Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-8bljj Total loading time: 0 Render date: 2024-06-23T11:58:19.968Z Has data issue: false hasContentIssue false

27 - Jun proteins and AP-1 in tumorigenesis

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Shira Anzi
Affiliation:
Department of Developmental Biology and Cancer Research, IMRIC, he Hebrew University - Hadassah Medical School, Jerusalem, Israel
Eitan Shaulian
Affiliation:
Department of Biochemistry and Molecular Biology, IMRIC, The Hebrew University - Hadassah Medical School, Jerusalem, Israel
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

AP-1

Activating protein 1 (AP-1) is a dimeric transcription factor which serves as an integrator of numerous signals to regulate gene expression affecting cellular proliferation, differentiation, apoptosis, autophagy, inflammation, cellular migration, and neoplastic transformation. AP-1 is composed of proteins from several families whose common denominator is the possession of basic leucine-zipper (bZIP) domains that are essential for dimerization and DNA binding. Jun (c-Jun, JunB, and JunD) and Fos (c-Fos, FosB, Fra1, and Fra2) subfamilies are the major AP-1 proteins. ATF proteins ATF2, LRF1/ATF3, B-ATF, JDP1, and JDP2 are also components of AP-1, and the Maf subfamily, which includes c-Maf, MafB, MafA, MafG/F/K, and Nrl are considered to be close relatives which form heterodimers with Fos and Jun, and occupy AP-1 binding sites. Jun–Fos heterodimers bind preferentially to a heptamer consensus sequence known as the TPA responsive element (TRE; 5ʹ-TGA(C/G)TCA-3ʹ), whereas Jun-ATF dimers bind with higher affinity to another consensus sequence known as the cyclic AMP responsive element (CRE; 5ʹ- TGACGTCA -3ʹ). The seven Fos-Jun family members can form 18 different homo- and heterodimers. The composition of the dimers varies among different tissues, regulated by exposure to different stimuli, and determines specificity of the binding site and consequently the identity of activated genes (1). Increased diversity of AP-1 and consequently increased complexity, is also generated by the ability of “core AP-1 proteins” Jun and Fos to interact with other leucine-zipper-containing proteins, sometimes resulting in binding to novel DNA elements (2). Additional layers of complexity may stem from interactions of AP-1 components with structurally unrelated proteins which recruit them to other divergent binding sites on the DNA in order to regulate gene transcription (1).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 319 - 327
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

Chinenov, Y, Kerppola, TK. Close encounters of many kinds: Fos-Jun interactions that mediate transcription regulatory specificity. Oncogene 2001;20:2438–52.CrossRef
Cai, DH, Wang, D, Keefer, J, et al. C/EBP alpha: AP-1 leucine zipper heterodimers bind novel DNA elements, activate the PU.1 promoter and direct monocyte lineage commitment more potently than C/EBP alpha homodimers or AP-1. Oncogene 2008;27:2772–9.CrossRef
Kouzarides, T, Ziff, E. The role of the leucine zipper in the fos-jun interaction. Nature 1988;336:646–51.CrossRef
Kallunki, T, Deng, T, Hibi, M, Karin, M. c-Jun can recruit JNK to phosphorylate dimerization partners via specific docking interactions. Cell 1996;87:929–39.CrossRef
Adler, J, Reuven, N, Kahana, C, Shaul, Y. c-Fos proteasomal degradation by default and its regulation by NQO1 determines c-Fos serum response kinetics. Molecular and Cellular Biology 2010;30:3767–78.CrossRef
Hibi, M, Lin, A, Smeal, T, Minden, A, Karin, M. Identification of an oncoprotein- and UV-responsive protein kinase that binds and potentiates the c-Jun activation domain. Genes and Development 1993;7:2135–48.CrossRef
Derijard, B, Hibi, M, Wu, IH, et al. JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain. Cell 1994;76:1025–37.CrossRef
Morton, S, Davis, RJ, McLaren, A, Cohen, P. A reinvestigation of the multisite phosphorylation of the transcription factor c-Jun. EMBO Journal 2003;22:3876–86.CrossRef
Lin, A, Frost, J, Deng, T, et al. Casein kinase II is a negative regulator of c-Jun DNA binding and AP-1 activity. Cell 1992;70:777–89.CrossRef
Boyle, WJ, Smeal, T, Defize, LH, et al. Activation of protein kinase C decreases phosphorylation of c-Jun at sites that negatively regulate its DNA-binding activity. Cell 1991;64:573–84.CrossRef
Barila, D, Mangano, R, Gonfloni, S, et al. A nuclear tyrosine phosphorylation circuit: c-Jun as an activator and substrate of c-Abl and JNK. EMBO Journal 2000;19:273–81.CrossRef
Zhu, F, Choi, BY, Ma, WY, et al. COOH-terminal Src kinase-mediated c-Jun phosphorylation promotes c-Jun degradation and inhibits cell transformation. Cancer Research 2006;66:5729–36.CrossRef
Weiss, C, Schneider, S, Wagner, EF, et al. JNK phosphorylation relieves HDAC3-dependent suppression of the transcriptional activity of c-Jun. EMBO Journal 2003;22:3686–95.CrossRef
Angel, P, Hattori, K, Smeal, T, Karin, M. The jun proto-oncogene is positively autoregulated by its product, Jun/AP-1. Cell 1988;55:875–85.CrossRef
Stein, B, Angel, P, van Dam, H, et al. Ultraviolet-radiation induced c-jun gene transcription: two AP-1 like binding sites mediate the response. Photochemistry and Photobiology 1992;55:409–15.CrossRef
Abate, C, Patel, L, Rauscher, FJ, 3rd, Curran T. Redox regulation of fos and jun DNA-binding activity in vitro. Science 1990;249:1157–61.CrossRef
Xanthoudakis, S, Curran, T. Redox regulation of AP-1: a link between transcription factor signaling and DNA repair. Advances in Experimental Medical Biology 1996;387:69–75.CrossRef
Fuchs, SY, Dolan, L, Davis, RJ, Ronai, Z. Phosphorylation-dependent targeting of c-Jun ubiquitination by Jun N-kinase. Oncogene 1996;13:1531–5.
Musti, AM, Treier, M, Bohmann, D. Reduced ubiquitin-dependent degradation of c-Jun after phosphorylation by MAP kinases. Science 1997;275:400–2.CrossRef
Wei, W, Jin, J, Schlisio, S, Harper, JW, Kaelin, WG The v-Jun point mutation allows c-Jun to escape GSK3-dependent recognition and destruction by the Fbw7 ubiquitin ligase. Cancer Cell 2005;8:25–33.CrossRef
Yogev, O, Saadon, K, Anzi, S, Inoue, K, Shaulian, E. DNA damage-dependent translocation of B23 and p19 ARF is regulated by the Jun N-terminal kinase pathway. Cancer Research 2008;68:1398–406.CrossRef
Shaulian, E, Karin, M. AP-1 as a regulator of cell life and death. Nature Cell Biology 2002;4:E131–6.
Greenberg, ME, Ziff EB. Stimulation of 3T3 cells induces transcription of the c-fos proto-oncogene. Nature 1984;311:433–8.CrossRef
Angel, P, Karin, M. The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation. Biochimica et Biophysica Acta 1991;1072:129–57.
Piette, J, Hirai, S, Yaniv, M. Constitutive synthesis of activator protein 1 transcription factor after viral transformation of mouse fibroblasts. Proceedings of the National Academy of Sciences USA 1988;85:3401–5.CrossRef
Smeal, T, Binetruy, B, Mercola, DA, Birrer, M, Karin, M. Oncogenic and transcriptional cooperation with Ha-Ras requires phosphorylation of c-Jun on serines 63 and 73. Nature 1991;354:494–6.CrossRef
Hernandez, JM, Floyd, DH, Weilbaecher, KN, Green, PL, Boris-Lawrie, K. Multiple facets of junD gene expression are atypical among AP-1 family members. Oncogene 2008;27:4757–67.CrossRef
Shaulian, E, Schreiber, M, Piu F, et al. The mammalian UV response: c-Jun induction is required for exit from p53-imposed growth arrest. Cell 2000;103:897–907.CrossRef
Devary, Y, Gottlieb, RA, Lau, LF, Karin, M. Rapid and preferential activation of the c-jun gene during the mammalian UV response. Molecular and Cellular Biology 1991;11:2804–11.CrossRef
Gupta, S, Campbell, D, Derijard, B, Davis, RJ. Transcription factor ATF2 regulation by the JNK signal transduction pathway. Science 1995;267:389–93.CrossRef
Yogev, O, Anzi, S, Inoue, K, Shaulian, E.Induction of transcriptionally active Jun proteins regulates drug-induced senescence. Journal of Biological Chemistry 2006;281:34 475–83.CrossRefGoogle ScholarPubMed
Macian, F, Lopez-Rodriguez, C, Rao, A. Partners in transcription: NFAT and AP-1. Oncogene 2001;20:2476–89.CrossRef
Shaulian, E, Karin, M. AP-1 in cell proliferation and survival. Oncogene 2001;20:2390–400.CrossRef
Schreiber, M, Kolbus, A, Piu, F, et al. Control of cell cycle progression by c-Jun is p53 dependent. Genes and Development 1999;13:607–19.CrossRef
Johnson, RS, van Lingen, B, Papaioannou, VE, Spiegelman, BM. A null mutation at the c-jun locus causes embryonic lethality and retarded cell growth in culture. Genes and Development 1993;7:1309–17.CrossRef
Weitzman, JB, Fiette, L, Matsuo, K, Yaniv, M. JunD protects cells from p53-dependent senescence and apoptosis. Molecular Cell 2000;6:1109–19.CrossRef
Johnson, AC, Murphy, BA, Matelis, CM, et al. Activator protein-1 mediates induced but not basal epidermal growth factor receptor gene expression. Molecular Medicine 2000;6:17–27.
Zenz, R, Scheuch, H, Martin, P, et al. c-Jun regulates eyelid closure and skin tumor development through EGFR signaling. Developmental Cell 2003;4:879–89.CrossRef
MacLaren, A, Black, EJ, Clark, W, Gillespie, DA. c-Jun-deficient cells undergo premature senescence as a result of spontaneous DNA damage accumulation. Molecular and Cellular Biology 2004;24:9006–18.CrossRef
Passegue, E, Wagner, EF. JunB suppresses cell proliferation by transcriptional activation of p16(INK4a) expression. EMBO Journal 2000;19:2969–79.CrossRef
Andrecht, S, Kolbus, A, Hartenstein, B, Angel, P, Schorpp-Kistner M. Cell cycle promoting activity of JunB through cyclin A activation. Journal of Biological Chemistry 2002;277:35 961–8.CrossRefGoogle ScholarPubMed
Passegue, E, Jochum, W, Behrens, A, Ricci, R, Wagner, EF. JunB can substitute for Jun in mouse development and cell proliferation. Nature Genetics 2002;30:158–66.CrossRef
Brown, JR, Nigh, E, Lee, RJ, et al. Fos family members induce cell cycle entry by activating cyclin D1. Molecular and Cellular Biology 1998;18:5609–19.CrossRef
Brusselbach, S, Mohle-Steinlein, U, Wang, ZQ, et al. Cell proliferation and cell cycle progression are not impaired in fibroblasts and ES cells lacking c-Fos. Oncogene 1995;10:79–86.
Eferl, R, Wagner, EF. AP-1: a double-edged sword in tumorigenesis. Nature Reviews Cancer 2003;3:859–68.CrossRef
Shaulian, E. AP-1–The Jun proteins: oncogenes or tumor suppressors in disguise? Cellular Signaling 2010;22:894–9.
Hilberg, F, Aguzzi, A, Howells, N, Wagner, EF. c-Jun is essential for normal mouse development and hepatogenesis. Nature 1993;365:179–81 [published erratum appears in Nature 1993;366:368].
Eferl, R, Ricci, R, Kenner, L, et al. Liver tumor development. c-Jun antagonizes the proapoptotic activity of p53. Cell 2003;112:181–92.
Le-Niculescu, H, Bonfoco, E, Kasuya, Y, et al. Withdrawal of survival factors results in activation of the JNK pathway in neuronal cells leading to Fas ligand induction and cell death. Molecular and Cellular Biology 1999;19:751–63.CrossRef
Behrens, A, Sibilia, M, Wagner, EF. Amino-terminal phosphorylation of c-Jun regulates stress-induced apoptosis and cellular proliferation. Nature Genetics 1999;21:326–9.CrossRef
Gurzov, EN, Ortis, F, Bakiri, L, Wagner, EF, Eizirik, DL. JunB inhibits ER stress and apoptosis in pancreatic beta cells. PLoS ONE 2008;3:e3030.
Yogev, O, Goldberg, R, Anzi, S, Shaulian, E. Jun proteins are starvation-regulated inhibitors of autophagy. Cancer Research 2010;70:2318–27.CrossRef
Yogev, O, Shaulian, E. Jun proteins inhibit autophagy and induce cell death. Autophagy 2010;6(4):566–7.CrossRef
Saez, E, Rutberg, SE, Mueller, E, et al. c-fos is required for malignant progression of skin tumors. Cell 1995;82:721–32.CrossRef
Ozanne, BW, Spence, HJ, McGarry, LC, Hennigan, RF. Transcription factors control invasion: AP-1 the first among equals. Oncogene 2007;26:1–10.CrossRef
Reichmann, E, Schwarz, H, Deiner, EM, et al. Activation of an inducible c-FosER fusion protein causes loss of epithelial polarity and triggers epithelial-fibroblastoid cell conversion. Cell 1992;71:1103–16.CrossRef
Fialka, I, Schwarz, H, Reichmann, E, et al. The estrogen-dependent c-JunER protein causes a reversible loss of mammary epithelial cell polarity involving a destabilization of adherens junctions. Journal of Cell Biology 1996;132:1115–32.CrossRefGoogle ScholarPubMed
Vleugel, MM, Greijer, AE, Bos, R, van der Wall, E, van Diest, PJ. c-Jun activation is associated with proliferation and angiogenesis in invasive breast cancer. Human Pathology 2006;37:668–74.CrossRef
Zhang, G, Dass, CR, Sumithran, E, et al. Effect of deoxyribozymes targeting c-Jun on solid tumor growth and angiogenesis in rodents. Journal of the National Cancer Institute 2004;96:683–96.CrossRefGoogle ScholarPubMed
Damert, A, Ikeda, E, Risau, W. Activator-protein-1 binding potentiates the hypoxia-inducible factor-1-mediated hypoxia-induced transcriptional activation of vascular-endothelial growth factor expression in C6 glioma cells. Biochemical Journal 1997;327:419–23.CrossRef
Schmidt, D, Textor, B, Pein, OT, et al. Critical role for NF-kappaB-induced JunB in VEGF regulation and tumor angiogenesis. EMBO Journal 2007;26:710–19.CrossRef
Gerald, D, Berra, E, Frapart, YM, et al. JunD reduces tumor angiogenesis by protecting cells from oxidative stress. Cell 2004;118:781–94.CrossRef
Ruther, U, Garber, C, Komitowski, D, Muller, R, Wagner, EF. Deregulated c-Fos expression interferes with normal bone development in transgenic mice. Nature 1987;325:412–16.CrossRef
Ruther, U, Komitowski, D, Schubert, FR, Wagner, EF. c-Fos expression induces bone tumors in transgenic mice. Oncogene 1989;4:861–5.
Young, MR, Li, JJ, Rincon, M, et al. Transgenic mice demonstrate AP-1 (activator protein-1) transactivation is required for tumor promotion. Proceedings of the National Academy of Sciences USA 1999;96:9827–32.CrossRef
Behrens, A, Jochum, W, Sibilia, M, Wagner, EF. Oncogenic transformation by ras and fos is mediated by c-Jun N-terminal phosphorylation. Oncogene 2000;19:2657–63.CrossRef
Chang, Q, Zhang, Y, Beezhold, KJ, et al. Sustained JNK1 activation is associated with altered histone H3 methylations in human liver cancer. Journal of Hepatology 2009;50:323–33.CrossRefGoogle ScholarPubMed
Hui, L, Zatloukal, K, Scheuch, H, Stepniak, E, Wagner, EF. Proliferation of human HCC cells and chemically induced mouse liver cancers requires JNK1-dependent p21 downregulation. Journal of Clinical Investigation 2008;118:3943–53.CrossRefGoogle ScholarPubMed
Toualbi, K, Guller, MC, Mauriz, JL, et al. Physical and functional cooperation between AP-1 and beta-catenin for the regulation of TCF-dependent genes. Oncogene 2007;26:3492–502.CrossRef
Sancho, R, Nateri, AS, de Vinuesa, AG, et al. JNK signalling modulates intestinal homeostasis and tumourigenesis in mice. EMBO Journal 2009;28:1843–54.CrossRef
Nateri, AS, Spencer-Dene, B, Behrens, A. Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development. Nature 2005;437:281–5.CrossRef
Passegue, E, Jochum, W, Schorpp-Kistner, M, Mohle-Steinlein, U, Wagner, EF. Chronic myeloid leukemia with increased granulocyte progenitors in mice lacking junB expression in the myeloid lineage. Cell 2001;104:21–32.CrossRef
Passegue, E, Wagner, EF, Weissman, IL. JunB deficiency leads to a myeloproliferative disorder arising from hematopoietic stem cells. Cell 2004;119:431–43.CrossRef
Santaguida, M, Schepers, K, King, B, et al. JunB protects against myeloid malignancies by limiting hematopoietic stem cell proliferation and differentiation without affecting self-renewal. Cancer Cell 2009;15:341–52.CrossRef
Milde-Langosch, K. The Fos family of transcription factors and their role in tumourigenesis. European Journal of Cancer 2005;41:2449–61.CrossRefGoogle ScholarPubMed
Verde, P, Casalino, L, Talotta, F, Yaniv, M, Weitzman, JB. Deciphering AP-1 function in tumorigenesis: fra-ternizing on target promoters. Cell Cycle 2007;6:2633–9.CrossRef
Mariani, O, Brennetot, C, Coindre, JM, et al. JUN oncogene amplification and overexpression block adipocytic differentiation in highly aggressive sarcomas. Cancer Cell 2007;11:361–74.CrossRef
Mathas, S, Hinz, M, Anagnostopoulos, I, et al. Aberrantly expressed c-Jun and JunB are a hallmark of Hodgkin lymphoma cells, stimulate proliferation and synergize with NF-kappa B. EMBO Journal 2002;21:4104–13.CrossRef
Rassidakis, GZ, Thomaides, A, Atwell, C, et al. JunB expression is a common feature of CD30+ lymphomas and lymphomatoid papulosis. Modern Pathology 2005;18:1365–70.CrossRef
Mao, X, Orchard, G, Lillington, DM, et al. BCL2 and JUNB abnormalities in primary cutaneous lymphomas. British Journal of Dermatology 2004;151:546–56.CrossRefGoogle ScholarPubMed
Mao, X, Orchard, G, Lillington, DM, et al. Amplification and overexpression of JUNB is associated with primary cutaneous T-cell lymphomas. Blood 2003;101:1513–19.CrossRef
Staber, PB, Vesely, P, Haq, N, et al. The oncoprotein NPM-ALK of anaplastic large-cell lymphoma induces JUNB transcription via ERK1/2 and JunB translation via mTOR signaling. Blood 2007;110:3374–83.CrossRef
Yang, MY, Liu, TC, Chang, JG, Lin, PM, Lin, SF. JunB gene expression is inactivated by methylation in chronic myeloid leukemia. Blood 2003;101:3205–11.CrossRef
Steidl, U, Rosenbauer, F, Verhaak, RG, et al. Essential role of Jun family transcription factors in PU.1 knockdown-induced leukemic stem cells. Nature Genetics 2006;38:1269–77.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.

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.

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.

Available formats
×