Curcumin has been shown to prevent and inhibit carcinogen-induced tumorigenesis in different organs of rodent carcinogenesis models. Our objective is to study global gene expression profiles elicited by curcumin in mouse liver and small intestine as well as to identify curcumin-regulated nuclear factor E2-related factor 2 (Nrf2)–dependent genes. Wild-type C57BL/6J and Nrf2 knockout C57BL/6J/Nrf2(−/−) mice were given a single oral dose of curcumin at 1,000 mg/kg. Liver and small intestine were collected at 3 and 12 hours after treatments. Total RNA was extracted and analyzed using Affymetrix (Santa Clara, CA) mouse genome 430 array (45K) and GeneSpring 6.1 software (Silicon Genetics, Redwood City, CA). Genes that were induced or suppressed >2-fold by curcumin treatments compared with vehicle in wild-type mice but not in knockout mice were filtered using GeneSpring software and regarded as Nrf2-dependent genes. Among those well-defined genes, 822 (664 induced and 158 suppressed) and 222 (154 induced and 68 suppressed) were curcumin-regulated Nrf2-dependent genes identified in the liver and small intestine, respectively. Based on their biological functions, these genes can be classified into the category of ubiquitination and proteolysis, electron transport, detoxification, transport, apoptosis and cell cycle control, cell adhesion, kinase and phosphatase, and transcription factor. Many phase II detoxification/antioxidant enzyme genes, which are regulated by Nrf2, are among the identified genes. The identification of curcumin-regulated Nrf2-dependent genes not only provides potential novel insights into the biological effects of curcumin on global gene expression and chemoprevention but also points to the potential role of Nrf2 in these processes. [Mol Cancer Ther 2006;5(1):39–51]

Cancer development is believed to be a multistage process, including initiation, promotion, and progression (1, 2). In 1976, Dr. Michael B. Sporn first coined the term “chemoprevention” and advocated using cancer chemopreventive agents to decrease the incidence of cancer (3). Since then, many natural products isolated from food and plants have been investigated for their potential as cancer chemopreventive agent. Curcumin, a naturally occurring flavonoid present in the spice turmeric, has been shown to prevent and inhibit carcinogen-induced tumorigenesis in different organs in rodent carcinogenesis models, and its cancer chemopreventive effects in these animal models have been reviewed previously (4, 5). In addition to its cancer chemopreventive activity, curcumin is also well known for its antioxidant and anti-inflammatory properties (6, 7). Therefore, numerous studies have been carried out to elucidate the molecular mechanisms of the above effects of curcumin. Based on these studies, the potential mechanisms or molecular targets of curcumin have been extensively reviewed recently (4, 5, 8, 9). These include the regulation of a variety of signal transduction pathways [such as epidermal growth factor receptor, nuclear factor-κB, activator protein-1, β-catenin/TCF, mitogen-activated protein kinase (MAPK), and Akt pathways] as well as the expression of many oncogenes (such as c-jun, c-fos, c-myc, cyclooxygenase-2, and NOS) that are involved in the cell proliferation, differentiation, apoptosis, and angiogenesis. However, the chemopreventive mechanism of curcumin, especially in vivo, is still not fully elucidated because the interactions between these different signal transduction pathways in response to curcumin treatment are not fully understood.

Basic leucine zipper family transcription factor nuclear factor E2-related factor 2 (Nrf2) involves the regulation of antioxidant response element (ARE)–mediated gene transcription. Under homeostasis condition, Nrf2 is sequestered in cytoplasm by Kelch-like ECH-associated protein 1 (10). Exposure of cells to oxidative stress or ARE inducers triggers the release of Nrf2 from Kelch-like ECH-associated protein 1 and facilitate its nuclear translocation (11). The nuclear translocation of Nrf2 and subsequent dimerization with small Maf protein and other coactivators, such as CBP, will drive the transcription of its target genes (12). One large group of these target genes is the phase II detoxification and antioxidant genes. By inducing these genes through the Nrf2/ARE pathway, chemopreventive agents could increase the detoxification of procarcinogens or carcinogens and protect normal cells from the DNA/protein damage caused by electrophiles and reactive oxygen intermediates, thus decreasing the incidence of tumor initiation and reducing the risk of cancer. The role of Nrf2 in preventing tumorigenesis is also supported by studies in which Nrf2 knockout mice were much more susceptible to carcinogen-induced carcinogenesis and failed to respond to certain cancer chemopreventive agents, which were effective in Nrf2 wild-type mice (1315). Therefore, Nrf2 has been considered as a molecular target of cancer chemoprevention (16). Previous studies have shown that chemopreventive agent sulforaphane and 3H-1,2-dithiole-3-thione could regulate a variety of genes, including phase II genes, in a Nrf2-dependent manner (17, 18). Curcumin has also been shown to be able to induce many phase II genes as well as ARE reporter gene activities (19). Therefore, studies investigating the role of Nrf2 in curcumin-regulated gene expression may help to identify new molecular mechanisms of the cancer protective effects of curcumin. Furthermore, it will also address other possible roles of Nrf2 in cancer chemoprevention in addition to the regulation of phase II detoxification enzyme and antioxidant enzyme genes.

Gene expression profiling using genome-based Affymetrix (Santa Clara, CA) microarray is an unbiased method to identify novel molecular targets of curcumin in vivo. In the current study, the global gene expression profiles elicited by oral administration of curcumin in wild-type and Nrf2-knockout C57BL/6J mice were compared by microarray analysis. The identification of curcumin-regulated Nrf2-dependent genes will yield valuable insights into the role of Nrf2 in the curcumin-mediated gene regulation and its cancer chemopreventive effects. The current study is also the first to investigate the global gene expression profiles elicited by curcumin in an in vivo mouse model where the role of Nrf2 is also examined.

Animal and Treatment

Nrf2 knockout mice Nrf2(−/−) (C57BL/SV129) were described previously (20). Nrf2(−/−) mice were back-crossed with C57BL/6J mice (The Jackson Laboratory, Bar Harbor, ME). Mice were genotyped for Nrf2 status by PCR amplification of genomic DNA extracted from tail. PCR amplification was carried out by using primers (3′-primer, 5′-GGAATGGAAAATAGCTCCTGCC-3′; 5′-primer, 5′-GCCTGAGAGCTGTAGGCCC-3′; and lacZ primer, 5′-GGGTTTTCCCAGTCACGAC-3′). Male C57BL/6J/Nrf2(−/−) mice from third generation of back-crossing were used in this study. Age-matched male C57BL/6J mice were purchased from The Jackson Laboratory. Mice 9 to 12 weeks old were used and housed at Rutgers Animal Facility. Mice were fed AIN-76A diet (Research Diets, Inc., New Brunswick, NJ) with free access to water ad libitum under 12-hour light/dark cycles. After 1 week of acclimatization, mice were treated with curcumin (Sigma, St. Louis, MO) at a dose of 1,000 mg/kg (dissolved in 50% polyethylene glycol 400 solution at concentration of 100 mg/mL) by oral gavages. The control groups were given vehicle only (50% polyethylene glycol 400 solution). Each treatment was administrated to a group of four animals for both C57BL/6J and C57BL/6J/Nrf2(−/−) mice. Mice were sacrificed 3 and 12 hours after curcumin treatment or 3 hours after vehicle treatment (control group; Fig. 1). Livers and small intestines were removed and stored in RNA Later (Ambion, Austin, TX) solution immediately.

Figure 1.

Schematic diagram of experimental design.

Figure 1.

Schematic diagram of experimental design.

Close modal

RNA Extraction, Microarray Hybridization, and Data Analysis

Total RNA from liver and small intestine were isolated by using a method of Trizol (Invitrogen, Carlsbad, CA) extraction coupled with the RNeasy Midi kit from Qiagen (Valencia, CA) according to the manufacturer's protocol. After RNA isolation, all the subsequent technical procedures, including quality control, concentration measurement of RNA, cDNA synthesis, and biotin labeling of cRNA, hybridization, and scanning of the arrays, were done at CINJ Core Expression Array Facility of Robert Wood Johnson Medical School (New Brunswick, NJ). Affymetrix mouse genome 430 2.0 array containing >45,101 probe sets was used to probe the global gene expression profile in mice following curcumin treatment. Each array was hybridized with cRNA derived from a pooled total RNA sample from four mice per treatment group, per time point, per organ, and per genotype (total 12 chips were used in this study; Fig. 1). After hybridization and washing, the intensity of the fluorescence of the array chips were measured by the Affymetrix GeneChip Scanner. The expression analysis file created from each sample (chip) scanning was imported into GeneSpring 6.1 software (Silicon Genetics, Redwood City, CA) for further data characterization. A new experiment was generated after importing data from the same organ in which data were normalized to the 50th percentile of all measurements on that array. Data filtration based on flags present in at least one of the samples was generated. Lists of genes that were either induced or suppressed >2-fold between treated and vehicle group of same genotype were created by filtration-on-fold function within the presented flag list. By using color-by-Venn-diagram function, lists of genes that were regulated >2-fold only in C57BL/6J mice in both liver and small intestine were created.

Quantitative Real-time PCR for Microarray Data Validation

To verify the microarray data, several genes (including the housekeeping gene glyceraldehyde-3-phosphate dehydrogenase) from different categories were chosen for quantitative real-time PCR analyses. The specific primers for these genes were listed in Table 1. Instead of using pooled RNA from each group, RNA samples isolated from individual mice as described above were used in real-time PCR analyses. First-strand cDNA was synthesized using 4 μg total RNA following the protocol of SuperScript III First-Strand cDNA Synthesis System (Invitrogen). Real-time PCR was done as described previously (21). The gene expression was determined by normalization with control gene glyceraldehyde-3-phosphate dehydrogenase. The correlation between corresponding microarray data and real-time PCR data was validated by Spearman rank correlation method.

Table 1.

Oligonucleotide primers used in quantitative real-time PCR

Gene nameGenbankForward primer (5′-3′)Reverse primer (5′-3′)
Rho-associated coiled-coil forming kinase 2 (ROCK2BB761686 TTCTGTGACCTTCAGATGGCC TTCCCAACCAGAGCACAGCT 
PKC, μ (PKCmNM_008858 AGCCCTTCAACGAGCAACAA ACCATCCACCCTTCCTTCATC 
Inhibitor of κB kinase γ (IKKgNM_010547 CTGAAAGTTGGCTGCCATGAG GAGTGGTGAGCTGGAGCAGG 
GST, μ (GSTmNM_010358 GAAGCCAGTGGCTGAATGAGA GATGGCATTGCTCTGGGTG 
ATPase, Cu2+transporting, α-polypeptide (ATP7AU03434 TTGTGGCGGCTGGTACTTCT CAAATGCGATGGTGGTTGC 
Heme oxygenase 1 (HO-1NM_010442 CCCACCAAGTTCAAACAGCTC AGGAAGGCGGTCTTAGCCTC 
Glyceraldehyde-3-phosphate dehydrogenase (GAPDHNM_008084 CACCAACTGCTTAGCCCCC TCTTCTGGGTGGCAGTGATG 
Gene nameGenbankForward primer (5′-3′)Reverse primer (5′-3′)
Rho-associated coiled-coil forming kinase 2 (ROCK2BB761686 TTCTGTGACCTTCAGATGGCC TTCCCAACCAGAGCACAGCT 
PKC, μ (PKCmNM_008858 AGCCCTTCAACGAGCAACAA ACCATCCACCCTTCCTTCATC 
Inhibitor of κB kinase γ (IKKgNM_010547 CTGAAAGTTGGCTGCCATGAG GAGTGGTGAGCTGGAGCAGG 
GST, μ (GSTmNM_010358 GAAGCCAGTGGCTGAATGAGA GATGGCATTGCTCTGGGTG 
ATPase, Cu2+transporting, α-polypeptide (ATP7AU03434 TTGTGGCGGCTGGTACTTCT CAAATGCGATGGTGGTTGC 
Heme oxygenase 1 (HO-1NM_010442 CCCACCAAGTTCAAACAGCTC AGGAAGGCGGTCTTAGCCTC 
Glyceraldehyde-3-phosphate dehydrogenase (GAPDHNM_008084 CACCAACTGCTTAGCCCCC TCTTCTGGGTGGCAGTGATG 

Curcumin-Altered Gene Expression Pattern in Mouse Liver and Small Intestine

Genes that were only regulated by curcumin in C57BL/6J mice but not in C57BL/6J/Nrf2(−/−) mice were regarded as curcumin-regulated Nrf2-dependent genes. Among these Nrf2-dependent genes, expression levels of 822 well-defined genes were either induced (664) or suppressed (158) >2-fold by curcumin only in wild-type mice liver at both time points (Fig. 2). Similar changes in gene expression profiles were also observed in the small intestine array data analysis. Compared with the results from liver sample arrays, an even smaller percentage of total probes on the array were either induced or suppressed >2-fold by curcumin regardless of Nrf2 status at both time points. Further analyses showed that 222 well-defined genes were regulated >2-fold (154 up-regulated and 68 down-regulated) in a Nrf2-dependent manner at both time points by curcumin (Fig. 2).

Figure 2.

Regulation of Nrf2-dependent gene expression by curcumin in mice liver and small intestine. Gene expression patterns in liver and small intestine were analyzed at 3 and 12 h after a single oral dose of curcumin at 1,000 mg/kg; Nrf2-dependent genes with well known functions and regulated >2-fold at both time points were selected. The positive numbers on the Y axis refer to the number of genes being induced; the negative numbers on the Y axis refer to the number of genes being suppressed.

Figure 2.

Regulation of Nrf2-dependent gene expression by curcumin in mice liver and small intestine. Gene expression patterns in liver and small intestine were analyzed at 3 and 12 h after a single oral dose of curcumin at 1,000 mg/kg; Nrf2-dependent genes with well known functions and regulated >2-fold at both time points were selected. The positive numbers on the Y axis refer to the number of genes being induced; the negative numbers on the Y axis refer to the number of genes being suppressed.

Close modal

Curcumin-Induced Nrf2-Dependent Genes in Liver and Small Intestine

Genes that were induced only in wild-type mice but not in Nrf2(−/−) mice by curcumin were considered curcumin-induced Nrf2-dependent genes. Based on their biological functions, these genes can be classified into categories, including heat shock protein, ubiquitination and proteolysis, electron transport, detoxification enzyme, transport, cell cycle control and apoptosis, cell adhesion, kinase and phosphatase, transcription, G protein-coupled receptor, and nuclear receptor (Table 2). Among these genes, a group of curcumin-induced Nrf2-dependent phase II detoxification and antioxidant genes was identified in both liver and small intestine microarray analysis. These include quinone reductase, catalytic subunit of glutamate-cysteine ligase (γ-GCS), and thioredoxin reductase 1 genes in liver and different isoforms of glutathione S-transferase (GST), heme oxygenase 1 (HO-1), and UDP-glucuronosyltransferase 2b5 genes in small intestine.

Table 2.

Curcumin-induced Nrf2-dependent gene lists in liver and small intestine

Gene descriptionSymbolGenbankLiver*
Small intestine
3 h12 h3 h12 h
Heat shock protein       
    Heat shock protein 2 Hspb2 AK012780 3.39 4.71   
    Heat shock protein 1A Hspa1a M12573 8.28 6.24 5.88 2.22 
    Heat shock protein 1A Hspa1a M12573 6.15 4.30 5.48 2.12 
    Crystallin, αB Cryab NM_009964 6.02 6.68   
Ubiquitination and proteolysis       
    A disintegrin and metalloproteinase domain 19 (meltrin β) Adam19 NM_009616 2.67 2.68   
    Cathepsin M Cstm NM_022326 3.68 2.14   
    Dipeptidylpeptidase 9 Dpp9 BB667346 2.15 4.83   
    Leishmanolysin-like (metallopeptidase M8 family) Lmln BB182358 5.17 5.70   
    Mucosa-associated lymphoid tissue lymphoma translocation gene 1 Malt1 BM239348 2.41 2.62   
    Procollagen C-proteinase enhancer protein Pcolce NM_008788 8.12 10.07   
    Proprotein convertase subtilisin/kexin type 5 Pcsk5 BC013068 11.83 18.73   
    Proteaseome (prosome, macropain) 28S subunit, 3 Psme3 U60330 2.30 2.88   
    Proteasome (prosome, macropain) 26S subunit, ATPase, 6 Psmc6 AW208944 2.31 3.02   
    Proteasome (prosome, macropain) 26S subunit, non-ATPase, 11 Psmd11 AA050796 2.67 3.41   
    Proteasome (prosome, macropain) 26S subunit, ATPase, 4 Psmc4 NM_011874 2.52 2.60   
    Proteasome (prosome, macropain) 26S subunit, non-ATPase, 9 Psmd9 BG092381 2.39 2.75   
    Ring finger protein 11 Rnf11 BI150320 2.12 2.24   
    Ubiquitin fusion degradation 1 like  BB500664 3.28 2.72   
    Ubiquitin-specific protease 30 Usp30 BG067690 3.37 3.93   
    Ubiquitin-specific protease 38 Usp38 BG064874 2.43 2.70   
    Ubiquitin-conjugating enzyme E2B  AK011961 2.31 2.13   
    Coagulation factor IX F9 M23109   5.06 8.86 
    Proteasome (prosome, macropain) activator, subunit 4  BB200981   8.21 3.24 
    Tolloid-like Tll1 NM_009390   7.80 3.61 
    Transferrin receptor 2 Trfr2 AV027486   3.86 4.20 
Electron transport       
    Aldehyde oxidase 1 Aox1 NM_009676 2.01 2.47   
    Cytochrome c oxidase, subunit VIIa 2 Cox7a2 BB745549 4.32 4.82   
    Cytochrome c oxidase, subunit VIIIb Cox8b NM_007751 3.14 2.38   
    Similar to cytochrome P450, 4a10 Cyp4a10 BC025936 2.02 2.42   
    Thioredoxin reductase 1 Txnrd1 NM_015762 2.12 2.16   
    Ubiquinol-cytochrome c reductase core protein 1  BG864756 2.08 2.57   
    Cytochrome P450, family 2, subfamily c, polypeptide 55 Cyp2c55 NM_028089   2.89 4.95 
Detoxification enzyme       
    Crystallin, ζ (quinone reductase) like 1 Cryzl1 AK010433 2.02 2.26   
    Fucosyltransferase 8 Fut8 NM_016893 10.78 10.62   
    Glutamate-cysteine ligase, catalytic subunit Gclc AW825835 2.41 4.26   
    Methyltransferase-like 1 Mettl1 AI838750 4.28 5.76   
    Sialyltransferase 10 Siat10 NM_018784 15.70 17.79   
    Steroid sulfatase Sts NM_009293 2.49 3.33   
    Thioredoxin interacting protein Txnip AF173681 2.94 2.34   
    Carbonyl reductase 3 Cbr3 AK003232   21.68 13.22 
    GST, μ1 Gstm1 NM_010358   4.42 11.65 
    GST, μ1 Gstm1 J03952   2.84 7.90 
    GST, μ3 Gstm3 J03953   3.72 5.91 
    GST, α2 (Yc2) Gsta2 NM_008182   2.79 9.58 
    GST, α3 Gsta3 AI172943   4.49 2.62 
    GST, α4 Gsta4 NM_010357   3.76 2.49 
    HO-1 (decycling) Hmox1 NM_010442   76.06 4.54 
    UDP-glucuronosyltransferase 2 family, member 5 Ugt2b5 NM_009467   2.07 6.70 
Transport       
    Aquaporin 7 Aqp7 AB056091 2.38 3.30   
    ATP synthase, H+ transporting, mitochondrial F0 complex, subunit c (subunit 9), isoform 2 Atp5g2 AW413339 4.03 5.12   
    ATPase, Ca2+ transporting, cardiac muscle, slow twitch 2 Atp2a2 AA245637 2.12 3.10   
    ATPase, class II, type 9A Atp9a AF011336 2.14 2.60   
    ATPase, Cu2+ transporting, α-polypeptide Atp7a U03434 4.06 3.10   
    ATP-binding cassette, subfamily B (MDR/TAP), member 1A Abcb1a M30697 2.23 2.05   
    ATP-binding cassette, subfamily B (MDR/TAP), member 1B Abcb1b NM_011075 7.06 5.18   
    ATP-binding cassette, subfamily D (ALD), member 3 Abcd3 BB042134 2.10 2.37   
    Cation channel, sperm associated 2 Catsper2 BB484902 2.18 2.15   
    Chloride channel 3 Clcn3 BB328803 12.31 15.49   
    Chloride channel calcium activated 1 Clca1 AF047838 7.22 3.48   
    Fatty acid–binding protein 4, adipocyte Fabp4 BC002148 5.62 3.24   
    FXYD domain-containing ion transport regulator 2 Fxyd2 NM_052823 3.62 11.19 4.50 6.00 
    Kinesin family member 5B Kif5b BI328541 2.27 2.41   
    Membrane targeting (tandem) C2 domain containing 1 Mtac2d1 AB062282 8.52 7.72   
    Myosin IC Myo1c NM_008659 3.28 2.79   
    N-ethylmaleimide-sensitive fusion protein Nsf BB400581 2.20 2.95   
    Potassium voltage-gated channel, Shab-related subfamily, member 1 Kcnb1 BB324482 6.09 3.17   
    Potassium voltage-gated channel, subfamily Q, member 2 Kcnq2 AB000502 11.48 2.10   
    Solute carrier family 12, member 2 Slc12a2 BG069505 2.27 2.71   
    Solute carrier family 13 (sodium-dependent dicarboxylate transporter), member 2 Slc13a2 BC013493 5.37 4.68   
    Solute carrier family 16 (monocarboxylic acid transporters), member 1 Slc16a1 NM_009196 2.25 8.09   
    Solute carrier family 18 (vesicular monoamine), member 2 Pdzk8 BB102308 3.38 3.12   
    Solute carrier family 2 (facilitated glucose transporter), member 9 BE197100 2.73 2.58    
    Solute carrier family 22 (organic cation transporter), member 3 Slc22a3 NM_011395 3.51 5.57   
    Solute carrier family 22 (organic cation transporter), member 5 Slc22a5 NM_011396 2.18 3.76   
    Solute carrier family 25 (mitochondrial carrier, palmitoylcarnitine transporter), member 29 Slc25a29 BC006711 2.06 2.20   
    Solute carrier family 37 (glycerol-3-phosphate transporter), member 3 Slc37a3 BC005744 2.09 2.49   
    Solute carrier family 39 (zinc transporter), member 14 Slc39a14 BB022806 3.78 2.11   
    Solute carrier family 6 (neurotransmitter transporter), member 14 Slc6a14 AF320226 23.50 19.96   
    Solute carrier family 9 (sodium/hydrogen exchanger), member 8 Slc9a8 AK018301 2.09 4.70   
    Src activating and signaling molecule Srcasm BC004710 2.08 2.04   
    Syntaxin 6 Stx6 BB492711 2.78 3.83   
    Transporter 2, ATP-binding cassette, subfamily B (MDR/TAP) Tap2 BE691515 2.90 2.12   
    ATPase, class V, type 10A Atp10a BM249532   5.13 3.11 
    Chemokine (C-C motif) ligand 7 Ccl7 AF128193   8.15 12.09 
    Hemopexin Hpxn BC011246   7.84 32.11 
    5-Hydroxytryptamine (serotonin) receptor 3A Htr3a NM_013561   2.74 2.31 
    Major urinary protein 3 Mup3 M27608   13.25 102.59 
    Solute carrier family 17 (sodium phosphate), member 1 Slc17a1 NM_009198   2.01 3.00 
    Solute carrier family 34 (sodium phosphate), member 2 Slc34a2 NM_011402   2.13 5.54 
    Solute carrier family 35 (UDP galactose transporter), member 2 AU080926   2.14 2.61  
    Solute carrier family 40 (iron-regulated transporter), member 1 Slc40a1 AF226613   2.22 2.43 
    Solute carrier family 6 (neurotransmitter transporter), member 14 Slc6a14 AF320226   3.50 3.29 
    Zinc finger protein 316 Zfp316 AV367169   2.34 2.00 
    Solute carrier family 4, sodium bicarbonate transporter-like, member 11 Slc4a11 BB498904   3.12 2.49 
    Sodium channel, voltage-gated, type IX, α-polypeptide  BB452274   2.72 4.16 
Apoptosis and cell cycle control       
    Apoptotic protease-activating factor 1 Apaf1 AK018076 16.54 14.58   
    Baculoviral IAP repeat-containing 1a Birc1a AF135491 3.02 4.02   
    Bcl-2-associated transcription factor 1 Bclaf1 BI965039 2.23 2.62   
    Bcl-2-like Bcl2l1 NM_009743 2.98 3.08   
    Cyclin-dependent kinase inhibitor 1A (p21) Cdkn1a AK007630 2.02 10.06   
    Cytotoxic granule-associated RNA-binding protein 1 Tia1 BG518542 4.18 4.22   
    Ring finger protein 7 Rnf7 AV047821 2.62 3.74   
    Tumor necrosis factor receptor-associated factor 3 Traf3 U21050 5.78 4.25   
    Transformation-related protein 53-inducible nuclear protein 1 Trp53inp1 AW495711 2.26 2.41   
    Tripartite motif-containing 35  BQ175280 3.80 3.27   
    Tumor differentially expressed 1 Tde1 NM_012032 3.15 4.29   
    CWF19-like 2, cell cycle control (Schizosaccharomyces pombeCwf19l2 AK014327 3.18 3.97   
    Bcl-2-interacting killer-like Biklk NM_007546   3.41 9.60 
    RAS like, estrogen regulated, growth inhibitor Rerg BC026463   3.81 2.60 
Cell adhesion       
    Cadherin 11 Cdh11 NM_009866 3.54 3.35   
    Cadherin 22 Cdh22 AB019618 2.00 2.54 2.37 2.77 
    Cadherin 4 Cdh4 NM_009867 15.48 10.96   
    Catenin α-like 1 Catnal1 BQ031240 4.49 4.74   
    Catenin src Catns NM_007615 2.15 3.08   
    Contactin-associated protein 1 Cntnap1 NM_016782 4.56 4.75   
    Integrin α8 Itga8 BB623587 6.71 6.44   
    Laminin, β3 Lamb3 NM_008484 2.41 5.53   
    Neurotrimin  AF282980 0.49 0.43   
    Osteomodulin Omd NM_012050 3.11 3.01   
    Protocadherin 18 Pcdh18 BM218630 2.83 3.78   
    Fibronectin leucine-rich transmembrane protein 2 Flrt2 BB817332   11.38 5.14 
    Procollagen, type IX, α1 Col9a1 AK004383   3.33 2.32 
    Thrombospondin 2 Thbs2 BB233297   2.06 2.51 
Kinase and phosphatase       
    Casein kinase II, α1 polypeptide Csnk2a1 BB283759 3.85 2.07   
    Induced in fatty liver dystrophy 2  BB508622 3.68 5.71   
    Keratin complex 2, basic, gene 8 Krt2-8 AW322280 2.49 2.56   
    MAPK-activated protein kinase 2 Mapkapk2 BG918951 2.07 2.84   
    Microtubule-associated serine/threonine kinase 2 Mast2 BB367890 5.34 12.50   
    MAPK kinase kinase 12 Map3k12 NM_009582 3.57 2.70   
    MAPK 8 interacting protein 3 Mapkip8 AF178636 3.41 4.28   
    MAPK kinase kinase kinase 4 Map4k4 NM_008696 3.71 4.24   
    MAPK kinase kinase kinase 5 Map4k5 BG067961 8.29 7.17   
    PCTAIRE-motif protein kinase 1 Pctk1 AW539955 6.66 5.81   
    PKC, α  BB355213 4.49 4.04   
    Protein kinase, cAMP-dependent regulatory, type IIβ Prkar2b BB216074 26.47 4.75   
    Proviral integration site 2  NM_138606 2.00 2.45   
    Regulator of G protein signaling 19 Rgs19 BC003838 2.09 2.49   
    Rho-associated coiled-coil forming kinase 1 Rock1 BI662863 2.15 3.07   
    Rho-associated coiled-coil forming kinase 2 Rock2 BB761686 3.07 2.54   
    Ribosomal protein S6 kinase, polypeptide 5 Rps6ka5 BQ174267 2.65 2.39   
    Serine/threonine kinase 17b (apoptosis inducing) Stk17b AV173139 3.05 2.53   
    Serum/glucocorticoid-regulated kinase 3 Sgk3 BB768208 6.68 6.67   
    SNF1-like kinase Snf1lk AI648260 7.55 9.82   
    Testis-specific protein kinase 1 Tesk1 NM_011571 2.05 2.28   
    Tousled-like kinase 2 (ArabidopsisTlk2 NM_011903 2.48 2.81   
    Dual specificity phosphatase 6 Dusp6 NM_026268 2.45 2.18   
    Eyes absent 3 homologue (DrosophilaEya3 BB428881 2.94 3.12   
    Paladin  NM_013753 2.14 2.53   
    Protein tyrosine phosphatase, nonreceptor type 21 Ptpn21 AW987375 7.44 13.85   
    PKC, μ Prkcm NM_008858   2.22 2.04 
    Keratin complex 2, basic, gene 8 Krt2-18 NM_016879   2.85 2.29 
    MAPK kinase kinase 10 Map3k10 AA789425   10.02 7.37 
    Dual specificity phosphatase 4 Dusp4 AK012530   2.65 2.00 
G protein-coupled receptors       
    G protein-coupled receptor 65 Gpr65 NM_008152 6.34 5.32   
    Endothelial differentiation, sphingolipid G protein-coupled receptor, 8 Edg8 NM_053190 4.92 3.92   
    Endothelin receptor type A Ednra BC008277 9.12 10.42   
    Endothelin receptor type A Ednra AW558570 7.84 7.60   
Transcription factors       
    Ankyrin repeat domain 1 (cardiac muscle) Ankrd1 AK009959 2.12 2.88   
    Basic transcription element-binding protein 1 Bteb1 NM_010638 2.90 3.54   
    cAMP-responsive element modulator Crem AU258667 9.73 6.29   
    cAMP-responsive element modulator Crem AI467599 2.58 2.56   
    cAMP-responsive element–binding protein–binding protein  BG076163 5.62 10.55   
    cAMP-responsive element–binding protein–binding protein/EP300 inhibitory protein 1 Cri1 BC010712 2.05 2.19   
    E4F transcription factor 1 E4f1 NM_007893 3.69 3.50   
    E74-like factor 1 Elf1 NM_007920 2.06 2.22   
    Early growth response 1 Egr1 NM_007913 4.19 5.06   
    Ewing sarcoma homologue Ewsr1 AW610680 10.50 5.63   
    Forkhead box N2 Foxn2 AV295543 3.34 2.75   
    Forkhead box P1 Foxp1 BG962849 2.71 3.65   
    Heterogeneous nuclear ribonucleoprotein A/B Hnrpab AK013709 6.55 7.28   
    Heterogeneous nuclear ribonucleoprotein R Hnrpr BB251000 2.50 2.10   
    Homeobox C8 Hoxc8 BB283726 4.83 2.57   
    Homeodomain leucine zipper-encoding gene  AV298304 7.72 8.52   
    Inhibitor of κB kinase γ Ikbkg BB147462 2.73 3.55   
    Inhibitor of κB kinase γ Ikbkg NM_010547 2.12 2.83   
    Kruppel-like factor 7 (ubiquitous) Klf7 BB524597 2.09 2.97   
    LIM homeobox protein 9 Lhx9 AK013209 3.23 5.88   
    Longevity assurance homologue 4 (Saccharomyces cerevisiaeLass4 BB006809 3.18 9.26   
    Nuclear factor, interleukin 3, regulated Nfil3 AY061760 2.13 3.52   
    Nuclear receptor subfamily 2, group C, member 2 Nr2c2 AU066920 2.22 2.60   
    Retinoid X receptor γ Rxrg NM_009107 2.63 2.94   
    SCAN-KRAB-zinc finger gene 1 Zpf306 BC007473 2.21 3.91   
    Suppressor of K+ transport defect 3 Skd3 NM_009191 2.80 2.76   
    TAF5 RNA polymerase II, TATA box-binding protein–associated factor Taf5 AV117817 2.96 3.90   
    TAR DNA-binding protein Tardbp BC012873 2.63 3.96   
    Transforming growth factor-β-inducible early growth response 1 Tieg1 NM_013692 2.76 3.52   
    Transcription factor 12 Tcf12 BB540782 2.79 2.86   
    Transcription factor 20 Tcf20 AW552808 2.22 2.14   
    Transcription factor 3 Tcf3 NM_009332 4.44 5.08   
    Zinc finger proliferation 1 Zipro1 AI326272 4.02 7.65   
    Zinc finger protein 148 Zfp148 X98096 2.11 2.29   
    Zinc finger protein 207 Zfp207 AV338324 2.16 2.11   
    Zinc finger protein 263 Zfp263 AI326880 3.02 2.96   
    Zinc finger protein 319 Zfp319 BB476317 5.45 5.41   
    Zinc finger protein 354C Zfp354c NM_013922 3.33 3.73   
    Zinc fingers and homeoboxes 3 Zhx3 BE952825 6.37 7.45   
    Ankyrin repeat domain 1 (cardiac muscle) Ankrd1 AK009959   3.82 16.77 
    CBFA2T1 identified gene homologue (human) Cbfa2t1h BG072085   19.43 12.24 
    E4F transcription factor 1  BB027397   7.90 9.35 
    Myeloid ecotropic viral integration site 1 Meis1 AW547821   4.31 3.67 
    POU domain, class 2, transcription factor 2 Pou2f2 X57938   12.27 12.47 
    Runt-related transcription factor 1 Runx1 NM_009821   2.24 2.64 
    Transforming growth factor-β1-induced transcript 4 Tgfbli4 AW413169   9.17 4.53 
    Zinc finger protein 2 Zfp2 NM_009550   6.84 3.90 
    Zinc finger protein 37 Zfp37 NM_009554   7.84 5.46 
    Zinc finger protein 68 Zfp68 NM_013844   2.03 3.15 
Others       
    Amyloid-β (A4) precursor protein binding, family B, member 3 Apbb3 BC024809 2.37 2.33   
    Aryl hydrocarbon receptor nuclear translocator like Arntl BC011080 5.12 21.26   
    Breast cancer metastasis suppressor 1 Brms1 NM_134155 2.04 4.57   
    Peroxisome proliferator-activated receptor–binding protein Pparbp NM_134027 2.56 2.63   
    Suppression of tumorigenicity 7 St7 NM_022332 2.08 3.75   
    Suppressor of cytokine signaling 6 Socs6 NM_018821 2.31 2.28   
    Suppressor of cytokine signaling 4 Socs4 AK014988 2.72 2.09   
    Tumor necrosis factor receptor-associated factor 6 Traf6 AV244412 2.22 2.14   
Gene descriptionSymbolGenbankLiver*
Small intestine
3 h12 h3 h12 h
Heat shock protein       
    Heat shock protein 2 Hspb2 AK012780 3.39 4.71   
    Heat shock protein 1A Hspa1a M12573 8.28 6.24 5.88 2.22 
    Heat shock protein 1A Hspa1a M12573 6.15 4.30 5.48 2.12 
    Crystallin, αB Cryab NM_009964 6.02 6.68   
Ubiquitination and proteolysis       
    A disintegrin and metalloproteinase domain 19 (meltrin β) Adam19 NM_009616 2.67 2.68   
    Cathepsin M Cstm NM_022326 3.68 2.14   
    Dipeptidylpeptidase 9 Dpp9 BB667346 2.15 4.83   
    Leishmanolysin-like (metallopeptidase M8 family) Lmln BB182358 5.17 5.70   
    Mucosa-associated lymphoid tissue lymphoma translocation gene 1 Malt1 BM239348 2.41 2.62   
    Procollagen C-proteinase enhancer protein Pcolce NM_008788 8.12 10.07   
    Proprotein convertase subtilisin/kexin type 5 Pcsk5 BC013068 11.83 18.73   
    Proteaseome (prosome, macropain) 28S subunit, 3 Psme3 U60330 2.30 2.88   
    Proteasome (prosome, macropain) 26S subunit, ATPase, 6 Psmc6 AW208944 2.31 3.02   
    Proteasome (prosome, macropain) 26S subunit, non-ATPase, 11 Psmd11 AA050796 2.67 3.41   
    Proteasome (prosome, macropain) 26S subunit, ATPase, 4 Psmc4 NM_011874 2.52 2.60   
    Proteasome (prosome, macropain) 26S subunit, non-ATPase, 9 Psmd9 BG092381 2.39 2.75   
    Ring finger protein 11 Rnf11 BI150320 2.12 2.24   
    Ubiquitin fusion degradation 1 like  BB500664 3.28 2.72   
    Ubiquitin-specific protease 30 Usp30 BG067690 3.37 3.93   
    Ubiquitin-specific protease 38 Usp38 BG064874 2.43 2.70   
    Ubiquitin-conjugating enzyme E2B  AK011961 2.31 2.13   
    Coagulation factor IX F9 M23109   5.06 8.86 
    Proteasome (prosome, macropain) activator, subunit 4  BB200981   8.21 3.24 
    Tolloid-like Tll1 NM_009390   7.80 3.61 
    Transferrin receptor 2 Trfr2 AV027486   3.86 4.20 
Electron transport       
    Aldehyde oxidase 1 Aox1 NM_009676 2.01 2.47   
    Cytochrome c oxidase, subunit VIIa 2 Cox7a2 BB745549 4.32 4.82   
    Cytochrome c oxidase, subunit VIIIb Cox8b NM_007751 3.14 2.38   
    Similar to cytochrome P450, 4a10 Cyp4a10 BC025936 2.02 2.42   
    Thioredoxin reductase 1 Txnrd1 NM_015762 2.12 2.16   
    Ubiquinol-cytochrome c reductase core protein 1  BG864756 2.08 2.57   
    Cytochrome P450, family 2, subfamily c, polypeptide 55 Cyp2c55 NM_028089   2.89 4.95 
Detoxification enzyme       
    Crystallin, ζ (quinone reductase) like 1 Cryzl1 AK010433 2.02 2.26   
    Fucosyltransferase 8 Fut8 NM_016893 10.78 10.62   
    Glutamate-cysteine ligase, catalytic subunit Gclc AW825835 2.41 4.26   
    Methyltransferase-like 1 Mettl1 AI838750 4.28 5.76   
    Sialyltransferase 10 Siat10 NM_018784 15.70 17.79   
    Steroid sulfatase Sts NM_009293 2.49 3.33   
    Thioredoxin interacting protein Txnip AF173681 2.94 2.34   
    Carbonyl reductase 3 Cbr3 AK003232   21.68 13.22 
    GST, μ1 Gstm1 NM_010358   4.42 11.65 
    GST, μ1 Gstm1 J03952   2.84 7.90 
    GST, μ3 Gstm3 J03953   3.72 5.91 
    GST, α2 (Yc2) Gsta2 NM_008182   2.79 9.58 
    GST, α3 Gsta3 AI172943   4.49 2.62 
    GST, α4 Gsta4 NM_010357   3.76 2.49 
    HO-1 (decycling) Hmox1 NM_010442   76.06 4.54 
    UDP-glucuronosyltransferase 2 family, member 5 Ugt2b5 NM_009467   2.07 6.70 
Transport       
    Aquaporin 7 Aqp7 AB056091 2.38 3.30   
    ATP synthase, H+ transporting, mitochondrial F0 complex, subunit c (subunit 9), isoform 2 Atp5g2 AW413339 4.03 5.12   
    ATPase, Ca2+ transporting, cardiac muscle, slow twitch 2 Atp2a2 AA245637 2.12 3.10   
    ATPase, class II, type 9A Atp9a AF011336 2.14 2.60   
    ATPase, Cu2+ transporting, α-polypeptide Atp7a U03434 4.06 3.10   
    ATP-binding cassette, subfamily B (MDR/TAP), member 1A Abcb1a M30697 2.23 2.05   
    ATP-binding cassette, subfamily B (MDR/TAP), member 1B Abcb1b NM_011075 7.06 5.18   
    ATP-binding cassette, subfamily D (ALD), member 3 Abcd3 BB042134 2.10 2.37   
    Cation channel, sperm associated 2 Catsper2 BB484902 2.18 2.15   
    Chloride channel 3 Clcn3 BB328803 12.31 15.49   
    Chloride channel calcium activated 1 Clca1 AF047838 7.22 3.48   
    Fatty acid–binding protein 4, adipocyte Fabp4 BC002148 5.62 3.24   
    FXYD domain-containing ion transport regulator 2 Fxyd2 NM_052823 3.62 11.19 4.50 6.00 
    Kinesin family member 5B Kif5b BI328541 2.27 2.41   
    Membrane targeting (tandem) C2 domain containing 1 Mtac2d1 AB062282 8.52 7.72   
    Myosin IC Myo1c NM_008659 3.28 2.79   
    N-ethylmaleimide-sensitive fusion protein Nsf BB400581 2.20 2.95   
    Potassium voltage-gated channel, Shab-related subfamily, member 1 Kcnb1 BB324482 6.09 3.17   
    Potassium voltage-gated channel, subfamily Q, member 2 Kcnq2 AB000502 11.48 2.10   
    Solute carrier family 12, member 2 Slc12a2 BG069505 2.27 2.71   
    Solute carrier family 13 (sodium-dependent dicarboxylate transporter), member 2 Slc13a2 BC013493 5.37 4.68   
    Solute carrier family 16 (monocarboxylic acid transporters), member 1 Slc16a1 NM_009196 2.25 8.09   
    Solute carrier family 18 (vesicular monoamine), member 2 Pdzk8 BB102308 3.38 3.12   
    Solute carrier family 2 (facilitated glucose transporter), member 9 BE197100 2.73 2.58    
    Solute carrier family 22 (organic cation transporter), member 3 Slc22a3 NM_011395 3.51 5.57   
    Solute carrier family 22 (organic cation transporter), member 5 Slc22a5 NM_011396 2.18 3.76   
    Solute carrier family 25 (mitochondrial carrier, palmitoylcarnitine transporter), member 29 Slc25a29 BC006711 2.06 2.20   
    Solute carrier family 37 (glycerol-3-phosphate transporter), member 3 Slc37a3 BC005744 2.09 2.49   
    Solute carrier family 39 (zinc transporter), member 14 Slc39a14 BB022806 3.78 2.11   
    Solute carrier family 6 (neurotransmitter transporter), member 14 Slc6a14 AF320226 23.50 19.96   
    Solute carrier family 9 (sodium/hydrogen exchanger), member 8 Slc9a8 AK018301 2.09 4.70   
    Src activating and signaling molecule Srcasm BC004710 2.08 2.04   
    Syntaxin 6 Stx6 BB492711 2.78 3.83   
    Transporter 2, ATP-binding cassette, subfamily B (MDR/TAP) Tap2 BE691515 2.90 2.12   
    ATPase, class V, type 10A Atp10a BM249532   5.13 3.11 
    Chemokine (C-C motif) ligand 7 Ccl7 AF128193   8.15 12.09 
    Hemopexin Hpxn BC011246   7.84 32.11 
    5-Hydroxytryptamine (serotonin) receptor 3A Htr3a NM_013561   2.74 2.31 
    Major urinary protein 3 Mup3 M27608   13.25 102.59 
    Solute carrier family 17 (sodium phosphate), member 1 Slc17a1 NM_009198   2.01 3.00 
    Solute carrier family 34 (sodium phosphate), member 2 Slc34a2 NM_011402   2.13 5.54 
    Solute carrier family 35 (UDP galactose transporter), member 2 AU080926   2.14 2.61  
    Solute carrier family 40 (iron-regulated transporter), member 1 Slc40a1 AF226613   2.22 2.43 
    Solute carrier family 6 (neurotransmitter transporter), member 14 Slc6a14 AF320226   3.50 3.29 
    Zinc finger protein 316 Zfp316 AV367169   2.34 2.00 
    Solute carrier family 4, sodium bicarbonate transporter-like, member 11 Slc4a11 BB498904   3.12 2.49 
    Sodium channel, voltage-gated, type IX, α-polypeptide  BB452274   2.72 4.16 
Apoptosis and cell cycle control       
    Apoptotic protease-activating factor 1 Apaf1 AK018076 16.54 14.58   
    Baculoviral IAP repeat-containing 1a Birc1a AF135491 3.02 4.02   
    Bcl-2-associated transcription factor 1 Bclaf1 BI965039 2.23 2.62   
    Bcl-2-like Bcl2l1 NM_009743 2.98 3.08   
    Cyclin-dependent kinase inhibitor 1A (p21) Cdkn1a AK007630 2.02 10.06   
    Cytotoxic granule-associated RNA-binding protein 1 Tia1 BG518542 4.18 4.22   
    Ring finger protein 7 Rnf7 AV047821 2.62 3.74   
    Tumor necrosis factor receptor-associated factor 3 Traf3 U21050 5.78 4.25   
    Transformation-related protein 53-inducible nuclear protein 1 Trp53inp1 AW495711 2.26 2.41   
    Tripartite motif-containing 35  BQ175280 3.80 3.27   
    Tumor differentially expressed 1 Tde1 NM_012032 3.15 4.29   
    CWF19-like 2, cell cycle control (Schizosaccharomyces pombeCwf19l2 AK014327 3.18 3.97   
    Bcl-2-interacting killer-like Biklk NM_007546   3.41 9.60 
    RAS like, estrogen regulated, growth inhibitor Rerg BC026463   3.81 2.60 
Cell adhesion       
    Cadherin 11 Cdh11 NM_009866 3.54 3.35   
    Cadherin 22 Cdh22 AB019618 2.00 2.54 2.37 2.77 
    Cadherin 4 Cdh4 NM_009867 15.48 10.96   
    Catenin α-like 1 Catnal1 BQ031240 4.49 4.74   
    Catenin src Catns NM_007615 2.15 3.08   
    Contactin-associated protein 1 Cntnap1 NM_016782 4.56 4.75   
    Integrin α8 Itga8 BB623587 6.71 6.44   
    Laminin, β3 Lamb3 NM_008484 2.41 5.53   
    Neurotrimin  AF282980 0.49 0.43   
    Osteomodulin Omd NM_012050 3.11 3.01   
    Protocadherin 18 Pcdh18 BM218630 2.83 3.78   
    Fibronectin leucine-rich transmembrane protein 2 Flrt2 BB817332   11.38 5.14 
    Procollagen, type IX, α1 Col9a1 AK004383   3.33 2.32 
    Thrombospondin 2 Thbs2 BB233297   2.06 2.51 
Kinase and phosphatase       
    Casein kinase II, α1 polypeptide Csnk2a1 BB283759 3.85 2.07   
    Induced in fatty liver dystrophy 2  BB508622 3.68 5.71   
    Keratin complex 2, basic, gene 8 Krt2-8 AW322280 2.49 2.56   
    MAPK-activated protein kinase 2 Mapkapk2 BG918951 2.07 2.84   
    Microtubule-associated serine/threonine kinase 2 Mast2 BB367890 5.34 12.50   
    MAPK kinase kinase 12 Map3k12 NM_009582 3.57 2.70   
    MAPK 8 interacting protein 3 Mapkip8 AF178636 3.41 4.28   
    MAPK kinase kinase kinase 4 Map4k4 NM_008696 3.71 4.24   
    MAPK kinase kinase kinase 5 Map4k5 BG067961 8.29 7.17   
    PCTAIRE-motif protein kinase 1 Pctk1 AW539955 6.66 5.81   
    PKC, α  BB355213 4.49 4.04   
    Protein kinase, cAMP-dependent regulatory, type IIβ Prkar2b BB216074 26.47 4.75   
    Proviral integration site 2  NM_138606 2.00 2.45   
    Regulator of G protein signaling 19 Rgs19 BC003838 2.09 2.49   
    Rho-associated coiled-coil forming kinase 1 Rock1 BI662863 2.15 3.07   
    Rho-associated coiled-coil forming kinase 2 Rock2 BB761686 3.07 2.54   
    Ribosomal protein S6 kinase, polypeptide 5 Rps6ka5 BQ174267 2.65 2.39   
    Serine/threonine kinase 17b (apoptosis inducing) Stk17b AV173139 3.05 2.53   
    Serum/glucocorticoid-regulated kinase 3 Sgk3 BB768208 6.68 6.67   
    SNF1-like kinase Snf1lk AI648260 7.55 9.82   
    Testis-specific protein kinase 1 Tesk1 NM_011571 2.05 2.28   
    Tousled-like kinase 2 (ArabidopsisTlk2 NM_011903 2.48 2.81   
    Dual specificity phosphatase 6 Dusp6 NM_026268 2.45 2.18   
    Eyes absent 3 homologue (DrosophilaEya3 BB428881 2.94 3.12   
    Paladin  NM_013753 2.14 2.53   
    Protein tyrosine phosphatase, nonreceptor type 21 Ptpn21 AW987375 7.44 13.85   
    PKC, μ Prkcm NM_008858   2.22 2.04 
    Keratin complex 2, basic, gene 8 Krt2-18 NM_016879   2.85 2.29 
    MAPK kinase kinase 10 Map3k10 AA789425   10.02 7.37 
    Dual specificity phosphatase 4 Dusp4 AK012530   2.65 2.00 
G protein-coupled receptors       
    G protein-coupled receptor 65 Gpr65 NM_008152 6.34 5.32   
    Endothelial differentiation, sphingolipid G protein-coupled receptor, 8 Edg8 NM_053190 4.92 3.92   
    Endothelin receptor type A Ednra BC008277 9.12 10.42   
    Endothelin receptor type A Ednra AW558570 7.84 7.60   
Transcription factors       
    Ankyrin repeat domain 1 (cardiac muscle) Ankrd1 AK009959 2.12 2.88   
    Basic transcription element-binding protein 1 Bteb1 NM_010638 2.90 3.54   
    cAMP-responsive element modulator Crem AU258667 9.73 6.29   
    cAMP-responsive element modulator Crem AI467599 2.58 2.56   
    cAMP-responsive element–binding protein–binding protein  BG076163 5.62 10.55   
    cAMP-responsive element–binding protein–binding protein/EP300 inhibitory protein 1 Cri1 BC010712 2.05 2.19   
    E4F transcription factor 1 E4f1 NM_007893 3.69 3.50   
    E74-like factor 1 Elf1 NM_007920 2.06 2.22   
    Early growth response 1 Egr1 NM_007913 4.19 5.06   
    Ewing sarcoma homologue Ewsr1 AW610680 10.50 5.63   
    Forkhead box N2 Foxn2 AV295543 3.34 2.75   
    Forkhead box P1 Foxp1 BG962849 2.71 3.65   
    Heterogeneous nuclear ribonucleoprotein A/B Hnrpab AK013709 6.55 7.28   
    Heterogeneous nuclear ribonucleoprotein R Hnrpr BB251000 2.50 2.10   
    Homeobox C8 Hoxc8 BB283726 4.83 2.57   
    Homeodomain leucine zipper-encoding gene  AV298304 7.72 8.52   
    Inhibitor of κB kinase γ Ikbkg BB147462 2.73 3.55   
    Inhibitor of κB kinase γ Ikbkg NM_010547 2.12 2.83   
    Kruppel-like factor 7 (ubiquitous) Klf7 BB524597 2.09 2.97   
    LIM homeobox protein 9 Lhx9 AK013209 3.23 5.88   
    Longevity assurance homologue 4 (Saccharomyces cerevisiaeLass4 BB006809 3.18 9.26   
    Nuclear factor, interleukin 3, regulated Nfil3 AY061760 2.13 3.52   
    Nuclear receptor subfamily 2, group C, member 2 Nr2c2 AU066920 2.22 2.60   
    Retinoid X receptor γ Rxrg NM_009107 2.63 2.94   
    SCAN-KRAB-zinc finger gene 1 Zpf306 BC007473 2.21 3.91   
    Suppressor of K+ transport defect 3 Skd3 NM_009191 2.80 2.76   
    TAF5 RNA polymerase II, TATA box-binding protein–associated factor Taf5 AV117817 2.96 3.90   
    TAR DNA-binding protein Tardbp BC012873 2.63 3.96   
    Transforming growth factor-β-inducible early growth response 1 Tieg1 NM_013692 2.76 3.52   
    Transcription factor 12 Tcf12 BB540782 2.79 2.86   
    Transcription factor 20 Tcf20 AW552808 2.22 2.14   
    Transcription factor 3 Tcf3 NM_009332 4.44 5.08   
    Zinc finger proliferation 1 Zipro1 AI326272 4.02 7.65   
    Zinc finger protein 148 Zfp148 X98096 2.11 2.29   
    Zinc finger protein 207 Zfp207 AV338324 2.16 2.11   
    Zinc finger protein 263 Zfp263 AI326880 3.02 2.96   
    Zinc finger protein 319 Zfp319 BB476317 5.45 5.41   
    Zinc finger protein 354C Zfp354c NM_013922 3.33 3.73   
    Zinc fingers and homeoboxes 3 Zhx3 BE952825 6.37 7.45   
    Ankyrin repeat domain 1 (cardiac muscle) Ankrd1 AK009959   3.82 16.77 
    CBFA2T1 identified gene homologue (human) Cbfa2t1h BG072085   19.43 12.24 
    E4F transcription factor 1  BB027397   7.90 9.35 
    Myeloid ecotropic viral integration site 1 Meis1 AW547821   4.31 3.67 
    POU domain, class 2, transcription factor 2 Pou2f2 X57938   12.27 12.47 
    Runt-related transcription factor 1 Runx1 NM_009821   2.24 2.64 
    Transforming growth factor-β1-induced transcript 4 Tgfbli4 AW413169   9.17 4.53 
    Zinc finger protein 2 Zfp2 NM_009550   6.84 3.90 
    Zinc finger protein 37 Zfp37 NM_009554   7.84 5.46 
    Zinc finger protein 68 Zfp68 NM_013844   2.03 3.15 
Others       
    Amyloid-β (A4) precursor protein binding, family B, member 3 Apbb3 BC024809 2.37 2.33   
    Aryl hydrocarbon receptor nuclear translocator like Arntl BC011080 5.12 21.26   
    Breast cancer metastasis suppressor 1 Brms1 NM_134155 2.04 4.57   
    Peroxisome proliferator-activated receptor–binding protein Pparbp NM_134027 2.56 2.63   
    Suppression of tumorigenicity 7 St7 NM_022332 2.08 3.75   
    Suppressor of cytokine signaling 6 Socs6 NM_018821 2.31 2.28   
    Suppressor of cytokine signaling 4 Socs4 AK014988 2.72 2.09   
    Tumor necrosis factor receptor-associated factor 6 Traf6 AV244412 2.22 2.14   
*

Genes that were induced >2-fold by curcumin only in liver of Nrf2 wild-type mice but not in liver of Nrf2 knockout mice comparing with vehicle treatment at both time points. The relative mRNA expression levels of each gene in treatment group over vehicle group (fold of change) were listed.

Genes that were induced >2-fold by curcumin only in small intestine of Nrf2 wild-type mice but not in small intestine of Nrf2 knockout mice comparing with vehicle treatment at both time points. The relative mRNA expression levels of each gene in treatment group over vehicle group (fold of change) were listed.

Surprisingly, curcumin treatment induced more genes that were not known previously as Nrf2/ARE pathway target genes than those related to Nrf2/ARE pathway in a Nrf2-dependent manner. For example, cytochrome P450 genes cyp4a10 and cyp2c55 were selectively induced in liver and small intestine, respectively. Many ubiquitination (Usp30 and Usp38) and proteolysis-related (Psmc4, Psmd9, Psme3, etc.) genes were also induced by curcumin in a Nrf2-dependent manner, especially in liver. Another major category of genes induced by curcumin in a Nrf2-dependent manner were transporter genes. Solute carrier family member genes were the major genes to be induced in both liver and small intestine. Interestingly, several ATP-binding cassette family transporter genes, such as Abcb1a, Abcb1b [multidrug resistance 1 (MDR1)], Abcd3, and Tap2 (Abcb3), also seem to be Nrf2 dependently induced by curcumin in liver. Transporter genes with function of transporting ions of Cu2+, K+, Cl, and H+ were also identified as Nrf2-dependent genes in liver. In addition to genes related to xenobiotic metabolism and excretion, genes involved in cell apoptosis, cell cycle control, cell adhesion, and signal transduction (kinase, phosphatase, and G protein-coupled receptor) were identified as targets of curcumin through Nrf2-dependent pathway. Representative genes affected in these categories include apoptotic protease-activating factor 1, cyclin-dependent kinase inhibitor 1A (p21), cadherin (Cdh4, Cdh11, and Cdh22), MAPK (Map3k12, Map4k4, and Map4k5), and G protein-coupled receptor 65, however, were mostly in liver. Curcumin treatment could also modulate many transcription-related genes in a Nrf2-dependent manner. These include cyclic AMP (cAMP)–responsive element modulator, cAMP-responsive element–binding protein–binding protein, inhibitor of κB kinase γ, and many zinc finger protein genes.

Curcumin-Suppressed Nrf2-Dependent Genes in Liver and Small Intestine

As shown in Table 3, curcumin treatment also inhibited the expression of many genes falling into similar functional categories in a Nrf2-dependent manner, although the number of genes was much smaller. Arachidonate 12-lipoxygenase gene was suppressed >2-fold by curcumin in liver. Cyp11a1 and Cyp2c50 genes were selectively inhibited in liver and small intestine, respectively. Solute carrier family genes were still the major ones in the category of transport to be suppressed in both liver and small intestine. In liver, transcription factor genes were another major category of genes being suppressed, such as forkhead box genes (Foxf2 and Foxm1), homeobox genes (Hoxb8 and Msx2), and Kruppel-like factor genes (Klf3 and Klf5).

Table 3.

Curcumin-suppressed Nrf2-dependent gene lists in liver and small intestine

Gene descriptionSymbolGenbankLiver*
Small intestine
3 h12 h3 h12 h
Ubiquitination and proteolysis       
    Cathepsin G Cstg NM_007800 0.27 0.06   
    Mast cell protease 6 Mcpt6 NM_010781 0.46 0.19   
    Matrix metalloproteinase 24 Mmp24 AB021226 0.24 0.35   
    Kallikrein 26 Klk13 NM_010115   0.49 0.25 
    Carboxypeptidase A4 Cpa4 AV294399   0.19 0.36 
Electron transport       
    Arachidonate 12-lipoxygenase Alox12 BB554189 0.33 0.21   
    Cytochrome P450, family 11, subfamily a, polypeptide 1 Cyp11a1 C87524 0.38 0.37   
    Interleukin-4 induced 1 Ll4i1 NM_010215 0.10 0.17   
    Phosducin Pdc NM_024458 0.45 0.46   
    Thioredoxin like 1 Txnl1 AV106191 0.33 0.20   
    Cytochrome P450, family 2, subfamily c, polypeptide 50 Cyp2c50 NM_134144   0.14 0.09 
Transport       
    ATPase, aminophospholipid transporter, class I, type 8A, member 1 Atp8a1 AW610650 0.36 0.21   
    ATPase, H+ transporting, V0 subunit D, isoform 2 Atp6v0d2 AV204216 0.49 0.43   
    ATP-binding cassette, subfamily C (CFTR/MDR-associated protein), member 8 Abcc8 BB515948 0.47 0.38   
    Fatty acid–binding protein 3, muscle and heart Fabp3 NM_010174 0.42 0.43   
    γ-Aminobutyric acid-A receptor, subunit δ Gabrd NM_008072 0.50 0.37   
    Glutamate receptor, ionotropic, AMPA4 (α4) Gria4 AV336506 0.11 0.37   
    Potassium channel, subfamily K, member 2 Kcnk2 NM_010607 0.41 0.23   
    Potassium inwardly rectifying channel, subfamily J, member 15 Kcnj15 BB533892 0.07 0.14   
    Retinaldehyde-binding protein 1 Rlbp1 NM_020599 0.27 0.06   
    Structural maintenance of chromosomes 2 like 1 (yeast) Smc2l1 BI684556 0.49 0.41   
    Solute carrier family 12, member 1 Slc12a1 NM_011389 0.40 0.39   
    Solute carrier family 15 (oligopeptide transporter), member 1 Slc15a1 NM_053079 0.46 0.30   
    Solute carrier family 2 (facilitated glucose transporter), member 10 Slc2a10 NM_130451 0.34 0.25   
    Solute carrier family 24 (sodium/potassium/calcium exchanger), member 1 Slc24a1 BC016094 0.44 0.41   
    Synaptogyrin 1 Syngr1 NM_009303 0.41 0.42   
    ATPase, H+/K+ transporting, nongastric, α-polypeptide Atp12a NM_138652   0.26 0.12 
    Glutamate receptor, ionotropic, kainate 2 (β2)  BB355480   0.23 0.36 
    Potassium intermediate/small conductance calcium-activated channel, subfamily N, member 2 Kcnn2 NM_080465   0.50 0.31 
    Solute carrier family 2 (facilitated glucose transporter), member 4 Slc2a4 AB008453   0.47 0.49 
    Solute carrier family 23 (nucleobase transporters), member 1 Slc23a1 AA276202   0.39 0.40 
    Solute carrier family 34 (sodium phosphate), member 1 Slc34a1 AK005930   0.13 0.45 
    Calcium channel, voltage-dependent, γ subunit 7 (Cacng7), mRNA  AF361349   0.42 0.11 
Cell adhesion       
    Desmocollin 1 Dsc1 NM_013504 0.07 0.24   
    Neurotrimin  AF282980 0.49 0.43   
    Protocadherin β16 Pcdhb16 BB131219 0.34 0.32   
    Cartilage link protein 1 Hapln1 AF098460   0.09 0.32 
    Putative neuronal cell adhesion molecule Punc BG067286   0.42 0.40 
Kinase and phosphatase       
    Interleukin-1 receptor-associated kinase 3 Irak3 BB497580 0.43 0.42   
    MAPK kinase 6  BB540608 0.50 0.30   
    Protein kinase, cyclic guanosine 3′,5′-monophosphate-dependent, type II Prkg2 BB823350 0.48 0.34   
    Protein tyrosine phosphatase, receptor type, E Ptpre U35368 0.35 0.44   
    Regulator of G protein signaling 18 Rgs18 BB139986 0.48 0.25   
    Tousled-like kinase 1 Tlk1 BM244995 0.35 0.32   
    Wingless-related mouse mammary tumor virus integration site 6 Wnt6 AV308073 0.35 0.34   
    Wingless-related mouse mammary tumor virus integration site 7A Wnt7a BB129109 0.36 0.45   
    Eph receptor A3 Epha3 M68515   0.39 0.26 
    Germ cell–specific gene 2 Gsg2 BE457839   0.49 0.34 
    Similar to PKCζ (LOC233024), mRNA PrkcZ BG143376   0.41 0.37 
G protein-coupled receptors       
    5-Hydroxytryptamine (serotonin) receptor 4 Htr4 Y09587 0.10 0.19   
    5-Hydroxytryptamine (serotonin) receptor 2C Htr2c BQ174268 0.42 0.38   
    G protein-coupled receptor 63 Gpr63 BB131092 0.23 0.19   
    Olfactory receptor 17 Olfr17 NM_020598 0.42 0.41   
    Platelet-derived growth factor receptor like Pdgfrl AK004179   0.35 0.47 
Transcription factors       
    Ankyrin repeat and SOCs box-containing protein 5 Asb5 NM_029569 0.10 0.44   
    Ankyrin repeat domain 6 Ankrd6 BM199504 0.50 0.39   
    Camello-like 3 Cml3 NM_053097 0.44 0.38   
    Forkhead box F2 Foxf2 NM_010225 0.24 0.35   
    Forkhead box M1 Foxm1 BB398835 0.32 0.34   
    Hairy/enhancer-of-split related with YRPW motif 2 Hey2 NM_013904 0.18 0.15   
    Homeobox B8 Hoxb8 X13721 0.45 0.05   
    Homeobox, msh-like 2 Msx2 AV297190 0.29 0.10   
    Jumonji domain containing 2C Jmjd2c BC020180 0.37 0.36   
    Kruppel-like factor 3 (basic) Klf3 BE687999 0.40 0.30   
    Kruppel-like factor 5 Klf5 BC006646 0.30 0.40   
    Paired like homeodomain factor 1 Prop1 NM_008936 0.48 0.30   
    Snail homologue 2 (DrosophilaSnai2 NM_011415 0.40 0.42   
    D site albumin promoter-binding protein Dbp BC018323   0.40 0.06 
    Insulin promoter factor 1, homeodomain transcription factor Lpf1 AK020261   0.37 0.26 
    Nuclear factor, erythroid derived 2, like 3 Nfe2l3 NM_010903   0.49 0.28 
    Period homologue 3 (DrosophilaPer3 NM_011067   0.46 0.15 
    Retinoic acid receptor–related orphan receptor β Rorb BB751387   0.16 0.38 
    SRY box–containing gene 11 Sox11 BG072739   0.49 0.31 
    SRY box–containing gene 4 Sox4 AI428101   0.31 2.42 
    Zinc finger protein 354C Zfp354c BB024472   0.28 0.31 
Others       
    Interleukin-4 induced 1 Il4i1 NM_010215 0.10 0.17   
    Chemokine (C-X-C motif) ligand 14 Cxcl14 AF252873 0.37 0.47   
    Vitamin D receptor Vdr AV290079 0.12 0.50   
Gene descriptionSymbolGenbankLiver*
Small intestine
3 h12 h3 h12 h
Ubiquitination and proteolysis       
    Cathepsin G Cstg NM_007800 0.27 0.06   
    Mast cell protease 6 Mcpt6 NM_010781 0.46 0.19   
    Matrix metalloproteinase 24 Mmp24 AB021226 0.24 0.35   
    Kallikrein 26 Klk13 NM_010115   0.49 0.25 
    Carboxypeptidase A4 Cpa4 AV294399   0.19 0.36 
Electron transport       
    Arachidonate 12-lipoxygenase Alox12 BB554189 0.33 0.21   
    Cytochrome P450, family 11, subfamily a, polypeptide 1 Cyp11a1 C87524 0.38 0.37   
    Interleukin-4 induced 1 Ll4i1 NM_010215 0.10 0.17   
    Phosducin Pdc NM_024458 0.45 0.46   
    Thioredoxin like 1 Txnl1 AV106191 0.33 0.20   
    Cytochrome P450, family 2, subfamily c, polypeptide 50 Cyp2c50 NM_134144   0.14 0.09 
Transport       
    ATPase, aminophospholipid transporter, class I, type 8A, member 1 Atp8a1 AW610650 0.36 0.21   
    ATPase, H+ transporting, V0 subunit D, isoform 2 Atp6v0d2 AV204216 0.49 0.43   
    ATP-binding cassette, subfamily C (CFTR/MDR-associated protein), member 8 Abcc8 BB515948 0.47 0.38   
    Fatty acid–binding protein 3, muscle and heart Fabp3 NM_010174 0.42 0.43   
    γ-Aminobutyric acid-A receptor, subunit δ Gabrd NM_008072 0.50 0.37   
    Glutamate receptor, ionotropic, AMPA4 (α4) Gria4 AV336506 0.11 0.37   
    Potassium channel, subfamily K, member 2 Kcnk2 NM_010607 0.41 0.23   
    Potassium inwardly rectifying channel, subfamily J, member 15 Kcnj15 BB533892 0.07 0.14   
    Retinaldehyde-binding protein 1 Rlbp1 NM_020599 0.27 0.06   
    Structural maintenance of chromosomes 2 like 1 (yeast) Smc2l1 BI684556 0.49 0.41   
    Solute carrier family 12, member 1 Slc12a1 NM_011389 0.40 0.39   
    Solute carrier family 15 (oligopeptide transporter), member 1 Slc15a1 NM_053079 0.46 0.30   
    Solute carrier family 2 (facilitated glucose transporter), member 10 Slc2a10 NM_130451 0.34 0.25   
    Solute carrier family 24 (sodium/potassium/calcium exchanger), member 1 Slc24a1 BC016094 0.44 0.41   
    Synaptogyrin 1 Syngr1 NM_009303 0.41 0.42   
    ATPase, H+/K+ transporting, nongastric, α-polypeptide Atp12a NM_138652   0.26 0.12 
    Glutamate receptor, ionotropic, kainate 2 (β2)  BB355480   0.23 0.36 
    Potassium intermediate/small conductance calcium-activated channel, subfamily N, member 2 Kcnn2 NM_080465   0.50 0.31 
    Solute carrier family 2 (facilitated glucose transporter), member 4 Slc2a4 AB008453   0.47 0.49 
    Solute carrier family 23 (nucleobase transporters), member 1 Slc23a1 AA276202   0.39 0.40 
    Solute carrier family 34 (sodium phosphate), member 1 Slc34a1 AK005930   0.13 0.45 
    Calcium channel, voltage-dependent, γ subunit 7 (Cacng7), mRNA  AF361349   0.42 0.11 
Cell adhesion       
    Desmocollin 1 Dsc1 NM_013504 0.07 0.24   
    Neurotrimin  AF282980 0.49 0.43   
    Protocadherin β16 Pcdhb16 BB131219 0.34 0.32   
    Cartilage link protein 1 Hapln1 AF098460   0.09 0.32 
    Putative neuronal cell adhesion molecule Punc BG067286   0.42 0.40 
Kinase and phosphatase       
    Interleukin-1 receptor-associated kinase 3 Irak3 BB497580 0.43 0.42   
    MAPK kinase 6  BB540608 0.50 0.30   
    Protein kinase, cyclic guanosine 3′,5′-monophosphate-dependent, type II Prkg2 BB823350 0.48 0.34   
    Protein tyrosine phosphatase, receptor type, E Ptpre U35368 0.35 0.44   
    Regulator of G protein signaling 18 Rgs18 BB139986 0.48 0.25   
    Tousled-like kinase 1 Tlk1 BM244995 0.35 0.32   
    Wingless-related mouse mammary tumor virus integration site 6 Wnt6 AV308073 0.35 0.34   
    Wingless-related mouse mammary tumor virus integration site 7A Wnt7a BB129109 0.36 0.45   
    Eph receptor A3 Epha3 M68515   0.39 0.26 
    Germ cell–specific gene 2 Gsg2 BE457839   0.49 0.34 
    Similar to PKCζ (LOC233024), mRNA PrkcZ BG143376   0.41 0.37 
G protein-coupled receptors       
    5-Hydroxytryptamine (serotonin) receptor 4 Htr4 Y09587 0.10 0.19   
    5-Hydroxytryptamine (serotonin) receptor 2C Htr2c BQ174268 0.42 0.38   
    G protein-coupled receptor 63 Gpr63 BB131092 0.23 0.19   
    Olfactory receptor 17 Olfr17 NM_020598 0.42 0.41   
    Platelet-derived growth factor receptor like Pdgfrl AK004179   0.35 0.47 
Transcription factors       
    Ankyrin repeat and SOCs box-containing protein 5 Asb5 NM_029569 0.10 0.44   
    Ankyrin repeat domain 6 Ankrd6 BM199504 0.50 0.39   
    Camello-like 3 Cml3 NM_053097 0.44 0.38   
    Forkhead box F2 Foxf2 NM_010225 0.24 0.35   
    Forkhead box M1 Foxm1 BB398835 0.32 0.34   
    Hairy/enhancer-of-split related with YRPW motif 2 Hey2 NM_013904 0.18 0.15   
    Homeobox B8 Hoxb8 X13721 0.45 0.05   
    Homeobox, msh-like 2 Msx2 AV297190 0.29 0.10   
    Jumonji domain containing 2C Jmjd2c BC020180 0.37 0.36   
    Kruppel-like factor 3 (basic) Klf3 BE687999 0.40 0.30   
    Kruppel-like factor 5 Klf5 BC006646 0.30 0.40   
    Paired like homeodomain factor 1 Prop1 NM_008936 0.48 0.30   
    Snail homologue 2 (DrosophilaSnai2 NM_011415 0.40 0.42   
    D site albumin promoter-binding protein Dbp BC018323   0.40 0.06 
    Insulin promoter factor 1, homeodomain transcription factor Lpf1 AK020261   0.37 0.26 
    Nuclear factor, erythroid derived 2, like 3 Nfe2l3 NM_010903   0.49 0.28 
    Period homologue 3 (DrosophilaPer3 NM_011067   0.46 0.15 
    Retinoic acid receptor–related orphan receptor β Rorb BB751387   0.16 0.38 
    SRY box–containing gene 11 Sox11 BG072739   0.49 0.31 
    SRY box–containing gene 4 Sox4 AI428101   0.31 2.42 
    Zinc finger protein 354C Zfp354c BB024472   0.28 0.31 
Others       
    Interleukin-4 induced 1 Il4i1 NM_010215 0.10 0.17   
    Chemokine (C-X-C motif) ligand 14 Cxcl14 AF252873 0.37 0.47   
    Vitamin D receptor Vdr AV290079 0.12 0.50   
*

Genes that were suppressed >2-fold by curcumin only in liver of Nrf2 wild-type mice but not in liver of Nrf2 knockout mice comparing with vehicle treatment at both time points. The relative mRNA expression levels of each gene in treatment group over vehicle group (fold of change) were listed.

Genes that were suppressed >2-fold by curcumin only in small intestine of Nrf2 wild-type mice but not in small intestine of Nrf2 knockout mice comparing with vehicle treatment at both time points. The relative mRNA expression levels of each gene in treatment group over vehicle group (fold of change) were listed.

Quantitative Real-time PCR Validation of Microarray Data

To verify the data generated from the microarray, seven genes from different categories (Table 1) were chosen to confirm the curcumin regulation effects by using quantitative real-time PCR analyses as described in Materials and Methods. Values for each gene were normalized by the values of corresponding glyceraldehyde-3-phosphate dehydrogenase gene and the ratios of treated/vehicle were calculated. The Spearman correlation was calculated and it showed that the data generated from microarray analyses are well correlated with the results obtained from quantitative real-time PCR (R2 = 0.74; Fig. 3).

Figure 3.

Correlation of microarray data and quantitative real-time PCR data. Fold of changes of gene expression measured by real-time PCR were plotted against the corresponding fold of changes in microarray data. The Spearman correlation was calculated as R2 = 0.74, which indicated the data from the two methods were in good correlation.

Figure 3.

Correlation of microarray data and quantitative real-time PCR data. Fold of changes of gene expression measured by real-time PCR were plotted against the corresponding fold of changes in microarray data. The Spearman correlation was calculated as R2 = 0.74, which indicated the data from the two methods were in good correlation.

Close modal

The major goal of this study is to identify cancer chemopreventive agent curcumin-regulated Nrf2-dependent genes in mice liver and small intestine by using Nrf2 wild-type/knockout mice and genome-scale microarray analysis. As a cancer chemopreventive agent, curcumin could function as a cancer-blocking agent to block the tumorigenesis process in many rodent carcinogenesis models (2224) by inducing phase II detoxification and antioxidant genes to enhance the elimination of carcinogen or reactive intermediates. During this process, Nrf2 is believed to play a central role because phase II detoxification and antioxidant genes are mainly regulated by Nrf2/ARE pathway in response to phase II inducer or chemopreventive agents. Because it is known that curcumin can induce several phase II detoxification enzyme genes and Nrf2 is critical in phase II gene induction and cancer chemoprevention, the identification of many phase II detoxification and antioxidant genes as curcumin-induced Nrf2-dependent genes in this study not only is consistent with previous studies (17, 19) but also validated the results from a biological perspective. For example, the induction of selective isoform of GST and oxidative-stress response gene HO-1 is consistent with previous findings in which curcumin could induce the expression of GST (25, 26) and HO-1 (19) through Nrf2/ARE pathway. The induction of cytochrome c oxidase subunits (Cox7a2 and Cox8b) and thioredoxin reductase 1 further supports the role of Nrf2 in curcumin-elicited gene expression because their promoter regions all contain putative Nrf2-binding sites.

Interestingly, many genes involved in phase I drug metabolism and phase III drug transporting process were also regulated by curcumin depending on Nrf2 status. Cyp4a10 and Cyp2c55 were induced in liver and small intestine, respectively, whereas Cyp11a1 and Cyp2c50 were suppressed; however, their roles in the cancer chemopreventive effect of curcumin remain unclear. The transport function-related genes were the major group of genes being regulated by curcumin in a Nrf2-dependent manner in both liver and small intestine. Although the interaction between curcumin and transporters, such as MDR1 and MDR-associated protein 1, has been investigated in vitro (27), the effects of curcumin on other transporters, especially their expression, have never been examined in vivo. Solute carrier family transporter genes were the major ones to be selectively regulated in liver and small intestine. Altered expression of these transporter genes could perturb the transporting of organic cation (Slc22a3), glycerol-3-phosphate (Slc37a3), and monocarboxylic acid (Slc16a1) and could affect sodium/hydrogen exchange (Slc9a8). The induction of four ATP-binding cassette transporter genes, such as Abcb1b (MDR1), suggested that Nrf2 also play a significant role in regulating ATP-binding cassette family transporter genes. Although PXR and CAR have been shown to play critical roles in regulating the expression of MDR1 and MDR-associated protein genes (28), the role of Nrf2 has not been excluded. Hemopexin was dramatically induced by curcumin in a Nrf2-dependent manner in small intestine. Hemopexin is critical in maintaining the homeostasis of metal ions by forming a complex with heme. As the major vehicle for the transportation of heme, hemopexin could prevent heme-mediated oxidative stress and heme-bound iron loss (29), functionally analogous to HO-1, which metabolizes heme and prevent oxidative stress. Taken together, our current study suggested that curcumin could coordinately regulate the phase I, II, and III xenobiotic metabolizing enzyme genes as well as antioxidative stress genes through Nrf2-dependent pathways in vivo. Such regulation (especially induction) of these genes could have significant effects on prevention of tumor initiation by enhancing the cellular defense system, preventing the activation of procarcinogens/reactive intermediates, and increasing the excretion of reactive carcinogen or metabolites.

Previous in vitro (3033) and in vivo (34, 35) studies have suggested that curcumin could also act as a tumor-suppressing agent by regulating many cellular signal transduction pathways in cancer cells. Therefore, modulation of signaling pathways (4, 8, 9) involved in cell proliferation, cell cycle control, apoptosis, adhesion, invasion and metastasis, angiogenesis, and inflammation by curcumin were linked to its strong cancer chemopreventive effects. However, the role of Nrf2 in curcumin-elicited alternation of signaling transduction pathways related to these cellular events has never been investigated. In the current study, apoptosis-related gene apoptotic protease-activating factor 1 was induced by curcumin >14-fold at both time points in the liver. Because the regulation of apoptotic protease-activating factor 1 by curcumin and Nrf2 has not been reported, our results suggested that curcumin-induced cancer cell apoptosis may result from its Nrf2-dependent regulation of apoptotic protease-activating factor 1–related pathways. Cell cycle control gene cyclin-dependent kinase inhibitor 1A (p21) was induced >10-fold at 12 hours on curcumin administration. This is supported by a previous study in which curcumin cause G1 arrest in PC-3 cells by induction of p21 (36). Curcumin has been shown to inhibit cancer cell invasion and metastasis (25, 37) by modulating integrin receptors, collagenase activity, and expression of E-cadherin. In the current study, several cadherin genes were also induced by curcumin, such as Cdh4, Cdh11, and Cdh22, in liver, although Cdh22 gene was also induced in small intestine. The cadherin family of transmembrane glycoproteins plays a critical role in cell-to-cell adhesion, and cadherin dysregulation is strongly associated with cancer metastasis and progression (38). Because impaired expression of cadherin genes were associated with cancer invasion and metastasis (39), the induction of cadherin genes through Nrf2/ARE pathway by curcumin could be another potential mechanism of exerting its cancer chemoprevention effects. Although microarray studies cannot provide information on the regulation of kinase phosphorylation by curcumin, our results indicated that the expression of many signaling pathway members was affected in a Nrf2-dependent manner after curcumin treatment. The induction of nuclear factor-κB signaling pathway component gene inhibitor of κB kinase γ and suppression of Wnt signaling pathway-related Wnt6 and Wnt7a genes were consistent with previous results (4042). The suppression of phosphatidylinositol 3-kinase downstream target protein kinase C (PKC) ζ–related gene Prkcz suggested that curcumin may intervene in the phosphatidylinositol 3-kinase signaling and nuclear factor-κB p65 subunit nuclear translocation in small intestine (43). Because PKCμ could phosphorylate E-cadherin and increase prostate cancer cell aggregation and decrease cellular motility (38), the induction of both PKCμ gene and several cadherin genes by curcumin in small intestine may contribute to its colon cancer chemopreventive effect.

Because of the protective role of Nrf2-mediated gene expression in response to carcinogen or reactive oxygen intermediate challenge, it is essential and important to identify novel Nrf2/ARE pathway target genes related to cancer chemoprevention in addition to phase II detoxification and antioxidant genes (2, 16). By comparing the gene expression patterns elicited by promising cancer chemopreventive agent curcumin between Nrf2 wild-type and knockout mice, we identified many novel curcumin-regulated Nrf2-dependent genes with a variety of biological functions in mice liver and small intestine. The identification of these genes clearly expanded our scope of understanding the role of Nrf2 in cancer chemoprevention as well as potential new mechanisms of cancer chemoprevention. Interestingly, two previous microarray studies using chemopreventive agent sulforaphane (18) and 3H-1,2-dithiole-3-thione (17) to compare their gene expression profiles between wild-type and Nrf2-deficient mice also identified some of the similar functional categories of Nrf2-dependent genes. Although the chemopreventive agents used in previous studies and our current study are different, the similar induction pattern and the regulation of many identical Nrf2-dependent genes strongly suggested a relationship between the sets of genes being regulated and the cancer chemopreventive effects of these compounds as well as the predominant role of Nrf2 in the regulation of these genes. It also suggested that an elicited similar global gene expression change rather than the regulation of individual pathways could lead to the overall cancer protective effect by these different classes of chemopreventive compounds. Future in vivo or in vitro studies to explore the roles of Nrf2-dependent genes related to ubiquitination, drug metabolism, cell growth and adhesion, phosphorylation, and transcription as uncovered in our current study will greatly extend our knowledge on cancer chemoprevention.

Grant support: NIH grant R01 CA094828.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Curtis Krier (CINJ Core Expression Array Facility) for assistance with the microarray analyses.

1
Surh YJ. Cancer chemoprevention with dietary phytochemicals.
Nat Rev Cancer
2003
;
3
:
768
–80.
2
Chen C, Kong AN. Dietary cancer-chemopreventive compounds: from signaling and gene expression to pharmacological effects.
Trends Pharmacol Sci
2005
;
26
:
318
–26.
3
Sporn MB, Dunlop NM, Newton DL, Smith JM. Prevention of chemical carcinogenesis by vitamin A and its synthetic analogs (retinoids).
Fed Proc
1976
;
35
:
1332
–8.
4
Huang MT, Newmark HL, Frenkel K. Inhibitory effects of curcumin on tumorigenesis in mice.
J Cell Biochem Suppl
1997
;
27
:
26
–34.
5
Chauhan DP. Chemotherapeutic potential of curcumin for colorectal cancer.
Curr Pharm Des
2002
;
8
:
1695
–706.
6
Plummer SM, Holloway KA, Manson MM, et al. Inhibition of cyclo-oxygenase 2 expression in colon cells by the chemopreventive agent curcumin involves inhibition of NF-κB activation via the NIK/IKK signalling complex.
Oncogene
1999
;
18
:
6013
–20.
7
Hong J, Bose M, Ju J, et al. Modulation of arachidonic acid metabolism by curcumin and related β-diketone derivatives: effects on cytosolic phospholipase A(2), cyclooxygenases and 5-lipoxygenase.
Carcinogenesis
2004
;
25
:
1671
–9.
8
Leu TH, Maa MC. The molecular mechanisms for the antitumorigenic effect of curcumin.
Curr Med Chem Anti-Canc Agents
2002
;
2
:
357
–70.
9
Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of curcumin: preclinical and clinical studies.
Anticancer Res
2003
;
23
:
363
–98.
10
Itoh K, Wakabayashi N, Katoh Y, et al. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain.
Genes Dev
1999
;
13
:
76
–86.
11
Dinkova-Kostova AT, Holtzclaw WD, Cole RN, et al. Direct evidence that sulfhydryl groups of Keap1 are the sensors regulating induction of phase 2 enzymes that protect against carcinogens and oxidants.
Proc Natl Acad Sci U S A
2002
;
99
:
11908
–13.
12
Shen G, Hebbar V, Nair S, et al. Regulation of Nrf2 transactivation domain activity. The differential effects of mitogen-activated protein kinase cascades and synergistic stimulatory effect of Raf and CREB-binding protein.
J Biol Chem
2004
;
279
:
23052
–60.
13
Aoki Y, Sato H, Nishimura N, et al. Accelerated DNA adduct formation in the lung of the Nrf2 knockout mouse exposed to diesel exhaust.
Toxicol Appl Pharmacol
2001
;
173
:
154
–60.
14
Enomoto A, Itoh K, Nagayoshi E, et al. High sensitivity of Nrf2 knockout mice to acetaminophen hepatotoxicity associated with decreased expression of ARE-regulated drug metabolizing enzymes and antioxidant genes.
Toxicol Sci
2001
;
59
:
169
–77.
15
Ramos-Gomez M, Kwak MK, Dolan PM, et al. Sensitivity to carcinogenesis is increased and chemoprotective efficacy of enzyme inducers is lost in nrf2 transcription factor-deficient mice.
Proc Natl Acad Sci U S A
2001
;
98
:
3410
–5.
16
Lee JS, Surh YJ. Nrf2 as a novel molecular target for chemoprevention.
Cancer Lett
2005
;
224
:
171
–84.
17
Kwak MK, Wakabayashi N, Itoh K, et al. Modulation of gene expression by cancer chemopreventive dithiolethiones through the Keap1-Nrf2 pathway. Identification of novel gene clusters for cell survival.
J Biol Chem
2003
;
278
:
8135
–45.
18
Thimmulappa RK, Mai KH, Srisuma S, et al. Identification of Nrf2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray.
Cancer Res
2002
;
62
:
5196
–203.
19
Balogun E, Hoque M, Gong P, et al. Curcumin activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive element.
Biochem J
2003
;
371
:
887
–95.
20
Chan K, Lu R, Chang JC, Kan YW. NRF2, a member of the NFE2 family of transcription factors, is not essential for murine erythropoiesis, growth, and development.
Proc Natl Acad Sci U S A
1996
;
93
:
13943
–8.
21
Hu R, Hebbar V, Kim BR, et al. In vivo pharmacokinetics and regulation of gene expression profiles by isothiocyanate sulforaphane in the rat.
J Pharmacol Exp Ther
2004
;
310
:
263
–71.
22
Huang MT, Ma W, Lu YP, et al. Effects of curcumin, demethoxycurcumin, bisdemethoxycurcumin and tetrahydrocurcumin on 12-O-tetradecanoylphorbol-13-acetate-induced tumor promotion.
Carcinogenesis
1995
;
16
:
2493
–7.
23
Huang MT, Wang ZY, Georgiadis CA, Laskin JD, Conney AH. Inhibitory effects of curcumin on tumor initiation by benzo[a]pyrene and 7,12-dimethylbenz[a]anthracene.
Carcinogenesis
1992
;
13
:
2183
–6.
24
Huang MT, Lou YR, Ma W, et al. Inhibitory effects of dietary curcumin on forestomach, duodenal, and colon carcinogenesis in mice.
Cancer Res
1994
;
54
:
5841
–7.
25
Piper JT, Singhal SS, Salameh MS, et al. Mechanisms of anticarcinogenic properties of curcumin: the effect of curcumin on glutathione linked detoxification enzymes in rat liver.
Int J Biochem Cell Biol
1998
;
30
:
445
–56.
26
Singhal SS, Awasthi S, Pandya U, et al. The effect of curcumin on glutathione-linked enzymes in K562 human leukemia cells.
Toxicol Lett
1999
;
109
:
87
–95.
27
Nabekura T, Kamiyama S, Kitagawa S. Effects of dietary chemopreventive phytochemicals on P-glycoprotein function.
Biochem Biophys Res Commun
2005
;
327
:
866
–70.
28
Xu C, Li CY, Kong AN. Induction of phase I, II and III drug metabolism/transport by xenobiotics.
Arch Pharm Res
2005
;
28
:
249
–68.
29
Tolosano E, Altruda F. Hemopexin: structure, function, and regulation.
DNA Cell Biol
2002
;
21
:
297
–306.
30
Jee SH, Shen SC, Tseng CR, Chiu HC, Kuo ML. Curcumin induces a p53-dependent apoptosis in human basal cell carcinoma cells.
J Invest Dermatol
1998
;
111
:
656
–61.
31
Han SS, Chung ST, Robertson DA, Ranjan D, Bondada S. Curcumin causes the growth arrest and apoptosis of B cell lymphoma by downregulation of egr-1, c-myc, bcl-XL, NF-κB, and p53.
Clin Immunol
1999
;
93
:
152
–61.
32
Choudhuri T, Pal S, Agwarwal ML, Das T, Sa G. Curcumin induces apoptosis in human breast cancer cells through p53-dependent Bax induction.
FEBS Lett
2002
;
512
:
334
–40.
33
Rashmi R, Santhosh Kumar TR, Karunagaran D. Human colon cancer cells differ in their sensitivity to curcumin-induced apoptosis and heat shock protects them by inhibiting the release of apoptosis-inducing factor and caspases.
FEBS Lett
2003
;
538
:
19
–24.
34
Goel A, Boland CR, Chauhan DP. Specific inhibition of cyclooxygenase-2 (COX2) expression by dietary curcumin in HT-29 human colon cancer cells.
Cancer Lett
2001
;
172
:
111
–8.
35
Kawamori T, Lubet R, Steele VE, et al. Chemopreventive effect of curcumin, a naturally occurring anti-inflammatory agent, during the promotion/progression stages of colon cancer.
Cancer Res
1999
;
59
:
597
–601.
36
Hour TC, Chen J, Huang CY, et al. Curcumin enhances cytotoxicity of chemotherapeutic agents in prostate cancer cells by inducing p21(WAF1/CIP1) and C/EBPβ expressions and suppressing NF-κB activation.
Prostate
2002
;
51
:
211
–8.
37
Ray S, Chattopadhyay N, Mitra A, Siddiqi M, Chatterjee A. Curcumin exhibits antimetastatic properties by modulating integrin receptors, collagenase activity, and expression of Nm23 and E-cadherin.
J Environ Pathol Toxicol Oncol
2003
;
22
:
49
–58.
38
Jaggi M, Rao PS, Smith DJ, et al. E-cadherin phosphorylation by protein kinase D1/protein kinase Cμ is associated with altered cellular aggregation and motility in prostate cancer.
Cancer Res
2005
;
65
:
483
–92.
39
Matsuura K, Kawanishi J, Fujii S, et al. Altered expression of E-cadherin in gastric cancer tissues and carcinomatous fluid.
Br J Cancer
1992
;
66
:
1122
–30.
40
Bharti AC, Donato N, Singh S, Aggarwal BB. Curcumin (diferuloylmethane) down-regulates the constitutive activation of nuclear factor-κB and IκBα kinase in human multiple myeloma cells, leading to suppression of proliferation and induction of apoptosis.
Blood
2003
;
101
:
1053
–62.
41
Surh YJ, Han SS, Keum YS, Seo HJ, Lee SS. Inhibitory effects of curcumin and capsaicin on phorbol ester-induced activation of eukaryotic transcription factors, NF-κB and AP-1.
Biofactors
2000
;
12
:
107
–12.
42
Narayan S. Curcumin, a multi-functional chemopreventive agent, blocks growth of colon cancer cells by targeting β-catenin-mediated transactivation and cell-cell adhesion pathways.
J Mol Histol
2004
;
35
:
301
–7.
43
Cataldi A, Di Pietro R, Centurione L, et al. Engagement of PI-3-kinase mediated protein kinase C ζ activation in protecting Friend cells from ionizing radiation-induced apoptosis.
Int J Oncol
2003
;
22
:
129
–35.