The Multifaceted Output of c-Jun Biological Activity: Focus at the Junction of CD8 T Cell Activation and Exhaustion
Abstract
:1. Introduction
2. Function and Regulation of the c-Jun/AP-1 Transcription Factor
2.1. AP-1 Complexity
2.2. AP-1 Functions: From Tissue Differentiation to Cancer Progression and Immune Response
2.3. Levels of c-Jun/AP-1 Regulation
2.3.1. Dimer Composition
2.3.2. Transcriptional and Post-Translational Modifications
2.4. Regulation of c-Jun Activity by N-Terminal and C-Terminal Phosphorylation
2.5. Impact of c-Jun N-Terminal Phosphorylation on c-Jun Ubiquitine-Depedendent Degradation
2.6. Role of c-Jun N-Terminal Phosphorylation in c-Jun Biological Outputs
2.7. Therapeutic Targeting of c-Jun/AP-1 Complexes
3. Role of c-Jun/Ap-1 in the Transcriptional and Epigenetic Regulation of Effector CD8T Cell Differentiation
3.1. The CD8 T Cell Response to Acute Infection
3.2. c-Jun/AP1 Cooperates with NFAT Downstream TCR and Costimulatory Signaling
3.3. c-Jun Cooperates with BATF and IRF4 during Effector CD8 T Differentiation
4. Regulation of c-Jun/Ap-1 in Exhausted CD8T Cells during Chronic Viral Infection and Cancer
4.1. CD8 T Cell Exhaustion in Chronic Infection
4.2. CD8 T Cell Exhaustion in Cancer
4.3. The Core Transcriptional Program Driving Exhaustion is Triggered by Partnerless NFAT
4.4. c-Jun as A Tool in CAR T Cell Therapy
5. Conclusions
Funding
Acknowledgments
Conflicts of Interest
References
- Bejjani, F.; Evanno, E.; Zibara, K.; Piechaczyk, M.; Jariel-Encontre, I. The AP-1 transcriptional complex: Local switch or remote command? Biochim. Biophys. Acta Rev. Cancer 2019, 1872, 11–23. [Google Scholar] [CrossRef] [PubMed]
- Hess, J.; Angel, P.; Schorpp-Kistner, M. AP-1 subunits: Quarrel and harmony among siblings. J. Cell Sci. 2004, 117 Pt 25, 5965–5973. [Google Scholar] [CrossRef] [Green Version]
- Rodríguez-Martínez, J.A.; Reinke, A.W.; Bhimsaria, D.; Keating, A.E.; Ansari, A.Z. Combinatorial bZIP dimers display complex DNA-binding specificity landscapes. Elife 2017, 6, e19272. [Google Scholar] [CrossRef] [PubMed]
- Macián, F.; García-Rodríguez, C.; Rao, A. Gene expression elicited by NFAT in the presence or absence of cooperative recruitment of Fos and Jun. EMBO J. 2000, 19, 4783–4795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Müller, M.R.; Rao, A. NFAT, immunity and cancer: A transcription factor comes of age. Nat. Rev. Immunol. 2010, 10, 645–656. [Google Scholar] [CrossRef]
- Kurachi, M.; Barnitz, R.A.; Yosef, N.; Odorizzi, P.M.; DiIorio, M.A.; Lemieux, M.E.; Yates, K.; Godec, J.; Klatt, M.G.; Regev, A.; et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8+ T cells. Nat. Immunol. 2014, 15, 373–383. [Google Scholar] [CrossRef]
- Chen, Y.; Zander, R.; Khatun, A.; Schauder, D.M.; Cui, W. Transcriptional and Epigenetic Regulation of Effector and Memory CD8 T Cell Differentiation. Front. Immunol. 2018, 9, 2826. [Google Scholar] [CrossRef] [Green Version]
- McLane, L.M.; Abdel-Hakeem, M.S.; Wherry, E.J. CD8 T Cell Exhaustion during Chronic Viral Infection and Cancer. Annu. Rev. Immunol. 2019, 37, 457–495. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Glover, J.N.; Hogan, P.G.; Rao, A.; Harrison, S.C. Structure of the DNA-binding domains from NFAT, Fos and Jun bound specifically to DNA. Nature 1998, 392, 42–48. [Google Scholar] [CrossRef]
- Jain, J.; McCaffrey, P.G.; Miner, Z.; Kerppola, T.K.; Lambert, J.N.; Verdine, G.L.; Curran, T.; Rao, A. The T-cell transcription factor NFATp is a substrate for calcineurin and interacts with Fos and Jun. Nature 1993, 365, 352–355. [Google Scholar] [CrossRef]
- Kel, A.; Kel-Margoulis, O.; Babenko, V.; Wingender, E. Recognition of NFATp/AP-1 composite elements within genes induced upon the activation of immune cells. J. Mol. Biol. 1999, 288, 353–376. [Google Scholar] [CrossRef] [PubMed]
- Kaminuma, O.; Elly, C.; Tanaka, Y.; Mori, A.; Liu, Y.C.; Altman, A.; Miyatake, S. Vav-induced activation of the human IFN-gamma gene promoter is mediated by upregulation of AP-1 activity. FEBS Lett. 2002, 514, 153–158. [Google Scholar] [CrossRef] [Green Version]
- Teixeira, L.K.; Fonseca, B.P.; Vieira-de-Abreu, A.; Barboza, B.A.; Robbs, B.K.; Bozza, P.T.; Viola, J.P. IFN-gamma production by CD8+ T cells depends on NFAT1 transcription factor and regulates Th differentiation. J. Immunol. 2005, 175, 5931–5939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bevington, S.L.; Cauchy, P.; Piper, J.; Bertrand, E.; Lalli, N.; Jarvis, R.C.; Gilding, L.N.; Ott, S.; Bonifer, C.; Cockerill, P.N. Inducible chromatin priming is associated with the establishment of immunological memory in T cells. EMBO J. 2016, 35, 515–535. [Google Scholar] [CrossRef] [PubMed]
- Rooney, J.W.; Sun, Y.L.; Glimcher, L.H.; Hoey, T. Novel NFAT sites that mediate activation of the interleukin-2 promoter in response to T-cell receptor stimulation. Mol. Cell. Biol. 1995, 15, 6299–6310. [Google Scholar] [CrossRef] [Green Version]
- McGuire, K.L.; Iacobelli, M. Involvement of Rel, Fos, and Jun proteins in binding activity to the IL-2 promoter CD28 response element/AP-1 sequence in human T cells. J. Immunol. 1997, 159, 1319–1327. [Google Scholar]
- Nguyen, T.N.; Kim, L.J.; Walters, R.D.; Drullinger, L.F.; Lively, T.N.; Kugel, J.F.; Goodrich, J.A. The C-terminal region of human NFATc2 binds cJun to synergistically activate interleukin-2 transcription. Mol. Immunol. 2010, 47, 2314–2322. [Google Scholar] [CrossRef] [Green Version]
- Walters, R.D.; Drullinger, L.F.; Kugel, J.F.; Goodrich, J.A. NFATc2 recruits cJun homodimers to an NFAT site to synergistically activate interleukin-2 transcription. Mol. Immunol. 2013, 56, 48–56. [Google Scholar] [CrossRef] [Green Version]
- Yu, B.; Zhang, K.; Milner, J.J.; Toma, C.; Chen, R.; Scott-Browne, J.P.; Pereira, R.M.; Crotty, S.; Chang, J.T.; Pipkin, M.E.; et al. Epigenetic landscapes reveal transcription factors that regulate CD8+ T cell differentiation. Nat. Immunol. 2017, 18, 573–582. [Google Scholar] [CrossRef] [Green Version]
- Moskowitz, D.M.; Zhang, D.W.; Hu, B.; Le Saux, S.; Yanes, R.E.; Ye, Z.; Buenrostro, J.D.; Weyand, C.M.; Greenleaf, W.J.; Goronzy, J.J. Epigenomics of human CD8 T cell differentiation and aging. Sci. Immunol. 2017, 2, eaag0192. [Google Scholar] [CrossRef] [Green Version]
- Martinez, G.J.; Pereira, R.M.; Aijo, T.; Kim, E.Y.; Marangoni, F.; Pipkin, M.E.; Togher, S.; Heissmeyer, V.; Zhang, Y.C.; Crotty, S.; et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 2015, 42, 265–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mognol, G.P.; Spreafico, R.; Wong, V.; Scott-Browne, J.P.; Togher, S.; Hoffmann, A.; Hogan, P.G.; Rao, A.; Trifari, S. Exhaustion-associated regulatory regions in CD8+ tumor-infiltrating T cells. Proc. Natl. Acad. Sci. USA 2017, 114, E2776–E2785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Z.; Liu, S.; Zhang, B.; Qiao, L.; Zhang, Y.; Zhang, Y. T Cell Dysfunction and Exhaustion in Cancer. Front. Cell Dev. Biol. 2020, 8, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schietinger, A.; Philip, M.; Krisnawan, V.E.; Chiu, E.Y.; Delrow, J.J.; Basom, R.S.; Lauer, P.; Brockstedt, D.G.; Knoblaugh, S.E.; Hammerling, G.J.; et al. Tumor-Specific T Cell Dysfunction Is a Dynamic Antigen-Driven Differentiation Program Initiated Early during Tumorigenesis. Immunity 2016, 45, 389–401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khan, O.; Giles, J.R.; McDonald, S.; Manne, S.; Ngiow, S.F.; Patel, K.P.; Werner, M.T.; Huang, A.C.; Alexander, K.A.; Wu, J.E.; et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 2019, 571, 211–218. [Google Scholar] [CrossRef] [PubMed]
- Yao, C.; Sun, H.W.; Lacey, N.E.; Ji, Y.; Moseman, E.A.; Shih, H.Y.; Heuston, E.F.; Kirby, M.; Anderson, S.; Cheng, J.; et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 2019, 20, 890–901. [Google Scholar] [CrossRef] [PubMed]
- Alfei, F.; Kanev, K.; Hofmann, M.; Wu, M.; Ghoneim, H.E.; Roelli, P.; Utzschneider, D.T.; von Hoesslin, M.; Cullen, J.G.; Fan, Y.; et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 2019, 571, 265–269. [Google Scholar] [CrossRef]
- Scott, A.C.; Dundar, F.; Zumbo, P.; Chandran, S.S.; Klebanoff, C.A.; Shakiba, M.; Trivedi, P.; Menocal, L.; Appleby, H.; Camara, S.; et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 2019, 571, 270–274. [Google Scholar] [CrossRef]
- Chen, J.; Lopez-Moyado, I.F.; Seo, H.; Lio, C.J.; Hempleman, L.J.; Sekiya, T.; Yoshimura, A.; Scott-Browne, J.P.; Rao, A. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 2019, 567, 530–534. [Google Scholar] [CrossRef]
- Lynn, R.C.; Weber, E.W.; Sotillo, E.; Gennert, D.; Xu, P.; Good, Z.; Anbunathan, H.; Lattin, J.; Jones, R.; Tieu, V.; et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 2019, 576, 293–300. [Google Scholar] [CrossRef]
- Seo, H.; Chen, J.; Gonzalez-Avalos, E.; Samaniego-Castruita, D.; Das, A.; Wang, Y.H.; Lopez-Moyado, I.F.; Georges, R.O.; Zhang, W.; Onodera, A.; et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl. Acad. Sci. USA 2019, 116, 12410–12415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ye, N.; Ding, Y.; Wild, C.; Shen, Q.; Zhou, J. Small molecule inhibitors targeting activator protein 1 (AP-1). J. Med. Chem. 2014, 57, 6930–6948. [Google Scholar] [CrossRef] [PubMed]
- Cohen, A.J.; Saiakhova, A.; Corradin, O.; Luppino, J.M.; Lovrenert, K.; Bartels, C.F.; Morrow, J.J.; Mack, S.C.; Dhillon, G.; Beard, L.; et al. Hotspots of aberrant enhancer activity punctuate the colorectal cancer epigenome. Nat. Commun. 2017, 8, 14400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Franco, H.L.; Nagari, A.; Malladi, V.S.; Li, W.; Xi, Y.; Richardson, D.; Allton, K.L.; Tanaka, K.; Li, J.; Murakami, S.; et al. Enhancer transcription reveals subtype-specific gene expression programs controlling breast cancer pathogenesis. Genome Res. 2018, 28, 159–170. [Google Scholar] [CrossRef] [PubMed]
- Oh, I.Y.; Albea, D.M.; Goodwin, Z.A.; Quiggle, A.M.; Baker, B.P.; Guggisberg, A.M.; Geahlen, J.H.; Kroner, G.M.; de Guzman Strong, C. Regulation of the dynamic chromatin architecture of the epidermal differentiation complex is mediated by a c-Jun/AP-1-modulated enhancer. J. Investig. Dermatol. 2014, 134, 2371–2380. [Google Scholar] [CrossRef] [Green Version]
- Vierbuchen, T.; Ling, E.; Cowley, C.J.; Couch, C.H.; Wang, X.; Harmin, D.A.; Roberts, C.W.M.; Greenberg, M.E. AP-1 Transcription Factors and the BAF Complex Mediate Signal-Dependent Enhancer Selection. Mol. Cell 2017, 68, 1067–1082.e12. [Google Scholar] [CrossRef] [Green Version]
- Zanconato, F.; Forcato, M.; Battilana, G.; Azzolin, L.; Quaranta, E.; Bodega, B.; Rosato, A.; Bicciato, S.; Cordenonsi, M.; Piccolo, S. Genome-wide association between YAP/TAZ/TEAD and AP-1 at enhancers drives oncogenic growth. Nat. Cell Biol. 2015, 17, 1218–1227. [Google Scholar] [CrossRef]
- Gazon, H.; Barbeau, B.; Mesnard, J.M.; Peloponese, J.M., Jr. Hijacking of the AP-1 Signaling Pathway during Development of ATL. Front. Microbiol. 2017, 8, 2686. [Google Scholar] [CrossRef] [Green Version]
- Bozec, A.; Bakiri, L.; Jimenez, M.; Schinke, T.; Amling, M.; Wagner, E.F. Fra-2/AP-1 controls bone formation by regulating osteoblast differentiation and collagen production. J. Cell Biol. 2010, 190, 1093–1106. [Google Scholar] [CrossRef] [Green Version]
- Obier, N.; Cauchy, P.; Assi, S.A.; Gilmour, J.; Lie, A.L.M.; Lichtinger, M.; Hoogenkamp, M.; Noailles, L.; Cockerill, P.N.; Lacaud, G.; et al. Cooperative binding of AP-1 and TEAD4 modulates the balance between vascular smooth muscle and hemogenic cell fate. Development 2016, 143, 4324–4340. [Google Scholar] [CrossRef] [Green Version]
- Malik, A.N.; Vierbuchen, T.; Hemberg, M.; Rubin, A.A.; Ling, E.; Couch, C.H.; Stroud, H.; Spiegel, I.; Farh, K.K.; Harmin, D.A.; et al. Genome-wide identification and characterization of functional neuronal activity-dependent enhancers. Nat. Neurosci. 2014, 17, 1330–1339. [Google Scholar] [CrossRef] [PubMed]
- Su, Y.; Shin, J.; Zhong, C.; Wang, S.; Roychowdhury, P.; Lim, J.; Kim, D.; Ming, G.L.; Song, H. Neuronal activity modifies the chromatin accessibility landscape in the adult brain. Nat. Neurosci. 2017, 20, 476–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roychoudhuri, R.; Clever, D.; Li, P.; Wakabayashi, Y.; Quinn, K.M.; Klebanoff, C.A.; Ji, Y.; Sukumar, M.; Eil, R.L.; Yu, Z.; et al. BACH2 regulates CD8+ T cell differentiation by controlling access of AP-1 factors to enhancers. Nat. Immunol. 2016, 17, 851–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kappelmann, M.; Bosserhoff, A.; Kuphal, S. AP-1/c-Jun transcription factors: Regulation and function in malignant melanoma. Eur. J. Cell Biol. 2014, 93, 76–81. [Google Scholar] [CrossRef]
- Shaulian, E. AP-1—The Jun proteins: Oncogenes or tumor suppressors in disguise? Cell. Signal. 2010, 22, 894–899. [Google Scholar] [CrossRef] [PubMed]
- El-Fattah Ibrahim, S.A.; Abudu, A.; Johnson, E.; Aftab, N.; Conrad, S.; Fluck, M. Correction: The role of AP-1 in self-sufficient proliferation and migration of cancer cells and its potential impact on an autocrine/paracrine loop. Oncotarget 2019, 10, 799. [Google Scholar] [CrossRef]
- Chang, H.; Liu, Y.; Xue, M.; Liu, H.; Du, S.; Zhang, L.; Wang, P. Synergistic action of master transcription factors controls epithelial-to-mesenchymal transition. Nucleic Acids Res. 2016, 44, 2514–2527. [Google Scholar] [CrossRef] [Green Version]
- Morrow, J.J.; Bayles, I.; Funnell, A.P.W.; Miller, T.E.; Saiakhova, A.; Lizardo, M.M.; Bartels, C.F.; Kapteijn, M.Y.; Hung, S.; Mendoza, A.; et al. Positively selected enhancer elements endow osteosarcoma cells with metastatic competence. Nat. Med. 2018, 24, 176–185. [Google Scholar] [CrossRef]
- Atsaves, V.; Leventaki, V.; Rassidakis, G.Z.; Claret, F.X. AP-1 Transcription Factors as Regulators of Immune Responses in Cancer. Cancers 2019, 11, 1037. [Google Scholar] [CrossRef] [Green Version]
- Shaffer, S.M.; Dunagin, M.C.; Torborg, S.R.; Torre, E.A.; Emert, B.; Krepler, C.; Beqiri, M.; Sproesser, K.; Brafford, P.A.; Xiao, M.; et al. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature 2017, 546, 431–435. [Google Scholar] [CrossRef] [Green Version]
- Halazonetis, T.D.; Georgopoulos, K.; Greenberg, M.E.; Leder, P. c-Jun dimerizes with itself and with c-Fos, forming complexes of different DNA binding affinities. Cell 1988, 55, 917–924. [Google Scholar] [CrossRef]
- Karamouzis, M.V.; Konstantinopoulos, P.A.; Papavassiliou, A.G. The activator protein-1 transcription factor in respiratory epithelium carcinogenesis. Mol. Cancer Res. 2007, 5, 109–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papavassiliou, A.G.; Treier, M.; Chavrier, C.; Bohmann, D. Targeted degradation of c-Fos, but not v-Fos, by a phosphorylation-dependent signal on c-Jun. Science 1992, 258, 1941–1944. [Google Scholar] [CrossRef] [PubMed]
- Meng, Q.; Xia, Y. c-Jun, at the crossroad of the signaling network. Protein Cell 2011, 2, 889–898. [Google Scholar] [CrossRef] [Green Version]
- Wagner, E.F.; Schonthaler, H.B.; Guinea-Viniegra, J.; Tschachler, E. Psoriasis: What we have learned from mouse models. Nat. Rev. Rheumatol. 2010, 6, 704–714. [Google Scholar] [CrossRef]
- Yadav, S.; Kalra, N.; Ganju, L.; Singh, M. Activator protein-1 (AP-1): A bridge between life and death in lung epithelial (A549) cells under hypoxia. Mol. Cell. Biochem. 2017, 436, 99–110. [Google Scholar] [CrossRef]
- Chiu, R.; Boyle, W.J.; Meek, J.; Smeal, T.; Hunter, T.; Karin, M. The c-Fos protein interacts with c-Jun/AP-1 to stimulate transcription of AP-1 responsive genes. Cell 1988, 54, 541–552. [Google Scholar] [CrossRef]
- Hai, T.; Curran, T. Cross-family dimerization of transcription factors Fos/Jun and ATF/CREB alters DNA binding specificity. Proc. Natl. Acad. Sci. USA 1991, 88, 3720–3724. [Google Scholar] [CrossRef] [Green Version]
- Van Dam, H.; Duyndam, M.; Rottier, R.; Bosch, A.; de Vries-Smits, L.; Herrlich, P.; Zantema, A.; Angel, P.; van der Eb, A.J. Heterodimer formation of cJun and ATF-2 is responsible for induction of c-jun by the 243 amino acid adenovirus E1A protein. EMBO J. 1993, 12, 479–487. [Google Scholar] [CrossRef]
- Vesely, P.W.; Staber, P.B.; Hoefler, G.; Kenner, L. Translational regulation mechanisms of AP-1 proteins. Mutat. Res. 2009, 682, 7–12. [Google Scholar] [CrossRef]
- Fu, L.; Peng, S.; Wu, W.; Ouyang, Y.; Tan, D.; Fu, X. LncRNA HOTAIRM1 promotes osteogenesis by controlling JNK/AP-1 signalling-mediated RUNX2 expression. J. Cell. Mol. Med. 2019, 23, 7517–7524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Knirsh, R.; Ben-Dror, I.; Modai, S.; Shomron, N.; Vardimon, L. MicroRNA 10b promotes abnormal expression of the proto-oncogene c-Jun in metastatic breast cancer cells. Oncotarget 2016, 7, 59932–59944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papavassiliou, A.G.; Chavrier, C.; Bohmann, D. Phosphorylation state and DNA-binding activity of c-Jun depend on the intracellular concentration of binding sites. Proc. Natl. Acad. Sci. USA 1992, 89, 11562–11565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vries, R.G.; Prudenziati, M.; Zwartjes, C.; Verlaan, M.; Kalkhoven, E.; Zantema, A. A specific lysine in c-Jun is required for transcriptional repression by E1A and is acetylated by p300. EMBO J. 2001, 20, 6095–6103. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, D.; Muller, S. Members of the PIAS family act as SUMO ligases for c-Jun and p53 and repress p53 activity. Proc. Natl. Acad. Sci. USA 2002, 99, 2872–2877. [Google Scholar] [CrossRef] [Green Version]
- Muller, S.; Berger, M.; Lehembre, F.; Seeler, J.S.; Haupt, Y.; Dejean, A. c-Jun and p53 activity is modulated by SUMO-1 modification. J. Biol. Chem. 2000, 275, 13321–13329. [Google Scholar] [CrossRef] [Green Version]
- Bossis, G.; Malnou, C.E.; Farras, R.; Andermarcher, E.; Hipskind, R.; Rodriguez, M.; Schmidt, D.; Muller, S.; Jariel-Encontre, I.; Piechaczyk, M. Down-regulation of c-Fos/c-Jun AP-1 dimer activity by sumoylation. Mol. Cell. Biol. 2005, 25, 6964–6979. [Google Scholar] [CrossRef] [Green Version]
- 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 Dev. 1993, 7, 2135–2148. [Google Scholar] [CrossRef] [Green Version]
- Smeal, T.; Binetruy, B.; Mercola, D.A.; Birrer, M.; Karin, M. Oncogenic and transcriptional cooperation with Ha-Ras requires phosphorylation of c-Jun on serines 63 and 73. Nature 1991, 354, 494–496. [Google Scholar] [CrossRef]
- Morton, S.; Davis, R.J.; McLaren, A.; Cohen, P. A reinvestigation of the multisite phosphorylation of the transcription factor c-Jun. EMBO J. 2003, 22, 3876–3886. [Google Scholar] [CrossRef] [Green Version]
- Papavassiliou, A.G.; Treier, M.; Bohmann, D. Intramolecular signal transduction in c-Jun. EMBO J. 1995, 14, 2014–2019. [Google Scholar] [CrossRef] [PubMed]
- Reddy, C.E.; Albanito, L.; De Marco, P.; Aiello, D.; Maggiolini, M.; Napoli, A.; Musti, A.M. Multisite phosphorylation of c-Jun at threonine 91/93/95 triggers the onset of c-Jun pro-apoptotic activity in cerebellar granule neurons. Cell Death Dis. 2013, 4, e852. [Google Scholar] [CrossRef] [PubMed]
- Vinciguerra, M.; Esposito, I.; Salzano, S.; Madeo, A.; Nagel, G.; Maggiolini, M.; Gallo, A.; Musti, A.M. Negative charged threonine 95 of c-Jun is essential for c-Jun N-terminal kinase-dependent phosphorylation of threonine 91/93 and stress-induced c-Jun biological activity. Int. J. Biochem. Cell Biol. 2008, 40, 307–316. [Google Scholar] [CrossRef] [PubMed]
- Wei, W.; Jin, J.; Schlisio, S.; Harper, J.W.; Kaelin, W.G., Jr. 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. [Google Scholar] [CrossRef] [Green Version]
- Liu, S.; Yu, S.; Hasegawa, Y.; Lapushin, R.; Xu, H.J.; Woodgett, J.R.; Mills, G.B.; Fang, X. Glycogen synthase kinase 3beta is a negative regulator of growth factor-induced activation of the c-Jun N-terminal kinase. J. Biol. Chem. 2004, 279, 51075–51081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, A.; Frost, J.; Deng, T.; Smeal, T.; al-Alawi, N.; Kikkawa, U.; Hunter, T.; Brenner, D.; Karin, M. Casein kinase II is a negative regulator of c-Jun DNA binding and AP-1 activity. Cell 1992, 70, 777–789. [Google Scholar] [CrossRef]
- Huang, C.C.; Wang, J.M.; Kikkawa, U.; Mukai, H.; Shen, M.R.; Morita, I.; Chen, B.K.; Chang, W.C. Calcineurin-mediated dephosphorylation of c-Jun Ser-243 is required for c-Jun protein stability and cell transformation. Oncogene 2008, 27, 2422–2429. [Google Scholar] [CrossRef] [Green Version]
- Treier, M.; Staszewski, L.M.; Bohmann, D. Ubiquitin-dependent c-Jun degradation in vivo is mediated by the delta domain. Cell 1994, 78, 787–798. [Google Scholar] [CrossRef]
- Nateri, A.S.; Riera-Sans, L.; Da Costa, C.; Behrens, A. The ubiquitin ligase SCFFbw7 antagonizes apoptotic JNK signaling. Science 2004, 303, 1374–1378. [Google Scholar] [CrossRef] [Green Version]
- Karin, M.; Liu, Z.; Zandi, E. AP-1 function and regulation. Curr. Opin. Cell Biol. 1997, 9, 240–246. [Google Scholar] [CrossRef]
- Woodgett, J.R.; Pulverer, B.J.; Nikolakaki, E.; Plyte, S.; Hughes, K.; Franklin, C.C.; Kraft, A.S. Regulation of jun/AP-1 oncoproteins by protein phosphorylation. Adv. Second Messenger Phosphoprot. Res. 1993, 28, 261–269. [Google Scholar]
- Weiss, C.; Schneider, S.; Wagner, E.F.; Zhang, X.; Seto, E.; Bohmann, D. JNK phosphorylation relieves HDAC3-dependent suppression of the transcriptional activity of c-Jun. EMBO J. 2003, 22, 3686–3695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bannister, A.J.; Oehler, T.; Wilhelm, D.; Angel, P.; Kouzarides, T. Stimulation of c-Jun activity by CBP: C-Jun residues Ser63/73 are required for CBP induced stimulation in vivo and CBP binding in vitro. Oncogene 1995, 11, 2509–2514. [Google Scholar] [PubMed]
- Bitomsky, N.; Bohm, M.; Klempnauer, K.H. Transformation suppressor protein Pdcd4 interferes with JNK-mediated phosphorylation of c-Jun and recruitment of the coactivator p300 by c-Jun. Oncogene 2004, 23, 7484–7493. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.S.; See, R.H.; Deng, T.; Shi, Y. Adenovirus E1A downregulates cJun- and JunB-mediated transcription by targeting their coactivator p300. Mol. Cell. Biol. 1996, 16, 4312–4326. [Google Scholar] [CrossRef] [Green Version]
- Sanna, M.D.; Galeotti, N. The HDAC1/c-JUN complex is essential in the promotion of nerve injury-induced neuropathic pain through JNK signaling. Eur. J. Pharmacol. 2018, 825, 99–106. [Google Scholar] [CrossRef]
- Nateri, A.S.; Spencer-Dene, B.; Behrens, A. Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development. Nature 2005, 437, 281–285. [Google Scholar] [CrossRef]
- Aguilera, C.; Nakagawa, K.; Sancho, R.; Chakraborty, A.; Hendrich, B.; Behrens, A. c-Jun N-terminal phosphorylation antagonises recruitment of the Mbd3/NuRD repressor complex. Nature 2011, 469, 231–235. [Google Scholar] [CrossRef]
- Boyle, W.J.; Smeal, T.; Defize, L.H.; Angel, P.; Woodgett, J.R.; Karin, M.; Hunter, T. Activation of protein kinase C decreases phosphorylation of c-Jun at sites that negatively regulate its DNA-binding activity. Cell 1991, 64, 573–584. [Google Scholar] [CrossRef]
- Marchand, B.; Tremblay, I.; Cagnol, S.; Boucher, M.J. Inhibition of glycogen synthase kinase-3 activity triggers an apoptotic response in pancreatic cancer cells through JNK-dependent mechanisms. Carcinogenesis 2012, 33, 529–537. [Google Scholar] [CrossRef]
- Musti, A.M.; Treier, M.; Bohmann, D. Reduced ubiquitin-dependent degradation of c-Jun after phosphorylation by MAP kinases. Science 1997, 275, 400–402. [Google Scholar] [CrossRef] [PubMed]
- Sabapathy, K.; Hochedlinger, K.; Nam, S.Y.; Bauer, A.; Karin, M.; Wagner, E.F. Distinct roles for JNK1 and JNK2 in regulating JNK activity and c-Jun-dependent cell proliferation. Mol. Cell 2004, 15, 713–725. [Google Scholar] [CrossRef] [PubMed]
- Coffey, E.T. Nuclear and cytosolic JNK signalling in neurons. Nat. Rev. Neurosci. 2014, 15, 285–299. [Google Scholar] [CrossRef] [PubMed]
- Ye, X.; Nalepa, G.; Welcker, M.; Kessler, B.M.; Spooner, E.; Qin, J.; Elledge, S.J.; Clurman, B.E.; Harper, J.W. Recognition of phosphodegron motifs in human cyclin E by the SCF(Fbw7) ubiquitin ligase. J. Biol. Chem. 2004, 279, 50110–50119. [Google Scholar] [CrossRef] [Green Version]
- Conze, D.; Krahl, T.; Kennedy, N.; Weiss, L.; Lumsden, J.; Hess, P.; Flavell, R.A.; Le Gros, G.; Davis, R.J.; Rincon, M. c-Jun NH(2)-terminal kinase (JNK)1 and JNK2 have distinct roles in CD8+ T cell activation. J. Exp. Med. 2002, 195, 811–823. [Google Scholar] [CrossRef] [Green Version]
- Ameyar, M.; Wisniewska, M.; Weitzman, J.B. A role for AP-1 in apoptosis: The case for and against. Biochimie 2003, 85, 747–752. [Google Scholar] [CrossRef]
- Bossy-Wetzel, E.; Bakiri, L.; Yaniv, M. Induction of apoptosis by the transcription factor c-Jun. EMBO J. 1997, 16, 1695–1709. [Google Scholar] [CrossRef] [Green Version]
- Wisdom, R.; Johnson, R.S.; Moore, C. c-Jun regulates cell cycle progression and apoptosis by distinct mechanisms. EMBO J. 1999, 18, 188–197. [Google Scholar] [CrossRef] [Green Version]
- Albanito, L.; Reddy, C.E.; Musti, A.M. c-Jun is essential for the induction of Il-1beta gene expression in in vitro activated Bergmann glial cells. Glia 2011, 59, 1879–1890. [Google Scholar] [CrossRef]
- Madeo, A.; Vinciguerra, M.; Lappano, R.; Galgani, M.; Gasperi-Campani, A.; Maggiolini, M.; Musti, A.M. c-Jun activation is required for 4-hydroxytamoxifen-induced cell death in breast cancer cells. Oncogene 2010, 29, 978–991. [Google Scholar] [CrossRef] [Green Version]
- Ansell, S.M.; Lesokhin, A.M.; Borrello, I.; Halwani, A.; Scott, E.C.; Gutierrez, M.; Schuster, S.J.; Millenson, M.M.; Cattry, D.; Freeman, G.J.; et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 2015, 372, 311–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, L.; Flies, D.B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242. [Google Scholar] [CrossRef] [PubMed]
- Rizvi, N.A.; Mazieres, J.; Planchard, D.; Stinchcombe, T.E.; Dy, G.K.; Antonia, S.J.; Horn, L.; Lena, H.; Minenza, E.; Mennecier, B.; et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): A phase 2, single-arm trial. Lancet Oncol. 2015, 16, 257–265. [Google Scholar] [CrossRef] [Green Version]
- Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; Maio, M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; Kalinka-Warzocha, E.; et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 2015, 372, 320–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baccam, M.; Woo, S.Y.; Vinson, C.; Bishop, G.A. CD40-mediated transcriptional regulation of the IL-6 gene in B lymphocytes: Involvement of NF-κB, AP-1, and C/EBP. J. Immunol. 2003, 170, 3099–3108. [Google Scholar] [CrossRef] [Green Version]
- Jiang, X.; Zhou, J.; Giobbie-Hurder, A.; Wargo, J.; Hodi, F.S. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD-L1 expression that is reversible by MEK and PI3K inhibition. Clin. Cancer Res. 2013, 19, 598–609. [Google Scholar] [CrossRef] [Green Version]
- Papavassiliou, K.A.; Papavassiliou, A.G. Transcription Factor Drug Targets. J. Cell. Biochem. 2016, 117, 2693–2696. [Google Scholar] [CrossRef]
- Kaech, S.M.; Tan, J.T.; Wherry, E.J.; Konieczny, B.T.; Surh, C.D.; Ahmed, R. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat. Immunol. 2003, 4, 1191–1198. [Google Scholar] [CrossRef]
- Joshi, N.S.; Cui, W.; Chandele, A.; Lee, H.K.; Urso, D.R.; Hagman, J.; Gapin, L.; Kaech, S.M. Inflammation directs memory precursor and short-lived effector CD8+ T cell fates via the graded expression of T-bet transcription factor. Immunity 2007, 27, 281–295. [Google Scholar] [CrossRef] [Green Version]
- Sarkar, S.; Kalia, V.; Haining, W.N.; Konieczny, B.T.; Subramaniam, S.; Ahmed, R. Functional and genomic profiling of effector CD8 T cell subsets with distinct memory fates. J. Exp. Med. 2008, 205, 625–640. [Google Scholar] [CrossRef] [Green Version]
- Cui, W.; Kaech, S.M. Generation of effector CD8+ T cells and their conversion to memory T cells. Immunol. Rev. 2010, 236, 151–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harding, F.A.; McArthur, J.G.; Gross, J.A.; Raulet, D.H.; Allison, J.P. CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones. Nature 1992, 356, 607–609. [Google Scholar] [CrossRef] [PubMed]
- Takemoto, N.; Intlekofer, A.M.; Northrup, J.T.; Wherry, E.J.; Reiner, S.L. Cutting Edge: IL-12 inversely regulates T-bet and eomesodermin expression during pathogen-induced CD8+ T cell differentiation. J. Immunol. 2006, 177, 7515–7519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agarwal, P.; Raghavan, A.; Nandiwada, S.L.; Curtsinger, J.M.; Bohjanen, P.R.; Mueller, D.L.; Mescher, M.F. Gene regulation and chromatin remodeling by IL-12 and type I IFN in programming for CD8 T cell effector function and memory. J. Immunol. 2009, 183, 1695–1704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rao, R.R.; Li, Q.; Odunsi, K.; Shrikant, P.A. The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin. Immunity 2010, 32, 67–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, J.T.; Wherry, E.J.; Goldrath, A.W. Molecular regulation of effector and memory T cell differentiation. Nat. Immunol. 2014, 15, 1104–1115. [Google Scholar] [CrossRef] [PubMed]
- Russ, B.E.; Olshanksy, M.; Smallwood, H.S.; Li, J.; Denton, A.E.; Prier, J.E.; Stock, A.T.; Croom, H.A.; Cullen, J.G.; Nguyen, M.L.; et al. Distinct epigenetic signatures delineate transcriptional programs during virus-specific CD8+ T cell differentiation. Immunity 2014, 41, 853–865. [Google Scholar] [CrossRef] [Green Version]
- Henning, A.N.; Roychoudhuri, R.; Restifo, N.P. Epigenetic control of CD8+ T cell differentiation. Nat. Rev. Immunol. 2018, 18, 340–356. [Google Scholar] [CrossRef]
- Vaeth, M.; Feske, S. NFAT control of immune function: New Frontiers for an Abiding Trooper. F1000Research 2018, 7, 260. [Google Scholar] [CrossRef] [Green Version]
- Vaeth, M.; Maus, M.; Klein-Hessling, S.; Freinkman, E.; Yang, J.; Eckstein, M.; Cameron, S.; Turvey, S.E.; Serfling, E.; Berberich-Siebelt, F.; et al. Store-Operated Ca2+ Entry Controls Clonal Expansion of T Cells through Metabolic Reprogramming. Immunity 2017, 47, 664–679.e6. [Google Scholar] [CrossRef] [Green Version]
- Gwack, Y.; Feske, S.; Srikanth, S.; Hogan, P.G.; Rao, A. Signalling to transcription: Store-operated Ca2+ entry and NFAT activation in lymphocytes. Cell Calcium 2007, 42, 145–156. [Google Scholar] [CrossRef] [PubMed]
- Brignall, R.; Cauchy, P.; Bevington, S.L.; Gorman, B.; Pisco, A.O.; Bagnall, J.; Boddington, C.; Rowe, W.; England, H.; Rich, K.; et al. Integration of Kinase and Calcium Signaling at the Level of Chromatin Underlies Inducible Gene Activation in T Cells. J. Immunol. 2017, 199, 2652–2667. [Google Scholar] [CrossRef] [PubMed]
- Boomer, J.S.; Green, J.M. An enigmatic tail of CD28 signaling. Cold Spring Harb. Perspect. Biol. 2010, 2, a002436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Esensten, J.H.; Helou, Y.A.; Chopra, G.; Weiss, A.; Bluestone, J.A. CD28 Costimulation: From Mechanism to Therapy. Immunity 2016, 44, 973–988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okkenhaug, K.; Rottapel, R. Grb2 forms an inducible protein complex with CD28 through a Src homology 3 domain-proline interaction. J. Biol. Chem. 1998, 273, 21194–21202. [Google Scholar] [CrossRef] [Green Version]
- Kaminuma, O.; Deckert, M.; Elly, C.; Liu, Y.C.; Altman, A. Vav-Rac1-mediated activation of the c-Jun N-terminal kinase/c-Jun/AP-1 pathway plays a major role in stimulation of the distal NFAT site in the interleukin-2 gene promoter. Mol. Cell. Biol. 2001, 21, 3126–3136. [Google Scholar] [CrossRef] [Green Version]
- Coso, O.A.; Chiariello, M.; Yu, J.C.; Teramoto, H.; Crespo, P.; Xu, N.; Miki, T.; Gutkind, J.S. The small GTP-binding proteins Rac1 and Cdc42 regulate the activity of the JNK/SAPK signaling pathway. Cell 1995, 81, 1137–1146. [Google Scholar] [CrossRef] [Green Version]
- Cockerill, P.N.; Bert, A.G.; Jenkins, F.; Ryan, G.R.; Shannon, M.F.; Vadas, M.A. Human granulocyte-macrophage colony-stimulating factor enhancer function is associated with cooperative interactions between AP-1 and NFATp/c. Mol. Cell. Biol. 1995, 15, 2071–2079. [Google Scholar] [CrossRef] [Green Version]
- Macian, F.; Lopez-Rodriguez, C.; Rao, A. Partners in transcription: NFAT and AP-1. Oncogene 2001, 20, 2476–2489. [Google Scholar] [CrossRef] [Green Version]
- Tsai, E.Y.; Jain, J.; Pesavento, P.A.; Rao, A.; Goldfeld, A.E. Tumor necrosis factor alpha gene regulation in activated T cells involves ATF-2/Jun and NFATp. Mol. Cell. Biol. 1996, 16, 459–467. [Google Scholar] [CrossRef] [Green Version]
- Klein-Hessling, S.; Muhammad, K.; Klein, M.; Pusch, T.; Rudolf, R.; Floter, J.; Qureischi, M.; Beilhack, A.; Vaeth, M.; Kummerow, C.; et al. NFATc1 controls the cytotoxicity of CD8+ T cells. Nat. Commun. 2017, 8, 511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnson, B.V.; Bert, A.G.; Ryan, G.R.; Condina, A.; Cockerill, P.N. Granulocyte-macrophage colony-stimulating factor enhancer activation requires cooperation between NFAT and AP-1 elements and is associated with extensive nucleosome reorganization. Mol. Cell. Biol. 2004, 24, 7914–7930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buenrostro, J.D.; Wu, B.; Chang, H.Y.; Greenleaf, W.J. ATAC-seq: A Method for Assaying Chromatin Accessibility Genome-Wide. Curr. Protoc. Mol. Biol. 2015, 109, 21.29.1–21.29.9. [Google Scholar] [CrossRef] [PubMed]
- Cockerill, P.N. Receptor Signaling Directs Global Recruitment of Pre-Existing Transcription Factors to Inducible Elements. Yale J. Biol. Med. 2016, 89, 591–596. [Google Scholar]
- Yukawa, M.; Jagannathan, S.; Vallabh, S.; Kartashov, A.V.; Chen, X.; Weirauch, M.T.; Barski, A. AP-1 activity induced by co-stimulation is required for chromatin opening during T cell activation. J. Exp. Med. 2020, 217, e20182009. [Google Scholar] [CrossRef]
- Scharer, C.D.; Bally, A.P.; Gandham, B.; Boss, J.M. Cutting Edge: Chromatin Accessibility Programs CD8 T Cell Memory. J. Immunol. 2017, 198, 2238–2243. [Google Scholar] [CrossRef] [Green Version]
- Feau, S.; Schoenberger, S.P.; Altman, A.; Becart, S. SLAT regulates CD8+ T cell clonal expansion in a Cdc42- and NFAT1-dependent manner. J. Immunol. 2013, 190, 174–183. [Google Scholar] [CrossRef] [Green Version]
- Man, K.; Gabriel, S.S.; Liao, Y.; Gloury, R.; Preston, S.; Henstridge, D.C.; Pellegrini, M.; Zehn, D.; Berberich-Siebelt, F.; Febbraio, M.A.; et al. Transcription Factor IRF4 Promotes CD8+ T Cell Exhaustion and Limits the Development of Memory-like T Cells during Chronic Infection. Immunity 2017, 47, 1129–1141.e5. [Google Scholar] [CrossRef] [Green Version]
- Chen, Z.; Ji, Z.; Ngiow, S.F.; Manne, S.; Cai, Z.; Huang, A.C.; Johnson, J.; Staupe, R.P.; Bengsch, B.; Xu, C.; et al. TCF-1-Centered Transcriptional Network Drives an Effector versus Exhausted CD8 T Cell-Fate Decision. Immunity 2019, 51, 840–855. [Google Scholar] [CrossRef]
- Danilo, M.; Chennupati, V.; Silva, J.G.; Siegert, S.; Held, W. Suppression of Tcf1 by Inflammatory Cytokines Facilitates Effector CD8 T Cell Differentiation. Cell Rep. 2018, 22, 2107–2117. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Wang, Y.; Lu, H.; Li, J.; Yan, X.; Xiao, M.; Hao, J.; Alekseev, A.; Khong, H.; Chen, T.; et al. Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction. Nature 2019, 567, 525–529. [Google Scholar] [CrossRef]
- Redeker, A.; Welten, S.P.; Baert, M.R.; Vloemans, S.A.; Tiemessen, M.M.; Staal, F.J.; Arens, R. The Quantity of Autocrine IL-2 Governs the Expansion Potential of CD8+ T Cells. J. Immunol. 2015, 195, 4792–4801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalia, V.; Sarkar, S. Regulation of Effector and Memory CD8 T Cell Differentiation by IL-2-A Balancing Act. Front. Immunol. 2018, 9, 2987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ishihara, S.; Schwartz, R.H. Two-step binding of transcription factors causes sequential chromatin structural changes at the activated IL-2 promoter. J. Immunol. 2011, 187, 3292–3299. [Google Scholar] [CrossRef] [PubMed]
- Scott-Browne, J.P.; Lopez-Moyado, I.F.; Trifari, S.; Wong, V.; Chavez, L.; Rao, A.; Pereira, R.M. Dynamic Changes in Chromatin Accessibility Occur in CD8+ T Cells Responding to Viral Infection. Immunity 2016, 45, 1327–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xin, A.; Masson, F.; Liao, Y.; Preston, S.; Guan, T.; Gloury, R.; Olshansky, M.; Lin, J.X.; Li, P.; Speed, T.P.; et al. A molecular threshold for effector CD8+ T cell differentiation controlled by transcription factors Blimp-1 and T-bet. Nat. Immunol. 2016, 17, 422–432. [Google Scholar] [CrossRef]
- Wang, D.; Diao, H.; Getzler, A.J.; Rogal, W.; Frederick, M.A.; Milner, J.; Yu, B.; Crotty, S.; Goldrath, A.W.; Pipkin, M.E. The Transcription Factor Runx3 Establishes Chromatin Accessibility of cis-Regulatory Landscapes that Drive Memory Cytotoxic T Lymphocyte Formation. Immunity 2018, 48, 659–674.e6. [Google Scholar] [CrossRef] [Green Version]
- Mittrucker, H.W.; Matsuyama, T.; Grossman, A.; Kundig, T.M.; Potter, J.; Shahinian, A.; Wakeham, A.; Patterson, B.; Ohashi, P.S.; Mak, T.W. Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science 1997, 275, 540–543. [Google Scholar] [CrossRef]
- Man, K.; Miasari, M.; Shi, W.; Xin, A.; Henstridge, D.C.; Preston, S.; Pellegrini, M.; Belz, G.T.; Smyth, G.K.; Febbraio, M.A.; et al. The transcription factor IRF4 is essential for TCR affinity-mediated metabolic programming and clonal expansion of T cells. Nat. Immunol. 2013, 14, 1155–1165. [Google Scholar] [CrossRef]
- Murphy, T.L.; Tussiwand, R.; Murphy, K.M. Specificity through cooperation: BATF-IRF interactions control immune-regulatory networks. Nat. Rev. Immunol. 2013, 13, 499–509. [Google Scholar] [CrossRef]
- Glasmacher, E.; Agrawal, S.; Chang, A.B.; Murphy, T.L.; Zeng, W.; Vander Lugt, B.; Khan, A.A.; Ciofani, M.; Spooner, C.J.; Rutz, S.; et al. A genomic regulatory element that directs assembly and function of immune-specific AP-1-IRF complexes. Science 2012, 338, 975–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciofani, M.; Madar, A.; Galan, C.; Sellars, M.; Mace, K.; Pauli, F.; Agarwal, A.; Huang, W.; Parkhurst, C.N.; Muratet, M.; et al. A validated regulatory network for Th17 cell specification. Cell 2012, 151, 289–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, P.; Spolski, R.; Liao, W.; Wang, L.; Murphy, T.L.; Murphy, K.M.; Leonard, W.J. BATF-JUN is critical for IRF4-mediated transcription in T cells. Nature 2012, 490, 543–546. [Google Scholar] [CrossRef] [PubMed]
- Martinez, G.J.; Rao, A. Immunology. Cooperative transcription factor complexes in control. Science 2012, 338, 891–892. [Google Scholar] [CrossRef] [Green Version]
- Ito, T.; Yamauchi, M.; Nishina, M.; Yamamichi, N.; Mizutani, T.; Ui, M.; Murakami, M.; Iba, H. Identification of SWI.SNF complex subunit BAF60a as a determinant of the transactivation potential of Fos/Jun dimers. J. Biol. Chem. 2001, 276, 2852–2857. [Google Scholar] [CrossRef] [Green Version]
- Ndlovu, M.N.; Van Lint, C.; Van Wesemael, K.; Callebert, P.; Chalbos, D.; Haegeman, G.; Vanden Berghe, W. Hyperactivated NF-κB and AP-1 transcription factors promote highly accessible chromatin and constitutive transcription across the interleukin-6 gene promoter in metastatic breast cancer cells. Mol. Cell. Biol. 2009, 29, 5488–5504. [Google Scholar] [CrossRef] [Green Version]
- Shin, H.; Blackburn, S.D.; Intlekofer, A.M.; Kao, C.; Angelosanto, J.M.; Reiner, S.L.; Wherry, E.J. A role for the transcriptional repressor Blimp-1 in CD8+ T cell exhaustion during chronic viral infection. Immunity 2009, 31, 309–320. [Google Scholar] [CrossRef] [Green Version]
- Cornberg, M.; Kenney, L.L.; Chen, A.T.; Waggoner, S.N.; Kim, S.K.; Dienes, H.P.; Welsh, R.M.; Selin, L.K. Clonal exhaustion as a mechanism to protect against severe immunopathology and death from an overwhelming CD8 T cell response. Front. Immunol. 2013, 4, 475. [Google Scholar] [CrossRef] [Green Version]
- Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
- Attanasio, J.; Wherry, E.J. Costimulatory and Coinhibitory Receptor Pathways in Infectious Disease. Immunity 2016, 44, 1052–1068. [Google Scholar] [CrossRef] [Green Version]
- Blackburn, S.D.; Shin, H.; Haining, W.N.; Zou, T.; Workman, C.J.; Polley, A.; Betts, M.R.; Freeman, G.J.; Vignali, D.A.; Wherry, E.J. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 2009, 10, 29–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wherry, E.J.; Ha, S.J.; Kaech, S.M.; Haining, W.N.; Sarkar, S.; Kalia, V.; Subramaniam, S.; Blattman, J.N.; Barber, D.L.; Ahmed, R. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 2007, 27, 670–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paley, M.A.; Kroy, D.C.; Odorizzi, P.M.; Johnnidis, J.B.; Dolfi, D.V.; Barnett, B.E.; Bikoff, E.K.; Robertson, E.J.; Lauer, G.M.; Reiner, S.L.; et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 2012, 338, 1220–1225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef] [PubMed]
- Utzschneider, D.T.; Charmoy, M.; Chennupati, V.; Pousse, L.; Ferreira, D.P.; Calderon-Copete, S.; Danilo, M.; Alfei, F.; Hofmann, M.; Wieland, D.; et al. T Cell Factor 1-Expressing Memory-like CD8+ T Cells Sustain the Immune Response to Chronic Viral Infections. Immunity 2016, 45, 415–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, R.; Hou, S.; Liu, C.; Zhang, A.; Bai, Q.; Han, M.; Yang, Y.; Wei, G.; Shen, T.; Yang, X.; et al. Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 2016, 537, 412–428. [Google Scholar] [CrossRef]
- Mule, J.J.; Ettinghausen, S.E.; Spiess, P.J.; Shu, S.; Rosenberg, S.A. Antitumor efficacy of lymphokine-activated killer cells and recombinant interleukin-2 in vivo: Survival benefit and mechanisms of tumor escape in mice undergoing immunotherapy. Cancer Res. 1986, 46, 676–683. [Google Scholar]
- Rosenberg, S.A.; Spiess, P.; Lafreniere, R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 1986, 233, 1318–1321. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Packard, B.S.; Aebersold, P.M.; Solomon, D.; Topalian, S.L.; Toy, S.T.; Simon, P.; Lotze, M.T.; Yang, J.C.; Seipp, C.A.; et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N. Engl. J. Med. 1988, 319, 1676–1680. [Google Scholar] [CrossRef]
- Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107–1111. [Google Scholar] [CrossRef]
- Dudley, M.E.; Wunderlich, J.R.; Yang, J.C.; Hwu, P.; Schwartzentruber, D.J.; Topalian, S.L.; Sherry, R.M.; Marincola, F.M.; Leitman, S.F.; Seipp, C.A.; et al. A phase I study of nonmyeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes in patients with metastatic melanoma. J. Immunother. 2002, 25, 243–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galon, J.; Costes, A.; Sanchez-Cabo, F.; Kirilovsky, A.; Mlecnik, B.; Lagorce-Pages, C.; Tosolini, M.; Camus, M.; Berger, A.; Wind, P.; et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006, 313, 1960–1964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mahmoud, S.M.; Paish, E.C.; Powe, D.G.; Macmillan, R.D.; Grainge, M.J.; Lee, A.H.; Ellis, I.O.; Green, A.R. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J. Clin. Oncol. 2011, 29, 1949–1955. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Lachapelle, J.; Leung, S.; Gao, D.; Foulkes, W.D.; Nielsen, T.O. CD8+ lymphocyte infiltration is an independent favorable prognostic indicator in basal-like breast cancer. Breast Cancer Res. 2012, 14, R48. [Google Scholar] [CrossRef] [Green Version]
- Denkert, C.; von Minckwitz, G.; Darb-Esfahani, S.; Lederer, B.; Heppner, B.I.; Weber, K.E.; Budczies, J.; Huober, J.; Klauschen, F.; Furlanetto, J.; et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: A pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018, 19, 40–50. [Google Scholar] [CrossRef]
- Thompson, E.D.; Enriquez, H.L.; Fu, Y.X.; Engelhard, V.H. Tumor masses support naive T cell infiltration, activation, and differentiation into effectors. J. Exp. Med. 2010, 207, 1791–1804. [Google Scholar] [CrossRef]
- Ahmadzadeh, M.; Johnson, L.A.; Heemskerk, B.; Wunderlich, J.R.; Dudley, M.E.; White, D.E.; Rosenberg, S.A. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009, 114, 1537–1544. [Google Scholar] [CrossRef]
- Sakuishi, K.; Apetoh, L.; Sullivan, J.M.; Blazar, B.R.; Kuchroo, V.K.; Anderson, A.C. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 2010, 207, 2187–2194. [Google Scholar] [CrossRef]
- Baitsch, L.; Baumgaertner, P.; Devevre, E.; Raghav, S.K.; Legat, A.; Barba, L.; Wieckowski, S.; Bouzourene, H.; Deplancke, B.; Romero, P.; et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Investig. 2011, 121, 2350–2360. [Google Scholar] [CrossRef] [Green Version]
- Thommen, D.S.; Schumacher, T.N. T Cell Dysfunction in Cancer. Cancer Cell 2018, 33, 547–562. [Google Scholar] [CrossRef] [Green Version]
- Bengsch, B.; Ohtani, T.; Khan, O.; Setty, M.; Manne, S.; O’Brien, S.; Gherardini, P.F.; Herati, R.S.; Huang, A.C.; Chang, K.M.; et al. Epigenomic-Guided Mass Cytometry Profiling Reveals Disease-Specific Features of Exhausted CD8 T Cells. Immunity 2018, 48, 1029–1045. [Google Scholar] [CrossRef] [Green Version]
- Miller, B.C.; Sen, D.R.; Al Abosy, R.; Bi, K.; Virkud, Y.V.; LaFleur, M.W.; Yates, K.B.; Lako, A.; Felt, K.; Naik, G.S.; et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 2019, 20, 326–336. [Google Scholar] [CrossRef]
- Philip, M.; Schietinger, A. Heterogeneity and fate choice: T cell exhaustion in cancer and chronic infections. Curr. Opin. Immunol. 2019, 58, 98–103. [Google Scholar] [CrossRef]
- Blank, C.U.; Haining, W.N.; Held, W.; Hogan, P.G.; Kallies, A.; Lugli, E.; Lynn, R.C.; Philip, M.; Rao, A.; Restifo, N.P.; et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 2019, 19, 665–674. [Google Scholar] [CrossRef]
- Doering, T.A.; Crawford, A.; Angelosanto, J.M.; Paley, M.A.; Ziegler, C.G.; Wherry, E.J. Network analysis reveals centrally connected genes and pathways involved in CD8+ T cell exhaustion versus memory. Immunity 2012, 37, 1130–1144. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.; Park, S.; Park, S.Y.; Kim, G.; Park, S.M.; Cho, J.W.; Kim, D.H.; Park, Y.M.; Koh, Y.W.; Kim, H.R.; et al. Single-cell transcriptome analysis reveals TOX as a promoting factor for T cell exhaustion and a predictor for anti-PD-1 responses in human cancer. Genome Med. 2020, 12, 22. [Google Scholar] [CrossRef] [Green Version]
- Zappasodi, R.; Merghoub, T.; Wolchok, J.D. Emerging Concepts for Immune Checkpoint Blockade-Based Combination Therapies. Cancer Cell 2018, 33, 581–598. [Google Scholar] [CrossRef] [Green Version]
- June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science 2018, 359, 1361–1365. [Google Scholar] [CrossRef] [Green Version]
- Calderon, H.; Mamonkin, M.; Guedan, S. Analysis of CAR-Mediated Tonic Signaling. Methods Mol. Biol. 2020, 2086, 223–236. [Google Scholar]
- Ravasi, T.; Suzuki, H.; Cannistraci, C.V.; Katayama, S.; Bajic, V.B.; Tan, K.; Akalin, A.; Schmeier, S.; Kanamori-Katayama, M.; Bertin, N.; et al. An atlas of combinatorial transcriptional regulation in mouse and man. Cell 2010, 140, 744–752. [Google Scholar] [CrossRef] [Green Version]
PTM | Modified Site | Modifying Enzyme | Functions | REF |
---|---|---|---|---|
Phosphorylation | Ser63; Ser73 | JNK1-2; ERK1-2 | Transcritpional activation | [68,69,70] |
Phosphorylation | Thr91; Thr93 | JNK1-2-3 | Transcriptional activation | [70,71] |
Phopshorylation | Thr95 | ND | Primimg Th93 phosphorylation | [72,73] |
Phosphorylation | Thr239 | GSK3 | Inhibition of DNA binding | [70,74] |
Phosphorylation | Thr243 | ND | Priming 239Thr phoshorylation | [70,75] |
Phosphorylation | Thr249 | Casein kinase II | Inhibition of DNA binding | [76] |
Dephopshorylation | Thr243 | Calcineurin | Increase of protein stability | [77] |
Ubiquitinotion | ND | Fbw7 | Ubiquitin-dependent degradation | [78,79] |
Acetylation | Lys271 | p300/CBP | Transcriptional repression | [64] |
Sumoylation | Lys229 | Pias1; Piasxb | Transcriptional repression | [65,66] |
Gene | Function in T cells | Transcription Factors | CD8 T Cell Differentiation State | RREF |
---|---|---|---|---|
IL-2 | Clonal expansion | NFAT/AP-1(c-Jun/c-Fos) complex | Effector | [4,17,18,126,137] |
INFg | Effector cytokine | NFAT/AP-1(c-Jun/c-Fos) complex | Effector | [12,13] |
TNFa | Effector cytokine | NFAT/AP-1(c-Jun/ATF2) complex | Effector | [130] |
Granzyme B | Cytotoxic | NFAT-2 | Effector/CTL | [131] |
Pdcd1 (PD-1) | Immunoregulatory receptor | NFAT; BATF; NFAT/BATF/IRF4 complex | Exhaustion | [21,138] |
Havcr2 (TM3) | Immunoregulatory receptor | NFAT/BATF/IRF4 comlex | Exhaustion | [138] |
Tbx21 (T-bet) | Lineage-specific TF | Jun/BATF/IRF4 complex | Effector | [6] |
Tcf7 (TCF1) | Lineage-specificTF; self-renewal | Repressed by Blmp1 | Memory; Progenitor- exhausted cells | [139,140] |
Prdm1 (Blmp1) | Transcritional repressor | Jun/BATF/IRF4 complex | Memory; Exhaustion | [6] |
Eomes | Lineage-specific TF | Jun/BATF/IRF4 complex | Memory; Exhaustion | [6] |
TOX | TF; chromatin accessibility | NFAT | Exhaustion | [25,31] |
NR4A | TF; chromatin accessibility | NFAT | Exhaustion | [29,141] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Papavassiliou, A.G.; Musti, A.M. The Multifaceted Output of c-Jun Biological Activity: Focus at the Junction of CD8 T Cell Activation and Exhaustion. Cells 2020, 9, 2470. https://doi.org/10.3390/cells9112470
Papavassiliou AG, Musti AM. The Multifaceted Output of c-Jun Biological Activity: Focus at the Junction of CD8 T Cell Activation and Exhaustion. Cells. 2020; 9(11):2470. https://doi.org/10.3390/cells9112470
Chicago/Turabian StylePapavassiliou, Athanasios G., and Anna Maria Musti. 2020. "The Multifaceted Output of c-Jun Biological Activity: Focus at the Junction of CD8 T Cell Activation and Exhaustion" Cells 9, no. 11: 2470. https://doi.org/10.3390/cells9112470
APA StylePapavassiliou, A. G., & Musti, A. M. (2020). The Multifaceted Output of c-Jun Biological Activity: Focus at the Junction of CD8 T Cell Activation and Exhaustion. Cells, 9(11), 2470. https://doi.org/10.3390/cells9112470