Hypoxia-inducible factor (HIF), a key modulator of the transcriptional response to hypoxia, is increased in colon cancer. However, the role of HIF in colon carcinogenesis in vivo remains unclear. In this study, we found that intestinal epithelium-specific disruption of the von Hippel–Lindau tumor suppressor protein (VHL) resulted in constitutive HIF signaling, and increased HIF expression augmented colon tumorigenesis in the Apcmin/+ intestinal tumor model. Intestine-specific disruption of Vhl increased colon tumor multiplicity and progression from adenomas to carcinomas. These effects were ameliorated in mice with double disruption of Vhl and HIF-2α. Activation of HIF signaling resulted in increased cell survival in normal colon tissue; however, tumor apoptosis was not affected. Interestingly, a robust activation of cyclin D1 was observed in tumors of Apcmin/+ mice in which HIF-2α was activated in the intestine. Consistent with this result, bromodeoxyuridine incorporation indicated that cellular proliferation was increased in colon tumors following HIF activation. Further analysis showed that dysregulation of the intestinal iron absorption transporter divalent metal transporter-1 (DMT-1) was a critical event in HIF-2α–mediated colon carcinogenesis. These data provide a mechanistic basis for the widely reported link between iron accumulation and colon cancer risk. Together, our findings show that a chronic increase in HIF-2α in the colon initiates protumorigenic signaling, which may have important implications in developing preventive and therapeutic strategies for colon cancer. Cancer Res; 72(9); 2285–93. ©2012 AACR.

Hypoxic microenvironment is a hallmark for solid tumors (1). In response to hypoxia, tumor cells activate genes that are critical in angiogenesis, cell survival, cell proliferation, and glucose metabolism (2). Hypoxia-induced signal transduction is transcriptionally mediated by hypoxia-inducible factor (HIF), a member of the Per-ARNT-Sim family of basic helix-loop-helix transcription factors that bind hypoxia response elements at target gene loci under hypoxic conditions (3–5). Functional HIF is a heterodimer that comprises a constitutive subunit, aryl hydrocarbon nuclear translocator (Arnt, also known as HIF-1β), and a hypoxia-inducible α-subunit (3, 4). Stabilization of the α-subunit is regulated by a family of oxygen and iron-dependent prolyl hydroxylase (PHD) enzymes. PHD enzymes hydroxylate the α-subunit enabling the binding of the von Hippel–Lindau tumor suppressor protein (VHL) coupled to the E3 ubiquitin ligase complex, which leads to proteasomal degradation of HIF-α subunits (6, 7).

Two highly homologous and transcriptionally active subunits have been identified, HIF-1α and HIF-2α (8, 9). Both are expressed in many of the same cells and regulate overlapping and distinct sets of genes critical in the adaptation to hypoxic environments, including cancer development (10). In cancer-derived cell lines and in renal carcinomas, HIF-1α and HIF-2α have opposing roles in cell proliferation. HIF-1α decreases cell proliferation, whereas HIF-2α induces proliferation via an increase in c-Myc activity (11–16). HIF-1α and HIF-2α are overexpressed in a variety of tumor tissues including colon cancer (17, 18). However, the role of HIF-α in colon carcinogenesis is not completely clear.

This study shows that Apcmin/+ mice with an intestine-specific activation of HIF signaling via disruption of Vhl using a villin-cre recombinase developed mainly colorectal tumors, with carcinomas seen in 8 of 10 (80%) mice followed for 6 months. Disruption of both Vhl and Hif-2α in intestinal epithelial cells prevented colon tumors indicating an HIF-2α–dependent mechanism. Through global gene expression analysis in the Apcmin/+ mice and human colorectal tumor samples, an HIF-2α–dependent dysregulation of colon iron homeostasis was observed. Increase in local iron levels have been implicated in the progression of colon carcinogenesis (19–25), and these data show the increase in tumor iron exacerbated cell proliferation and was critical in colon tumor formation following HIF-2α activation. Together, these data reveal a novel role for HIF-2α in initiating a coordinated process that is critical in the progression of colon cancer.

Animals and diets

The floxed or compound floxed mice hemizygous for villin-cre were mated to each other to generate VhlF/F, VhlΔIE, VhlF/F/Hif-2αF/F, VhlΔIE/Hif-2αΔIE mice and were described previously (26–28). Apcmin/+ mice were purchased from The Jackson Laboratory. To investigate the role of HIF-2α in colorectal cancer, VhlΔIE and VhlΔIE/Hif-2αΔIE mice were crossed with Apcmin/+ mice to generate VhlF/F/Apcmin/+, VhlΔIE/Apcmin/+, VhlΔIE/Hif-2αΔIE/Apcmin/+, and VhlF/F/Hif-2αF/F/Apcmin/+ mice. All mice were 129S6/SvEv background and maintained in standard cages in a light- and temperature-controlled room and were allowed standard chow and water ad libitum. For low iron study, the 6-week-old VhlΔIE/Apcmin/+ and VhlF/F/Apcmin/+ mice were given iron-replete AIN93G diet containing 350 ppm of iron or iron-deficient AIN93G diet (>5 ppm of iron) for 8 weeks (Dyets). All animal studies were carried out in accordance with Institute of Laboratory Animal Resources Guidelines and approved by the University Committee on the Use and Care of Animals (UCUCA) at the University of Michigan (approval number: 10299).

Histology, immunohistochemistry, and immunofluorescence

For bromodeoxyuridine (BrdUrd) staining, animals were sacrificed following a 2-hour treatment of 100 mg/kg of BrdUrd (Sigma). Paraffin-embedded tissue sections (5 μm) were deparaffinized in xylene and rehydrated in ethanol gradient. Immunohistochemical analysis was conducted with antibodies for BrdUrd (Bu20a; eBioscience Inc.) followed by detection with Alexa Fluor 488 goat anti-mouse immunoglobulin G (Molecular Probes Inc.). Terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling (TUNEL) assay (Roche Diagnostics) was conducted according to the supplier's instructions. Briefly, deparaffinized sections were labeled with TdT and biotinylated dUTP and then were examined under a fluorescence microscope (29). Tissue iron detection was carried out in paraffin-embedded sections stained with Perls Prussian blue and enhanced with 3,3′-diaminobenzidine and H2O2 (30). Histologic analysis was done on hematoxylin and eosin (H&E)–stained paraffin sections and microscopically analyzed by a gastrointestinal pathologist.

Quantitative real-time reverse transcriptase PCR

RNA was isolated from fresh or frozen tissue with Isol-RNA lysis reagent (3 Prime). After quantification with NanoDrop 2000 (NanoDrop products), RNA with a purity of approximately 2.0 (260/280 ratio) was reverse transcribed using M-MLV Reverse Transcriptase (Fisher Scientific). cDNA was quantified by SYBR green dye and run on a 7900HT Fast Real Time RT-PCR system (Life Technologies; primers listed in Supplementary Table S1). Ct values were normalized to β-actin and expressed as fold difference from controls.

Western blot analysis

Tissues were homogenized and lysed in radioimmunoprecipitation assay buffer (RIPA; 50 mmol/L Tris-HCl pH 7.5, 150 mmol/L NaCl, 2 mmol/L EDTA, 1% NP-40, 0.1% SDS) for whole-cell extracts. Proteins were separated and transferred to nitrocellulose membranes using standard methods. Membranes were incubated with antibodies against cleaved keratin 18 (K18; kindly provided by Prof. Bishr Omary, University of Michigan, Ann Arbor, MI), total caspase 3, cleaved caspase 3 (Cell Signaling Technology Inc.), HIF-2α (Novus Biologicals), divalent metal transporter-1 (DMT-1; Alpha Diagonostic International) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH; Santa Cruz Biotechnology) followed by incubation with appropriate horseradish peroxidase–conjugated secondary antibodies (Cell Signaling Technology, Inc.). Blots were detected by the Enhanced Chemiluminescence Detection kit (Thermo Scientific).

Primary colon organoid and other cell culture

For organoid whole-colon cultures, colons were opened longitudinally and were rinsed gently and quickly placed in culture medium, RPMI-1640 plus 10% heat inactivated FBS and 1% antibiotic/antimycotic (1 unit/mL of penicillin, 1 μg/mL of streptomycin, and 2.5 ng/mL of amphotericin B; Life Technologies). The colons were cut into 5 to 7 mm pieces and incubated in the culture medium described above for 2 hours at 37°C, 5% CO2/21% O2. After incubation, tissues were homogenized and lysed in RIPA buffer to conduct Western blot analysis as described above. HCT116 cells were obtained from American Type Culture Collection and maintained at 37°C in 5% CO2 and 21% O2. Cells were cultured in Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 10% FBS and 1% antibiotic/antimycotic. Early passaged (P10) stable HIF-2α and parental HCT116 cells were generated by transfection of a normoxically stable HIF-2α or pIRES puro 3 empty vector with Lipofectamine 2000 (Life Technologies) according to manufacturer's instructions and selected by 2 μg/mL puromycin. The cells were maintained in growth media described above containing 1 μg/mL of puromycin.

PolyHema apoptosis assay

Cell adhesion to proper extracelluar matrix is essential for epithelial cell survival. Failure of attachment leads to a form of apoptosis termed anoikis (31). Poly(hydroxy ethyl methacrylate) (PolyHema) is a polymer which can form a surface that prevents cell adherence when coated on cell culture plate (32). Twenty-four–well plates were coated with 250 μL per well PolyHema (20 mg/mL) to achieve nonadherent conditions. HIF-2α overexpressing or parental HCT116 cells were plated at a concentration of 1 × 104 cells/mL in DMEM supplemented with 10% FBS and 1% antibiotic/antimycotic into PolyHema coated plates or noncoated control plates. After 24- or 48-hour culture, 125 μL 5 mg/mL MTT (Sigma) reagent was added to each well and incubated for 30 minutes. Dimethyl sulfoxide was added and absorbance was measured at 570 nm.

cDNA microarray analysis

RNA was extracted from colon mucosal scrapings from 5-week-old VhlF/F, VhlΔIE, VhlF/F/Apcmin/+, and VhlΔIE/Apcmin/+ mice. Isolated RNA with RNA integrity number values more than 6.5 examined by Agilent 2100 bioanalyzer (Agilent Technologies, Inc.) was amplified, reverse transcribed, labeled, and hybridized to mouse 430 2.0 Affymetrix GeneChips (Affymetrix), and data were analyzed as previously described (28). The full data set is available on the Gene Expression Omnibus database accession number GSE36091.

Statistics

Results are expressed as mean ± SD. P values were calculated by independent t test, one-way ANOVA, Dunnett t test, and two-way ANOVA. P < 0.05 was considered significant.

Activation of HIF-2α potentiates colon carcinogenesis following adenomatous polyposis coli mutation

Mice with an intestine-specific disruption of Vhl (VhlΔIE) activate HIF signaling, whereas no induction of HIF signaling is observed in littermate controls (VhlF/F; refs. 28, 33). To assess the role of HIF in intestinal tumorigenesis, these mice were crossed to the Apcmin/+ mice (Supplementary Fig. S1A). Apcmin/+ mice have a nonsense mutation at codon 850 of the murine Apc gene leading to a truncated protein (34). Adenomatous polyposis coli (APC) is a tumor suppressor protein highly relevant in colon cancer; more than 80% of patients with sporadic colon cancer have a somatic mutation of the Apc gene (35), and mutations are observed in early and late colitis–associated neoplasms (36–38). In patients with Apc mutations, the predominant cancer is colon cancer and very rarely (∼10-fold less) are small intestinal tumors observed (39). However, Apcmin/+ mice develop mostly small intestinal tumors (34). Consistent with previous data, VhlF/F/Apcmin/+ mice developed predominantly small intestinal tumors. In the small intestine, tumors were found in 17 of 17 3-month-old VhlF/F/Apcmin/+ mice, with a tumor multiplicity of 32.24, whereas 4 of 17 VhlF/F/Apcmin/+ mice developed tumors in the colon with a tumor multiplicity of 0.59. Interestingly, VhlΔIE mice crossed to the Apcmin/+ background had a dramatic shift in the tumor localization to the colon in 3-month-old mice (Fig. 1A–C). Small intestinal tumors were significantly decreased in the 3-month-old VhlΔIE/Apcmin/+ compared with VhlF/F/Apcmin/+ mice (tumor incidence: 11 of 15 mice; tumor multiplicity: 2.80). However, a significant increase in tumor incidence and numbers were observed in the colon of 3-month-old VhlΔIE/Apcmin/+ mice (tumor incidence: 15 of 15 mice; tumor multiplicity: 13.87). Consistent with this, Western blot analysis showed an increase of HIF-2α protein expression in both normal colon and tumor tissues from VhlΔIE/Apcmin/+ mice (Supplementary Fig. S2). To assess whether the changes in tumor number and incidence were dependent on HIF-2α, a double knockout mouse model of Vhl and Hif-2α on an Apcmin/+ background (VhlΔIE/Hif-2αΔIE/Apcmin/+) was generated and compared with littermate controls (VhlF/F/Hif-2αF/F/Apcmin/+; Supplementary Fig. S1B). Interestingly, small intestinal tumors in VhlΔIE/Hif-2αΔIE/Apcmin/+ mice were significantly repressed compared with the VhlF/F/Hif-2αF/F/Apcmin/+ mice. These data were similar to that observed in the VhlΔIE/Apcmin/+ mice indicating that VHL-mediated decrease in small intestinal tumorigenesis is independent of HIF-2α (Fig. 1B and C). However, the increase in the colon tumorigenesis in VhlΔIE/Apcmin/+ mice was completely ablated following compound knockout of Vhl and Hif-2α in the Apcmin/+ background, showing that HIF-2α plays a critical role in the development of colon cancer (Fig. 1B and C). These results were further confirmed in the 6-month-old VhlΔIE/Apcmin/+ and VhlΔIE//Hif-2αΔIE/Apcmin/+ mice (Fig. 1D and E). Compared with 3-month-old VhlF/F/Apcmin/+ mice, the tumor multiplicities in 6-month-old VhlF/F/Apcmin/+ mice were increased in the small intestine (tumor multiplicity: 62.50, tumor incidence: 6 of 6 mice) and colon (tumor multiplicity: 1.33, tumor incidence: 3 of 6 mice). However, small intestinal tumors were still significantly decreased in the 6-month-old VhlΔIE/Apcmin/+ (tumor incidence: 10 of 10 mice; tumor multiplicity: 15.10) compared with 6-month-old VhlF/F/Apcmin/+ mice. The tumor incidence and numbers observed in the colon of 6-month-old VhlΔIE/Apcmin/+ mice (tumor incidence: 10 of 10 mice; tumor multiplicity: 14.50) were not further increased compared with 3-month-old VhlΔIE/Apcmin/+ mice. However, they were still significantly increased compared with 6-month-old VhlF/F/Apcmin/+ mice. Assessing the double knockout mice (VhlΔIE/Hif-2αΔIE/Apcmin/+) at 6 months further showed that the decrease in small intestinal tumorigenesis is independent of HIF-2α, whereas the increase in colon carcinogenesis is completely dependent on HIF-2α.

Figure 1.

HIF-2α modulates intestinal tumorigenesis in Apcmin/+ mice. A, representative colons from VhlF/F/Apcmin/+ (n = 24) and VhlΔIE/Apcmin/+ mice (n = 25). Tumor counting in the small intestine (B) and colon (C) from 3-month-old VhlF/F/Apcmin/+ (n = 17), VhlΔIE/Apcmin/+ (n = 15), VhlF/F/Hif-2αF/F/Apcmin/+ (n = 13), and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice (n = 11). Tumor counting in the small intestine (D) and colon (E) from 6-month-old VhlF/F/Apcmin/+ (n = 7), VhlΔIE/Apcmin/+ (n = 10), VhlF/F/Hif-2αF/F/Apcmin/+ (n = 6), and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice (n = 6). *, P < 0.05 and ***, P < 0.001; NS, not significant.

Figure 1.

HIF-2α modulates intestinal tumorigenesis in Apcmin/+ mice. A, representative colons from VhlF/F/Apcmin/+ (n = 24) and VhlΔIE/Apcmin/+ mice (n = 25). Tumor counting in the small intestine (B) and colon (C) from 3-month-old VhlF/F/Apcmin/+ (n = 17), VhlΔIE/Apcmin/+ (n = 15), VhlF/F/Hif-2αF/F/Apcmin/+ (n = 13), and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice (n = 11). Tumor counting in the small intestine (D) and colon (E) from 6-month-old VhlF/F/Apcmin/+ (n = 7), VhlΔIE/Apcmin/+ (n = 10), VhlF/F/Hif-2αF/F/Apcmin/+ (n = 6), and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice (n = 6). *, P < 0.05 and ***, P < 0.001; NS, not significant.

Close modal

Activation of HIF-2α increases tumor progression in the Apcmin/+ mice

All tumors assessed in the small intestine or colon from 3- and 6-month-old VhlF/F/Apcmin/+ mice showed well-organized glandular structures and were classified as adenomas (Fig. 2A). However, 2 out of 15 tumors from 3-month-old VhlΔIE/Apcmin/+ mice showed early signs of carcinoma formation (Fig. 2B and D). In 6-month-old VhlΔIE/Apcmin/+ mice, 8 out of 10 mice had colon tumors that displayed complex glands with cribriform architecture and desmoplastic stroma, and thus were classified as carcinomas (Fig. 2C and D). To assess whether the increase in colon cancer progression was HIF-2α dependent, tumor analysis was conducted on 6-month-old VhlΔIE/Hif-2αΔIE/Apcmin/+ mice (Fig. 2D). Although adenomas were observed, no tumors progressed to carcinomas. These data show that activation of HIF-2α is critical for the progression of colon tumors.

Figure 2.

HIF-2α promotes carcinoma formation in Apcmin/+ mice. A, H&E staining of representative duodenum and colon from 3-month-old VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice. B, H&E staining of representative colon from 3-month-old VhlΔIE/Apcmin/+ mice with an adenoma and progression to early-stage carcinoma. C, H&E staining of representative colon carcinoma from 6-month-old VhlΔIE/Apcmin/+ mice. D, incidence of carcinoma in 3- and 6-month-old VhlF/F/Apcmin/+, VhlΔIE/Apcmin/+, VhlF/F/Hif-2αF/F/Apcmin/+, and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice.

Figure 2.

HIF-2α promotes carcinoma formation in Apcmin/+ mice. A, H&E staining of representative duodenum and colon from 3-month-old VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice. B, H&E staining of representative colon from 3-month-old VhlΔIE/Apcmin/+ mice with an adenoma and progression to early-stage carcinoma. C, H&E staining of representative colon carcinoma from 6-month-old VhlΔIE/Apcmin/+ mice. D, incidence of carcinoma in 3- and 6-month-old VhlF/F/Apcmin/+, VhlΔIE/Apcmin/+, VhlF/F/Hif-2αF/F/Apcmin/+, and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice.

Close modal

HIF-2α regulates cell survival in the colon

Analysis of apoptosis in colon tumors between VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ was conducted with TUNEL assay. No significant difference was observed in tumors from VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice (Supplementary Fig. S3). Interestingly, a dramatic increase in cell survival was observed in normal colon epithelial cells of VhlΔIE mice compared with VhlF/F mice. Intestinal epithelial cells undergo a spontaneous form of apoptosis termed anoikis as the differentiated cells reach the villus tips and are shed (40). Capturing this by TUNEL or caspase staining is difficult. Therefore, the apoptotic response of primary colon organoids generated from VhlF/F and VhlΔIE mice were compared. The colons from VhlF/F and VhlΔIE mice were isolated and snap frozen immediately or cultured for 2 hours. Upon culturing, the colon undergoes a dramatic induction of anoikis-induced apoptosis as seen with increased expressions of cleaved K18 and cleaved caspase 3 in colons isolated from VhlF/F mice (Fig. 3A). However, in colons cultured from VhlΔIE mice, significant decreases in cleaved K18 and cleaved caspase 3 expressions were observed (Fig. 3A). In contrast, cleaved K18 and cleaved caspase 3 expressions were partially restored in colons cultured from VhlΔIE/Hif-2αΔIE mice (Fig. 3A). To further confirm these results, a stable HIF-2α overexpressing colon-derived HCT116 cell line was generated. As expected, in parental HCT116 cells, HIF-2α was not detectable, whereas in the stable overexpressing cells, a dramatic increase in HIF-2α expression was observed (Fig. 3B). Cell survival was assessed in an anoikis-induced apoptotic assay. Cells were incubated in PolyHema-coated plates to inhibit attachment, leading to increased apoptosis. Parental cells showed decreased survival rate following 24 and 48 hours of incubation. The survival rate was significantly increased in HCT116 cells overexpressing HIF-2α (Fig. 3C) with a concordant decrease in the activation of caspase 3 following 24 hours of incubation on PolyHema-coated plates (Fig. 3D). Although no significant difference is observed in tumor apoptosis (Supplementary Fig. S3), activation of HIF-2α induces cell survival that may play an important role in colon tumor progression.

Figure 3.

HIF-2α increases cell survival in the colon. A, Western blot for cleaved caspase 3 (cCasp 3), cleaved K18, and GAPDH in colon organoid cultures from VhlF/F, VhlΔIE, VhlF/F/Hif-2αF/F, and VhlΔIE/Hif-2αΔIE mice incubated for 0 or 2 hours at 37°C. B, Western blot analysis for HIF-2α and GAPDH from HCT116 stably transfected with empty vector (EV) or HIF-2α plasmid. C, cell survival rate of HCT116 cells stably transfected with EV or HIF-2α plasmid in an anoikis-induced apoptosis assay. Survival rates were compared with uncoated control plates. Each bar represents the mean value ± SD (n = 3). *, P < 0.05; and **, P < 0.01 versus EV. D, Western blot analysis for cCasp3, total caspase 3 (Casp3), and GAPDH from HCT116 cells stably transfected with EV or HIF-2α plasmid 24 hours following induction of anoikis.

Figure 3.

HIF-2α increases cell survival in the colon. A, Western blot for cleaved caspase 3 (cCasp 3), cleaved K18, and GAPDH in colon organoid cultures from VhlF/F, VhlΔIE, VhlF/F/Hif-2αF/F, and VhlΔIE/Hif-2αΔIE mice incubated for 0 or 2 hours at 37°C. B, Western blot analysis for HIF-2α and GAPDH from HCT116 stably transfected with empty vector (EV) or HIF-2α plasmid. C, cell survival rate of HCT116 cells stably transfected with EV or HIF-2α plasmid in an anoikis-induced apoptosis assay. Survival rates were compared with uncoated control plates. Each bar represents the mean value ± SD (n = 3). *, P < 0.05; and **, P < 0.01 versus EV. D, Western blot analysis for cCasp3, total caspase 3 (Casp3), and GAPDH from HCT116 cells stably transfected with EV or HIF-2α plasmid 24 hours following induction of anoikis.

Close modal

HIF-2α regulates cell proliferation in colon cancer

Because apoptosis could not fully explain the increase in colon tumor formation in the VhlΔIE/Apcmin/+ mice, the contribution of cell proliferation to HIF-2α–induced colon cancer was assessed. Cyclin D1 expression was assessed in VhlΔIE/Apcmin/+ and VhlF/F/Apcmin/+ mice by quantitative PCR (qPCR) analysis. No significant difference of cyclin D1 expression was observed in the normal colon tissues of VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice. However, cyclin D1 was significantly induced in the colon tumors of VhlΔIE/Apcmin/+ mice compared with tumors isolated from VhlF/F/Apcmin/+ mice (Fig. 4A). Consistent with cyclin D1 expression, BrdUrd incorporation revealed no significant difference in the normal colon tissues of VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice (Fig. 4B and C). However, the numbers of BrdUrd positive proliferating epithelial cells in the colon tumors of VhlΔIE/Apcmin/+ mice were significantly higher than those in tumors of VhlF/F/Apcmin/+ mice (Fig. 4B and C). Elevated tumor proliferation was ablated in the colon tumors of VhlΔIE/Hif-2αΔIE/Apcmin/+ mice (Fig. 4D). These data show that HIF-2α increases tumor cell proliferation, which may be critical for the increase in colon tumor formation and progression.

Figure 4.

HIF-2α increases cell proliferation in the colon tumors. A, qPCR analysis of cyclin D1 (Ccnd1) in normal and tumor colon tissues from VhlF/F/Apcmin/+, VhlΔIE/Apcmin/+, and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice. Each bar represents the mean value ± SD (n = 4). Expression was normalized to β-actin. BrdUrd staining (B) and quantification in normal and tumor colon tissues from 6-month-old VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice 2 hours following 100 mg/kg BrdUrd intraperitoneal injection (C). Each bar represents the mean value ± SD (n = 4). D, BrdUrd staining and quantification in normal and tumor colon tissues from VhlF/F/Hif-2αF/F/Apcmin/+ and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice 2 hours following 100 mg/kg BrdUrd intraperitoneal injection. N, normal tissue; T, tumor tissue; NS, not significant; *, P < 0.05; **, P < 0.01 versus normal tissue; ##, P < 0.01 versus VhlF/F/Apcmin/+ mice. Each bar represents the mean value ± SD (n = 4).

Figure 4.

HIF-2α increases cell proliferation in the colon tumors. A, qPCR analysis of cyclin D1 (Ccnd1) in normal and tumor colon tissues from VhlF/F/Apcmin/+, VhlΔIE/Apcmin/+, and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice. Each bar represents the mean value ± SD (n = 4). Expression was normalized to β-actin. BrdUrd staining (B) and quantification in normal and tumor colon tissues from 6-month-old VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice 2 hours following 100 mg/kg BrdUrd intraperitoneal injection (C). Each bar represents the mean value ± SD (n = 4). D, BrdUrd staining and quantification in normal and tumor colon tissues from VhlF/F/Hif-2αF/F/Apcmin/+ and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice 2 hours following 100 mg/kg BrdUrd intraperitoneal injection. N, normal tissue; T, tumor tissue; NS, not significant; *, P < 0.05; **, P < 0.01 versus normal tissue; ##, P < 0.01 versus VhlF/F/Apcmin/+ mice. Each bar represents the mean value ± SD (n = 4).

Close modal

DMT-1 and iron uptake represent a cellular target and regulator of HIF-2α–induced colon cancer

To identify the precise molecular mechanisms that could contribute to the increase in colon carcinogenesis, microarray gene expression analysis was conducted on colon RNA isolated from 5-week-old VhlF/F and VhlΔIE, VhlΔIE/Apcmin/+ and VhlF/F/Apcmin/+ mice. The data identified 469 genes that were significantly increased/decreased in both VhlΔIE and VhlΔIE/Apcmin/+ compared with their littermate controls and 29 genes that were significantly increased/decreased in both VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ compared with their littermate Apc wild-type controls (Fig. 5A). The top 10 genes that were regulated in an HIF-2α–dependent manner were further assessed using ONCOMINE (Supplementary Table S2), a cancer microarray data mining platform (41). Those genes that were increased in the colons of VhlΔIE/Apcmin/+ mice and in human colon cancer samples were identified. This analysis showed a specific increase in DMT-1 expression in colon adenomas and carcinomas isolated from patients compared with normal controls in 3 independent studies (refs. 42–44; Fig. 5B). DMT-1 is the major apical iron transporter in the intestine (45–47). It is essential for iron absorption and its expression in the small intestine is directly regulated by HIF-2α (26). Colon tumors have been shown to accumulate iron (23). Moreover, mouse models of colon cancer and population-based studies show a direct correlation between an increase in intestinal iron and increased incidence of colon cancer (24, 25). These studies suggest that dysregulation of local iron homeostasis is critical in colon carcinogenesis. Consistent with these results, measuring tissue iron using enhanced Prussian iron staining showed a significant increase in iron accumulation in colon tumors compared with normal tissue (Fig. 5C). To assess whether HIF-2α was critical for DMT-1 regulation during colon tumorigenesis, DMT-1 expression was measured by qPCR. DMT-1 was significantly induced in the colons of VhlF/F/Apcmin/+ mice compared with wild-type littermates (VhlF/F), which was further potentiated in VhlΔIE/Apcmin/+ mice compared with wild-type littermates in an HIF-2α–dependent manner (Fig. 5D). Similar expression patterns were also noted in the small intestine (Supplementary Fig. S4). Western blot analysis further showed that DMT-1 protein expression was increased in an HIF-2α-dependent manner in colon tumors (Fig. 5E).

Figure 5.

DMT-1expression in colon tumors. A, global gene expression profiling in colon RNAs isolated from 5-week-old VhlF/F (n = 4), VhlF/F/Apcmin/+ (n = 3), VhlΔIE (n = 3), and VhlΔIE/Apcmin/+ mice (n = 5). B, DMT1 gene expression levels in 3 independent human colon cancer studies were analyzed by ONCOMINE. **, P < 0.01; ***, P < 0.001 versus normal colon tissue. C, enhanced Prussian blue staining for iron in normal colon and colon cancer tissue. Arrows indicate iron staining. D, qPCR analysis of Dmt1 in colon epithelial cells from VhlF/F (n = 6), VhlF/F/Apcmin/+ (n = 5), VhlΔIE/Apcmin/+ (n = 5), and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice (n = 3). Expression was normalized to β-actin. *, P < 0.05; ***, P < 0.001. E, Western blot analysis for DMT-1 and GAPDH from normal and tumor colon tissues of 3-month-old VhlF/F/Apcmin/+, VhlΔIE/Apcmin/+, VhlF/F/Hif-2αF/F/Apcmin/+, and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice. N, normal tissue; T, tumor tissue.

Figure 5.

DMT-1expression in colon tumors. A, global gene expression profiling in colon RNAs isolated from 5-week-old VhlF/F (n = 4), VhlF/F/Apcmin/+ (n = 3), VhlΔIE (n = 3), and VhlΔIE/Apcmin/+ mice (n = 5). B, DMT1 gene expression levels in 3 independent human colon cancer studies were analyzed by ONCOMINE. **, P < 0.01; ***, P < 0.001 versus normal colon tissue. C, enhanced Prussian blue staining for iron in normal colon and colon cancer tissue. Arrows indicate iron staining. D, qPCR analysis of Dmt1 in colon epithelial cells from VhlF/F (n = 6), VhlF/F/Apcmin/+ (n = 5), VhlΔIE/Apcmin/+ (n = 5), and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice (n = 3). Expression was normalized to β-actin. *, P < 0.05; ***, P < 0.001. E, Western blot analysis for DMT-1 and GAPDH from normal and tumor colon tissues of 3-month-old VhlF/F/Apcmin/+, VhlΔIE/Apcmin/+, VhlF/F/Hif-2αF/F/Apcmin/+, and VhlΔIE/Hif-2αΔIE/Apcmin/+ mice. N, normal tissue; T, tumor tissue.

Close modal

Low iron treatment reduces HIF-2α–induced colon tumor formation

To verify that the increase in iron contributed to the increase in colon carcinogenesis, VhlΔIE/Apcmin/+ and VhlF/F/Apcmin/+ mice were placed on control or low iron diet. As expected, the VhlΔIE/Apcmin/+ mice on control diet showed increased colon tumor numbers compared with littermate controls (Fig. 6A). The increase in colon tumors was completely ablated in VhlΔIE/Apcmin/+ mice on low iron diet for 2 months (Fig. 6A). Analysis of BrdUrd incorporation revealed no significant difference in the normal colon tissues of VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice treated with control diet. The number of BrdUrd-positive proliferating epithelial cells in the colon tumors of VhlΔIE/Apcmin/+ mice was dramatically higher than that in tumors of VhlF/F/Apcmin/+ mice, whereas low iron diet treatment greatly decreased the number of BrdUrd-positive epithelial cells in the tumor colon tissues of both VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice (Fig. 6B). These results show that tumoral iron is essential for colon cancer progression and a critical pathway by which HIF-2α signaling contributes to colon tumorigensis.

Figure 6.

Low iron diet decreases HIF-2α–mediated intestinal tumorigenesis and cellular proliferation. A, tumor numbers in the colon from VhlF/F/Apcmin/+ (n = 11) and VhlΔIE/Apcmin/+ mice (n = 10) treated with control or low iron diet for 2 months. **, P < 0.01; NS, not significant. B, BrdUrd staining of colon tumors from VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice treated with control or low iron diet for 2 months.

Figure 6.

Low iron diet decreases HIF-2α–mediated intestinal tumorigenesis and cellular proliferation. A, tumor numbers in the colon from VhlF/F/Apcmin/+ (n = 11) and VhlΔIE/Apcmin/+ mice (n = 10) treated with control or low iron diet for 2 months. **, P < 0.01; NS, not significant. B, BrdUrd staining of colon tumors from VhlF/F/Apcmin/+ and VhlΔIE/Apcmin/+ mice treated with control or low iron diet for 2 months.

Close modal

Hypoxia is an adaptive response in many solid tumors. However, its role in tumor development in vivo is still not clear. This study found that the activation of HIF signaling through HIF-2α enhanced colon cancer incidence and progression in the Apcmin/+ mice. The Apc tumor suppressor is a gatekeeper gene mutated in a majority of patients with familial, sporadic, and colitis-associated colon cancer (35–38). However, Apcmin/+ mice develop mostly small intestinal tumors, and colon tumors are observed with low incidence and multiplicity (34). HIF-2α activation in the intestines of VhlΔIE/Apcmin/+ mice dramatically increased colon tumor multiplicity and incidence. Moreover, at 6 months of age the majority of colon tumors observed were histologically defined as carcinomas, whereas no tumors identified in the VhlF/F/Apcmin/+ mice progressed further than adenomas, showing that HIF-2α may be a critical transcription factor involved in colon cancer progression.

Interestingly, small intestinal tumors observed in the VhlΔIE/Apcmin/+ mice were rare and this decrease in tumorigenesis was independent of HIF-2α expression. Despite the profound decrease in tumorigenesis, no changes in small intestinal proliferation were observed between VhlΔIE/Apcmin/+ and VhlF/F/Apcmin/+ mice (Supplementary Fig. S5). Moreover, the VhlΔIE/Apcmin/+ mice had increased DMT-1 expression in the small intestine, suggesting that decreased small intestinal tumorigenesis by the HIF-2α–independent mechanism is downstream of the iron accumulation effects. HIF-1α was shown to have an antagonistic role in cell growth and VHL has several HIF-independent functions (11, 48–52), and these pathways are currently being assessed in the small intestine. There is some evidence that colon tumors in the Apcmin/+ do not form because of the large tumor load of the small intestine compromise the mice, therefore the slower forming colon tumors do not develop. However, in the VhlΔIE/Hif-2αΔIE/Apcmin/+ mice, the decrease in small intestinal tumors did not lead to a refractory increase in colon tumors. Confirming that the increase in colon tumors in the VhlΔIE/Apcmin/+ mice is mediated by HIF-2α activation. The VhlΔIE/Apcmin/+ mice, through HIF-2α–dependent and HIF-2α–independent mechanisms, can recapitulate downstream events following Apc mutation that is observed in sporadic, familial, and colitis-associated colon cancer and is an optimal preclinical animal model to study colon cancer.

The mechanisms that decrease small intestinal tumorigenesis are currently not known. However, the increases in colon tumor incidence, multiplicity, and progression in VhlΔIE/Apcmin/+ mice was completely ablated following double knockout of Vhl and Hif-2α in the Apcmin/+ background, showing that HIF-2α plays a critical role in the development of colon cancer following Apcmin/+ mutation. Activation of HIF-2α in the intestine increased epithelial cell survival. This was measured with colon organoids that may be in undergoing apoptosis very early following the culture of the colons and using colon cancer–derived cells lines. Therefore, the anoikis detected may not be a physiologic response, and a more detailed analysis of HIF-2α–mediated cell survival is needed. However, this study clearly shows a role of HIF-2α in tumor proliferation. To understand the mechanisms involved in the increase in colon tumorigenesis, the HIF-2α–dependent gene expression profile was analyzed by the ONCOMINE database (41). The HIF-2α target gene DMT-1 was highly induced in colon adenomas and carcinomas compared with normal colon tissue isolated from patients. DMT-1 is a critical intestinal iron transporter, and its overexpression can lead to iron accumulation (45, 46, 53, 54). Iron accumulation is a critical factor in colon carcinogenesis and studies have shown that both body iron stores and dietary iron intake are positively associated with subsequent risk of colon cancer (22). In mouse models of colon cancer, iron-enriched diets increase the development of colon tumors (21). Iron activates free radical formation, which have a pleiotropic effect in carcinogenesis, including increase in cell survival and proliferation, which is consistent with this study. Together the data suggest that HIF-2α–mediated dysregulation of intestinal iron transport is critical for the increase in colon tumors observed in the VhlΔIE/Apcmin/+ mice. Therefore, inhibition of HIF-2α activation in colon tumors may represent a potential strategy for the prevention and treatment of colon cancer. Moreover, our data suggest that iron chelators will be beneficial for patients with colon cancer. It is important to note that HIF-2α activates a large battery of genes in the intestine and its mechanistic role in tumor development is most likely through multiple pathways. However, in this study, we clearly show that the activation of HIF-2α contributes to the increase in cell proliferation, dysregulation of local iron homeostasis, and eventually an increase in the incidence of colon cancer.

No potential conflicts of interest were disclosed.

Conception and design: X. Xue, A. Qu, E.M. Zimmermann, F.J. Gonzalez, Y.M. Shah

Development of methodology: X. Xue, E. Anderson, C. Hao, Y.M. Shah

Acquisition of data: X. Xue, M. Taylor, E. Anderson, A. Qu, Y.M. Shah

Analysis and interpretation of data: X. Xue, A. Qu, J.K. Greenson, Y.M. Shah

Writing, review, and/or revision of the manuscript: X. Xue, M. Taylor, E. Anderson, C. Hao, E.M. Zimmermann, F.J. Gonzalez, Y.M. Shah

Administrative, technical, or material support: Y.M. Shah

Study supervision: F.J. Gonzalez, Y.M. Shah

Provided mice: F.J. Gonzalez

The authors thank Dr. Bishr Omary for providing cleaved K18 antibody and The University of Michigan Cancer Center Microarray Core for Affymetrix gene array services and analysis.

This study was supported by grants to Y.M. Shah from the NIH (CA148828), The University of Michigan Gastrointestinal Peptide Center, and Jeffrey A. Colby Colon Cancer Research and the Tom Liu Memorial Funds of the University of Michigan Comprehensive Cancer Center. F.J. Gonzalez was supported by the Intramural Research Program of the National Cancer Institute.

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.

1.
Hockel
M
,
Vaupel
P
. 
Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects
.
J Natl Cancer Inst
2001
;
93
:
266
76
.
2.
Bertout
JA
,
Patel
SA
,
Simon
MC
. 
The impact of O2 availability on human cancer
.
Nat Rev Cancer
2008
;
8
:
967
75
.
3.
Semenza
GL
,
Wang
GL
. 
A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation
.
Mol Cell Biol
1992
;
12
:
5447
54
.
4.
Wang
GL
,
Semenza
GL
. 
Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia
.
J Biol Chem
1993
;
268
:
21513
8
.
5.
Wang
GL
,
Semenza
GL
. 
General involvement of hypoxia-inducible factor 1 in transcriptional response to hypoxia
.
Proc Natl Acad Sci U S A
1993
;
90
:
4304
8
.
6.
Ivan
M
,
Kondo
K
,
Yang
H
,
Kim
W
,
Valiando
J
,
Ohh
M
, et al
HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing
.
Science
2001
;
292
:
464
8
.
7.
Jaakkola
P
,
Mole
DR
,
Tian
YM
,
Wilson
MI
,
Gielbert
J
,
Gaskell
SJ
, et al
Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation
.
Science
2001
;
292
:
468
72
.
8.
Wang
GL
,
Jiang
BH
,
Rue
EA
,
Semenza
GL
. 
Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension
.
Proc Natl Acad Sci U S A
1995
;
92
:
5510
4
.
9.
Tian
H
,
McKnight
SL
,
Russell
DW
. 
Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells
.
Genes Dev
1997
;
11
:
72
82
.
10.
Loboda
A
,
Jozkowicz
A
,
Dulak
J
. 
HIF-1 and HIF-2 transcription factors–similar but not identical
.
Mol Cells
2010
;
29
:
435
42
.
11.
Gordan
JD
,
Bertout
JA
,
Hu
CJ
,
Diehl
JA
,
Simon
MC
. 
HIF-2alpha promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity
.
Cancer Cell
2007
;
11
:
335
47
.
12.
Gordan
JD
,
Lal
P
,
Dondeti
VR
,
Letrero
R
,
Parekh
KN
,
Oquendo
CE
, et al
HIF-alpha effects on c-Myc distinguish two subtypes of sporadic VHL-deficient clear cell renal carcinoma
.
Cancer Cell
2008
;
14
:
435
46
.
13.
Wen
W
,
Ding
J
,
Sun
W
,
Wu
K
,
Ning
B
,
Gong
W
, et al
Suppression of cyclin D1 by hypoxia-inducible factor-1 via direct mechanism inhibits the proliferation and 5-fluorouracil-induced apoptosis of A549 cells
.
Cancer Res
2010
;
70
:
2010
9
.
14.
Goda
N
,
Ryan
HE
,
Khadivi
B
,
McNulty
W
,
Rickert
RC
,
Johnson
RS
. 
Hypoxia-inducible factor 1alpha is essential for cell cycle arrest during hypoxia
.
Mol Cell Biol
2003
;
23
:
359
69
.
15.
Koshiji
M
,
Kageyama
Y
,
Pete
EA
,
Horikawa
I
,
Barrett
JC
,
Huang
LE
. 
HIF-1alpha induces cell cycle arrest by functionally counteracting Myc
.
EMBO J
2004
;
23
:
1949
56
.
16.
Hubbi
ME
,
Luo
W
,
Baek
JH
,
Semenza
GL
. 
MCM proteins are negative regulators of hypoxia-inducible factor 1
.
Mol Cell
2011
;
42
:
700
12
.
17.
Talks
KL
,
Turley
H
,
Gatter
KC
,
Maxwell
PH
,
Pugh
CW
,
Ratcliffe
PJ
, et al
The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages
.
Am J Pathol
2000
;
157
:
411
21
.
18.
Zhong
H
,
De Marzo
AM
,
Laughner
E
,
Lim
M
,
Hilton
DA
,
Zagzag
D
, et al
Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases
.
Cancer Res
1999
;
59
:
5830
5
.
19.
Sesink
AL
,
Termont
DS
,
Kleibeuker
JH
,
Van der Meer
R
. 
Red meat and colon cancer: the cytotoxic and hyperproliferative effects of dietary heme
.
Cancer Res
1999
;
59
:
5704
9
.
20.
Bastide
NM
,
Pierre
FH
,
Corpet
DE
. 
Heme iron from meat and risk of colorectal cancer: a meta-analysis and a review of the mechanisms involved
.
Cancer Prev Res (Phila)
2011
;
4
:
177
84
.
21.
Siegers
CP
,
Bumann
D
,
Baretton
G
,
Younes
M
. 
Dietary iron enhances the tumor rate in dimethylhydrazine-induced colon carcinogenesis in mice
.
Cancer Lett
1988
;
41
:
251
6
.
22.
Nelson
RL
,
Davis
FG
,
Sutter
E
,
Sobin
LH
,
Kikendall
JW
,
Bowen
P
. 
Body iron stores and risk of colonic neoplasia
.
J Natl Cancer Inst
1994
;
86
:
455
60
.
23.
Siegers
CP
,
Bumann
D
,
Trepkau
HD
,
Schadwinkel
B
,
Baretton
G
. 
Influence of dietary iron overload on cell proliferation and intestinal tumorigenesis in mice
.
Cancer Lett
1992
;
65
:
245
9
.
24.
Nelson
RL
. 
Iron and colorectal cancer risk: human studies
.
Nutr Rev
2001
;
59
:
140
8
.
25.
Seril
DN
,
Liao
J
,
Ho
KL
,
Warsi
A
,
Yang
CS
,
Yang
GY
. 
Dietary iron supplementation enhances DSS-induced colitis and associated colorectal carcinoma development in mice
.
Dig Dis Sci
2002
;
47
:
1266
78
.
26.
Shah
YM
,
Matsubara
T
,
Ito
S
,
Yim
SH
,
Gonzalez
FJ
. 
Intestinal hypoxia-inducible transcription factors are essential for iron absorption following iron deficiency
.
Cell Metab
2009
;
9
:
152
64
.
27.
Haase
VH
,
Glickman
JN
,
Socolovsky
M
,
Jaenisch
R
. 
Vascular tumors in livers with targeted inactivation of the von Hippel-Lindau tumor suppressor
.
Proc Natl Acad Sci U S A
2001
;
98
:
1583
8
.
28.
Taylor
M
,
Qu
A
,
Anderson
ER
,
Matsubara
T
,
Martin
A
,
Gonzalez
FJ
, et al
Hypoxia-inducible factor-2alpha mediates the adaptive increase of intestinal ferroportin during iron deficiency in mice
.
Gastroenterology
2011
;
140
:
2044
55
.
29.
Xiao
Y
,
Xue
X
,
Wu
YF
,
Xin
GZ
,
Qian
Y
,
Xie
TP
, et al
beta-Naphthoflavone protects mice from aristolochic acid-I-induced acute kidney injury in a CYP1A dependent mechanism
.
Acta Pharmacol Sin
2009
;
30
:
1559
65
.
30.
Meguro
R
,
Asano
Y
,
Iwatsuki
H
,
Shoumura
K
. 
Perfusion-Perls and -Turnbull methods supplemented by DAB intensification for nonheme iron histochemistry: demonstration of the superior sensitivity of the methods in the liver, spleen, and stomach of the rat
.
Histochem Cell Biol
2003
;
120
:
73
82
.
31.
Frisch
SM
,
Francis
H
. 
Disruption of epithelial cell-matrix interactions induces apoptosis
.
J Cell Biol
1994
;
124
:
619
26
.
32.
Folkman
J
,
Moscona
A
. 
Role of cell shape in growth control
.
Nature
1978
;
273
:
345
9
.
33.
Shah
YM
,
Ito
S
,
Morimura
K
,
Chen
C
,
Yim
SH
,
Haase
VH
, et al
Hypoxia-inducible factor augments experimental colitis through an MIF-dependent inflammatory signaling cascade
.
Gastroenterology
2008
;
134
:
2036
48
,
2048.e1-3
.
34.
Su
LK
,
Kinzler
KW
,
Vogelstein
B
,
Preisinger
AC
,
Moser
AR
,
Luongo
C
, et al
Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene
.
Science
1992
;
256
:
668
70
.
35.
Fearnhead
NS
,
Britton
MP
,
Bodmer
WF
. 
The ABC of APC
.
Hum Mol Genet
2001
;
10
:
721
33
.
36.
Cooper
HS
,
Everley
L
,
Chang
WC
,
Pfeiffer
G
,
Lee
B
,
Murthy
S
, et al
The role of mutant Apc in the development of dysplasia and cancer in the mouse model of dextran sulfate sodium-induced colitis
.
Gastroenterology
2001
;
121
:
1407
16
.
37.
Pastorelli
L
,
Garg
RR
,
Hoang
SB
,
Spina
L
,
Mattioli
B
,
Scarpa
M
, et al
Epithelial-derived IL-33 and its receptor ST2 are dysregulated in ulcerative colitis and in experimental Th1/Th2 driven enteritis
.
Proc Natl Acad Sci U S A
2010
;
107
:
8017
22
.
38.
Redston
MS
,
Papadopoulos
N
,
Caldas
C
,
Kinzler
KW
,
Kern
SE
. 
Common occurrence of APC and K-ras gene mutations in the spectrum of colitis-associated neoplasias
.
Gastroenterology
1995
;
108
:
383
92
.
39.
Jemal
A
,
Siegel
R
,
Ward
E
,
Hao
Y
,
Xu
J
,
Murray
T
, et al
Cancer statistics, 2008
.
CA Cancer J Clin
2008
;
58
:
71
96
.
40.
Vachon
PH
,
Harnois
C
,
Grenier
A
,
Dufour
G
,
Bouchard
V
,
Han
J
, et al
Differentiation state-selective roles of p38 isoforms in human intestinal epithelial cell anoikis
.
Gastroenterology
2002
;
123
:
1980
91
.
41.
Rhodes
DR
,
Chinnaiyan
AM
. 
Integrative analysis of the cancer transcriptome
.
Nat Genet
2005
;
37
Suppl
:
S31
7
.
42.
Zou
TT
,
Selaru
FM
,
Xu
Y
,
Shustova
V
,
Yin
J
,
Mori
Y
, et al
Application of cDNA microarrays to generate a molecular taxonomy capable of distinguishing between colon cancer and normal colon
.
Oncogene
2002
;
21
:
4855
62
.
43.
Skrzypczak
M
,
Goryca
K
,
Rubel
T
,
Paziewska
A
,
Mikula
M
,
Jarosz
D
, et al
Modeling oncogenic signaling in colon tumors by multidirectional analyses of microarray data directed for maximization of analytical reliability
.
PLoS One
2010
;
5
.
44.
Sabates-Bellver
J
,
Van der Flier
LG
,
de Palo
M
,
Cattaneo
E
,
Maake
C
,
Rehrauer
H
, et al
Transcriptome profile of human colorectal adenomas
.
Mol Cancer Res
2007
;
5
:
1263
75
.
45.
Gunshin
H
,
Mackenzie
B
,
Berger
UV
,
Gunshin
Y
,
Romero
MF
,
Boron
WF
, et al
Cloning and characterization of a mammalian proton-coupled metal-ion transporter
.
Nature
1997
;
388
:
482
8
.
46.
Fleming
MD
,
Trenor
CC
 3rd
,
Su
MA
,
Foernzler
D
,
Beier
DR
,
Dietrich
WF
, et al
Microcytic anaemia mice have a mutation in Nramp2, a candidate iron transporter gene
.
Nat Genet
1997
;
16
:
383
6
.
47.
Canonne-Hergaux
F
,
Gruenheid
S
,
Ponka
P
,
Gros
P
. 
Cellular and subcellular localization of the Nramp2 iron transporter in the intestinal brush border and regulation by dietary iron
.
Blood
1999
;
93
:
4406
17
.
48.
Kaelin
WG
 Jr
. 
The von Hippel-Lindau tumour suppressor protein: O2 sensing and cancer
.
Nat Rev Cancer
2008
;
8
:
865
73
.
49.
Young
AP
,
Schlisio
S
,
Minamishima
YA
,
Zhang
Q
,
Li
L
,
Grisanzio
C
, et al
VHL loss actuates a HIF-independent senescence programme mediated by Rb and p400
.
Nat Cell Biol
2008
;
10
:
361
9
.
50.
Kurban
G
,
Hudon
V
,
Duplan
E
,
Ohh
M
,
Pause
A
. 
Characterization of a von Hippel Lindau pathway involved in extracellular matrix remodeling, cell invasion, and angiogenesis
.
Cancer Res
2006
;
66
:
1313
9
.
51.
Zhou
MI
,
Wang
H
,
Ross
JJ
,
Kuzmin
I
,
Xu
C
,
Cohen
HT
. 
The von Hippel-Lindau tumor suppressor stabilizes novel plant homeodomain protein Jade-1
.
J Biol Chem
2002
;
277
:
39887
98
.
52.
Russell
RC
,
Sufan
RI
,
Zhou
B
,
Heir
P
,
Bunda
S
,
Sybingco
SS
, et al
Loss of JAK2 regulation via a heterodimeric VHL-SOCS1 E3 ubiquitin ligase underlies Chuvash polycythemia
.
Nat Med
2011
;
17
:
845
53
.
53.
Foot
NJ
,
Leong
YA
,
Dorstyn
LE
,
Dalton
HE
,
Ho
K
,
Zhao
L
, et al
Ndfip1-deficient mice have impaired DMT1 regulation and iron homeostasis
.
Blood
2011
;
117
:
638
46
.
54.
Foot
NJ
,
Dalton
HE
,
Shearwin-Whyatt
LM
,
Dorstyn
L
,
Tan
SS
,
Yang
B
, et al
Regulation of the divalent metal ion transporter DMT1 and iron homeostasis by a ubiquitin-dependent mechanism involving Ndfips and WWP2
.
Blood
2008
;
112
:
4268
75
.