1,995
Views
24
CrossRef citations to date
0
Altmetric
Research Paper

Fusaric acid decreases p53 expression by altering promoter methylation and m6A RNA methylation in human hepatocellular carcinoma (HepG2) cells

, & ORCID Icon
Pages 79-91 | Received 13 Feb 2020, Accepted 02 Jun 2020, Published online: 07 Jul 2020

References

  • Ghazi T, Nagiah S, Naidoo P, et al. Fusaric acid-induced promoter methylation of DNA methyltransferases triggers DNA hypomethylation in human hepatocellular carcinoma (HepG2) cells. Epigenetics. 2019 Aug;14(8):1–14.
  • D’Alton A, Etherton B. Effects of fusaric acid on tomato root hair membrane potentials and ATP levels. Plant Physiol. 1984 Jan;74(1):39–42.
  • Diniz S, Oliveira R. Effects of fusaric acid on Zea mays L. seedlings. Phyton Int J Exp Bot. 2009 Jan;78:155–160.
  • Pavlovkin J, Mistrik I, Prokop M. Some aspects of the phytotoxic action of fusaric acid on primary ricinus roots. Plant Soil Environ. 2004 Sept;50(9):397–401.
  • Singh VK, Singh HB, Upadhyay RS. Role of fusaric acid in the development of ‘Fusarium wilt’ symptoms in tomato: physiological, biochemical and proteomic perspectives. Plant Physiol Biochem. 2017 Sept; 118:320–332.
  • Hidaka H, Asano M. Relaxation of isolated rabbit arteries by fusaric (5-butylpicolinic) acid. J Pharmacol Exp Ther. 1976 Dec;199(3):620–629.
  • Hidaka H, Nagatsu T, Takeya K, et al. Fusaric acid, a hypotensive agent produced by fungi. J Antibiot. 1969 May;22(5):228–230.
  • Yin ES, Rakhmankulova M, Kucera K, et al. Fusaric acid induces a notochord malformation in zebrafish via copper chelation. Biometals. 2015 Aug;28(4):783–789.
  • Bungo T, Shimojo M, Masuda Y, et al. Induction of food intake by a noradrenergic system using clonidine and fusaric acid in the neonatal chick. Brain Res. 1999 May;826(2):313–316.
  • Devnarain N, Tiloke C, Nagiah S, et al. Fusaric acid induces oxidative stress and apoptosis in human cancerous oesophageal SNO cells. Toxicon. 2017 Feb;126:4–11.
  • Abdul NS, Nagiah S, Chuturgoon AA. Fusaric acid induces mitochondrial stress in human hepatocellular carcinoma (HepG2) cells. Toxicon. 2016 Sept;119:336–344.
  • Ghazi T, Nagiah S, Tiloke C, et al. Fusaric acid induces DNA damage and post‐translational modifications of p53 in human hepatocellular carcinoma (HepG2) cells. J Cell Biochem. 2017 Nov;118(11):3866–3874.
  • Mamur S, Ünal F, Yılmaz S, et al. Evaluation of the cytotoxic and genotoxic effects of mycotoxin fusaric acid. Drug Chem Toxicol. 2020 Mar;43(2):149–157.
  • Dhani S, Nagiah S, Naidoo DB, et al. Fusaric acid immunotoxicity and MAPK activation in normal peripheral blood mononuclear cells and Thp-1 cells. Sci Rep. 2017 Jun;7(1):3051–3060.
  • Ogata S, Inoue K, Iwata K, et al. Apoptosis induced by picolinic acid-related compounds in HL-60 cells. Biosci Biotechnol Biochem. 2001 Oct;65(10):2337–2339.
  • Diringer MN, Kramarcy NR, Brown JW, et al. Effect of fusaric acid on aggression, motor activity, and brain monoamines in mice. Pharmacol Biochem Behav. 1982 Jan;16(1):73–79.
  • Porter JK, Bacon CW, Wray EM, et al. Fusaric acid in Fusarium moniliforme cultures, corn, and feeds toxic to livestock and the neurochemical effects in the brain and pineal gland of rats. Nat Toxins. 1995;3(2):91–100.
  • Smith T, MacDonald E. Effect of fusaric acid on brain regional neurochemistry and vomiting behavior in swine. J Anim Sci. 1991 May;69(5):2044–2049.
  • Swamy H, Smith T, MacDonald E, et al. Effects of feeding a blend of grains naturally contaminated with Fusarium mycotoxins on swine performance, brain regional neurochemistry, and serum chemistry and the efficacy of a polymeric glucomannan mycotoxin adsorbent. J Anim Sci. 2002 Dec;80(12):3257–3267.
  • Devaraja S, Girish KS, Santhosh MS, et al. Fusaric acid, a mycotoxin, and its influence on blood coagulation and platelet function. Blood Coagul Fibrinolysis. 2013 Jun;24(4):419–423.
  • Reddy R, Larson C, Brimer G, et al. Developmental toxic effects of fusaric acid in CD1 mice. Bull Environ Contam Toxicol. 1996 Sept;57(3):354–360.
  • Terasawa F, Kameyama M. The clinical trial of a new hypotensive agent, fusaric acid (5-butylpicolinic acid): the preliminary report. Japanese Circ J. 1971 Apr;35(3):339–357.
  • Smith TK, McMillan EG, Castillo JB. Effect of feeding blends of Fusarium mycotoxin-contaminated grains containing deoxynivalenol and fusaric acid on growth and feed consumption of immature swine. J Anim Sci. 1997 Aug;75(8):2184–2191.
  • Bacon CW, Porter JK, Norred WP. Toxic interaction of fumonisin B1 and fusaric acid measured by injection into fertile chicken egg. Mycopathologia. 1995 Jan;129(1):29–35.
  • Fairchild A, Grimes J, Porter J, et al. Effects of diacetoxyscirpenol and fusaric acid on poults: individual and combined effects of dietary diacetoxyscirpenol and fusaric acid on turkey poult performance. Int J Poult Sci. 2005 Jun;4(3):350–355.
  • Laptenko O, Prives C. Transcriptional regulation by p53: one protein, many possibilities. Cell Death Differ. 2006 Jun;13(6):951–961.
  • Prives C, Hall PA. The p53 pathway. J Pathol. 1999 Jan;187(1):112–126.
  • Kruiswijk F, Labuschagne CF, Vousden KH. p53 in survival, death and metabolic health: a lifeguard with a licence to kill. Nat Rev Mol Cell Biol. 2015 Jul;16(7):393–405.
  • Vousden KH, Prives C. Blinded by the light: the growing complexity of p53. Cell. 2009 May;137(3):413–431.
  • Chang JR, Ghafouri M, Mukerjee R, et al. Role of p53 in neurodegenerative diseases. Neurodegener Dis. 2012 Feb;9(2):68–80.
  • Szybińska A, Leśniak W. P53 dysfunction in neurodegenerative diseases-the cause or effect of pathological changes? Aging Dis. 2017 Jul;8(4):506–518.
  • Mitsudomi T, Steinberg SM, Nau MM, et al. p53 gene mutations in non-small-cell lung cancer cell lines and their correlation with the presence of ras mutations and clinical features. Oncogene. 1992 Jan;7(1):171–180.
  • Barlev NA, Liu L, Chehab NH, et al. Acetylation of p53 activates transcription through recruitment of coactivators/histone acetyltransferases. Mol Cell. 2001 Dec;8(6):1243–1254.
  • Shieh SY, Ikeda M, Taya Y, et al. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell. 1997 Oct;91(3):325–334.
  • Tang Y, Zhao W, Chen Y, et al. Acetylation is indispensable for p53 activation. Cell. 2008 May;133(4):612–626.
  • Tuck SP, Crawford L. Characterization of the human p53 gene promoter. Mol Cell Biol. 1989 May;9(5):2163–2172.
  • Chmelarova M, Krepinska E, Spacek J, et al. Methylation in the p53 promoter in epithelial ovarian cancer. Clin Transl Oncol. 2013 Feb;15(2):160–163.
  • Kang JH, Kim SJ, Noh DY, et al. Methylation in the p53 promoter is a supplementary route to breast carcinogenesis: correlation between CpG methylation in the p53 promoter and the mutation of the p53 gene in the progression from ductal carcinoma in situ to invasive ductal carcinoma. Lab Invest. 2001 Apr;81(4):573–579.
  • Woo HH, Chambers SK. Human ALKBH3-induced m1A demethylation increases the CSF-1 mRNA stability in breast and ovarian cancer cells. Biochim Biophys Acta Gene Regul Mech. 2019 Jan;1862(1):35–46.
  • Fu Y, Dominissini D, Rechavi G, et al. Gene expression regulation mediated through reversible m6A RNA methylation. Nat Rev Genet. 2014 May;15(5):293–306.
  • Geula S, Moshitch-Moshkovitz S, Dominissini D, et al. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science. 2015 Feb;347(6225):1002–1006.
  • Meyer KD, Patil DP, Zhou J, et al. 5′ UTR m6A promotes cap-independent translation. Cell. 2015 Nov;163(4):999–1010.
  • Wang X, Zhao BS, Roundtree IA, et al. N6-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015 Jun;161(6):1388–1399.
  • Wang X, Lu Z, Gomez A, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014 Jan;505(7481):117–120.
  • Xiao W, Adhikari S, Dahal U, et al. Nuclear m6A reader YTHDC1 regulates mRNA splicing. Mol Cell. 2016 Feb;61(4):507–519.
  • Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013 Jan;49(1):18–29.
  • Batista PJ, Molinie B, Wang J, et al. m6A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell. 2014 Dec;15(6):707–719.
  • Wang CX, Cui GS, Liu X, et al. METTL3-mediated m6A modification is required for cerebellar development. PLoS Biol. 2018;16:1–29.
  • Wang X, Zhu L, Chen J, et al. mRNA m6A methylation downregulates adipogenesis in porcine adipocytes. Biochem Biophys Res Commun. 2015 Apr;459(2):201–207.
  • Dina C, Meyre D, Gallina S, et al. Variation in FTO contributes to childhood obesity and severe adult obesity. Nat Genet. 2007 Jun;39(6):724–726.
  • Yang Y, Shen F, Huang W, et al. Glucose is involved in the dynamic regulation of m6A in patients with type 2 diabetes. J Clin Endocrinol Metab. 2019 Mar;104(3):665–673.
  • Lin S, Choe J, Du P, et al. The m6A methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 2016 May;62(3):335–345.
  • Panneerdoss S, Eedunuri VK, Yadav P, et al. Cross-talk among writers, readers, and erasers of m6A regulates cancer growth and progression. Sci Adv. 2018 Oct;4(10):1–15.
  • Dai D, Wang H, Zhu L, et al. N6-methyladenosine links RNA metabolism to cancer progression. Cell Death Dis. 2018 Jan;9(2):124–136.
  • Wei W, Ji X, Guo X, et al. Regulatory role of N6‐methyladenosine (m6A) methylation in RNA processing and human diseases. J Cell Biochem. 2017 Sept;118(9):2534–2543.
  • Wang X, He C. Dynamic RNA modifications in posttranscriptional regulation. Mol Cell. 2014 Oct;56(1):5–12.
  • Liu J, Yue Y, Han D, et al. A METTL3–METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014 Feb;10(2):93–95.
  • Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 2011 Dec;7(12):885–887.
  • Heo SH, Kwak J, Jang KL. All-trans retinoic acid induces p53-depenent apoptosis in human hepatocytes by activating p14 expression via promoter hypomethylation. Cancer Lett. 2015 Jun;362(1):139–148.
  • Qing Y, Hu H, Liu Y, et al. Berberine induces apoptosis in human multiple myeloma cell line U266 through hypomethylation of p53 promoter. Cell Biol Int. 2014 May;38(5):563–570.
  • Köhler K, Bentrup FW. The effect of fusaric acid upon electrical membrane properties and ATP level in photoautotrophic cell suspension cultures of chenopodium rubrum L. Zeitschrift für Pflanzenphysiologie. 1983 Mar;109(4):355–361.
  • Chuturgoon A, Phulukdaree A, Moodley D. Fumonisin B1 induces global DNA hypomethylation in HepG2 cells–an alternative mechanism of action. Toxicology. 2014 Jan;315:65–69.
  • So MY, Tian Z, Phoon YS, et al. Gene expression profile and toxic effects in human bronchial epithelial cells exposed to zearalenone. PLoS One. 2014 May;9(5):65–69.
  • Zhu CC, Hou YJ, Han J, et al. Zearalenone exposure affects epigenetic modifications of mouse eggs. Mutagenesis. 2014 Nov;29(6):489–495.
  • Dominissini D, Moshitch-Moshkovitz S, Schwartz S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012 May;485(7397):201–206.
  • Li J, Han Y, Zhang H, et al. The m6A demethylase FTO promotes the growth of lung cancer cells by regulating the m6A level of USP7 mRNA. Biochem Biophys Res Commun. 2019 May;512(3):479–485.
  • Zhong X, Yu J, Frazier K, et al. Circadian clock regulation of hepatic lipid metabolism by modulation of m6A mRNA methylation. Cell Rep. 2018 Nov;25(7):1816–1828.
  • Li Z, Weng H, Su R, et al. FTO plays an oncogenic role in acute myeloid leukemia as a N6-methyladenosine RNA demethylase. Cancer Cell. 2017 Jan;31(1):127–141.
  • Kwok CT, Marshall AD, Rasko JE, et al. Genetic alterations of m6A regulators predict poorer survival in acute myeloid leukemia. J Hematol Oncol. 2017 Dec;10(1):39–44.
  • Zhang C, Samanta D, Lu H, et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proc Natl Acad Sci. 2016 Apr;113(14):2047–2056.
  • Chen M, Wei L, Law CT, et al. RNA N6‐methyladenosine methyltransferase‐like 3 promotes liver cancer progression through YTHDF2‐dependent posttranscriptional silencing of SOCS2. Hepatology. 2018 Jun;67(6):2254–2270.
  • Li X, Yang J, Zhu Y, et al. Mouse maternal high-fat intake dynamically programmed mRNA m6A modifications in adipose and skeletal muscle tissues in offspring. Int J Mol Sci. 2016 Aug;17(8):1336–1344.
  • Lu N, Li X, Yu J, et al. Curcumin attenuates lipopolysaccharide‐induced hepatic lipid metabolism disorder by modification of m6A RNA methylation in piglets. Lipids. 2018 Jan;53(1):53–63.
  • Uddin MB, Roy KR, Hosain SB, et al. An N6-methyladenosine at the transited codon 273 of p53 pre-mRNA promotes the expression of R273H mutant protein and drug resistance of cancer cells. Biochem Pharmacol. 2019 Feb;160:134–145.
  • Zhao T, Sun D, Zhao M, et al. N6-methyladenosine mediates arsenite-induced human keratinocyte transformation by suppressing p53 activation. Environ Pollut. 2020 Jan;259:113908–113920.
  • Cui Q, Shi H, Ye P, et al. m6A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 2017 Mar;18(11):2622–2634.
  • Xu D, Shao W, Jiang Y, et al. FTO expression is associated with the occurrence of gastric cancer and prognosis. Oncol Rep. 2017 Oct;38(4):2285–2292.
  • Liu JE, Li K, Cai J, et al. Landscape and regulation of m6A and m6Am methylome across human and mouse tissues. Mol Cell. 2020 Jan;77(2):426–440.
  • Huang B, Ding C, Zhou Q, et al. Cyclophosphamide regulates N6-methyladenosine and m6A RNA enzyme levels in human granulosa cells and in ovaries of a premature ovarian aging mouse model. Front Endocrinol. 2019 Jun;10:415–428.
  • Liu J, Ren D, Du Z, et al. M6A demethylase FTO facilitates tumor progression in lung squamous cell carcinoma by regulating MZF1 expression. Biochem Biophys Res Commun. 2018 Aug;502(4):456–464.
  • Zhang Y, Wang X, Zhang X, et al. RNA-binding protein YTHDF3 suppresses interferon-dependent antiviral responses by promoting FOXO3 translation. Proc Natl Acad Sci. 2019 Jan;116(3):976–981.
  • Li A, Chen YS, Ping XL, et al. Cytoplasmic m6A reader YTHDF3 promotes mRNA translation. Cell Res. 2017 Mar;27(3):444–447.
  • Shi H, Wang X, Lu Z, et al. YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res. 2017 Mar;27(3):315–328.
  • Tanabe A, Tanikawa K, Tsunetomi M, et al. RNA helicase YTHDC2 promotes cancer metastasis via the enhancement of the efficiency by which HIF-1α mRNA is translated. Cancer Lett. 2016 Jun;376(1):34–42.
  • Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 2001 Dec;25(4):402–408.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.