473
Views
15
CrossRef citations to date
0
Altmetric
Review Articles

Impact of Phytochemicals and Dietary Patterns on Epigenome and Cancer

&
Pages 184-200 | Received 12 Feb 2016, Accepted 17 Nov 2016, Published online: 17 Jan 2017

References

  • Waddington CH: The epigenotype. Int J Epidemiol 41, 10–13, 2012.
  • Herceg Z: Epigenetics and cancer: towards an evaluation of the impact of environmental and dietary factors. Mutagenesis 22, 91–103, 2007.
  • Choi JD and Lee JS: Interplay between epigenetics and genetics in cancer. Genomics Inform 11, 164–173, 2013.
  • Knorre DG, Kudryashova NV, and Godovikova TS: Chemical and functional aspects of posttranslational modification of proteins. Acta Naturae 1, 29–51, 2009.
  • Stallcup MR: Role of protein methylation in chromatin remodeling and transcriptional regulation. Oncogene 20, 3014–3020, 2001.
  • Freeman WH: Covalent modification is a means of regulating enzyme activity. In: Biochemistry, 5th edition, Berg JM, Tymoczko JL, and Stryer L (eds.). New York: Freeman and Company, 2002.
  • Strahl BD and Allis CD: The language of covalent histone modifications. Nature 403, 41–45, 2000.
  • Kouzarides T: Chromatin modifications and their function. Cell 128, 693–705, 2007.
  • Spange S, Wagner T, Heinzel, T and Krämer OH: Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int J Biochem Cell Biol 41, 185–198, 2009.
  • Das C and Kundu TK: Transcriptional regulation by the acetylation of nonhistone proteins in humans – a new target for therapeutics. IUBMB Life 57, 137–149, 2005.
  • Woo JH, Kim YH, Choi YJ, Kim DG, Lee KS, et al.: Molecular mechanisms of curcumin-induced cytotoxicity: induction of apoptosis through generation of reactive oxygen species, down-regulation of Bcl-XL and IAP, the release of cytochrome c and inhibition of Akt. Carcinogenesis 24, 1199–208, 2003.
  • Ropero S and Esteller M: The role of histone deacetylases (HDACs) in human cancer. Mol Oncology 1, 19–25, 2007.
  • Minucci S and Pelicci PG: Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6, 38–51, 2006.
  • Lin HY, Chen CS, Lin SP, Weng JR, and Chen CS: Targeting histone deacetylase in cancer therapy. Med Res Rev 26, 397–413, 2006.
  • Schwartz C, Beck K, and Mink S: Recruitment of p300 by C/EBPbeta triggers phosphorylation of p300 and modulates coactivator activity. EMBO J 22, 882–892, 2003.
  • Matsuzaki H, Daitoku H, Hatta M, Aoyama H, Yoshimochi K, et al.: Acetylation of FoxO1 alters its DNA-binding ability and sensitivity to phosphorylation. Proc Natl Acad Sci USA 102, 11278–11283, 2005.
  • Yang WM, Yao YL, Sun JM, Davie JR, and Seto E: Isolation and characterization of cDNAs corresponding to an additional member of the human histone deacetylase gene family. J Biol Chem 272, 28001–28007, 1997.
  • Galasinski SC, Resing KA, Goodrich JA, and Ahn NG: Phosphatase inhibition leads to histone deacetylases 1 and 2 phosphorylation and disruption of corepressor interactions. J Biol Chem 277, 19618–19626, 2002.
  • http://www.who.int/mediacentre/factsheets/fs297/en/
  • Wang J, Wu Z, Li D, Li N, Dindot SV, et al.: Nutrition, epigenetics, and metabolic syndrome. Antioxid Redox Signal 17, 282–301, 2012.
  • Bianchini F and Vainio H: Allium vegetables and organosulfur compounds: do they help prevent cancer? Environ Health Perspect 109, 893–902, 2001.
  • Druesne-Pecollo N and Latino-Martel P: Modulation of histone acetylation by garlic sulfur compounds. Anticancer Agents Med Chem 11, 254–259, 2011.
  • Lea MA and Randolph VM: Induction of histone acetylation in rat liver and hepatoma by organosulfur compounds including diallyl disulfide. Anticancer Res 21, 2841–2845, 2001.
  • Lea MA, Randolph VM, Lee JE, and desBordes C: Induction of histone acetylation in mouse erythroleukemia cells by some organosulfur compounds including allyl isothiocyanate. Int J Cancer 92, 784–789, 2001.
  • Wallace GC, Haar CP, Vandergrift WA, Giglio P, Dixon-Mah YN, et al.: Multi-targeted DATS prevents tumor progression and promotes apoptosis in ectopic glioblastoma xenografts in SCID mice via HDAC inhibition. J Neurooncol 114, 43–50, 2013.
  • Chandra-Kuntal K and Singh SV: Diallyl trisulfide inhibits activation of signal transducer and activator of transcription 3 in prostate cancer cells in culture and in vivo. Cancer Prev Res (Phila) 3, 1473–1483, 2010.
  • Wang YB, Qin J, Zheng XY, Bai Y, Yang K, et al.: Diallyl trisulfide induces Bcl-2 and caspase-3-dependent apoptosis via downregulation of Akt phosphorylation in human T24 bladder cancer cells. Phytomedicine 17, 363–368, 2010.
  • McKay DL and Blumberg JB: A review of the bioactivity and potential health benefits of chamomile tea (Matricaria recutita L.). Phytother Res 20, 519–530, 2006.
  • Pandey M, Kaur P, Shukla S, Abbas A, Fu P, et al.: Plant flavone apigenin inhibits HDAC and remodels chromatin to induce growth arrest and apoptosis in human prostate cancer cells: In vitro and in vivo study. Mol Carcinog 51, 952–962, 2012.
  • Kaur P, Shukla S and Gupta S: Plant flavonoid apigenin inactivates Akt to trigger apoptosis in human prostate cancer: an in vitro and in vivo study. Carcinogenesis 29, 2210–2217, 2008.
  • Shukla S and Gupta S: Apigenin suppresses insulin-like growth factor I receptor signaling in human prostate cancer: an in vitro and in vivo study. Mol Carcinog 48, 243–252, 2009.
  • King JC, Lu QY, Li G, Moro A, Takahashi H, et al.: Evidence for activation of mutated p53 by apigenin in human pancreatic cancer. Biochim Biophys Acta 1823, 593–604, 2012.
  • Ujiki MB, Ding XZ, Salabat MR, Bentrem DJ, Golkar L, et al.: Apigenin inhibits pancreatic cancer cell proliferation through G2/M cell cycle arrest. Mol Cancer 5, 76, 2006.
  • Wu DG, Yu P, Li JW, Jiang P, Sun J, et al.: Apigenin potentiates the growth inhibitory effects by IKK-β-mediated NF-κB activation in pancreatic cancer cells. Toxicol Lett 224, 157–164, 2014.
  • Yin F, Giuliano AE, and Van Herle AJ: Signal pathways involved in apigenin inhibition of growth and induction of apoptosis of human anaplastic thyroid cancer cells (ARO). Anticancer Res 19, 4297–4303, 1999.
  • Harrison ME, Power Coombs MR, Delaney LM, and Hoskin DW: Exposure of breast cancer cells to a subcytotoxic dose of apigenin causes growth inhibition, oxidative stress, and hypophosphorylation of Akt. Exp Mol Pathol 97, 211–217, 2014.
  • Yin F, Giuliano AE, Law RE, and Van Herle AJ: Apigenin inhibits growth and induces G2/M arrest by modulating cyclin-CDK regulators and ERK MAP kinase activation in breast carcinoma cells. Anticancer Res 21, 413–420, 2001.
  • Tseng TH, Chien MH, Lin WL, Wen YC, Chow JM, et al.: Inhibition of MDA-MB-231 breast cancer cell proliferation and tumor growth by apigenin through induction of G2/M arrest and histone H3 acetylation-mediated p21WAF1/CIP1 expression. Environ Toxicol 2016.
  • Lee WJ, Chen WK, Wang CJ, Lin WL, and Tseng TH: Apigenin inhibits HGF-promoted invasive growth and metastasis involving blocking PI3K/Akt pathway and beta 4 integrin function in MDA-MB-231 breast cancer cells. Toxicol Appl Pharmacol 226, 178–191, 2008.
  • Lu HF, Chie YJ, Yang MS, Lee CS, Fu JJ, et al.: Apigenin induces caspase-dependent apoptosis in human lung cancer A549 cells through Bax- and Bcl-2-triggered mitochondrial pathway. Int J Oncol 36, 1477–1484, 2010.
  • Zhao M, Ma J, Zhu HY, Zhang XH, Du ZY, et al.: Apigenin inhibits proliferation and induces apoptosis in human multiple myeloma cells through targeting the trinity of CK2, Cdc37 and Hsp90. Mol Cancer 10, 104, 2011.
  • Chunhua L, Donglan L, Xiuqiong F, Lihua Z, Qin F, et al.: Apigenin up-regulates transgelin and inhibits invasion and migration of colorectal cancer through decreased phosphorylation of AKT. J Nutr Biochem 24, 1766–1775, 2013.
  • Middleton E Jr, Kandaswami C, and Theoharides TC: The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacol Rev 52, 673–751, 2000.
  • Chang H, Mi M, Ling W, Zhu J, Zhang Q, et al.: Structurally related cytotoxic effects of flavonoids on human cancer cells in vitro. Arch Pharm Res 31, 1137–1144, 2008.
  • Pal-Bhadra M, Ramaiah MJ, Reddy TL, Krishnan A, Pushpavalli SNCVL, et al.: Plant HDAC inhibitor chrysin arrest cell growth and induce p21WAF1 by altering chromatin of STAT response element in A375 cells. BMC Cancer 12, 180, 2012.
  • Lirdprapamongkol K, Sakurai H, Abdelhamed S, Yokoyama S, Athikomkulchai S, et al.: Chrysin overcomes TRAIL resistance of cancer cells through Mcl-1 downregulation by inhibiting STAT3 phosphorylation. Int J Oncol 43, 329–337, 2013.
  • Xia Y, Lian S, Khoi PN, Yoon HJ, Joo YE, et al.: Chrysin inhibits tumor promoter-induced MMP-9 expression by blocking AP-1 via suppression of ERK and JNK pathways in gastric cancer cells. PLoS One 10, e0124007, 2015.
  • Woo KJ, Jeong YJ, Park JW, and Kwon TK: Chrysin-induced apoptosis is mediated through caspase activation and Akt inactivation in U937 leukemia cells. Biochem Biophys Res Commun 325, 1215–1222, 2004.
  • Liu H, Hwang JS, Li W, Choi TW, Liu K, et al.: A derivative of chrysin suppresses two-stage skin carcinogenesis by inhibiting mitogen- and stress-activated kinase 1. Cancer Prev Res (Phila) 7, 74–85, 2014.
  • Kristal AR and Lampe JW: Brassica vegetables and prostate cancer risk: a review of the epidemiological evidence. Nutr Cancer 42, 1–9, 2002.
  • Murillo G and Mehta RG: Cruciferous vegetables and cancer prevention. Nutr Cancer 41, 17–28, 2001.
  • Higdon JV, Delage B, Williams DE, and Dashwood RH: Cruciferous vegetables and human cancer risk: epidemiologic evidence and mechanistic basis. Pharmacol Res 55, 224–236, 2007.
  • Tang L, Zirpoli GR, Guru K, Moysich KB, Zhang Y, et al.: Consumption of raw cruciferous vegetables is inversely associated with bladder cancer risk. Cancer Epidemiol Biomarkers Prev 17, 938–944, 2008.
  • Hecht SS: Inhibition of carcinogenesis by isothiocyanates. Drug Metab Rev 32, 395–411, 2000.
  • Emily Ho, John D., Clarke,  , and Roderick H: Dashwood. Dietary Sulforaphane, a Histone Deacetylase Inhibitor for Cancer Prevention. J Nutr 139, 2393–2396, 2009.
  • Myzak MC, Karplus PA, Chung FL, and Dashwood RH: A novel mechanism of chemoprotection by sulforaphane: inhibition of histone deacetylase. Cancer Res 64, 5767–5774, 2004.
  • Navarro SL, Li F, and Lampe JW: Mechanisms of action of isothiocyanates in cancer chemoprevention: an update. Food Funct 2, 579–587, 2011.
  • Su Z-Y, Zhang C, Lee JH, Shu L, Wu TY, et al.: Requirement and epigenetics reprogramming of Nrf2 in suppression of tumor promoter TPA-induced mouse skin cell transformation by sulforaphane. Cancer Prev Res (Phila) 7, 319–329, 2014.
  • Wiczk A, Hofman D, Konopa G, Herman-Antosiewicz A: Sulforaphane, a cruciferous vegetable-derived isothiocyanate, inhibits protein synthesis in human prostate cancer cells. Biochim Biophys Acta 1823, 1295–1305, 2012.
  • Choi S, Lew KL, Xiao H, Herman-Antosiewicz A, Xiao D, et al.: D,L-Sulforaphane-induced cell death in human prostate cancer cells is regulated by inhibitor of apoptosis family proteins and Apaf-1. Carcinogenesis 28, 151–162, 2007.
  • Stehlik C, de Martin R, Kumabashiri I, Schmid JA, Binder BR, et al.: Nuclear factor (NF)-kappaB-regulated X-chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor alpha-induced apoptosis. J Exp Med 188, 211–216, 1998.
  • Xu C, Shen G, Chen C, Gélinas C, and Kong AN: Suppression of NF-kappaB and NF-kappaB-regulated gene expression by sulforaphane and PEITC through IkappaBalpha, IKK pathway in human prostate cancer PC-3 cells. Oncogene 24, 4486–4495, 2005.
  • Jeong WS, Kim IW, Hu R, and Kong AN: Modulatory properties of various natural chemopreventive agents on the activation of NF-kappaB signaling pathway. Pharm Res 21, 661–670, 2004.
  • Chinni SR, Li Y, Upadhyay S, Koppolu PK, and Sarkar FH: Indole-3-carbinol (I3C) induced cell growth inhibition, G1 cell cycle arrest and apoptosis in prostate cancer cells. Oncogene 20, 2927–2936, 2001.
  • Yan Chen, Wenxiu Shu, Weihua Chen, Qing Wu, Hongli Liu, et al.: Curcumin, both histone deacetylase and p300/CBP-specific inhibitor, represses the activity of nuclear factor kappa B and notch 1 in Raji cells. Basic Clin Pharmacol Toxicol 101, 427–433, 2007.
  • Swamy MV, Citineni B, Patlolla JM, Mohammed A, Zhang Y, et al.: Prevention and treatment of pancreatic cancer by curcumin in combination with omega-3 fatty acids. Nutr Cancer 60, 81–89, 2008.
  • Jiao Y, Wilkinson J 4th, Di X, Wang W, Hatcher H, et al.: Curcumin, a cancer chemopreventive and chemotherapeutic agent, is a biologically active iron chelator. Blood 113, 462–469, 2009.
  • Half E and Arber N: Colon cancer: preventive agents and the present status of chemoprevention. Expert Opin Pharmacother 10, 211–219, 2009.
  • Lv BH, Zhang L, Zhu CC, and Liu J: Inhibition of curcumin on histone deacetylase and expression promotion of P21 (WAF1/CIP1) in HepG2 cells. Zhongguo Zhong Yao Za Zhi 32, 2051–2055, 2007.
  • Liu HL, Chen Y, Cui GH, and Zhou JF: Curcumin, a potent anti-tumor reagent, is a novel histone deacetylase inhibitor regulating B-NHL cell line Raji proliferation. Acta Pharmacol Sin 26, 603–609, 2005.
  • Wang L, Shen Y, Song R, Sun Y, Xu J, et al.: An anticancer effect of curcumin mediated by down-regulating phosphatase of regenerating liver-3 expression on highly metastatic melanoma cells. Mol Pharmacol 76, 1238–1245, 2009.
  • Lee SJ, Krauthauser C, Maduskuie V, Fawcett PT, Olson JM, et al.: Curcumin-induced HDAC inhibition and attenuation of medulloblastoma growth in vitro and in vivo. BMC Cancer 11, 144, 2011.
  • Qian Y, Ma J, Guo X, Sun J, Yu Y, et al.: Curcumin Enhances the Radiosensitivity of U87 Cells by Inducing DUSP-2 Up-Regulation. Cell Physiol Biochem 35, 1381–1393, 2015.
  • Wang SH, Lin PY, Chiu YC, Huang JS, Kuo YT, et al.: Curcumin-Mediated HDAC Inhibition Suppresses the DNA Damage Response and Contributes to Increased DNA Damage Sensitivity. PLoS One 10, e0134110, 2015.
  • Yu S, Shen G, Khor TO, Kim JH, and Kong AN: Curcumin inhibits Akt/mammalian target of rapamycin signaling through protein phosphatase-dependent mechanism. Mol Cancer Ther 7, 2609–2620, 2008.
  • Squires MS, Hudson EA, Howells L, Sale S, Houghton CE, et al.: Relevance of mitogen activated protein kinase (MAPK) and phosphotidylinositol-3-kinase/protein kinase B (PI3K/PKB) pathways to induction of apoptosis by curcumin in breast cells. Biochem Pharmacol 65, 361–376, 2003.
  • Liu HS, Ke CS, Cheng HC, Huang CY, and Su CL: Curcumin-induced mitotic spindle defect and cell cycle arrest in human bladder cancer cells occurs partly through inhibition of aurora A. Mol Pharmacol 80, 638–646, 2011.
  • Tao Y, Zhang P, Frascogna V, Lecluse Y, Auperin A, et al.: Enhancement of radiation response by inhibition of Aurora A kinase using siRNA or a selective Aurora kinase inhibitor PHA680632 in p53-deficient cancer cells. Br J Cancer 97, 1664–1672, 2007.
  • Kundu JK, Na HK, and Surh YJ: Ginger-derived phenolic substances with cancer preventive and therapeutic potential. Forum Nutr 61, 182–192, 2009.
  • Jolad SD, Lantz RC, Chen GJ, Bates RB, and Timmermann BN: Commercially processed dry ginger (Zingiber officinale): composition and effects on LPS-stimulated PGE2 production. Phytochemistry 66, 1614–1635, 2005.
  • Shim S, Kim S, Choi DS, Kwon YB, and Kwon J: Anti-inflammatory effects of [6]-shogaol: potential roles of HDAC inhibition and HSP70 induction. Food Chem Toxicol 49, 2734–2740, 2011.
  • Ling H, Yang H, Tan SH, Chui WK, and Chew EH: 6-Shogaol, an active constituent of ginger, inhibits breast cancer cell invasion by reducing matrix metalloproteinase-9 expression via blockade of nuclear factor-κB activation. Br J Pharmacol 161, 1763–1777, 2010.
  • Kim SO, Kundu JK, Shin YK, Park JH, Cho MH, et al.: [6]-gingerol inhibits COX-2 expression by blocking the activation of p38 MAP kinase and NF-kappaB in phorbol ester-stimulated mouse skin. Oncogene 24, 2558–2567, 2005.
  • Park YJ, Wen J, Bang S, Park SW, and Song SY: [6]-Gingerol induces cell cycle arrest and cell death of mutant p53-expressing pancreatic cancer cells. Yonsei Med J 47, 688–697, 2006.
  • Sung B, Murakami A, Oyajobi BO, and Aggarwal BB: Zerumbone abolishes RANKL-induced NF-kappaB activation, inhibits osteoclastogenesis, and suppresses human breast cancer-induced bone loss in athymic nude mice. Cancer Res 69, 1477–1484, 2009.
  • Takada Y, Murakami A, and Aggarwal BB: Zerumbone abolishes NF-kappaB and IkappaBalpha kinase activation leading to suppression of antiapoptotic and metastatic gene expression, upregulation of apoptosis, and downregulation of invasion. Oncogene 24, 6957–6969, 2005.
  • Chung IM, Kim MY, Park WH, and Moon HI: Histone deacetylase inhibitors from the rhizomes of Zingiber zerumbet. Pharmazie 63, 774–776, 2008.
  • Martín AR, Villegas I, La Casa C, and de la Lastra CA: Resveratrol, a polyphenol found in grapes, suppresses oxidative damage and stimulates apoptosis during early colonic inflammation in rats. Biochem Pharmacol 67, 1399–1410, 2004.
  • Venturelli S, Berger A, Böcker A, Busch C, Weiland T, et al.: Resveratrol as a pan-HDAC inhibitor alters the acetylation status of histone [corrected] proteins in human-derived hepatoblastoma cells. PLoS One 8, e73097, 2013.
  • Kai L, Samuel SK, and Levenson AS: Resveratrol enhances p53 acetylation and apoptosis in prostate cancer by inhibiting MTA1/NuRD complex. Int J Cancer 126, 1538–1548, 2010.
  • Ai Z, Li C, Li L, and He G: Resveratrol inhibits β-amyloid-induced neuronal apoptosis via regulation of p53 acetylation in PC12 cells. Mol Med Rep 11, 2429–2434, 2015.
  • Wang Y, Romigh T, He X, Orloff MS, Silverman RH, et al.: Resveratrol regulates the PTEN/AKT pathway through androgen receptor-dependent and -independent mechanisms in prostate cancer cell lines. Hum Mol Genet 19, 4319–4329, 2010.
  • Chen Q, Ganapathy S, Singh KP, Shankar S, and Srivastava RK: Resveratrol induces growth arrest and apoptosis through activation of FOXO transcription factors in prostate cancer cells. PLoS One 5, e15288, 2010.
  • Frazzi R, Valli R, Tamagnini I, Casali B, Latruffe N, et al.: Resveratrol-mediated apoptosis of hodgkin lymphoma cells involves SIRT1 inhibition and FOXO3a hyperacetylation. Int J Cancer 132, 1013–1021, 2013.
  • He X, Wang Y, Zhu J, Orloff M, and Eng C: Resveratrol enhances the anti-tumor activity of the mTOR inhibitor rapamycin in multiple breast cancer cell lines mainly by suppressing rapamycin-induced AKT signaling. Cancer Lett 301, 168–176, 2011.
  • Li W, Ma J, Ma Q, Li B, Han L, et al.: Resveratrol inhibits the epithelial-mesenchymal transition of pancreatic cancer cells via suppression of the PI-3K/Akt/NF-κB pathway. Curr Med Chem 20, 4185–4194, 2013.
  • Jiang H, Shang X, Wu H, Gautam SC, Al-Holou S, et al.: Resveratrol downregulates PI3K/Akt/mTOR signaling pathways in human U251 glioma cells. J Exp Ther Oncol 8, 25–33, 2009.
  • Zhang P, Li H, Yang B, Yang F, Zhang LL, et al.: Biological significance and therapeutic implication of resveratrol-inhibited Wnt, Notch and STAT3 signaling in cervical cancer cells. Genes Cancer 5, 154–164, 2014.
  • Liu X, Zhang DY, Zhang W, Zhao X, Yuan C, et al.: The effect of green tea extract and EGCG on the signaling network in squamous cell carcinoma. Nutr Cancer 63, 466–475, 2011.
  • Hu Q, Chang X, Yan R, Rong C, Yang C, et al.: (2)-Epigallocatechin-3-gallate induces cancer cell apoptosis via acetylation of amyloid precursor protein. Med Oncol 32, 390, 2015.
  • Pandey M, Shukla S, and Gupta S: Promoter demethylation and chromatin remodeling by green tea polyphenols leads to re-expression of GSTP1 in human prostate cancer cells. Int J Cancer 126, 2520–2533, 2010.
  • Adhami VM, Siddiqui IA, Ahmad N, Gupta S, and Mukhtar H: Oral consumption of green tea polyphenols inhibits insulin-like growth factor-I-induced signaling in an autochthonous mouse model of prostate cancer. Cancer Res 64, 8715–8722, 2004.
  • Klein RD and Fischer SM: Black tea polyphenols inhibit IGF-I-induced signaling through Akt in normal prostate epithelial cells and Du145 prostate carcinoma cells. Carcinogenesis 23, 217–221, 2002.
  • Kiss AK, Granica S, Stolarczyk M, and Melzig MF: Epigenetic modulation of mechanisms involved in inflammation: Influence of selected polyphenolic substances on histone acetylation state. Food Chem 131, 1015–1020, 2012.
  • Nandakumar V, Vaid M, and Katiyar SK: (-)-Epigallocatechin-3-gallate reactivates silenced tumor suppressor genes, Cip1/p21 and p16INK4a, by reducing DNA methylation and increasing histones acetylation in human skin cancer cells. Carcinogenesis 32, 537–544, 2011.
  • Roy SK, Srivastava RK, and Shankar S: Inhibition of PI3K/AKT and MAPK/ERK pathways causes activation of FOXO transcription factor, leading to cell cycle arrest and apoptosis in pancreatic cancer. J Mol Signal 5, 10, 2010.
  • Shimizu M, Deguchi A, Hara Y, Moriwaki H, and Weinstein IB: EGCG inhibits activation of the insulin-like growth factor-1 receptor in human colon cancer cells. Biochem Biophys Res Commun 334, 947–953, 2005.
  • Milligan SA, Burke P, Coleman DT, Bigelow RL, Steffan JJ, et al.: The green tea polyphenol EGCG potentiates the antiproliferative activity of c-Met and epidermal growth factor receptor inhibitors in non-small cell lung cancer cells. Clin Cancer Res 15, 4885–4894, 2009.
  • Kim SO and Kim MR: (-)-Epigallocatechin 3-gallate inhibits invasion by inducing the expression of Raf kinase inhibitor protein in AsPC-1 human pancreatic adenocarcinoma cells through the modulation of histone deacetylase activity. Int J Oncol 42, 349–358, 2013.
  • Masuda M, Suzui M, Lim JT, and Weinstein IB: Epigallocatechin-3-gallate inhibits activation of HER-2/neu and downstream signaling pathways in human head and neck and breast carcinoma cells. Clin Cancer Res 9, 3486–3491, 2003.
  • Pan MH, Lin CC, Lin JK, and Chen WJ: Tea polyphenol (-)-epigallocatechin 3-gallate suppresses heregulin-beta1-induced fatty acid synthase expression in human breast cancer cells by inhibiting phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase cascade signaling. J Agric Food Chem 55, 5030–5037, 2007.
  • Liang YC, Lin-Shiau SY, Chen CF, and Lin JK: Inhibition of cyclin-dependent kinases 2 and 4 activities as well as induction of Cdk inhibitors p21 and p27 during growth arrest of human breast carcinoma cells by (-)-epigallocatechin-3-gallate. J Cell Biochem 75, 1–12, 1999.
  • Tang Y, Zhao DY, Elliott S, Zhao W, Curiel TJ, et al.: Epigallocatechin-3 gallate induces growth inhibition and apoptosis in human breast cancer cells through survivin suppression. Int J Oncol 31, 705–711, 2007.
  • Qin J, Xie LP, Zheng XY, Wang YB, Bai Y, et al.: A component of green tea, (-)-epigallocatechin-3-gallate, promotes apoptosis in T24 human bladder cancer cells via modulation of the PI3K/Akt pathway and Bcl-2 family proteins. Biochem Biophys Res Commun 354, 852–857, 2007.
  • Noguchi M, Yokoyama M, Watanabe S, Uchiyama M, Nakao Y, et al.: Inhibitory effect of the tea polyphenol, (-)-epigallocatechin gallate, on growth of cervical adenocarcinoma cell lines. Cancer Lett 234, 135–142, 2006.
  • Lee JI, Nian H, Cooper AJ, Sinha R, Dai J, et al.: Alpha-keto acid metabolites of naturally occurring organoselenium compounds as inhibitors of histone deacetylase in human prostate cancer cells. Cancer Prev Res (Phila) 2, 683–693, 2009.
  • Suzana S, Cham BG, Ahmad Rohi G, Mohd Rizal R, Fairulnizal MN, et al.: Relationship between selenium and breast cancer: a case-control study in the Klang Valley. Singapore Med J 50, 265–269, 2009.
  • Suzuki M, Endo M, Shinohara F, Echigo S, and Rikiishi H: Differential apoptotic response of human cancer cells to organoselenium compounds. Cancer Chemother Pharmacol 66, 475–484, 2010.
  • Hu H, Jiang C, Li G, and Lu J: PKB/AKT and ERK regulation of caspase-mediated apoptosis by methylseleninic acid in LNCaP prostate cancer cells. Carcinogenesis 26, 1374–1381, 2005.
  • Nian H, Bisson WH, Dashwood WM, Pinto JT, and Dashwood RH: Alpha-keto acid metabolites of organoselenium compounds inhibit histone deacetylase activity in human colon cancer cells. Carcinogenesis 30, 1416–1423, 2009.
  • Vanommeslaeghe K, Loverix S, Geerlings P, and Tourwé D: DFT-based ranking of zinc-binding groups in histone deacetylase inhibitors. Bioorg Med Chem 13, 6070–6082, 2005.
  • Gasparian AV, Yao YJ, Lü J, Yemelyanov AY, Lyakh LA, et al.: Selenium compounds inhibit I kappa B kinase (IKK) and nuclear factor-kappa B (NF-kappa B) in prostate cancer cells. Mol Cancer Ther 1, 1079–1087, 2002.
  • Facompre ND, Sinha I, El-Bayoumy K, Pinto JT, and Sinha R: Remarkable inhibition of mTOR signaling by the combination of rapamycin and 1,4-phenylenebis(methylene)selenocyanate in human prostate cancer cells. Int J Cancer 131, 2134–2142, 2012.
  • Wang L, Hu H, Wang Z, Xiong H, Cheng Y, et al.: Methylseleninic acid suppresses pancreatic cancer growth involving multiple pathways. Nutr Cancer 66, 295–307, 2014.
  • Khanduja KL, Gandhi RK, Pathania V, and Syal N: Prevention of N-nitrosodiethylamine-induced lung tumorigenesis by ellagic acid and quercetin in mice. Food Chem Toxicol 37, 313–318, 1999.
  • Devipriya S, Ganapathy V, and Shyamaladevi CS: Suppression of tumour growth and invasion in 9,10-dimethylbenz[a]anthracene induced mammary carcinoma by the plant bioflavonoid quercetin. Chem Biol Interact 162, 106–113, 2006.
  • Shan BE, Wang MX, and Li RQ: Quercetin inhibit human SW480 colon cancer growth in association with inhibition of cyclin D(1) and surviving expression through Wnt/beta-catenin signaling pathway. Cancer Invest 27, 604–612, 2009.
  • Jeong JH, An JY, Kwon YT, Rhee JG, and Lee YJ: Effects of low dose quercetin: Cancer cell-specific inhibition of cell cycle progression. J Cell Biochem 106, 73–82, 2009.
  • Notoya M, Tsukamoto Y, Nishimura H, Woo JT, Nagai K, et al.: Quercetin, a flavonoid, inhibits the proliferation, differentiation, and mineralization of osteoblasts in vitro. Eur J Pharmacol 485, 89–96, 2004.
  • Bishayee K, Khuda-Bukhsh AR, and Huh SO: PLGA-loaded gold-nanoparticles precipitated with quercetin downregulate HDAC-Akt activities controlling proliferation and activate p53-ROS crosstalk to induce apoptosis in hepatocarcinoma cells. Mol Cells 38, 518–527, 2015.
  • Priyadarsini RV, Vinothini G, Murugan RS, Manikandan P, and Nagini S: The flavonoid quercetin modulates the hallmark capabilities of hamster buccal pouch tumors. Nutr Cancer 63, 218–226, 2011.
  • Kim YH, Lee DH, Jeong JH, Guo ZS, and Lee YJ: Quercetin augments TRAIL-induced apoptotic death: involvement of the ERK signal transduction pathway. Biochem Pharmacol 75, 1946–1958, 2008.
  • Lee WJ, Chen YR, and Tseng TH: Quercetin induces FasL-related apoptosis, in part, through promotion of histone H3 acetylation in human leukemia HL-60 cells. Oncol Rep 25, 583–591, 2011.
  • Wang Y, Han A, Chen E, Singh RK, Chichester CO, et al.: The cranberry flavonoids PAC DP-9 and quercetin aglycone induce cytotoxicity and cell cycle arrest and increase cisplatin sensitivity in ovarian cancer cells. Int J Oncol 46, 1924–1934, 2015.
  • Delmonte P, Perry J, and Rader JI: Determination of isoflavones in dietary supplements containing soy, Red Clover and kudzu: extraction followed by basic or acid. J Chromatogr A 1107, 59–69, 2006.
  • Kikuno N, Shiina H, Urakami S, Kawamoto K, Hirata H, et al.: Genistein mediated histone acetylation and demethylation activates tumor suppressor genes in prostate cancer cells. Int J Cancer 123, 552–560, 2008.
  • Majid S, Dar AA, Ahmad AE, Hirata H, Kawakami K, et al.: BTG3 tumor suppressor gene promoter demethylation, histone modification and cell cycle arrest by genistein in renal cancer. Carcinogenesis 30, 662–670, 2009.
  • Majid S, Dar AA, Shahryari V, Hirata H, Ahmad A, et al.: Genistein reverses hypermethylation and induces active histone modifications in tumor suppressor gene B-Cell translocation gene 3 in prostate cancer. Cancer 116, 66–76, 2010.
  • Phillip CJ, Giardina CK, Bilir B, et al.: Genistein cooperates with the histone deacetylase inhibitor vorinostat to induce cell death in prostate cancer cells. BMC Cancer 12, 145, 2012.
  • Wang H, Li Q, and Chen H: Genistein affects histone modifications on Dickkopf-Related Protein 1 (DKK1) gene in SW480 human colon cancer cell line. PLoS One 7, e40955, 2012.
  • Li Y and Sarkar FH: Inhibition of nuclear factor kappaB activation in PC3 cells by genistein is mediated via Akt signaling pathway. Clin Cancer Res 8, 2369–2377, 2002.
  • Ferenc P, Solár P, Kleban J, Mikes J, and Fedorocko P: Down-regulation of Bcl-2 and Akt induced by combination of photoactivated hypericin and genistein in human breast cancer cells. J Photochem Photobiol B 98, 25–34, 2010.
  • Nakamura Y, Yogosawa S, Izutani Y, Watanabe H, Otsuji E, et al.: A combination of indol-3-carbinol and genistein synergistically induces apoptosis in human colon cancer HT-29 cells by inhibiting Akt phosphorylation and progression of autophagy. Mol Cancer 8, 100, 2009.
  • Sakla MS, Shenouda NS, Ansell PJ, Macdonald RS, and Lubahn DB: Genistein affects HER2 protein concentration, activation, and promoter regulation in BT-474 human breast cancer cells. Endocrine 32, 69–78, 2007.
  • Messing E, Gee JR, Saltzstein DR, Kim K, diSant'Agnese A, et al.: A phase 2 cancer chemoprevention biomarker trial of isoflavone G-2535 (genistein) in presurgical bladder cancer patients. Cancer Prev Res (Phila) 5, 621–630, 2012.
  • Basak S, Pookot D, Noonan EJ, and Dahiya R: Genistein down-regulates androgen receptor by modulating HDAC6-Hsp90 chaperone function. Mol Cancer Ther 7, 3195–3202, 2008.
  • Renehan AG, Zwahlen M, Minder C, O'Dwyer ST, Shalet SM, et al.: Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: systematic review and meta-regression analysis. Lancet 363, 1346–1353, 2004.
  • Lee J, Ju J, Park S, Hong SJ, and Yoon S: Inhibition of IGF-1 signaling by genistein: modulation of E-cadherin expression and downregulation of β-catenin signaling in hormone refractory PC-3 prostate cancer cells. Nutr Cancer 64, 153–162, 2012.
  • Li Y, Wang Z, Kong D, Li R, Sarkar SH, et al.: Regulation of Akt/FOXO3a/GSK-3beta/AR signaling network by isoflavone in prostate cancer cells. J Biol Chem 283, 27707–27716, 2008.
  • Singh-Gupta V, Zhang H, Banerjee S, Kong D, Raffoul JJ, et al.: Radiation-induced HIF-1α cell survival pathway is inhibited by soy isoflavones in prostate cancer cells. Int J Cancer 124, 1675–1684, 2009.
  • Trujillo E, Davis C, and Milner J: Nutrigenomics, proteomics, metabolomics, and the practice of dietetics. J Am Diet Assoc 106, 403–413, 2006.
  • Venkateswaran V, Haddad AQ, Fleshner NE, Fan R, Sugar LM, et al.: Association of diet-induced hyperinsulinemia with accelerated growth of prostate cancer (LNCaP) xenografts. J Natl Cancer Inst 99, 1793–1800, 2007.
  • Honma K, Mochizuki K, and Goda T: Carbohydrate/fat ratio in the diet alters histone acetylation on the sucrase-isomaltase gene and its expression in mouse small intestine. Biochem Biophys Res Commun 357, 1124–1129, 2007.
  • Honma K, Mochizuki K, and Goda T: Inductions of histone H3 acetylation at lysine 9 on SGLT1 gene and its expression by feeding mice a high carbohydrate/fat ratio diet. Nutrition 25, 40–44, 2009.
  • Inoue S, Mochizuki K, and Goda T: Jejunal induction of SI and SGLT1 genes in rats by high-starch/low-fat diet is associated with histone acetylation and binding of GCN5 on the genes. J Nutr Sci Vitaminol (Tokyo) 57, 162–169, 2011.
  • Kitada M, Kume S, Takeda-Watanabe A, Tsuda S, Kanasaki K, et al.: Calorie restriction in overweight males ameliorates obesity-related metabolic alterations and cellular adaptations through anti-aging effects, possibly including AMPK and SIRT1 activation. Biochim Biophys Acta 1830, 4820–4827, 2013.
  • Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, et al.: Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339, 211–214, 2013.
  • Draznin B, Wang C, Adochio R, Leitner JW, and Cornier MA: Effect of dietary macronutrient composition on AMPK and SIRT1 expression and activity in human skeletal muscle. Horm Metab Res 44, 650–655, 2012.
  • Bergouignan A, Gozansky WS, Barry DW, Leitner W, MacLean PS, et al.: Increasing dietary fat elicits similar changes in fat oxidation and markers of muscle oxidative capacity in lean and obese humans. PLoS One 7, e30164, 2012.
  • Fontana L, Adelaiye RM, Rastelli AL, Miles KM, Ciamporcero E, et al.: Dietary protein restriction inhibits tumor growth in human xenograft models of prostate and breast cancer. Oncotarget 4, 2451–2461, 2013.
  • Zheng S, Rollet M, Yang K, and Pan YX: A gestational low-protein diet represses p21(WAF1/Cip1) expression in the mammary gland of offspring rats through promoter histone modifications. Br J Nutr 108, 998–1007, 2012.
  • Kobayashi N, Barnard RJ, Said J, Hong-Gonzalez J, Corman DM, et al.: Effect of low-fat diet on development of prostate cancer and Akt phosphorylation in the Hi-Myc transgenic mouse model. Cancer Res 68, 3066–3073, 2008.
  • Bassett JK, Severi G, Hodge AM, MacInnis RJ, Gibson RA, et al.: Plasma phospholipid fatty acids, dietary fatty acids and prostate cancer risk. Int J Cancer 133, 1882–1891, 2013.
  • Cummings JH and Englyst HN: Fermentation in the human large intestine and the available substrates. Am J Clin Nutr 45, 1243–1255, 1987.
  • Wong JM, de Souza R, Kendall CW, Emam A, and Jenkins DJ: Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol 40, 235–243, 2006.
  • Sekhavat A, Sun JM, and Davie JR: Competitive inhibition of histone deacetylase activity by trichostatin A and butyrate. Biochem Cell Biol 85, 751–758, 2007.
  • Blottière HM, Buecher B, Galmiche JP, and Cherbut C: Molecular analysis of the effect of short-chain fatty acids on intestinal cell proliferation. Proc Nutr Soc 62, 101–106, 2003.
  • Hassig CA, Tong JK, and Schreiber SL: Fiber-derived butyrate and the prevention of colon cancer. Chem Biol 4, 783–789, 1997.
  • Li GX, Lee MJ, Liu AB, Yang Z, Lin Y, et al.: δ-tocopherol is more active than α - or γ -tocopherol in inhibiting lung tumorigenesis in vivo. Cancer Prev Res (Phila) 4, 404–413, 2011.
  • Wang H, Hong J, and Yang CS: δ-Tocopherol inhibits receptor tyrosine kinase-induced AKT activation in prostate cancer cells. Mol Carcinog 2015.
  • Lee HJ, Ju J, Paul S, So JY, DeCastro A, et al.: Mixed tocopherols prevent mammary tumorigenesis by inhibiting estrogen action and activating PPAR-γ. Clin Cancer Res 15, 4242–4249, 2009.
  • Ahn KS, Sethi G, Krishnan K, and Aggarwal BB: Gamma-tocotrienol inhibits nuclear factor-kappaB signaling pathway through inhibition of receptor-interacting protein and TAK1 leading to suppression of antiapoptotic gene products and potentiation of apoptosis. J Biol Chem 282, 809–820, 2007.
  • Kunnumakkara AB, Sung B, Ravindran J, Diagaradjane P, Deorukhkar A, et al.: {Gamma}-tocotrienol inhibits pancreatic tumors and sensitizes them to gemcitabine treatment by modulating the inflammatory microenvironment. Cancer Res 70, 8695–8705, 2010.
  • Samant GV and Sylvester PW: Gamma-Tocotrienol inhibits ErbB3-dependent PI3K/Akt mitogenic signalling in neoplastic mammary epithelial cells. Cell Prolif 39, 563–574, 2006.
  • Ye C, Zhao W, Li M, Zhuang J, Yan X, et al.: δ-tocotrienol induces human bladder cancer cell growth arrest, apoptosis and chemosensitization through inhibitionof STAT3 pathway. PLoS One 10, e0122712, 2015.
  • Birringer M, Pfluger P, Kluth D, Landes N, and Brigelius-Flohé R: Identities and differences in the metabolism of tocotrienols and tocopherols in HepG2 cells. J Nutr 132, 3113–3118, 2002.
  • Majumdar AP, Banerjee S, Nautiyal J, Patel BB, Patel V, et al.: Curcumin synergizes with resveratrol to inhibit colon cancer. Nutr Cancer 61(4), 544–553, 2009.
  • Verma SP, Salamone E, and Goldin B: Curcumin and genistein, plant natural products, show synergistic inhibitory effects on the growth of human breast cancer MCF-7 cells induced by estrogenic pesticides. Biochem Biophys Res Commun 233, 692–696, 1997.
  • Khafif A, Schantz SP, Chou TC, Edelstein D, and Sacks PG: Quantitation of chemopreventive synergism between (−)-epigallocatechin-3-gallate and curcumin in not normal, premalignant and malignant human oral epithelial cells. Carcinogenesis 19, 419–424, 1998.
  • Cruz-Correa M, Shoskes DA, Sanchez P, Zhao R, Hylind LM, et al.: Combination treatment with curcumin and quercetin of adenomas in familial adenomatous polyposis. Clin Gastroenterol Hepatol 4(8), 1035–1038, 2006.
  • Mertens-Talcott SU and Percival SS: Ellagic acid and quercetin interact synergistically with resveratrol in the induction of apoptosis and cause transient cell cycle arrest in human leukemia cells. Cancer Lett 218(2), 141–151, 2005.
  • Kowalczyk MC, Kowalczyk P, Tolstykh O, Hanausek M, and Walaszek Z: Synergistic effects of combined phytochemicals and skin cancer prevention in SENCAR mice. Cancer Prev Res (Phila) 3(2), 170–178, 2010.
  • Pappa G, Strathmann J, Löwinger M, Bartsch H, and Gerhäuser C: Quantitative combination effects between sulforaphane and 3,3′-diindolylmethane on proliferation of human colon cancer cells in vitro. Carcinogenesis 28, 1471–1477, 2007.
  • Hsieh TC and Wu JM: Suppression of cell proliferation and gene expression by combinatorial synergy of EGCG, resveratrol and gamma-tocotrienol in estrogen receptor-positive MCF-7 breast cancer cells. Int J Oncol 33(4), 851–859, 2008.
  • Russo GL: Ins and outs of dietary phytochemicals in cancer chemoprevention. Biochem Pharmacol 74, 533–544, 2007.
  • Gupta S, Hastak K, Ahmad N, Lewin JS, and Mukhtar H: Inhibition of prostate carcinogenesis in TRAMP mice by oral infusion of green tea polyphenols. Proc Natl Acad Sci USA 98(18), 10350–10355, 2001.
  • Shanafelt TD, Lee YK, Call TG, Nowakowski GS, Dingli D, et al.: Clinical effects of oral green tea extracts in four patients with low grade B-cell malignancies. Leuk Res 30(6), 707–712, 2006.
  • hang SM, Hunter DJ, Rosner BA, Giovannucci EL, Colditz GA, et al.: Intakes of fruits, vegetables, and related nutrients and the risk of non-Hodgkin's lymphoma among women. Cancer Epidemiol Biomarkers Prev 9, 477–485, 2000.
  • Michaud DS, Spiegelman D, Clinton SK, Rimm EB, Willett WC, et al.: Fruit and vegetable intake and incidence of bladder cancer in a male prospective cohort. J Natl Cancer Inst 91, 605–613, 1999.
  • Cohen JH, Kristal AR, and Stanford JL: Fruit and vegetable intakes and prostate cancer risk. J Natl Cancer Inst 92, 61–68, 2000.
  • Zhou Y, Zhuang W, Hu W, Liu GJ, Wu TX, et al.: Consumption of large amounts of Allium vegetables reduces risk for gastric cancer in a meta-analysis. Gastroenterology 141, 80–89, 2011.
  • Galeone C, Pelucchi C, Levi F, Negri E, Franceschi S, Talamini R, et al.: Onion and garlic use and human cancer. Am J Clin Nutr 84, 1027–1032, 2006.

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.