon: an oncogene and emerging tumor biomarker. Molecular Cancer 2009;8:9.CrossRef
Fantl, WJ, Muslin, AJ, Kikuchi, A, et al. Activation of Raf-1 by 14-3-3 proteins. Nature 1994;371:612–14.CrossRef
Freed, E, Symons, M, Macdonald, SG, McCormick F, Ruggieri R. Binding of 14-3-3 proteins to the protein kinase Raf and effects on its activation. Science 1994;265:1713–16.CrossRef
Fu, H, Xia, K, Pallas, DC, et al. Interaction of the protein kinase Raf-1 with 14-3-3 proteins. Science 1994;266:126–9.CrossRef
Wellbrock, C, Karasarides, M, Marais, R. The RAF proteins take centre stage. Nature Reviews Molecular and Cellular Biology 2004;5:875–85.CrossRef
Garnett, MJ, Rana, S, Paterson, H, Barford, D, Marais, R. Wild-type and mutant B-RAF activate C-RAF through distinct mechanisms involving heterodimerization. Molecular Cell 2005;20:963–9.CrossRef
Rushworth, LK, Hindley, AD, O’Neill, E, Kolch, W. Regulation and role of Raf-1/B-Raf heterodimerization. Molecular and Cellular Biology 2006;26:2262–72.CrossRef
Noble, C, Mercer, K, Hussain, J, et al. CRAF autophosphorylation of serine 621 is required to prevent its proteasome-mediated degradation. Molecular Cell 2008;31:862–72.CrossRef
Wan, PT, Garnett, MJ, Roe, SM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 2004;116:855–67.CrossRef
Heidorn, SJ, Milagre, C, Whittaker, S, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell;140:209–21.
Burgering, BM, Medema, RH. Decisions on life and death: FOXO Forkhead transcription factors are in command when PKB/Akt is off duty. Journal of Leukocyte Biology 2003;73:689–701.CrossRefGoogle ScholarPubMed
Macdonald, A, Campbell, DG, Toth, R, et al. Pim kinases phosphorylate multiple sites on Bad and promote 14-3-3 binding and dissociation from Bcl-XL. BMC Cell Biology 2006;7:1.CrossRef
Yoshida, K, Yamaguchi, T, Natsume, T, Kufe, D, Miki, Y. JNK phosphorylation of 14-3-3 proteins regulates nuclear targeting of c-Abl in the apoptotic response to DNA damage. Nature Cell Biology 2005;7:278–85.CrossRef
Bajpai, U, Sharma, R, Kausar, T, et al. Clinical significance of 14-3-3 zeta in human esophageal cancer. International Journal of Biological Markers 2008;23:231–7.
Keshamouni, VG, Michailidis, G, Grasso, CS, et al. Differential protein expression profiling by iTRAQ-2DLC-MS/MS of lung cancer cells undergoing epithelial-mesenchymal transition reveals a migratory/invasive phenotype. Journal of Proteome Research 2006;5:1143–54.CrossRefGoogle ScholarPubMed
Li, Z, Zhao, J, Du, Y, et al. Down-regulation of 14–3–3zeta suppresses anchorage-independent growth of lung cancer cells through anoikis activation. Proceedings of the National Academy of Sciences USA 2008;105:162–7.CrossRef
Ostergaard, M, Rasmussen, HH, Nielsen, HV, et al. Proteome profiling of bladder squamous cell carcinomas: identification of markers that define their degree of differentiation. Cancer Research 1997;57:4111–17.
Ferguson, AT, Evron, E, Umbricht, CB, et al. High frequency of hypermethylation at the 14-3-3 sigma locus leads to gene silencing in breast cancer. Proceedings of the National Academy of Sciences USA 2000;97:6049–54.CrossRef
Iwata, N, Yamamoto, H, Sasaki, S, et al. Frequent hypermethylation of CpG islands and loss of expression of the 14-3-3 sigma gene in human hepatocellular carcinoma. Oncogene 2000;19:5298–302.CrossRef
Suzuki, H, Itoh, F, Toyota, M, et al. Inactivation of the 14-3-3 sigma gene is associated with 5’ CpG island hypermethylation in human cancers. Cancer Research 2000;60:4353–7.
Umbricht, CB, Evron, E, Gabrielson, E, et al. Hypermethylation of 14-3-3 sigma (stratifin) is an early event in breast cancer. Oncogene 2001;20:3348–53.CrossRef
Vercoutter-Edouart, AS, Lemoine, J, Le Bourhis, X, et al. Proteomic analysis reveals that 14–3–3sigma is down-regulated in human breast cancer cells. Cancer Research 2001;61:76–80.
Bortner, JD., Das, A, Umstead, TM, et al. Down-regulation of 14-3-3 isoforms and annexin A5 proteins in lung adenocarcinoma induced by the tobacco-specific nitrosamine NNK in the A/J mouse revealed by proteomic analysis. Journal of Proteome Research 2009;8:4050–61.CrossRefGoogle Scholar
Li, DQ, Wang, L, Fei, F, et al. Identification of breast cancer metastasis-associated proteins in an isogenic tumor metastasis model using two-dimensional gel electrophoresis and liquid chromatography-ion trap-mass spectrometry. Proteomics 2006;6:3352–68.CrossRef
Niemantsverdriet, M, Wagner, K, Visser, M, Backendorf, C. Cellular functions of 14-3-3 zeta in apoptosis and cell adhesion emphasize its oncogenic character. Oncogene 2008;27:1315–19.CrossRef
Lu, J, Guo, H, Treekitkarnmongkol, W, et al. 14–3–3zeta Cooperates with ERBB2 to promote ductal carcinoma in situ progression to invasive breast cancer by inducing epithelial-mesenchymal transition. Cancer Cell 2009;16:195–207.CrossRef
Danes, CG, Wyszomierski, SL, Lu, J, et al. 14-3-3 zeta down-regulates p53 in mammary epithelial cells and confers luminal filling. Cancer Research 2008;68:1760–7.CrossRef
Somiari, RI, Somiari, S, Russell, S, Shriver, CD. Proteomics of breast carcinoma. Journal of Chromatography B Analytical Technology Biomedical Life Science 2005;815:215–25.CrossRefGoogle ScholarPubMed
Zang, L, Palmer Toy, D, et al. Proteomic analysis of ductal carcinoma of the breast using laser capture microdissection, LC-MS, and 16O/18O isotopic labeling. Journal of Proteome Research 2004;3:604–12.CrossRefGoogle ScholarPubMed
Jang, JS, Cho, HY, Lee, YJ, Ha, WS, Kim, HW. The differential proteome profile of stomach cancer: identification of the biomarker candidates. Oncology Research 2004;14:491–9.CrossRef
Qi, W, Liu, X, Qiao, D, Martinez, JD. Isoform-specific expression of 14-3-3 proteins in human lung cancer tissues. International Journal of Cancer 2005;113:359–63.CrossRefGoogle ScholarPubMed
Shoji, M, Kawamoto, S, Setoguchi, Y, et al. The 14-3-3 protein as the antigen for lung cancer-associated human monoclonal antibody AE6F4. Human Antibodies and Hybridomas 1994;5:123–30.
Arora, S, Matta, A, Shukla, NK, Deo, SV, Ralhan, R. Identification of differentially expressed genes in oral squamous cell carcinoma. Molecular Carcinogenesis 2005;42:97–108.CrossRef
Huber, E, Vlasny, D, Jeckel, S, Stubenrauch, F, Iftner, T. Gene profiling of cottontail rabbit papillomavirus-induced carcinomas identifies upregulated genes directly involved in stroma invasion as shown by small interfering RNA-mediated gene silencing. Journal of Virology 2004;78:7478–89.CrossRefGoogle ScholarPubMed
Hermeking, H, Lengauer, C, Polyak, K, et al. 14-3-3 sigma is a p53-regulated inhibitor of G2/M progression. Molecular Cell 1997;1:3–11.CrossRef
Aprelikova, O, Pace, AJ, Fang, B, Koller, BH, Liu, ET. BRCA1 is a selective co-activator of 14-3-3 sigma gene transcription in mouse embryonic stem cells. Journal of Biological Chemistry 2001;276:25 647–50.CrossRefGoogle ScholarPubMed
Henrique, R, Jeronimo, C, Hoque, MO, et al. Frequent 14-3-3 sigma promoter methylation in benign and malignant prostate lesions. DNA and Cell Biology 2005;24:264–9.CrossRef
Kang, GH, Lee, S, Kim, WH, et al. Epstein-barr virus-positive gastric carcinoma demonstrates frequent aberrant methylation of multiple genes and constitutes CpG island methylator phenotype-positive gastric carcinoma. American Journal of Pathology 2002;160:787–94.CrossRefGoogle ScholarPubMed
Gasco, M, Bell, AK, Heath, V, et al. Epigenetic inactivation of 14-3-3 sigma in oral carcinoma: association with p16(INK4a) silencing and human papillomavirus negativity. Cancer Research 2002;62:2072–6.
Gasco, M, Sullivan, A, Repellin, C, et al. Coincident inactivation of 14–3–3sigma and p16INK4a is an early event in vulval squamous neoplasia. Oncogene 2002;21:1876–81.CrossRef
Kaneuchi, M, Sasaki, M, Tanaka, Y, et al. Expression and methylation status of 14-3-3 sigma gene can characterize the different histological features of ovarian cancer. Biochemical and Biophysical Research Communications 2004;316:1156–62.CrossRef
Uchida, D, Begum, NM, Almofti, A, et al. Frequent downregulation of 14-3-3 sigma protein and hypermethylation of 14-3-3 sigma gene in salivary gland adenoid cystic carcinoma. British Journal of Cancer 2004;91:1131–8.CrossRefGoogle ScholarPubMed
Bhawal, UK, Tsukinoki, K, Sasahira, T, et al. Methylation and intratumoural heterogeneity of 14-3-3 sigma in oral cancer. Oncology Report 2007;18:817–24.
Kunze, E, Schlott, T. High frequency of promoter methylation of the 14-3-3 sigma and CAGE-1 genes, but lack of hypermethylation of the caveolin-1 gene, in primary adenocarcinomas and signet ring cell carcinomas of the urinary bladder. International Journal of Molecular Medicine 2007;20:557–63.Google Scholar
Zhu, F, Xia, X, Liu, B, et al. IKKalpha shields 14–3–3sigma, a G(2)/M cell cycle checkpoint gene, from hypermethylation, preventing its silencing. Molecular Cell 2007;27:214–27.CrossRef
Urano, T, Saito, T, Tsukui, T, et al. Efp targets 14-3-3 sigma for proteolysis and promotes breast tumour growth. Nature 2002;417:871–5.CrossRef
Chan, TA, Hermeking, H, Lengauer, C, Kinzler, KW, Vogelstein, B. 14–3–3Sigma is required to prevent mitotic catastrophe after DNA damage. Nature 1999;401:616–20.CrossRef
Dellambra, E, Golisano, O, Bondanza, S, et al. Downregulation of 14–3–3sigma prevents clonal evolution and leads to immortalization of primary human keratinocytes. Journal of Cell Biology 2000;149:1117–30.CrossRefGoogle ScholarPubMed
Fujiwara, T, Bandi, M, Nitta, M, et al. Cytokinesis failure generating tetraploids promotes tumorigenesis in p53-null cells. Nature 2005;437:1043–7.CrossRef
Shi, Q, King, RW. Chromosome nondisjunction yields tetraploid rather than aneuploid cells in human cell lines. Nature 2005;437:1038–42.CrossRef
Hanahan, D, Weinberg, RA. The hallmarks of cancer. Cell 2000;100:57–70.CrossRef
Luo, J, Solimini, NL, Elledge, SJ. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 2009;136:823–37.CrossRef
Maxwell, SA, Li, Z, Jaya, D, et al. 14–3–3zeta mediates resistance of diffuse large B cell lymphoma to an anthracycline-based chemotherapeutic regimen. Journal of Biological Chemistry 2009;284:22 379–89.CrossRefGoogle Scholar
Guweidhi, A, Kleeff, J, Giese, N, et al. Enhanced expression of 14–3–3 sigma in pancreatic cancer and its role in cell cycle regulation and apoptosis. Carcinogenesis 2004;25:1575–85.CrossRef
Neupane, D, Korc, M. 14–3–3sigma Modulates pancreatic cancer cell survival and invasiveness. Clinical Cancer Research 2008;14:7614–23.CrossRef
Wang, B, Liu, K, Lin, HY, et al. 14–3–3Tau regulates ubiquitin-independent proteasomal degradation of p21, a novel mechanism of p21 downregulation in breast cancer. Molecular and Cell Biology; 30:1508–27.
Wang, B, Yang, H, Liu, YC, et al. Isolation of high-affinity peptide antagonists of 14-3-3 proteins by phage display. Biochemistry 1999;38:12 499–504.
Masters, SC, Fu, H. 14-3-3 proteins mediate an essential anti-apoptotic signal. Journal of Biological Chemistry 2001;276:45 193–200.CrossRefGoogle ScholarPubMed
Nguyen, A, Rothman, DM, Stehn, J, Imperiali, B, Yaffe, MB. Caged phosphopeptides reveal a temporal role for 14-3-3 in G1 arrest and S-phase checkpoint function. Nature Biotechnology 2004;22:993–1000.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • 14-3-3 proteins in cancer
    • By Alexandra K. Gardino, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA, Michael B. Yaffe, David H. Koch Institute for Integrative Cancer Research and Department of Biological Engineering,Massachusetts Institute of Technology, Cambridge, MA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.025
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • 14-3-3 proteins in cancer
    • By Alexandra K. Gardino, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA, Michael B. Yaffe, David H. Koch Institute for Integrative Cancer Research and Department of Biological Engineering,Massachusetts Institute of Technology, Cambridge, MA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.025
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • 14-3-3 proteins in cancer
    • By Alexandra K. Gardino, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA, Michael B. Yaffe, David H. Koch Institute for Integrative Cancer Research and Department of Biological Engineering,Massachusetts Institute of Technology, Cambridge, MA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.025
Available formats
×