110
Views
0
CrossRef citations to date
0
Altmetric
Original Research

The Age-Specific Features and Clinical Significance of NRF2 and MAPK10 Expression in HCC Patients

, ORCID Icon, , , , , , & show all
Pages 737-748 | Published online: 20 Jan 2022

References

  • United Nations, Department of Economic and Social Affairs, Population division. World Population Prospects 2019: highlights [Internet]; 2019 [ cited August 21, 2019]. Available from: https://www.un.org/development/desa/publications/world-population-prospects-2019-highlights.html. Accessed January 15, 2022.
  • Pilleron S, Soto-perez-de-celis E, Vignat J, et al. Estimated global cancer incidence in the oldest adults in 2018 and projections to 2050. Int J Cancer. 2021;148(3):601–608. doi:10.1002/ijc.33232
  • Gouverneur A, Salvo F, Berdaï D, Moore N, Fourrier-Réglat A, Noize P. Inclusion of elderly or frail patients in randomized controlled trials of targeted therapies for the treatment of metastatic colorectal cancer: a systematic review. J Geriatr Oncol. 2018;9(1):15–23. doi:10.1016/j.jgo.2017.08.001
  • Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. doi:10.3322/caac.21492
  • Zhang S, Zheng R, Zeng H, Chen W. [The incidence differences among sex, geographical areas and mean age of diagnosis for liver cancer in China, 1989–2008]. Zhonghua Yu Fang Yi Xue Za Zhi. 2014;48(5):355–360. in Chinese.
  • Akinyemiju T, Abera S, Ahmed M, et al.; Global Burden of Disease Liver Cancer Collaboration. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: results from the Global Burden of Disease Study 2015. JAMA Oncol. 2017;3(12):1683–1691. doi:10.1001/jamaoncol.2017.3055.
  • López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–1217. doi:10.1016/j.cell.2013.05.039
  • Schmidlin CJ, Dodson MB, Madhavan L, Zhang DD. Redox regulation by NRF2 in aging and disease. Free Radic Biol Med. 2019;134:702–707. doi:10.1016/j.freeradbiomed.2019.01.016
  • Lewis KN, Wason E, Edrey YH, Kristan DM, Nevo E, Buffenstein R. Regulation of Nrf2 signaling and longevity in naturally long-lived rodents. Proc Natl Acad Sci U S A. 2015;112(12):3722–3727. doi:10.1073/pnas.1417566112
  • Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci. 2014;39(4):199–218. doi:10.1016/j.tibs.2014.02.002
  • Reddy NM, Kleeberger SR, Yamamoto M, et al. Genetic dissection of the Nrf2-dependent redox signaling-regulated transcriptional programs of cell proliferation and cytoprotection. Physiol Genomics. 2007;32(1):74–81. doi:10.1152/physiolgenomics.00126.2007
  • Jaiswal AK. Regulation of genes encoding NAD(P)H: quinone oxidoreductases. Free Radic Biol Med. 2000;29(3–4):254–262. doi:10.1016/S0891-5849(00)00306-3
  • Kitamura H, Motohashi H. NRF2 addiction in cancer cells. Cancer Sci. 2018;109(4):900–911. doi:10.1111/cas.13537
  • Satoh H, Moriguchi T, Taguchi K, et al. Nrf2-deficiency creates a responsive microenvironment for metastasis to the lung. Carcinogenesis. 2010;31(10):1833–1843. doi:10.1093/carcin/bgq105
  • Hiramoto K, Satoh H, Suzuki T, et al. Myeloid lineage-specific deletion of antioxidant system enhances tumor metastasis. Cancer Prev Res. 2014;7(8):835–844. doi:10.1158/1940-6207.CAPR-14-0094
  • Maj T, Wang W, Crespo J, et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol. 2017;18(12):1332–1341. doi:10.1038/ni.3868
  • Slenter DN, Kutmon M, Hanspers K, et al. WikiPathways: a multifaceted pathway database bridging metabolomics to other omics research. Nucleic Acids Res. 2018;46(D1):D661–D667. doi:10.1093/nar/gkx1064
  • Martens M, Ammar A, Riutta A, et al. WikiPathways: connecting communities. Nucleic Acids Res. 2021;49(D1):D613–D621. doi:10.1093/nar/gkaa1024
  • Bak MJ, Truong VL, Ko SY, et al. Induction of Nrf2/ARE-mediated cytoprotective genes by red ginseng oil through ASK1-MKK4/7-JNK and p38 MAPK signaling pathways in HepG2 cells. J Ginseng Res. 2016;40(4):423–430. doi:10.1016/j.jgr.2016.07.003
  • Tseng CK, Hsu SP, Lin CK, Wu YH, Lee JC, Young KC. Celastrol inhibits hepatitis C virus replication by upregulating heme oxygenase-1 via the JNK MAPK/Nrf2 pathway in human hepatoma cells. Antiviral Res. 2017;146:191–200. doi:10.1016/j.antiviral.2017.09.010
  • Jang HY, Hong OY, Chung EY, Park KH, Kim JS. Roles of JNK/Nrf2 pathway on hemin-induced heme oxygenase-1 activation in MCF-7 human breast cancer cells. Medicina. 2020;56(6):268. doi:10.3390/medicina56060268
  • Done AJ, Gage MJ, Nieto NC, Traustadóttir T. Exercise-induced Nrf2-signaling is impaired in aging. Free Radic Biol Med. 2016;96:130–138. doi:10.1016/j.freeradbiomed.2016.04.024
  • Suh JH, Shenvi SV, Dixon BM, et al. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid. Proc Natl Acad Sci U S A. 2004;101(10):3381–3386. doi:10.1073/pnas.0400282101
  • Dodson M, de la Vega MR, Cholanians A, Schmidlin CJ, Chapman E, Zhang DD. Modulating NRF2 in disease: timing is everything. Annu Rev Pharmacol Toxicol. 2019;59(1):555–575. doi:10.1146/annurev-pharmtox-010818-021856
  • Romero R, Sayin VI, Davidson SM, et al. Keap1 loss promotes Kras-driven lung cancer and results in dependence on glutaminolysis. Nat Med. 2017;23(11):1362–1368. doi:10.1038/nm.4407
  • Shibata T, Kokubu A, Saito S, et al. NRF2 mutation confers malignant potential and resistance to chemoradiation therapy in advanced esophageal squamous cancer. Neoplasia. 2011;13(9):864–873. doi:10.1593/neo.11750
  • Martinez VD, Vucic EA, Thu KL, Pikor LA, Lam S, Lam WL. Disruption of KEAP1/CUL3/RBX1 E3-ubiquitin ligase complex components by multiple genetic mechanisms: association with poor prognosis in head and neck cancer. Head Neck. 2015;37(5):727–734. doi:10.1002/hed.23663
  • Raghunath A, Sundarraj K, Arfuso F, Sethi G, Perumal E. Dysregulation of Nrf2 in hepatocellular carcinoma: role in cancer progression and chemoresistance. Cancers. 2018;10(12):481. doi:10.3390/cancers10120481
  • Cheng ML, Lu YF, Chen H, Shen ZY, Liu J. Liver expression of Nrf2-related genes in different liver diseases. Hepatobiliary Pancreat Dis Int. 2015;14(5):485–491. doi:10.1016/S1499-3872(15)60425-8
  • Zhang M, Zhang C, Zhang L, et al. Nrf2 is a potential prognostic marker and promotes proliferation and invasion in human hepatocellular carcinoma. BMC Cancer. 2015;15(1):531. doi:10.1186/s12885-015-1541-1
  • Zhang J, Bajari R, Andric D, et al. The international cancer genome consortium data portal. Nat Biotechnol. 2019;37(4):367–369. doi:10.1038/s41587-019-0055-9
  • Zhang J, Baran J, Cros A, et al. International cancer genome consortium data portal–a one-stop shop for cancer genomics data. Database. 2011;2011:bar026. doi:10.1093/database/bar026
  • Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1. doi:10.1126/scisignal.2004088
  • Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–404. doi:10.1158/2159-8290.CD-12-0095
  • Uhlén M, Fagerberg L, Hallström BM, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347(6220):1260419. doi:10.1126/science.1260419
  • Uhlen M, Zhang C, Lee S, et al. A pathology atlas of the human cancer transcriptome. Science. 2017;357(6352):eaan2507. doi:10.1126/science.aan2507
  • Hu B, Ma X, Fu P, et al. miRNA-mRNA regulatory network and factors associated with prediction of prognosis in hepatocellular carcinoma. Genomics Proteomics Bioinformatics. 2021. doi:10.1016/j.gpb.2021.03.001
  • Wu CC, Li MN, Meng HB, et al. Analysis of status and countermeasures of cancer incidence and mortality in China. Sci China Life Sci. 2019;62(5):640–647. doi:10.1007/s11427-018-9461-5
  • Nozawa A, Kubo S, Takemura S, et al. Hepatic resection for hepatocellular carcinoma in super-elderly patients aged 80 years and older in the first decade of the 21st century. Surg Today. 2015;45(7):851–857. doi:10.1007/s00595-014-0994-1
  • Oishi K, Itamoto T, Kohashi T, Matsugu Y, Nakahara H, Kitamoto M. Safety of hepatectomy for elderly patients with hepatocellular carcinoma. World J Gastroenterol. 2014;20(41):15028–15036. doi:10.3748/wjg.v20.i41.15028
  • Mauri G, Sartore-Bianchi A, Russo AG, Marsoni S, Bardelli A, Siena S. Early-onset colorectal cancer in young individuals. Mol Oncol. 2019;13(2):109–131. doi:10.1002/1878-0261.12417
  • The Lancet Oncology. Colorectal cancer: a disease of the young? Lancet Oncol. 2017;18(4):413.
  • Patel SG, Ahnen DJ. Colorectal cancer in the young. Curr Gastroenterol Rep. 2018;20(4):15. doi:10.1007/s11894-018-0618-9
  • Inra JA, Syngal S. Colorectal cancer in young adults. Dig Dis Sci. 2015;60(3):722–733. doi:10.1007/s10620-014-3464-0
  • Song M, Chan AT. Environmental factors, gut microbiota, and colorectal cancer prevention. Clin Gastroenterol Hepatol. 2019;17(2):275–289. doi:10.1016/j.cgh.2018.07.012
  • Murphy CC. Colorectal cancer in the young: Does screening make sense? Curr Gastroenterol Rep. 2019;21(7):28. doi:10.1007/s11894-019-0695-4
  • Poon RT, Fan ST, Lo CM, Liu CL, Wong J. Long-term survival and pattern of recurrence after resection of small hepatocellular carcinoma in patients with preserved liver function: implications for a strategy of salvage transplantation. Ann Surg. 2002;235(3):373–382. doi:10.1097/00000658-200203000-00009
  • Ng KK, Vauthey JN, Pawlik TM, et al.; International Cooperative Study Group on Hepatocellular Carcinoma. Is hepatic resection for large or multinodular hepatocellular carcinoma justified? Results from a multi-institutional database. Ann Surg Oncol. 2005;12(5):364–373. doi:10.1245/ASO.2005.06.004
  • European Association for the Study of the Liver. Electronic address: [email protected]; European Association for the Study of the Liver. EASL Clinical Practice Guidelines: management of hepatocellular carcinoma. J Hepatol. 2018;69(1):182–236. doi:10.1016/j.jhep.2018.03.019
  • Heimbach JK, Kulik LM, Finn RS, et al. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology. 2018;67(1):358–380. doi:10.1002/hep.29086