504
Views
2
CrossRef citations to date
0
Altmetric
Editorial

p53 further extends its reach

&
Article: e24959 | Received 01 May 2013, Accepted 01 May 2013, Published online: 01 May 2013
This article refers to:

p53 is a central sensor of severe cellular insults, including genotoxic, oxidative and oncogenic stress. Depending on the context, p53 cell-autonomously orchestrates responses of temporary cell cycle arrest coupled to DNA repair or permanent senescence, or it induces apoptotic, autophagic and necrotic forms of cell death.Citation1-Citation3 However, little is known about the non-cell autonomous effects of p53. Recent findings from Lujambio et al. uncover a new paracrine oncosuppressor function of p53 in chronic liver damage, a cytokine-dependent process that promotes an anti-tumorigenic microenvironment (at least in part) by modulating immunosurveillance.Citation4

Upon liver injury, stromal hepatic stellate cells (HSC) become activated and drive the fibrotic scarring of the liver parenchyma. Chronic liver injury coupled to smoldering fibrosis can result in cirrhosis, a main risk factor for hepatocellular carcinoma (HCC). The same authors had previously shown that p53 mediates a senescence program in HSCs that limits fibrosis and maintains tissue homeostasis. Such a p53-dependent senescence program is executed in cooperation with NFκB and induces the so-called senescence-associated secretory phenotype (SASP). The SASP is marked by the downregulation of extracellular matrix (ECM) proteins including collagen, by the upregulation of ECM-degrading enzymes and by production of immunomodulatory factors and inflammatory cytokines.Citation5,Citation6

The fibrosis-inhibitory effect of damage-activated p53 in HSCs was confirmed by Lujambio et al. in different stages of liver damage, including extended liver cirrhosis. Moreover, the authors demonstrated that the HSC-specific ablation of Trp53 (the murine gene coding for p53) exacerbates the malignant transformation of hepatocytes as induced by carbon tetrachloride (CCl4) and diethylnitrosamine (DEN). This strongly suggests that a non-cell autonomous mechanism of tumor suppression is at work in p53-competent livers, operating via a p53-mediated senescence/SASP response. Lujambio et al. also showed that genes associated with cell proliferation and ECM are downregulated by p53, while genes involved in cell cycle arrest, exocytosis/vesicle transport and activation of immune signaling are upregulated. By analyzing the medium conditioned by p53-activated (senescent) or p53-ablated (proliferative) HCSs, the authors could determine the factors preferentially secreted in these two conditions, namely, interleukin (IL)-6, intercellular adhesion molecule 1 (ICAM1) and interferon γ (IFNγ) by senescent HSCs, and IL-3, IL-4, and IL-5 by proliferative HSCs. Interestingly, the most consistent changes upon p53 depletion were observed in the levels of cytokines known to promote the activation and differentiation of macrophages (i.e., IL-4, IFNγ).

Macrophages are known for their ability to modulate liver fibrosis. Tumor-associated macrophages (TAMs), which are the most abundant immune cells within the tumor microenvironment, exert a major influence on tumorigenesis.Citation7 TAMs can exist in two distinct phenotypes, M1 and M2, which are quite plastic and are mainly defined by the associated gene expression profile. Besides exerting a broad anti-pathogen activity, M1 macrophages are activated by lipopolysaccharide (LPS) and IFNγ and exert three main antitumor functions: (1) they secrete immunostimulatory cytokines including tumor necrosis factor α (TNFα), IL-1, IL-12 and IL-23; (2) they exert prominent antigen-presenting functions; and (3) they can direct kill malignant cells, at least in selected circumstances. In contrast, pro-tumorigenic M2 macrophages, upon activation by IL-4, IL-10 and IL-13, downregulate MHC expression while secreting immunosuppressive cytokines such as IL-10, arginase, growth factors including transforming growth factor β (TGFβ) and platelet-derived growth factor (PDGF) as well as angiogenic mediators like vascular endothelial growth factor (VEGF). Taken together, these bioactive molecules promote tumor growth, angiogenesis, tissue remodeling and metastasis.Citation8-Citation10 Since the M2 phenotype often predominates among TAMs, elevated TAM levels generally correlate with poor prognosis.Citation11

Intriguingly, Lujambio and colleagues showed that liver-derived macrophages cultured in the medium conditioned by proliferating HSCs predominantly exhibit an M2 phenotype. Conversely, when cultured in the medium conditioned by senescent HSCs, liver-derived macrophages are skewed toward an M1 phenotype. Moreover, in co-culture experiments, senescent HSCs were destroyed by (presumably M1) macrophages (generating an anti-fibrogenic environment), while proliferating HSCs were not. Even more interesting, the incubation of (presumably M2) macrophages with the medium conditioned by proliferative HSCs significantly increased the proliferation of co-cultured premalignant hepatocytes. This suggests that the ablation of Trp53 stimulates a proliferative and supposedly tumorigenic microenvironment (at least in part) as it skews macrophage differentiation toward an M2 phenotype. The switch to an M1 macrophage phenotype upon p53 activation was also observed in CCl4-damaged livers in vivo. In contrast, Trp53-ablated livers showed a severe overall reduction in the abundance of macrophages, with a relative increase in M2 polarization. As for the molecular mechanism underpinning the p53-induced SASP of HSCs, the authors confirm previous data pointing to a cooperativity between p53 and NFκB. Indeed, the knockdown of NFκB phenocopies Trp53 ablation in this setting.

It is known that hepatic carcinogenesis is intimately linked to inflammationCitation12 and that macrophage polarization, especially in relationship with TAM-derived cytokines, chemokines and growth factors, plays an important role in HCC progression.Citation13 However, the molecular links that connect inflammation and HCC tumorigenesis are not fully understood. For example, IL-6 and TNFα favor tumor proliferation. TGFβ, a critical growth factor for hepatic carcinogenesis, is secreted by M2 macrophages. Increased signaling via the TGFβ-SMAD3 axis promotes HCC progression in mouse models by inducing the epithelial-to-mesenchymal transition (EMT), which directly promotes invasion and metastasis.Citation14-Citation17 Likewise, high levels of tumor-infiltrating M2 macrophages are associated with poor prognosis in HCC patients.Citation18 On the other hand, IFNγ (which favors the accumulation of M1 macrophage and was shown to be downregulated by p53 depletion by Lujambio et al.)Citation4 has previously been involved in HCC progression. Nevertheless, IFNγ upregulation has also been found to correlate with improved prognosis among HCC patients.Citation19 One of the critical components in the inflammation/tumorigenesis connection that eventually drives HCC is NFκB. NFκB can exert both pro- and anti-tumorigenic effects, depending on the tumor type and stage at which it is activated. Thus, NFκB activation in liver parenchymal cells mediates oncosuppressive functions (analogous to the findings of Lujambio et al. in HSCs),Citation4 while it exerts a pro-tumorigenic effect in TAMs.Citation20-Citation22

Overall, the novel insights into the regulation of macrophage polarization by the p53-SASP axis in HSCs provided by Lujambio et al. represent a significant step forward toward understanding of HCC initiation and progression. These findings extend our knowledge of the well-established cell-autonomous p53 oncosuppressive network to a minor, yet crucial, organ-specific mesenchymal cell type that acts non-cell autonomously to establish an anticancer immune microenvironment and restrain fibrogenesis. The results by Lujambio and colleagues also generated several interesting questions. For example, how does exactly p53 control NF-κB activation in HSCs? How the p53-dependent polarization of macrophages affects the tumor environment and thus influence tumor progression also warrants further exploration. Most importantly, does this mechanism apply to hepatic carcinogenesis in humans? How frequent are mutational or regulatory disruptions of the p53 program in viral, chemical or alcoholic hepatitis-driven human HCCs? If confirmed, this novel role of p53 in the remodeling of the tissue microenvironment may pave novel avenues for the development of anticancer drugs.

References

  • Lane DP. Cancer. p53, guardian of the genome. Nature 1992; 358:15 - 6; http://dx.doi.org/10.1038/358015a0; PMID: 1614522
  • Levine AJ. p53, the cellular gatekeeper for growth and division. Cell 1997; 88:323 - 31; http://dx.doi.org/10.1016/S0092-8674(00)81871-1; PMID: 9039259
  • Wahl GM, Linke SP, Paulson TG, Huang LC. Maintaining genetic stability through TP53 mediated checkpoint control. Cancer Surv 1997; 29:183 - 219; PMID: 9338102
  • Lujambio A, Akkari L, Simon J, Grace D, Tschaharganeh DF, Bolden JE, et al. Non-Cell-Autonomous Tumor Suppression by p53. Cell 2013; 153:449 - 60; http://dx.doi.org/10.1016/j.cell.2013.03.020; PMID: 23562644
  • Coppé JP, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol 2008; 6:2853 - 68; http://dx.doi.org/10.1371/journal.pbio.0060301; PMID: 19053174
  • Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, et al. Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity. Genes Dev 2011; 25:2125 - 36; http://dx.doi.org/10.1101/gad.17276711; PMID: 21979375
  • Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature 2008; 454:436 - 44; http://dx.doi.org/10.1038/nature07205; PMID: 18650914
  • Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010; 140:883 - 99; http://dx.doi.org/10.1016/j.cell.2010.01.025; PMID: 20303878
  • Cassetta L, Cassol E, Poli G. Macrophage polarization in health and disease. ScientificWorldJournal 2011; 11:2391 - 402; http://dx.doi.org/10.1100/2011/213962; PMID: 22194670
  • Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 2012; 122:787 - 95; http://dx.doi.org/10.1172/JCI59643; PMID: 22378047
  • Sica A, Allavena P, Mantovani A. Cancer related inflammation: the macrophage connection. Cancer Lett 2008; 267:204 - 15; http://dx.doi.org/10.1016/j.canlet.2008.03.028; PMID: 18448242
  • Nakagawa H, Maeda S. Inflammation- and stress-related signaling pathways in hepatocarcinogenesis. World J Gastroenterol 2012; 18:4071 - 81; http://dx.doi.org/10.3748/wjg.v18.i31.4071; PMID: 22919237
  • Shirabe K, Mano Y, Muto J, Matono R, Motomura T, Toshima T, et al. Role of tumor-associated macrophages in the progression of hepatocellular carcinoma. Surg Today 2012; 42:1 - 7; http://dx.doi.org/10.1007/s00595-011-0058-8; PMID: 22116397
  • Yang MH, Chen CL, Chau GY, Chiou SH, Su CW, Chou TY, et al. Comprehensive analysis of the independent effect of twist and snail in promoting metastasis of hepatocellular carcinoma. Hepatology 2009; 50:1464 - 74; http://dx.doi.org/10.1002/hep.23221; PMID: 19821482
  • Jing Y, Han Z, Liu Y, Sun K, Zhang S, Jiang G, et al. Mesenchymal stem cells in inflammation microenvironment accelerates hepatocellular carcinoma metastasis by inducing epithelial-mesenchymal transition. PLoS One 2012; 7:e43272; http://dx.doi.org/10.1371/journal.pone.0043272; PMID: 22952657
  • van Zijl F, Mair M, Csiszar A, Schneller D, Zulehner G, Huber H, et al. Hepatic tumor-stroma crosstalk guides epithelial to mesenchymal transition at the tumor edge. Oncogene 2009; 28:4022 - 33; http://dx.doi.org/10.1038/onc.2009.253; PMID: 19718050
  • Brandon-Warner E, Walling TL, Schrum LW, McKillop IH. Chronic ethanol feeding accelerates hepatocellular carcinoma progression in a sex-dependent manner in a mouse model of hepatocarcinogenesis. Alcohol Clin Exp Res 2012; 36:641 - 53; http://dx.doi.org/10.1111/j.1530-0277.2011.01660.x; PMID: 22017344
  • Capece D, Fischietti M, Verzella D, Gaggiano A, Cicciarelli G, Tessitore A, et al. The inflammatory microenvironment in hepatocellular carcinoma: a pivotal role for tumor-associated macrophages. Biomed Res Int 2013; 2013:187204; http://dx.doi.org/10.1155/2013/187204; PMID: 23533994
  • Breuhahn K, Vreden S, Haddad R, Beckebaum S, Stippel D, Flemming P, et al. Molecular profiling of human hepatocellular carcinoma defines mutually exclusive interferon regulation and insulin-like growth factor II overexpression. Cancer Res 2004; 64:6058 - 64; http://dx.doi.org/10.1158/0008-5472.CAN-04-0292; PMID: 15342387
  • Maeda S, Kamata H, Luo JL, Leffert H, Karin M. IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 2005; 121:977 - 90; http://dx.doi.org/10.1016/j.cell.2005.04.014; PMID: 15989949
  • Luedde T, Beraza N, Kotsikoris V, van Loo G, Nenci A, De Vos R, et al. Deletion of NEMO/IKKgamma in liver parenchymal cells causes steatohepatitis and hepatocellular carcinoma. Cancer Cell 2007; 11:119 - 32; http://dx.doi.org/10.1016/j.ccr.2006.12.016; PMID: 17292824
  • Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, et al. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004; 431:461 - 6; http://dx.doi.org/10.1038/nature02924; PMID: 15329734