期刊论文详细信息
Clinical Epigenetics
Nrf2 status affects tumor growth, HDAC3 gene promoter associations, and the response to sulforaphane in the colon
Roderick H. Dashwood4  David A. Lieberman5  Masayuki Yamamoto8  Emily Ho2  David E. Williams1  Christiane V. Löhr6  Kay A. Fischer6  Gavin Johnson3  Eunah Kim3  Yuki Kang7  Li Li3  Wan-Mohaiza Dashwood3  Praveen Rajendran3 
[1] Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA;College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA;Center for Epigenetics & Disease Prevention, Texas A&M Health Science Center, Houston, TX, USA;Department of Molecular & Cellular Medicine, Texas A&M University, College Station, TX, USA;Department of Medicine, Oregon Health & Science University, Portland, OR, USA;College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA;Linus Pauling Institute, Oregon State University, Corvallis, OR, USA;Division of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
关键词: Broccoli;    Sulforaphane;    Colon cancer;    Nrf2;    p16;    HDAC3;   
Others  :  1225826
DOI  :  10.1186/s13148-015-0132-y
 received in 2015-06-30, accepted in 2015-09-02,  发布年份 2015
PDF
【 摘 要 】

Background

The dietary agent sulforaphane (SFN) has been reported to induce nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2)-dependent pathways as well as inhibiting histone deacetylase (HDAC) activity. The current investigation sought to examine the relationships between Nrf2 status and HDAC expression in preclinical and translational studies.

Results

Wild type (WT) and Nrf2-deficient (Nrf2 −/+ ) mice were treated with the colon carcinogen 1,2-dimethylhydrazine (DMH) followed by 400 ppm SFN in the diet (n = 35 mice/group). WT mice were more susceptible than Nrf2 −/+mice to tumor induction in the colon. Tumors from WT mice had higher HDAC levels globally and locally on genes such as cyclin-dependant kinase inhibitor 2a (Cdkn2a/p16) that were dysregulated during tumor development. The average tumor burden was reduced by SFN from 62.7 to 26.0 mm 3in WT mice and from 14.6 to 11.7 mm 3in Nrf2 −/+mice. The decreased antitumor activity of SFN in Nrf2 −/+mice coincided with attenuated Cdkn2a promoter interactions involving HDAC3. HDAC3 knockdown in human colon cancer cells recapitulated the effects of SFN on p16 induction. Human subjects given a broccoli sprout extract supplement (200 μmol SFN equivalents), or reporting more than five cruciferous vegetable servings per week, had increased p16 expression that was inversely associated with HDAC3 in circulating peripheral blood mononuclear cells (PBMCs) and in biopsies obtained during screening colonoscopy.

Conclusions

Nrf2 expression varies widely in both normal human colon and human colon cancers and likely contributes to the overall rate of tumor growth in the large intestine. It remains to be determined whether this influences global HDAC protein expression levels, as well as local HDAC interactions on genes dysregulated during human colon tumor development. If corroborated in future studies, Nrf2 status might serve as a biomarker of HDAC inhibitor efficacy in clinical trials using single agent or combination modalities to slow, halt, or regress the progression to later stages of solid tumors and hematological malignancies.

【 授权许可】

   
2015 Rajendran et al.

【 预 览 】
附件列表
Files Size Format View
20150922032554982.pdf 2110KB PDF download
Fig. 9. 27KB Image download
Fig. 8. 52KB Image download
Fig. 7. 26KB Image download
Fig. 6. 62KB Image download
Fig. 5. 64KB Image download
Fig. 4. 54KB Image download
Fig. 3. 45KB Image download
Fig. 2. 28KB Image download
Fig. 1. 43KB Image download
【 图 表 】

Fig. 1.

Fig. 2.

Fig. 3.

Fig. 4.

Fig. 5.

Fig. 6.

Fig. 7.

Fig. 8.

Fig. 9.

【 参考文献 】
  • [1]Fredly H, Gjertsen B, Bruserud Ø: Histone deacetylase inhibition in the treatment of acute myeloid leukemia: the effects of valproic acid on leukemic cells, and the clinical and experimental evidence for combining valproic acid with other antileukemic agents. Clin Epigenetics. 2013, 5:12. BioMed Central Full Text
  • [2]Mack GS: Epigenetic cancer therapy makes headway. J Natl Cancer Inst. 2006, 98:1443-4.
  • [3]Wilson AJ, Byun DS, Popova N, Murray LB, L'Italien K, Sowa Y, et al.: Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. J Biol Chem. 2006, 281:13548-58.
  • [4]Marks PA, Xu W-S: Histone deacetylase inhibitors: potential in cancer therapy. J Cell Biochem. 2009, 107:600-8.
  • [5]Delcuve GP, Khan DH, Davie JR: Roles of histone deacetylases in epigenetic regulation: emerging paradigms from studies with inhibitors. Clin Epigenetics. 2012, 4:5. BioMed Central Full Text
  • [6]Dashwood RH, Myzak MC, Ho E: Dietary HDAC inhibitors: time to rethink weak ligands in cancer chemoprevention? Carcinogenesis. 2006, 27:344-9.
  • [7]Rajendran P, Ho E, Williams DE, Dashwood RH: Dietary phytochemicals, HDAC inhibition, and DNA damage/repair defects in cancer cells. Clin Epigenetics. 2011, 3:4. BioMed Central Full Text
  • [8]Rajendran P, Williams DE, Ho E, Dashwood RH: Metabolism as a key to histone deacetylase inhibition. Crit Rev Biochem Mol Biol. 2011, 46:181-99.
  • [9]Myzak MC, Dashwood RH: Chemoprotection by sulforaphane: keep one eye beyond Keap1. Cancer Lett. 2006, 233:208-18.
  • [10]Myzak MC, Dashwood WM, Orner GA, Ho E, Dashwood RH: Sulforaphane inhibits histone deacetylase in vivo and suppresses tumorigenesis in Apc min mice. FASEB J. 2006, 20:506-8.
  • [11]Myzak MC, Karplus PA, Chung F-L, Dashwood RH: A novel mechanism of chemoprotection by sulforaphane: inhibition of histone deacetylase. Cancer Res. 2004, 64:5767-74.
  • [12]Myzak MC, Tong P, Dashwood W-M, Dashwood RH, Ho E: Sulforaphane retards the growth of human PC-3 xenografts and inhibits HDAC activity in human subjects. Exp Biol Med. 2007, 232:227-34.
  • [13]Fahey JW, Zhang Y, Talalay P: Broccoli sprouts: an exceptionally rich source of inducers of enzymes that protect against chemical carcinogens. Proc Natl Acad Sci USA 1997, 94:10367-72.
  • [14]Zhang Y, Talalay P, Cho CG, Posner GH: A major inducer of anticarcinogenic protective enzymes from broccoli: isolation and elucidation of structure. Proc Natl Acad Sci USA 1992, 89:2399-403.
  • [15]Kensler TW, Egner PA, Agyeman AS, Visvanathan K, Groopman JD, Chen JG, et al.: Keap1-nrf2 signaling: a target for cancer prevention by sulforaphane. Top Curr Chem. 2013, 329:163-77.
  • [16]Satoh H, Moriguchi T, Takai J, Ebina M, Yamamoto M: Nrf2 prevents initiation but accelerates progression through the Kras signaling pathway during lung carcinogenesis. Cancer Res. 2013, 73:4158-68.
  • [17]Kensler TW, Wakabayashi N: Nrf2: friend or foe for chemoprevention? Carcinogenesis. 2010, 31:90-9.
  • [18]Corpet DE, Pierre F: How good are rodent models of carcinogenesis in predicting efficacy in humans? A systematic review and meta-analysis of colon chemoprevention in rats, mice and men. Eur J Cancer. 2005, 41:1911-22.
  • [19]Sumiyoshi H, Wargovich MJ: Chemoprevention of 1,2-dimethylhydrazine-induced colon cancer in mice by naturally occurring organosulfur compounds. Cancer Res. 1990, 50:5084-7.
  • [20]Rajendran P, Delage B, Dashwood WM, Yu TW, Wuth B, Williams DE, et al.: Histone deacetylase turnover and recovery in sulforaphane-treated colon cancer cells: competing actions of 14-3-3 and Pin1 in HDAC3/SMRT corepressor complex dissociation/reassembly. Mol Cancer. 2011, 10:68. BioMed Central Full Text
  • [21]Rajendran P, Kidane AI, Yu TW, Dashwood WM, Bisson WH, Löhr CV, et al.: HDAC turnover, CtIP acetylation and dysregulated DNA damage signaling in colon cancer cells treated with sulforaphane and related dietary isothiocyanates. Epigenetics. 2013, 8:612-23.
  • [22]Clarke JD, Hsu A, Williams DE, Dashwood RH, Stevens JF, Yamamoto M, et al.: Metabolism and tissue distribution of sulforaphane in Nrf2 knockout and wild-type mice. Pharm Res. 2011, 28:3171-9.
  • [23]Feng Y, Wang X, Xu L, Pan H, Zhu S, Liang Q, et al.: The transcription factor ZBP-89 suppresses p16 expression through a histone modification mechanism to affect cell senescence. FEBS J. 2009, 276:4197-206.
  • [24]Wang X, Feng Y, Xu L, Chen Y, Zhang Y, Su D, et al.: YY1 restrained cell senescence through repressing the transcription of p16. Biochim Biophys Acta. 1783, 2008:1876-83.
  • [25]Zheng S, Li Q, Zhang Y, Balluff Z, Pan Y-X: Histone deacetylase 3 (HDAC3) participates in the transcriptional repression of the p16 (INK4a) gene in mammary gland of the female rat offspring exposed to an early-life high-fat diet. Epigenetics. 2012, 7:183-90.
  • [26]Linggi B, Müller-Tidow C, van de Locht L, Hu M, Nip J, Serve H, et al.: The t(8;21) fusion protein, AML1 ETO, specifically represses the transcription of the p14(ARF) tumor suppressor in acute myeloid leukemia. Nat Med. 2002, 8:743-50.
  • [27]Atwell LL, Hsu A, Wong CP, Stevens JF, Bella D, Yu TW, et al.: Absorption and chemopreventive targets of sulforaphane in humans following consumption of broccoli sprouts or a myrosinase-treated broccoli sprout extract. Mol Nutr Food Res. 2015, 59:424-33.
  • [28]Thomson CA, Rock CL, Caan BJ, Flatt SW, Al-Delaimy WA, Newman VA, et al.: Increase in cruciferous vegetable intake in women previously treated for breast cancer participating in a dietary intervention trial. Nutr Cancer. 2007, 57:11-19.
  • [29]Chung FL, Conaway CC, Rao CV, Reddy BS: Chemoprevention of colonic aberrant crypt foci in Fischer rats by sulforaphane and phenethyl isothiocyanate. Carcinogenesis. 2000, 21:2287-91.
  • [30]Glauben R, Batra A, Stroh T, Erben U, Fedke I, Lehr HA, et al.: Histone deacetylases: novel targets for prevention of colitis-associated cancer in mice. Gut. 2008, 57:613-22.
  • [31]Hu R, Khor TO, Shen G, Jeong WS, Hebbar V, Chen C, et al.: Cancer chemoprevention of intestinal polyposis in ApcMin/+ mice by sulforaphane, a natural product derived from cruciferous vegetables. Carcinogenesis. 2006, 27:2038-46.
  • [32]Cheung KL, Lee JH, Khor TO, Wu TY, Li GX, Chan J, et al.: Nrf2 knockout enhances intestinal tumorigenesis in Apc(min/+) mice due to attenuation of anti-oxidative stress pathway while potentiates inflammation. Mol Carcinog. 2014, 53:77-84.
  • [33]Khor TO, Huang M-T, Kwon KH, Chan JY, Reddy BS, Kong A-N: Nrf2-deficient mice have an increased susceptibility to dextran sulfate sodium-induced colitis. Cancer Res. 2006, 66:11580-4.
  • [34]Khor TO, Huang MT, Prawan A, Liu Y, Hao X, Yu S, et al.: Increased susceptibility of Nrf2 knockout mice to colitis-associated colorectal cancer. Cancer Prev Res. 2008, 1:187-91.
  • [35]Osburn WO, Karim B, Dolan PM, Liu G, Yamamoto M, Huso DL, et al.: Increased colonic inflammatory injury and formation of aberrant crypt foci in Nrf2-deficient mice upon dextran sulfate treatment. Int J Cancer. 2007, 121:1883-91.
  • [36]Sporn MB, Liby KT: NRF2 and cancer: the good, the bad, and the importance of context. Nat Rev Cancer. 2012, 12:564-71.
  • [37]Wang QS, Papanikolaou A, Nambiar PR, Rosenberg DW: Differential expression of p16(INK4a) in azoxymethane-induced mouse colon tumorigenesis. Mol Carcinog. 2000, 28:139-47.
  • [38]Romagosa C, Simonetti S, López-Vicente L, Mazo A, Lleonart ME, Castellvi J, et al.: p16(Ink4a) overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors. Oncogene. 2011, 30:2087-97.
  • [39]Chen M, Voeller D, Marquez VE, Kaye FJ, Steeg PS, Giaccone G, et al.: Enhanced growth inhibition by combined DNA methylation/HDAC inhibitors in lung tumor cells with silenced CDKN2A. Int J Oncol. 2010, 37:963-71.
  • [40]Yamaguchi J, Sasaki M, Sato Y, Itatsu K, Harada K, Zen Y, et al.: Histone deacetylase inhibitor (SAHA) and repression of EZH2 synergistically inhibit proliferation of gallbladder carcinoma. Cancer Sci. 2010, 101:355-62.
  • [41]Valentini A, Gravina P, Federici G, Bernardini S: Valproic acid induces apoptosis, p16INK4A upregulation, and sensitization to chemotherapy in human melanoma cells. Cancer Biol Ther. 2007, 6:185-91.
  • [42]Chien WW, Ffrench M: Regulation of p16INK4a, senescence and oncogenesis. Med Sci. 2006, 22:865-71.
  • [43]Ohtani N, Zebedee Z, Huot TJ, Stinson JA, Sugimoto M, Ohashi Y, et al.: Opposing effects of Ets and Id proteins on p16INK4a expression during cellular senescence. Nature. 2001, 409:1067-70.
  • [44]Wang W, Pan K, Chen Y, Huang C, Zhang X: The acetylation of transcription factor HBP1 by p300/CBP enhances p16INK4A expression. Nucleic Acids Res. 2012, 40:981-95.
  • [45]Borinstein SC, Conerly M, Dzieciatkowski S, Biswas S, Washington MK, Trobridge P, et al.: Aberrant DNA methylation occurs in colon neoplasms arising in the azoxymethane colon cancer model. Mol Carcinog. 2010, 49:94-103.
  • [46]Meyer LR, Zweig AS, Hinrichs AS, Karolchik D, Kuhn RM, Wong M, et al.: The UCSC Genome Browser database: extensions and updates 2013. Nucleic Acids Res. 2013, 41:D64-9.
  • [47]Tuma RS: Epigenetic therapies move into new territory, but how exactly do they work? J Natl Cancer Inst 2009, 101(19):1300-1.
  • [48]Wang R, Dashwood WM, Nian H, Löhr CV, Fischer KA, Tsuchiya N, et al.: NADPH oxidase overexpression in human colon cancers and rat colon tumors induced by 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP). Int J Cancer. 2011, 128:2581-90.
  • [49]Nian H, Delage B, Pinto JT, Dashwood RH: Allyl mercaptan, a garlic-derived organosulfur compound, inhibits histone deacetylase and enhances Sp3 binding on the P21WAF1 promoter. Carcinogenesis. 2008, 29:1816-24.
  • [50]Parasramka M, Dashwood WM, Wang R, Saeed HH, Williams DE, Ho E, et al.: A role for low-abundance miRNAs in colon cancer: the miR-206/Krüppel-like factor 4 (KLF4) axis. Clin Epigenetics. 2012, 4:16. BioMed Central Full Text
  文献评价指标  
  下载次数:23次 浏览次数:11次