J Bacteriol Virol.  2014 Dec;44(4):297-304. 10.4167/jbv.2014.44.4.297.

Mitogen-activated Protein Kinase Signaling in Inflammation-related Carcinogenesis

Affiliations
  • 1Department of Microbiology and Immunology, School of Medicine and Brain Korea 21 PLUS Program, and Institute of Medical Science, Jeju National University, Jeju, Korea. yskoh7@jejunu.ac.kr

Abstract

The role of mitogen-activated protein kinases (MAPKs) in regulation of inflammation is well known. MAPK family is activated by various stimuli and involved in transmitting extracellular signals to nucleus leading to gene regulation. Inflammation is primary defensive response of host against microbes. Controlled inflammation is helpful and indispensable for host defense. However, uncontrolled inflammatory response leads to various inflammatory diseases and cancer. Persistent inflammation leads to cell proliferation and survival that plays crucial role in tumorigenesis. In this review, we recapitulate the recent knowledge of MAPK signaling and its roles in inflammation-associated carcinogenesis.

Keyword

Cancer; Inflammation; Mitogen-activated protein kinase

MeSH Terms

Carcinogenesis*
Cell Proliferation
Humans
Inflammation
Mitogen-Activated Protein Kinases
Protein Kinases*
Mitogen-Activated Protein Kinases
Protein Kinases

Figure

  • Figure 1. MAPK signaling pathways and cancer development. The MAPK signaling pathways are stimulated by pathogen-associated molecular patterns (PAMPs), growth factors (GF), tumor necrosis factor (TNF), stress, and inflammatory cytokines. Stimulation of MEK1/2 results in activation of MAPK (ERK1/2) activity. Activation of MEK4/7 is responsible for the activation of JNKs. The phosphorylation of p38 is regulated by MKK3/6. ERK1/2, JNK, and p38 activate various transcription factors such as TCF/ELK1, AP-1, ATF-2, and others. Different transcription factors including these result in expression of genes encoding pro-inflammatory cytokines, cell proliferation related proteins, growth factors, and anti-apoptotic factors. These factors are related with cancer development.


Cited by  1 articles

Acrosorium polyneurum Extract Inhibits the LPS-Induced Inflammatory Response by Impairing the MAPK and NF-κB Pathways
Zahid Manzoor, Irshad Ali, Doobyeong Chae, Young-Sang Koh
J Bacteriol Virol. 2016;46(4):288-294.    doi: 10.4167/jbv.2016.46.4.288.


Reference

1). Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008; 454:428–35.
Article
2). Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010; 140:805–20.
Article
3). O'Neill LA, Bowie AG. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol. 2007; 7:353–64.
4). Trinchieri G, Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol. 2007; 7:179–90.
Article
5). Miao EA, Leaf IA, Treuting PM, Mao DP, Dors M, Sarkar A, et al. Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat Immunol. 2010; 11:1136–42.
Article
6). Yuk JM, Jo EK. Host immune responses to mycobacterial antigens and their implications for the development of a vaccine to control tuberculosis. Clin Exp Vaccine Res. 2014; 3:155–67.
Article
7). Hong S, Park S, Yu JW. Pyrin domain (PYD)-containing inflammasome in innate immunity. J Bacteriol Virol. 2011; 41:133–46.
Article
8). Manzoor Z, Koh YS. Caspase-11, the main executioner in non-canonical inflammasome. J Bacteriol Virol. 2012; 42:169–71.
Article
9). Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. 2007; 449:819–26.
Article
10). Ishii KJ, Koyama S, Nakagawa A, Coban C, Akira S. Host innate immune receptors and beyond: making sense of microbial infections. Cell Host Microbe. 2008; 3:352–63.
Article
11). Koh YS. Nucleic acid recognition and signaling by Toll-like receptor 9: compartment-dependent regulation. J Bacteriol Virol. 2011; 41:131–2.
Article
12). Kawai T, Akira S. Signaling to NF-kappaB by Toll-like receptors. Trends Mol Med. 2007; 13:460–9.
13). Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010; 11:373–84.
Article
14). Sun SC, Ley SC. New insights into NF-kappaB regulation and function. Trends Immunol. 2008; 29:469–78.
15). Clevers H. At the crossroads of inflammation and cancer. Cell. 2004; 118:671–4.
Article
16). Balkwill F, Joffroy C. TNF: a tumor-suppressing factor or a tumor-promoting factor? Future Oncol. 2010; 6:1833–6.
Article
17). Wagner EF, Nebreda AR. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 2009; 9:537–49.
Article
18). Bromberg J, Wang TC. Inflammation and cancer: IL-6 and STAT3 complete the link. Cancer Cell. 2009; 15:79–80.
Article
19). Manzoor Z, Koh YS. Mitogen-activated protein kinases in inflammation. J Bacteriol Virol. 2012; 42:189–95.
Article
20). Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer. 2004; 4:937–47.
Article
21). Huang P, Han J, Hui L. MAPK signaling in inflammation-associated cancer development. Protein Cell. 2010; 1:218–26.
Article
22). Martinez E. Multi-protein complexes in eukaryotic gene transcription. Plant Mol Biol. 2002; 50:925–47.
23). Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berman K, et al. Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev. 2001; 22:153–83.
Article
24). Chen Z, Gibson TB, Robinson F, Silvestro L, Pearson G, Xu B, et al. MAP kinases. Chem Rev. 2001; 101:2449–76.
Article
25). Pagès G, Guérin S, Grall D, Bonino F, Smith A, Anjuere F, et al. Defective thymocyte maturation in p44 MAP kinase (Erk 1) knockout mice. Science. 1999; 286:1374–7.
Article
26). Hatano N, Mori Y, Oh-hora M, Kosugi A, Fujikawa T, Nakai N, et al. Essential role for ERK2 mitogen-activated protein kinase in placental development. Genes Cells. 2003; 8:847–56.
Article
27). Zebisch A, Troppmair J. Back to the roots: the remarkable RAF oncogene story. Cell Mol Life Sci. 2006; 63:1314–30.
Article
28). Campbell SL, Khosravi-Far R, Rossman KL, Clark GJ, Der CJ. Increasing complexity of Ras signaling. Oncogene. 1998; 17:1395–413.
Article
29). Hallberg B, Rayter SI, Downward J. Interaction of Ras and Raf in intact mammalian cells upon extracellular stimulation. J Biol Chem. 1994; 269:3913–6.
Article
30). Chambard JC, Lefloch R, Pouysségur J, Lenormand P. ERK implication in cell cycle regulation. Biochim Biophys Acta. 2007; 1773:1299–310.
Article
31). Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004; 116:855–67.
Article
32). Ascierto PA, Kirkwood JM, Grob JJ, Simeone E, Grimaldi AM, Maio M, et al. The role of BRAF V600 mutation in melanoma. J Transl Med. 2012; 10:85.
Article
33). Sullivan RJ, Flaherty K. MAP kinase signaling and inhibition in melanoma. Oncogene. 2013; 32:2373–9.
Article
34). Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE, Fagin JA. High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res. 2003; 63:1454–7.
35). Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, et al. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol. 2008; 26:5705–12.
Article
36). Guerra C, Schuhmacher AJ, Cañamero M, Grippo PJ, Verdaguer L, Pérez-Gallego L, et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007; 11:291–302.
Article
37). Kim K, Kim G, Kim JY, Yun HJ, Lim SC, Choi HS. Interleukin-22 promotes epithelial cell transformation and breast tumorigenesis via MAP3K8 activation. Carcinogenesis. 2014; 35:1352–61.
Article
38). Kyriakis JM, Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev. 2001; 81:807–69.
39). Johnson GL, Nakamura K. The c-jun kinase/stress-activated pathway: regulation, function and role in human disease. Biochim Biophys Acta. 2007; 1773:1341–8.
Article
40). Akira S, Nishio Y, Inoue M, Wang XJ, Wei S, Matsusaka T, et al. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell. 1994; 77:63–71.
Article
41). Shaulian E, Karin M. AP-1 in cell proliferation and survival. Oncogene. 2001; 20:2390–400.
Article
42). Nishina H, Wada T, Katada T. Physiological roles of SAPK/JNK signaling pathway. J Biochem. 2004; 136:123–6.
Article
43). Ventura JJ, Hübner A, Zhang C, Flavell RA, Shokat KM, Davis RJ. Chemical genetic analysis of the time course of signal transduction by JNK. Mol Cell. 2006; 21:701–10.
Article
44). Rincón M, Davis RJ. Regulation of the immune response by stress-activated protein kinases. Immunol Rev. 2009; 228:212–24.
Article
45). Mitsuyama K, Suzuki A, Tomiyasu N, Tsuruta O, Kitazaki S, Takeda T, et al. Pro-inflammatory signaling by Jun-N-terminal kinase in inflammatory bowel disease. Int J Mol Med. 2006; 17:449–55.
Article
46). Triantafillidis JK, Nasioulas G, Kosmidis PA. Colorectal cancer and inflammatory bowel disease: epidemiology, risk factors, mechanisms of carcinogenesis and prevention strategies. Anticancer Res. 2009; 29:2727–37.
47). 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. J Clin Invest. 2008; 118:3943–53.
Article
48). Hui L, Bakiri L, Mairhorfer A, Schweifer N, Haslinger C, Kenner L, et al. p38alpha suppresses normal and cancer cell proliferation by antagonizing the JNK-c-Jun pathway. Nat Genet. 2007; 39:741–9.
49). Maeda S, Chang L, Li ZW, Luo JL, Leffert H, Karin M. IKKbeta is required for prevention of apoptosis mediated by cell-bound but not by circulating TNFalpha. Immunity. 2003; 19:725–37.
50). Hasselblatt P, Rath M, Komnenovic V, Zatloukal K, Wagner EF. Hepatocyte survival in acute hepatitis is due to c-Jun/AP-1-dependent expression of inducible nitric oxide synthase. Proc Natl Acad Sci U S A. 2007; 104:17105–10.
Article
51). Chang Q, Zhang Y, Beezhold KJ, Bhatia D, Zhao H, Chen J, et al. Sustained JNK1 activation is associated with altered histone H3 methylations in human liver cancer. J Hepatol. 2009; 50:323–33.
Article
52). Das M, Sabio G, Jiang F, Rincón M, Flavell RA, Davis RJ. Induction of hepatitis by JNK-mediated expression of TNF-alpha. Cell. 2009; 136:249–60.
53). Ouyang X, Jessen WJ, Al-Ahmadie H, Serio AM, Lin Y, Shih WJ, et al. Activator protein-1 transcription factors are associated with progression and recurrence of prostate cancer. Cancer Res. 2008; 68:2132–44.
Article
54). Wang X, McGowan CH, Zhao M, He L, Downey JS, Fearns C, et al. Involvement of the MKK6-p38gamma cascade in gamma-radiation-induced cell cycle arrest. Mol Cell Biol. 2000; 20:4543–52.
55). Hui L, Bakiri L, Stepniak E, Wagner EF. p38alpha: a suppressor of cell proliferation and tumorigenesis. Cell Cycle. 2007; 6:2429–33.
Article
56). Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumar S, Green D, et al. A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature. 1994; 372:739–46.
Article
57). Roux PP, Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev. 2004; 68:320–44.
Article
58). Han J, Sun P. The pathways to tumor suppression via route p38. Trends Biochem Sci. 2007; 32:364–71.
Article
59). Kang YJ, Chen J, Otsuka M, Mols J, Ren S, Wang Y, et al. Macrophage deletion of p38alpha partially impairs lipopolysaccharide-induced cellular activation. J Immunol. 2008; 180:5075–82.
60). Koul HK, Pal M, Koul S. Role of p38 MAP Kinase Signal Transduction in Solid Tumors. Genes Cancer. 2013; 4:342–59.
Article
61). Campbell J, Ciesielski CJ, Hunt AE, Horwood NJ, Beech JT, Hayes LA, et al. A novel mechanism for TNF-alpha regulation by p38 MAPK: involvement of NF-kappa B with implications for therapy in rheumatoid arthritis. J Immunol. 2004; 173:6928–37.
62). Wajant H. The role of TNF in cancer. Results Probl Cell Differ. 2009; 49:1–15.
Article
63). Scott KA, Moore RJ, Arnott CH, East N, Thompson RG, Scallon BJ, et al. An anti-tumor necrosis factor-alpha antibody inhibits the development of experimental skin tumors. Mol Cancer Ther. 2003; 2:445–51.
64). Lee YJ, Lee DH, Cho CK, Chung HY, Bae S, Jhon GJ, et al. HSP25 inhibits radiation-induced apoptosis through reduction of PKCdelta-mediated ROS production. Oncogene. 2005; 24:3715–25.
65). Sakurai T, He G, Matsuzawa A, Yu GY, Maeda S, Hardiman G, et al. Hepatocyte necrosis induced by oxidative stress and IL-1 alpha release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell. 2008; 14:156–65.
66). Halawani D, Mondeh R, Stanton LA, Beier F. p38 MAP kinase signaling is necessary for rat chondrosarcoma cell proliferation. Oncogene. 2004; 23:3726–31.
Article
67). Ricote M, García-Tuñón I, Bethencourt F, Fraile B, Onsurbe P, Paniagua R, et al. The p38 transduction pathway in prostatic neoplasia. J Pathol. 2006; 208:401–7.
Article
68). Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008; 454:436–44.
Article
69). Mager DL. Bacteria and cancer: cause, coincidence or cure? A review. J Transl Med. 2006; 4:14.
Article
70). Parsonnet J. Bacterial infection as a cause of cancer. Environ Health Perspect. 1995; 103(Suppl 8):263–8.
Article
71). Ellmerich S, Schöller M, Duranton B, Gossé F, Galluser M, Klein JP, et al. Promotion of intestinal carcinogenesis by Streptococcus bovis. Carcinogenesis. 2000; 21:753–6.
72). Waris G, Ahsan H. Reactive oxygen species: role in the development of cancer and various chronic conditions. J Carcinog. 2006; 5:14.
73). Terzić J, Grivennikov S, Karin E, Karin M. Inflammation and colon cancer. Gastroenterology. 2010; 138:2101–14.
Article
74). Chun KS, Surh YJ. Signal transduction pathways regulating cyclooxygenase-2 expression: potential molecular targets for chemoprevention. Biochem Pharmacol. 2004; 68:1089–100.
Article
75). Liu CH, Chang SH, Narko K, Trifan OC, Wu MT, Smith E, et al. Overexpression of cyclooxygenase-2 is sufficient to induce tumorigenesis in transgenic mice. J Biol Chem. 2001; 276:18563–9.
Article
Full Text Links
  • JBV
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr