Nrf2 status affects tumor growth, HDAC3 gene promoter associations, and the response to sulforaphane in the colon

Praveen Rajendran, Wan-Mohaiza Dashwood, Li Li, Yuki Kang, Eunah Kim, Gavin Johnson, Kay A Fischer, Christiane V Löhr, David E Williams, Emily Ho, Masayuki Yamamoto, David A Lieberman, Roderick H Dashwood, Praveen Rajendran, Wan-Mohaiza Dashwood, Li Li, Yuki Kang, Eunah Kim, Gavin Johnson, Kay A Fischer, Christiane V Löhr, David E Williams, Emily Ho, Masayuki Yamamoto, David A Lieberman, Roderick H Dashwood

Abstract

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(3) in WT mice and from 14.6 to 11.7 mm(3) in 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.

Keywords: Broccoli; Colon cancer; HDAC3; Nrf2; Sulforaphane; p16.

Figures

Fig. 1
Fig. 1
Continuous and alternating dosing schedules for dietary SFN in the DMH model. a Male ICR mice were injected i.p. with DMH (20 mg/kg), once per week for 10 weeks (arrows). One week after completing the DMH treatment, mice were continued on standard AIN93 diet (no SFN) or given AIN93 diet containing 400 ppm SFN either continuously (daily SFN, green box) or on alternate days (Alt SFN, stripes). b Tumor multiplicity and c total tumor burden, i.e., the sum of individual tumor volumes, were determined for each animal at 25 weeks, and mean values were calculated (dotted line). *P < 0.05 vs. no SFN controls
Fig. 2
Fig. 2
SFN decreased HDAC activity and HDAC3 protein expression in mouse colon tumors. a HDAC activity in colon tumor lysates was determined as described in “Methods” section. Mean values were calculated in each group (dotted line). *P < 0.05 vs. no SFN controls. b HDAC expression and histone acetylation determined by immunoblotting. c Densitometry data for HDACs were normalized to β-actin, whereas acetylated histone H4K12 (AcH4K12) was normalized to histone H4. Data = mean ± SD (n = 3); *P < 0.05 vs. no SFN controls
Fig. 3
Fig. 3
Antitumor activity of dietary SFN in Nrf2+/+ and Nrf2−/+ mice. a Nrf2+/+ (WT) or Nrf2−/+ mice were injected i.p. with DMH (20 mg/kg), once per week for 8 weeks (arrows). One week after completing the carcinogen treatment, mice were continued on standard AIN93 diet (−SFN) or AIN93 diet containing 400 ppm SFN (+SFN). b Total tumor burden/mouse indicates the sum of individual tumor volumes after 35 weeks, plotted for each animal; dotted line represents the mean value in each group; P < 0.05 as indicated by the asterisk. c Relative mRNA expression levels of Nrf2 normalized to the β-actin gene, Actb. Data = mean ± SD (n = 3)
Fig. 4
Fig. 4
SFN decreased HDAC3 protein expression in colon tumors of Nrf2+/+ but not Nrf2−/+ mice. a HDAC expression was immunoblotted in tumor (T) and adjacent normal-looking colon (N) of WT mice, with densitometry data normalized to β-actin. b The corresponding data for Nrf2−/+ mice. Data are given as mean ± SD (n = 3); *P < 0.05, **P < 0.01 for control diet vs. SFN treatment. In addition to global reductions in HDAC3, marked loss of HDAC3 on gene targets was examined (see Fig. 6)
Fig. 5
Fig. 5
Differential gene expression in mouse colon tumors and normal colon. a Heat map representing the expression of differentially expressed genes, relative to Actb. Each vertical column represents the gene expression profile of pooled colon tumors or adjacent normal colon (n = 6) of mice on standard AIN93 diet (−SFN) or AIN93 diet containing 400 ppm SFN (+SFN), according to the groups indicated. Each row in the heat map represents a single named gene. Red represents high expression, green designates low expression. be Scatter plots compared the normalized expression of genes in the array by plotting log10 transformed 2−ΔCt values between selected groups. The most significantly altered genes, with an arbitrary cutoff of fivefold in either direction (upregulated or downregulated), are identified by the name adjacent to the corresponding data point. The red arrow designates Cdkn2a (p16)
Fig. 6
Fig. 6
Nrf2 status affects p16 expression and HDAC3 promoter interactions in mouse colon tumors. ap16 mRNA expression levels in mouse colon tumors were normalized to Actb. Data = mean ± SD (n = 3); *P < 0.05 for control diet vs. SFN treatment. be Representative immunohistochemical staining (×10) of p16 in mouse colon tumor and adjacent normal colon; tumor (T), normal (N). A magnified (×40) image is shown for each dotted box, with a 50-μm scale bar included in b, inset. f In vivo ChIP assays on mouse colon tumors, interrogating HDAC3 interactions on p16. Data = mean ± SD and are representative of three independent experiments. A region 10 Kb upstream of the p16 promoter, containing a Runx1 binding site, served as a negative control for the ChIP assays
Fig. 7
Fig. 7
HDAC3 knockdown induces p16 expression in human colon cancer cells. a HCT116 cells were transfected with non-specific scrambled negative control siRNA (Neg siRNA), HDAC3 siRNA, or Keap1 siRNA for 48 h, followed by vehicle (−SFN) or 15-μM SFN treatment for 24 h. p16 mRNA levels were normalized to ACTB; data = mean ± SD (n = 3), *P < 0.05 compared to untreated Neg siRNA. b The corresponding whole cell lysates were immunoblotted for p16, HDAC3, HDAC6 and Nrf2. HDAC6 was unchanged and served as loading control. c The same samples were analyzed for changes in heme oxygenase-1 (HO1), a known Nrf2 target gene. de Confirmation of Keap1 and HDAC3 knockdown. Kelch-like ECH-associated protein 1 (Keap1)
Fig. 8
Fig. 8
Human subjects given a broccoli sprout extract, or consuming high levels of cruciferous vegetables, have altered HDAC3 and p16 expression. a Human subjects (n = 5, each arm of the study) consumed a broccoli sprout extract supplement (BSE, 200 μmol SFN equivalent, IND #111736) or a placebo for 7 days, and blood was drawn at the times indicated. b Total levels of SFN and its metabolites in plasma (black symbols), and b, c HDAC3 and p16 protein expression changes in circulating PBMCs. Results are from a single volunteer and are representative of other subjects who took the BSE supplement. No such changes occurred after taking the placebo (data not shown). Individual SFN metabolites, noted in Additional file 3: Table S2, were determined as reported [27]. d Study design in screening colonoscopy patients. e Relative mRNA expression of p16 in subjects reporting low (0–1 servings/week) vs. high (>5 servings/week) cruciferous vegetable intake. f Immunoblotting of colon biopsies, arranged left to right according to cruciferous vegetable intake. gi Associations between p16, HDAC3, AcH4K12, and Nrf2 normalized to β-actin (relative densitometry of proteins in panel f)
Fig. 9
Fig. 9
Nrf2 and p16 are inversely associated in human colon tumors and normal colon. The Cancer Genome Atlas (TCGA, https://tcga-data.nci.nih.gov/tcga/) was mined for NFE2L2 (Nrf2) and CDKN2A (p16) mRNA expression data in colon tumors (black) and normal colon (red)

References

    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. doi: 10.1186/1868-7083-5-12.
    1. Mack GS. Epigenetic cancer therapy makes headway. J Natl Cancer Inst. 2006;98:1443–4. doi: 10.1093/jnci/djj447.
    1. 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. doi: 10.1074/jbc.M510023200.
    1. Marks PA, Xu W-S. Histone deacetylase inhibitors: potential in cancer therapy. J Cell Biochem. 2009;107:600–8. doi: 10.1002/jcb.22185.
    1. Delcuve GP, Khan DH, Davie JR. Roles of histone deacetylases in epigenetic regulation: emerging paradigms from studies with inhibitors. Clin Epigenetics. 2012;4:5. doi: 10.1186/1868-7083-4-5.
    1. Dashwood RH, Myzak MC, Ho E. Dietary HDAC inhibitors: time to rethink weak ligands in cancer chemoprevention? Carcinogenesis. 2006;27:344–9. doi: 10.1093/carcin/bgi253.
    1. 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. doi: 10.1186/1868-7083-3-4.
    1. 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. doi: 10.3109/10409238.2011.557713.
    1. Myzak MC, Dashwood RH. Chemoprotection by sulforaphane: keep one eye beyond Keap1. Cancer Lett. 2006;233:208–18. doi: 10.1016/j.canlet.2005.02.033.
    1. Myzak MC, Dashwood WM, Orner GA, Ho E, Dashwood RH. Sulforaphane inhibits histone deacetylase in vivo and suppresses tumorigenesis in Apcmin mice. FASEB J. 2006;20:506–8.
    1. 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. doi: 10.1158/0008-5472.CAN-04-1326.
    1. 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.
    1. 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. doi: 10.1073/pnas.94.19.10367.
    1. 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. doi: 10.1073/pnas.89.6.2399.
    1. 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. doi: 10.1007/128_2012_339.
    1. 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. doi: 10.1158/0008-5472.CAN-12-4499.
    1. Kensler TW, Wakabayashi N. Nrf2: friend or foe for chemoprevention? Carcinogenesis. 2010;31:90–9. doi: 10.1093/carcin/bgp231.
    1. 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. doi: 10.1016/j.ejca.2005.06.006.
    1. 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.
    1. 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. doi: 10.1186/1476-4598-10-68.
    1. 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. doi: 10.4161/epi.24710.
    1. 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. doi: 10.1007/s11095-011-0500-z.
    1. 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. doi: 10.1111/j.1742-4658.2009.07128.x.
    1. 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.
    1. 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. doi: 10.4161/epi.7.2.18972.
    1. 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. doi: 10.1038/nm726.
    1. 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. doi: 10.1002/mnfr.201400674.
    1. 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. doi: 10.1080/01635580701267875.
    1. 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. doi: 10.1093/carcin/21.12.2287.
    1. 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. doi: 10.1136/gut.2007.134650.
    1. 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. doi: 10.1093/carcin/bgl049.
    1. 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. doi: 10.1002/mc.21950.
    1. 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. doi: 10.1158/0008-5472.CAN-06-3562.
    1. 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. doi: 10.1158/1940-6207.CAPR-08-0028.
    1. 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. doi: 10.1002/ijc.22943.
    1. Sporn MB, Liby KT. NRF2 and cancer: the good, the bad, and the importance of context. Nat Rev Cancer. 2012;12:564–71. doi: 10.1038/nrc3278.
    1. 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. doi: 10.1002/1098-2744(200007)28:3<139::AID-MC2>;2-V.
    1. 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. doi: 10.1038/onc.2010.614.
    1. 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.
    1. 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. doi: 10.1111/j.1349-7006.2009.01387.x.
    1. 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. doi: 10.4161/cbt.6.2.3578.
    1. Chien WW, Ffrench M. Regulation of p16INK4a, senescence and oncogenesis. Med Sci. 2006;22:865–71.
    1. 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. doi: 10.1038/35059131.
    1. 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. doi: 10.1093/nar/gkr818.
    1. 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.
    1. 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. doi: 10.1093/nar/gks1048.
    1. Tuma RS. Epigenetic therapies move into new territory, but how exactly do they work? J Natl Cancer Inst. 2009;101(19):1300–1. doi: 10.1093/jnci/djp342.
    1. 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. doi: 10.1002/ijc.25610.
    1. 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. doi: 10.1093/carcin/bgn165.
    1. 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. doi: 10.1186/1868-7083-4-16.

Source: PubMed

3
Sottoscrivi