Abstract
The most active vitamin D metabolite, 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3), is a pleiotropic hormone with wide regulatory actions. Classically, vitamin D deficiency was known to alter calcium and phosphate metabolism and bone biology. In addition, recent epidemiological and experimental studies support the association of vitamin D deficiency with a large variety of human diseases, and particularly with the high risk of colorectal cancer. By regulating the expression of many genes via several mechanisms, 1,25(OH)2D3 induces differentiation, controls the detoxification metabolism and cell phenotype, sensitises cells to apoptosis and inhibits the proliferation of cultured human colon carcinoma cells. Consistently, 1,25(OH)2D3 and several of its analogues decrease intestinal tumourigenesis in animal models. Molecular, genetic and clinical data in humans are scarce but they suggest that vitamin D is protective against colon cancer. Clearly, the available evidence warrants new, well-designed, large-scale trials to clarify the role of vitamin D in the prevention and/or therapy of this important neoplasia.
The vitamin D system
Vitamin D belongs to a group of steroids with a broken ring known as secosteroids. The most important forms of vitamin D are vitamin D3 (cholecalciferol), produced in the human skin; and vitamin D2 (ergocalciferol), derived from plants (Horst et al. 2005). Nowadays, vitamin D3 is not categorised as a vitamin, but rather as a prohormone, given its synthesis in the skin and the multiple systemic actions of its metabolites.
The most active metabolite of vitamin D3, 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3, calcitriol), is synthesised in a highly regulated multi-step process. When human skin is exposed to sunlight, u.v.-B irradiation causes the photolysis of provitamin D3 (7-dehydrocholesterol) to pre-vitamin D3, a thermodynamically unstable isomer that is rapidly converted into vitamin D3. This passes into the bloodstream and is transported to the liver bound to the vitamin D-binding protein (DBP, encoded by the GC gene) and, to a lesser extent, to albumin. Alternatively, vitamin D3 can be absorbed in the intestine from dietary sources (Holick 2005, Norman 2008). Vitamin D3 is first hydroxylated in the liver by several mitochondrial and microsomal vitamin D3-25-hydroxylases. The resultant 25-hydroxyvitamin D3 (25(OH)D3, calcidiol) is the main circulating form of vitamin D3, and it is further hydroxylated in the kidney by mitochondrial 25(OH)D3-1α-hydroxylase (encoded by the gene CYP27B1), yielding the hormonally active form 1,25(OH)2D3 (Lamprecht & Lipkin 2003, Deeb et al. 2007). Importantly, CYP27B1 is also expressed in other human tissues and cell types, such as colon, brain, prostate, endothelial and immune cells, suggesting an extrarenal production of 1,25(OH)2D3 with paracrine/autocrine action (Townsend et al. 2005, Norman & Bouillon 2010). The rate-limiting step in vitamin D3 metabolism is the transformation of 25(OH)D3 and 1,25(OH)2D3 to the less active compounds 24,25(OH)2D3 and 1,24,25(OH)3D3 respectively through 24-hydroxylation by the ubiquitous CYP24A1 enzyme (Deeb et al. 2007).
Mechanism of action of 1,25(OH)2D3
1,25(OH)2D3 exerts its biological effects by binding to the vitamin D receptor (VDR), which is a member of the nuclear receptor super-family of transcription factors, and regulates gene expression in a ligand-dependent manner (Deeb et al. 2007, Thorne & Campbell 2008). VDR dimerises with another member of the same family, the retinoid X receptor (RXR), whose ligand is 9-cis-retinoic acid or, at least in the mouse brain, docosahexaenoic acid (Heyman et al. 1992, Levin et al. 1992, de Urquiza et al. 2000). The VDR–RXR heterodimer binds to specific sequences of its target genes termed vitamin D response elements (VDREs). Curiously, studies on a few genes indicate a variable effect (additive, synergistic or none) of 9-cis-retinoic acid on the action of 1,25(OH)2D3. Results from gel-shift and transactivation assays using DNA fragments close to transcription start sites (TSS) suggested that VDREs usually comprise two hexanucleotide direct repeats separated by three or four nucleotides (AGTTCAnnnAGTTCA, being n any nucleotide; DR3 or DR4) or two everted repeats inter-spaced by six to nine nucleotides (TGAACTnnnnnnAGTTCA; ER6–ER9) (Carlberg & Dunlop 2006, Carlberg & Seuter 2009).
However, this setting appears to be more complex in the light of the three chromatin immunoprecipitation sequencing (ChIP-Seq) studies published so far using human Epstein–Barr virus-transformed lymphoblastoid, THP-1 monocytic leukaemia and LS180 colon cancer cells (Ramagopalan et al. 2010, Heikkinen et al. 2011, Meyer et al. 2012). Although performed in different cell types and using different durations and doses of 1,25(OH)2D3 treatment, these genome-wide studies coincide in concluding that there are hundreds (623, 520 or 262, depending on the study) of genomic VDR-binding sites in the absence of 1,25(OH)2D3, which increases to thousands (2776, 1820 or 2209) after 1,25(OH)2D3 treatment. As expected due to the different conditions analysed, only a few of the identified VDR-binding sites overlap in the three studies (see also Carlberg et al. (2012)). Common findings were also the presence of most VDR-binding sites far from the TSS, the preference for the DR3-type in those located close to the TSS, and the shift from non-DR3 type to DR3-type upon ligand activation (Carlberg et al. 2012, Meyer et al. 2012). These studies revealed that though the DR3-type is the most common VDR-binding site, it represents <30% of all peaks, and that a high proportion of the peaks do not contain any classical VDRE. Very few genes have only one VDR-binding site: more complex scenarios are common, such as genes with multiple VDR-binding sites, two closely located target genes sharing one or more VDR-binding sites or even a cluster of target genes containing each of these multiple VDR-binding sites (Carlberg et al. 2012). In line with previous data, in LS180 cells RXR co-occupied a high proportion of VDR-binding sites, which were most commonly of the DR3-type (Meyer et al. 2012). Curiously, in this system, 1073 (out of 1674) 1,25(OH)2D3-induced VDR–RXR-binding sites are located within repetitive DNA (long interdispersed nuclear elements) (Meyer et al. 2012). The characterisation of the functional relevance of data from ChIP-Seq studies will require the combination with transcriptomic results and subsequent validation analyses.
The prevailing model for the mechanism of action of 1,25(OH)2D3/VDR predicts that in the absence of 1,25(OH)2D3 the VDR–RXR heterodimer is bound to VDREs and to transcriptional corepressors, such as nuclear corepressors (NCoRs) and the silencing mediator of retinoid and thyroid hormone receptors (SMRT), that recruit complexes with histone deacetylase (HDAC) activity and maintain the chromatin in a transcriptionally repressed state. In this model, 1,25(OH)2D3 binding to VDR induces conformational changes in VDR that causes the release of corepressors and binding of co-activators, such as steroid receptor co-activators and the CBP-p300 coregulator, and chromatin remodelers such as switch-sucrose non-fermenting (SWI-SNF), that acetylate nucleosome histones to untie DNA for transcription. Once the chromatin is derepressed, binding of the VDR-interacting protein 205 (DRIP205) to the VDR attracts other components of the DRIP–TRIP complex, which acts as a bridge with RNA polymerase II and the transcriptional machinery, thus permitting the initiation of transcription (Haussler et al. 1998, Deeb et al. 2007, Carlberg & Seuter 2009, Pike & Meyer 2010).
The mechanism by which 1,25(OH)2D3 represses gene expression is less understood, although this process is probably equally important for its action, as around one-third of the target genes are inhibited (Pálmer et al. 2003, Heikkinen et al. 2011). Several possibilities have been proposed, such as direct VDRE-dependent repression, competition for DNA binding between VDR and other transcription factors or indirect repression via intermediate proteins (Kato et al. 2007, Kouzmenko et al. 2011). The best characterised transcriptional repression mechanism was elucidated for the CYP27B1 gene. In this case, the VDR–RXR heterodimer represses gene transcription in a 1,25(OH)2D3-dependent manner through E-box-type negative VDREs (nVDREs: CANNTG). When bound to nVDREs, the VDR-interacting repressor (VDIR, also known as TCF3 or E47) induces the transcriptional activation of the CYP27B1 gene. However, upon ligand binding VDR interacts with VDIR, promoting the dissociation of co-activators and the recruitment of corepressors and complexes with HDAC activity, which results in gene repression (Murayama et al. 2004, Kouzmenko et al. 2011). In addition, the possibility that 1,25(OH)2D3 represses gene expression via the up-regulation of microRNAs (miRNAs) has recently emerged (Gocek et al. 2011, Thorne et al. 2011, Álvarez-Díaz et al. 2012). miRNAs are short non-coding RNAs that bind to the 3′-untranslated region of target mRNAs causing mRNA degradation and/or translational inhibition (Fabian et al. 2010). Thus, the induction of miRNAs constitutes a novel mechanism of gene repression by 1,25(OH)2D3.
Non-genomic actions of 1,25(OH)2D3 have also been described. They are rapid and transcription-independent effects that include the opening of ion channels and the activation or inhibition of kinases, phosphatases and phospholipases, and are probably relevant for the biological effects of 1,25(OH)2D3 (Norman et al. 2004, Haussler et al. 2011). Thus, in SW480-ADH colon cancer cells and in other cell types, 1,25(OH)2D3 causes a rapid increase in the cytosolic calcium concentration followed by the transient activation of the RhoA small GTPase, its effector ROCK and the p38MAPK and MSK-1 kinases that is required for the induction of its target genes (Ordóñez-Morán et al. 2008, 2010). This shows the convergence of non-genomic and genomic 1,25(OH)2D3 effects, at least in this system (Ordóñez-Morán & Muñoz 2009).
1,25(OH)2D3 is a pleiotropic hormone
The most widely and classically accepted physiological role for vitamin D, mediated mostly by 1,25(OH)2D3, is the regulation of calcium and phosphate homeostasis and bone mineralisation (Holick 2007, Verstuyf et al. 2010). 1,25(OH)2D3 enhances intestinal calcium absorption in the small intestine and affects the absorption of dietary phosphate possibly by increasing its influx through the enterocyte brush border membrane (Ajibade et al. 2010). In concert with these effects, 1,25(OH)2D3 induces the maturation of pre-osteoclasts into osteoclasts, which remove calcium and phosphate from bone (Holick 2007, Verstuyf et al. 2010).
In the past decades, evidence has emerged that the importance of 1,25(OH)2D3 for health extends far beyond bone. The discovery in 1981 that 1,25(OH)2D3 induces the differentiation of myeloid leukaemia cells and inhibits the growth of melanoma cells triggered the interest of cancer researchers (Abe et al. 1981, Colston et al. 1981). Since then, numerous observations have indicated a much broader range of action for 1,25(OH)2D3, including the regulation of cell differentiation, proliferation, apoptosis, invasion and angiogenesis in several types of tumour cells and animal models of cancer (reviewed in Ordóñez-Morán et al. (2005), Deeb et al. (2007) and Krishnan & Feldman (2011)). These 1,25(OH)2D3 actions suggest a potential therapeutic role against hyper-proliferative disorders such as cancer. Nevertheless, the use of 1,25(OH)2D3 is restricted by its hypercalcaemic effect at therapeutic doses. This can be putatively overcome by the use of analogues that retain the anti-tumoural action but have less calcaemic effect. Currently, there are numerous clinical trials ongoing using 1,25(OH)2D3 or its analogues, alone or in combination with other drugs, against several neoplasias (www.clinicaltrials.gov) (Deeb et al. 2007).
Vitamin D and colon cancer: observational and epidemiological studies
According to the International Agency for Research on Cancer (IARC) of the World Health Organization (WHO), about 12.7 million cancer cases and 7.6 million cancer deaths worldwide occurred in 2008. Colorectal cancer (CRC) is the third most frequent malignancy and the fourth leading cause of death from cancer worldwide, with 1 233 000 cases diagnosed and 608 000 deaths in 2008 (Ferlay et al. 2010).
The role of vitamin D in CRC prevention was first hypothesised by Frank and Cedric Garland based on ecological studies (Garland & Garland 1980). These authors proposed that the inverse relation between solar radiation (latitude) and CRC mortality and incidence in USA might be due to vitamin D synthesis. Later, the same authors reported an inverse association between vitamin D status and CRC in the USA (Garland et al. 1989). Since then, numerous studies have addressed the relationship between vitamin D status and colorectal adenoma and carcinoma incidence or mortality (Platz et al. 2000, Wu et al. 2007, Gandini et al. 2011). However, the integration and the overall analysis of all epidemiological studies is complex and thwarted by the fact that many of these studies do not take into account endogenous vitamin D production from the sun exposure and are limited by measurement error from the various dietary assessment methods and food composition tables used to calculate dietary intake. Such limitations can be overcome by measuring circulating 25(OH)D concentration, a biomarker that provides an overall estimate of vitamin D status (both from endogenous production and from dietary intake). Nonetheless, the effective colonic 1,25(OH)2D3 concentration is determined not only by 25(OH)D and 1,25(OH)2D3 serum levels but also by their uptake by the cell and by the expression and activity of CYP27B1 and CYP24A1 enzymes in the colon (Cross et al. 2011). In fact, it has been reported that the expression/activity of these enzymes is regulated by several factors (17β-estradiol, phytoestrogens, folate, calcium, 1,25(OH)2D3 itself, etc.) (Lechner et al. 2005, Cross et al. 2011) and also during colon cancer progression (discussed below). Therefore, several conditions may affect colonic 1,25(OH)2D3 levels and provoke that they do not correlate with 25(OH)D serum levels in some individuals.
The substantial body of literature that examines the relationship between 25(OH)D level and CRC risk generally supports an inverse association. A prospective study in the USA with 16 818 participants found that CRC mortality was inversely related to serum 25(OH)D level (Freedman et al. 2007). A similar association was obtained in a case–control study that included an ethnically diverse population of Japanese, Latino, African-American, Caucasian and Native Hawaiian participants (Woolcott et al. 2010). Accordingly, a large observational nested case–control study (520 000 participants from ten Western European countries) conducted within the European Prospective Investigation into Cancer and Nutrition concluded that there is a strong inverse association between 25(OH)D concentration and CRC (Jenab et al. 2010). A recent meta-analysis of 35 independent studies confirmed a consistent inverse relationship between serum 25(OH)D levels and CRC risk (Gandini et al. 2011). Ma et al. (2011) conducted a systematic review of 18 prospective studies assessing the association of vitamin D intake or serum levels of 25(OH)D and the risk of CRC in ∼1 000 000 individuals and found an inverse correlation between CRC risk and both serum 25(OH)D and vitamin D intake.
Accepting the problems of using different approaches and techniques to measure vitamin D status, the population diversity and the potential presence of confounding factors, the consistency of the association between vitamin D status and CRC risk indicates a causal association (Giovannucci 2011). Accordingly, the IARC stated that the epidemiological evidence for a relationship between serum 25(OH)D levels and the incidence of CRC is consistent and persuasive. However, such a causal relationship awaits large, adequate prospective clinical trials (IARC 2008).
Anti-tumoural actions of 1,25(OH)2D3 in CRC cells
CRC arises as a consequence of the progressive accumulation of genetic and epigenetic alterations that induce the transformation and malignant progression of normal colonic epithelial cells. The initial genetic change in most colorectal adenomas, the earliest step in CRC, is the somatic mutation in the tumour suppressor gene APC (or, to a lesser extent, AXIN2 or CTNNB1/β-catenin genes). Each of these mutually exclusive mutations leads to aberrant activation of the canonical WNT/β-catenin signalling pathway (Clevers 2006, Klaus & Birchmeier 2008). In addition, a large proportion of adenomas carry activating mutations in KRAS or BRAF, whereas inactivating mutations in the transforming growth factor (TGF)-β pathway (mainly in SMAD2, SMAD4 or TGFBR2) confer additional malignant features to adenoma cells (Markowitz & Bertagnolli 2009). The inactivation of the TP53 tumour suppressor gene matches the adenoma–carcinoma transition in around 50% of CRC tumours (Iacopetta 2003). The molecular mechanisms of carcinoma progression and the acquisition of metastatic ability remain to be fully elucidated. Recent studies indicate that the epithelial–mesenchymal transition (EMT) endows epithelial cells with the capacity to invade the surrounding tissue and then to migrate to distant organs (Thiery et al. 2009, Chaffer & Weinberg 2011).
Numerous studies have shown that the anti-tumoural action of 1,25(OH)2D3 in CRC relies on several mechanisms at the cellular level, such as inhibition of cell proliferation, sensitisation to apoptosis, induction of epithelial differentiation and cell detoxification metabolism and inhibition of angiogenesis (Fig. 1). The combined effect of these mechanisms, in a cell-type- and cell-context-dependent manner, may determine the anti-tumoural action of 1,25(OH)2D3 (reviewed in Lamprecht & Lipkin (2003), Deeb et al. (2007), Larriba et al. (2008) and Krishnan & Feldman (2011)). A detailed list of the most important 1,25(OH)2D3 target genes in human CRC cells and their role in the anti-tumoural actions of the hormone is shown in Table 1.
1,25(OH)2D3 anti-tumoural actions and target genes in human CRC cells
Proliferation
The growth-inhibitory effect of 1,25(OH)2D3 on CRC cells results from the accumulation of cells in the G0/G1 phase of the cell cycle. 1,25(OH)2D3 induces the expression of the cyclin-dependent kinase inhibitors p21CIP1 and p27KIP1, and represses that of cyclin A and cyclin F (Evans et al. 1999, Gaschott et al. 2002, Pálmer et al. 2003, Fernández-García et al. 2005). A recent report by Liu et al. (2010) indicates that the induction of p21CIP1 and the inhibition of proliferation by 1,25(OH)2D3 in CRC cell lines are dependent on the expression of the calcium-sensing receptor, which is in turn induced by 1,25(OH)2D3 (Chakrabarty et al. 2005). 1,25(OH)2D3 also induces the expression of the growth-arrest and DNA damage 45α (GADD45A) gene (Pálmer et al. 2003), which is involved in cell cycle arrest after DNA damage and is required for the maintenance of genomic stability. In addition, 1,25(OH)2D3 regulates many other genes related to proliferation, including MYC, FOS and JUN (Pálmer et al. 2001, 2003, Meyer et al. 2012). The c-MYC oncogene is deregulated and over-expressed in most cancers, and its down-regulation by 1,25(OH)2D3 occurs directly, via multiple VDREs located in the promoter and intragenic regions, and indirectly, by increasing the expression and promoting the binding of intermediate proteins to regulatory regions of the gene (Pan et al. 1996, Pan & Simpson 1999, Toropainen et al. 2010, Meyer et al. 2012). As MYC is a target of the WNT/β-catenin pathway, another indirect mechanism of down-regulation of this gene by 1,25(OH)2D3 is the antagonism of this signalling pathway (Pálmer et al. 2001, discussed below).
Apoptosis
Apoptosis sensitisation by 1,25(OH)2D3 in colorectal adenoma and carcinoma cells involves the up-regulation of the proapoptotic protein BAK1 and the down-regulation of the nuclear anti-apoptotic protein BAG1 (Díaz et al. 2000, Barnes et al. 2005). The effect of 1,25(OH)2D3 on the expression of other proapoptotic (BAX) or anti-apoptotic (BCL-2, BCL-XL) proteins, which are regulated by 1,25(OH)2D3 in other cell systems, is not clear in CRC, and appears to be cell-specific (Díaz et al. 2000, Hansen et al. 2001). In addition, 1,25(OH)2D3 induces the expression of G0S2 (Pálmer et al. 2003), a mitochondrial protein that interacts with BCL-2 and induces apoptosis in CRC cells by preventing BCL-2 from forming anti-apoptotic heterodimers with BAX (Welch et al. 2009). Although previous reports indicated that the apoptosis potentiated by 1,25(OH)2D3 does not require an intact TP53 tumour suppressor gene (Díaz et al. 2000, Hansen et al. 2001), a recent study by Stambolsky et al. (2010) has revealed that mutant p53 protein interacts with VDR and modulates the transcriptional activity of 1,25(OH)2D3 leading to the increase of the expression of survival-promoting genes and the decrease of that of proapoptotic genes, thus converting 1,25(OH)2D3 into an anti-apoptotic agent. In CRC cells, 1,25(OH)2D3 promotes sensitivity to the chemotherapeutic agent 5-fluorouracil by down-regulating the expression of survivin, an anti-apoptotic protein, and that of thymidylate synthase, an enzyme involved in DNA de novo synthesis and the molecular target of 5-fluorouracil (Liu et al. 2010). Moreover, in an in vitro model developed to evaluate the cross talk between tumour-associated macrophages and CRC cells, 1,25(OH)2D3 restores the sensitivity of CRC cells to TRAIL-induced apoptosis by interfering with the release of IL1β by macrophages (Kaler et al. 2010).
Differentiation
1,25(OH)2D3 has specific and multiple prodifferentiation effects in CRC cells. It increases the expression and/or activity of several brush border enzymes, such as alkaline phosphatase and maltase, and enhances the formation of microvilli (Brehier & Thomasset 1988, Giuliano et al. 1991, Halline et al. 1994, Díaz et al. 2000). In addition, 1,25(OH)2D3 increases the expression of several components of cell adhesion structures that are essential for the maintenance of the epithelial phenotype: i) occludin, zonula occludens (ZO)-1 and -2, and claudin-2 and -12 in tight junctions (Pálmer et al. 2001, Fujita et al. 2008); ii) E-cadherin in adherens junctions (Pálmer et al. 2001); and iii) plectin in hemidesmosomes (Pálmer et al. 2003). Moreover, 1,25(OH)2D3 induces the expression of proteins associated with the actin cytoskeleton and intermediate filaments such as keratin-13, vinculin and filamin A (Pálmer et al. 2001, 2003). Filamin A has been implicated in cell motility, adhesion and invasion (Nakamura et al. 2011). However, novel functions of filamin A have recently been described within the cell nucleus, such as the regulation of nuclear shape during EMT and of DNA double-strand break repair (Yue et al. 2009, Gay et al. 2011).
Angiogenesis
The angiogenic capacity of CRC cells may be affected by 1,25(OH)2D3, as it represses the expression and transcriptional activity of hypoxia-inducible factor (HIF)-1α, a key transcription factor involved in hypoxia-induced angiogenesis (Ben-Shoshan et al. 2007). In addition, 1,25(OH)2D3 regulates the expression of vascular endothelial growth factor (VEGFA) and thrombospondin-1 (THBS1), two major opposing factors that control tumour angiogenesis, leading to a balanced change in the angiogenic potential of SW480-ADH cells (Fernández-García et al. 2005). Further, 1,25(OH)2D3 strongly represses DICKKOPF-4 (DKK4), a weak WNT antagonist that promotes invasion and angiogenesis in cultured CRC cells and is up-regulated in human colon tumours (Pendás-Franco et al. 2008).
Detoxification
Intestinal epithelial cells express a variety of enzymes involved in the detoxification and metabolism of xenobiotics from dietary sources and of deleterious endobiotic compounds that may contribute to CRC development. Phase I enzymes, most of which belong to the cytochrome P450 (CYP) super-family, initiate the enzymatic detoxification of these compounds by oxidation, hydrolysis or reduction (Kaminsky & Zhang 2003). In CRC cells, 1,25(OH)2D3 induces the expression of CYP3A4, the member of the CYP super-family most expressed in the intestine (Thummel et al. 2001, Thompson et al. 2002, Pfrunder et al. 2003). Likewise, several members of the CYP2C sub-family contain VDREs in their promoters and are regulated by 1,25(OH)2D3 in hepatic tissues. As CYP2C is the second most abundant CYP sub-family in the human intestine, its putative regulation by 1,25(OH)2D3 in this tissue could be highly relevant (Drocourt et al. 2002, Chen & Goldstein 2009).
Phase II enzymes conjugate different charged groups to the compounds previously modified by phase I enzymes, which increases their solubility in water and facilitate their excretion (Kaminsky & Zhang 2003). In CaCo-2 cells, 1,25(OH)2D3 activates the transcription of SULT2A1, a phase II sulphotransferase that acts preferentially on hydroxysteroids and cholesterol-derived sterol bile acids, but may also modify several drugs and other xenobiotics (Echchgadda et al. 2004).
In addition, 1,25(OH)2D3 regulates the expression of several transporters responsible for the efflux of phase II conjugates to the extracellular medium. One of them is the drug efflux pump multi-drug resistance protein (MDR)-1, which is located in the enterocyte apical membrane (Pfrunder et al. 2003, Aiba et al. 2005, Fan et al. 2009). 1,25(OH)2D3 also modulates the expression of members of the multi-drug resistance-associated protein (MRP) family, which are involved in the transport of conjugated and unconjugated bile acids, and also in that of glutathione and sulphate conjugates (McCarthy et al. 2005, Fan et al. 2009).
Thus, 1,25(OH)2D3 has a wide-ranging effect on the expression of several proteins involved in intestinal detoxification, which is consistent with a preventive and therapeutic action of this hormone against CRC.
Interference with the WNT/β-catenin pathway and inter-play with other signalling pathways
1,25(OH)2D3 interferes with several signalling pathways, which may partially mediate its anti-tumoural effects. Results from our group and others have demonstrated that 1,25(OH)2D3 inhibits the WNT/β-catenin pathway and the activation of its target genes in CRC cells, which contributes to the inhibition of cell proliferation and to the maintenance of the differentiated phenotype (Pálmer et al. (2001) and reviewed in González-Sancho et al. (2011)). 1,25(OH)2D3 inhibits the WNT/β-catenin pathway through several mechanisms. First, it rapidly increases the amount of VDR bound to β-catenin, thus reducing the interaction between β-catenin and the transcription factors of the TCF/LEF family and leading to the repression of its target genes (Pálmer et al. 2001). Shah et al. (2006) confirmed the VDR/β-catenin interaction and characterised the protein domains involved, while Egan et al. (2010) reported that wild-type APC enhances the inhibition of β-catenin/TCF transcriptional activity by 1,25(OH)2D3. Second, 1,25(OH)2D3 induces β-catenin nuclear export linked to E-cadherin accumulation at the plasma membrane adherens junctions (Pálmer et al. 2001). Third, it induces the expression of DKK1, an extracellular WNT inhibitor (Aguilera et al. 2007). In addition, 1,25(OH)2D3 impedes the production of IL1β by THP-1 monocytic leukaemia cells and, if extrapolated to tumour-associated macrophages, this would represent another mechanism of inhibition of β-catenin/TCF-dependent gene activation in CRC cells, as IL1β inhibits β-catenin phosphorylation and labelling for degradation by GSK-3β (Kaler et al. 2009). Thus, 1,25(OH)2D3 exerts a complex set of regulatory actions leading to the inhibition of the WNT/β-catenin pathway. As this pathway is aberrantly activated in most adenomas and colorectal tumours and is considered the main driving force in this neoplasia, its inhibition is probably crucial for the anti-tumoural action of 1,25(OH)2D3 in CRC.
Additionally, 1,25(OH)2D3 sensitises CRC cells to the growth inhibitory action of TGF-β by up-regulating the expression of its type I receptor (TGFBR1) (Chen et al. 2002). Moreover, 1,25(OH)2D3 inhibits the stimulation of cell proliferation by EGF via the reduction of EGF receptor (EGFR) expression and the induction of its internalisation upon ligand binding (Tong et al. 1999). 1,25(OH)2D3 may also inhibit EGFR activity by inducing the expression of E-cadherin (Pálmer et al. 2001) and by repressing that of SPROUTY-2 (Barbáchano et al. 2010, see below), which are respectively negative and positive regulators of EGFR activity (Andl & Rustgi 2005, Cabrita & Christofori 2008). Likewise, 1,25(OH)2D3 interferes with the growth-stimulatory effect of insulin growth factor 2 (IGF2) through the inhibition of IGF2 secretion and the modulation of the expression of several genes encoding IGF-binding proteins (IGFBP; Oh et al. 2001, Pálmer et al. 2003).
Immunomodulation
1,25(OH)2D3 has the potential to be an important regulator of the immune system in the colon as epithelial and immune cells of the gut mucosa express VDR and CYP27B1 (Verstuyf et al. 2010). Several studies have shown that 1,25(OH)2D3 and VDR play a role in the innate immunity against bacterial infections in the colon. It has been observed that bacterial infiltration of the colon is higher in vitamin D-deficient animals than in controls (Lagishetty et al. 2010). Accordingly, in CRC cells 1,25(OH)2D3 induces the expression of cathelicidin anti-microbial peptide in a VDRE-dependent mechanism (Gombart et al. 2005) and VDR mediates the up-regulation of the anti-microbial peptide β-defensin-2 by dietary sulphoraphane (Schwab et al. 2008). Besides their anti-microbial activity, these peptides exert other functions such as chemoattraction of immune cells, degranulation of mast cells, stimulation of wound vascularisation and induction of cancer cells apoptosis and cytotoxicity (Kamysz et al. 2003, Oppenheim et al. 2003, Okumura et al. 2004, De Smet & Contreras 2005).
Several evidences suggest that 1,25(OH)2D3 could play a role in the prevention and therapy of inflammatory bowel disease (IBD), and thus may also reduce the risk of colitis-associated CRC. Cyp27b1 knockout mice are more susceptible to colitis, which is associated with increased levels of IL1 and 17 in the colon (Liu et al. 2008). In addition, vitamin D deficiency accelerates the development of the IBD symptoms that spontaneously occur in Il10 knockout mice (Cantorna et al. 2000, 2004). Accordingly, administration of 1,25(OH)2D3 to these mice inhibits the TNF-α pathway and significantly prevents disease progression and alleviates the symptoms of established IBD (Cantorna et al. 2000, 2004, Zhu et al. 2005). Additionally, work by Froicu et al. (2006) showed that VDR is required to control gastrointestinal inflammation in Il10 knockout mice, as Il10/Vdr double knockout mice develop exacerbated IBD symptoms associated with high local expression of IL2, IFN-γ, IL1β, TNF-α and IL12. In line with this, 1,25(OH)2D3 was shown to antagonize the NF-κB pathway and the production of IL8 in CRC cells treated with IL1β (Sun et al. 2008). These results suggest that 1,25(OH)2D3/VDR signalling has a potent anti-inflammatory action in vitro and in vivo that may influence the colonic inflammatory scenario that pre-dispose to the development of CRC.
1,25(OH)2D3 target genes in CRC cells
The development of high-throughput techniques for gene expression profiling, such as microarrays, has allowed a more detailed study of 1,25(OH)2D3 effects in the transcriptome of cancer cells (reviewed in Kriebitzsch et al. (2009)). We and others performed oligonucleotide microarrays in CRC cells treated with 1,25(OH)2D3 (Pálmer et al. 2003, Wood et al. 2004). Our results showed that around two-thirds of 1,25(OH)2D3 target genes are up-regulated, while one-third of these genes are down-regulated after 1,25(OH)2D3 treatment. A high proportion of the regulated genes are related to transcription, cell adhesion and metabolism, although genes related to many different cell functions were also found (Pálmer et al. 2003). In the past decade, our laboratory has dedicated much effort to characterizing the role of several of these genes in the anti-tumoural action of 1,25(OH)2D3 in CRC.
One of the genes regulated by 1,25(OH)2D3 in our transcriptomic analysis was CST5, encoding cystatin D, an inhibitor of cysteine proteases of the cathepsin family. We validated the strong induction of cystatin D by 1,25(OH)2D3 and found that it mediates the anti-proliferative and prodifferentiation effects of 1,25(OH)2D3 in CRC cells (Álvarez-Díaz et al. 2009). Cystatin D regulation by 1,25(OH)2D3 also occurs in xenografted mice treated with the 1,25(OH)2D3 analogue EB1089/Seocalcitol, and a direct correlation between cystatin D and VDR protein levels was observed in human CRC biopsies, suggesting that 1,25(OH)2D3 also regulates cystatin D in vivo (Álvarez-Díaz et al. 2009). In addition, ectopic cystatin D inhibits proliferation, migration and anchorage-independent growth of cultured CRC cells and growth of xenografted tumours in mice. Moreover, cystatin D expression is down-regulated during colorectal tumourigenesis and is associated with tumour dedifferentiation. All together, these data led us to propose CST5 as a candidate tumour suppressor gene in CRC (Álvarez-Díaz et al. 2009). The mechanism by which this protease inhibitor mediates its actions is not fully understood. Experiments using mutant cystatin D proteins with reduced anti-proteolytic activity indicate that at least some of its anti-tumour effects may be independent of cathepsin inhibition (Álvarez-Díaz et al. 2009). In line with this, 1,25(OH)2D3 regulates a number of genes related to the protein degradation machinery, such as proteases, other protease inhibitors, and components of the ubiquitin–proteasome system (Álvarez-Díaz et al. 2010).
SPRY2 is another gene shown by transcriptomic assays to be regulated by 1,25(OH)2D3 (5.6-fold repression at 4 h of treatment) (Pálmer et al. 2003). SPRY2 encodes SPROUTY-2, an intracellular modulator of EGFR and other growth factor tyrosine kinase receptors involved in the regulation of cell growth, migration and angiogenesis (Cabrita & Christofori 2008). SPROUTY-2 strongly inhibits the inter-cellular adhesion induced by 1,25(OH)2D3, and gain- and loss-of-function experiments showed that SPROUTY-2 and E-cadherin repress reciprocally in CRC cell lines and have opposite actions on cell differentiation (Barbáchano et al. 2010). Accordingly, the expressions of SPROUTY-2 and E-cadherin are mutually exclusive in xenografted tumours and correlate inversely in human CRC biopsies, where SPROUTY-2 is up-regulated in undifferentiated high-grade tumours and at the invasive front of low-grade carcinomas (Barbáchano et al. 2010). Therefore, SPROUTY-2 is inhibited by 1,25(OH)2D3 and is a potential novel marker of malignancy and a therapeutic target in CRC.
Several candidate 1,25(OH)2D3 target genes encode for proteins implicated in epigenetic regulation of gene expression. We have recently reported that 1,25(OH)2D3 induces the expression of the JMJD3 (KDM6B) gene (Pereira et al. 2011), which codes for a histone demethylase of the trimethyl repressive mark on lysine 27 of histone H3, and which may have an important tumour suppressor function (De Santa et al. 2007, Agger et al. 2009). JMJD3 partially mediates the prodifferentiation, anti-proliferative and gene regulatory actions of 1,25(OH)2D3, and also its antagonism of the WNT/β-catenin pathway (Pereira et al. 2011). Moreover, JMJD3 depletion up-regulates the expression of EMT inducers and mesenchymal markers, while it down-regulates that of epithelial proteins. Importantly, and in line with data in cultured cells, the expression of JMJD3 RNA was lower in tumour tissue than in normal tissue in 56% of human colorectal tumours and directly correlates with that of VDR (Pereira et al. 2011). Recent data indicate that 1,25(OH)2D3 modulates the expression of several genes coding for histone demethylases of the Jumonji C domain and lysine-specific demethylase families (Pereira et al. 2012). These findings link chromatin epigenetic regulation with the transcriptional activity of 1,25(OH)2D3.
In addition, our group has recently performed an miRNA-specific microarray study to identify miRNAs under the control of 1,25(OH)2D3 in SW480-ADH cells. We have found that several miRNA species are up-regulated or down-regulated by 1,25(OH)2D3 treatment (Álvarez-Díaz et al. 2012). miR-22 (MIR22) is induced by 1,25(OH)2D3 in a time-, dose- and VDR-dependent manner and is required for the anti-proliferative and anti-migratory effects of 1,25(OH)2D3 in colon cancer cells (Álvarez-Díaz et al. 2012). Consistently, miR-22 expression is lower in tumour than in the matched normal tissue in 78% CRC patients and correlates directly with VDR RNA expression, suggesting that miR-22 is also a 1,25(OH)2D3 target in vivo and may contribute to the anti-tumoural action of 1,25(OH)2D3 in human colon cancer (Álvarez-Díaz et al. 2012).
The new generation of ChIP-Seq studies mentioned above are expected to contribute to our understanding of 1,25(OH)2D3 actions on a genome-wide scale, and to the identification of its target genes. In their work, Meyer et al. (2012) report new genes activated by 1,25(OH)2D3 in LS180 colon cancer cells, such as several members of the peptidylarginine deiminase family, a class of enzymes that convert histone methylated residues of arginine to citrulline (Klose & Zhang 2007), thus expanding the number of epigenetic modifiers that are regulated by 1,25(OH)2D3 in CRC cells.
Proteomic approaches have also identified 1,25(OH)2D3-regulated proteins in CRC cells. Our laboratory has used 2D-DIGE electrophoresis combined with MALDI-TOF-TOF mass spectrometry to identify nuclear proteins regulated by 1,25(OH)2D3 in SW480-ADH cells (Cristobo et al. 2011). Interestingly, several of these proteins (SFPQ, SMARCE, KHSRP, TARDBP and PARP1) are involved in the regulation of the spliceosome machinery, suggesting a role of 1,25(OH)2D3 in alternative splicing, which is frequently altered in cancer (David & Manley 2011, Miura et al. 2011). In addition, 1,25(OH)2D3 also regulates the expression of a few cytoskeletal and actin-binding proteins (EZR, RDX and CORO1C), some of which perform transcription-related functions in the cell nucleus in addition to modulating cell morphology and adhesion (Cristobo et al. 2011).
Mechanisms of resistance to 1,25(OH)2D3 action in CRC
CRC cell responsiveness to 1,25(OH)2D3 depends mainly on the expression of VDR and on the bioavailability of 1,25(OH)2D3 within the cell. The intracellular level of 1,25(OH)2D3 is determined by the circulating levels of 25(OH)D3 and 1,25(OH)2D3, and by the activity of CYP27B1 and CYP24A1 within the cell. CYP27B1 expression and activity are up-regulated at early stages of colorectal tumourigenesis and drastically decreased in advanced CRC, while those of CYP24A1 are elevated in colorectal carcinomas. These changes lead to attenuated 1,25(OH)2D3 synthesis and accelerated 1,25(OH)2D3 catabolism in advanced CRC, causing resistance to 1,25(OH)2D3 (Bareis et al. 2001, Cross et al. 2001, Bises et al. 2004). As discussed above, several compounds (17β-estradiol, phytoestrogens, folate, calcium, etc.) are known to up-regulate the colonic expression and activity of CYP27B1 and/or to inhibit those of CYP24A1 (Lechner et al. 2005, Cross et al. 2011). In addition, several CYP24A1 inhibitors have been developed to prolong 1,25(OH)2D3 biological lifetime (Schuster et al. 2003). The use of these compounds in combination with 1,25(OH)2D3 or its analogues could circumvent the 1,25(OH)2D3 resistance caused by the aberrant expression and/or activity of these enzymes.
Several studies have shown that VDR is expressed by normal and certain tumour colon epithelial cells (Sheinin et al. 2000, González-Sancho et al. 2006, Modica et al. 2010) and is associated with a high degree of cell differentiation (Shabahang et al. 1993, Zhao & Feldman 1993). VDR expression is enhanced in early stages of colorectal tumourigenesis (adenomas, polyps), whereas it decreases in advanced stages (Sheinin et al. 2000, Cross et al. 2001, Larriba & Muñoz 2005, Matusiak et al. 2005, Anderson et al. 2006). Accordingly, elevated VDR expression is associated with high tumour differentiation, absence of node involvement and good prognosis in CRC (Cross et al. 1996, Evans et al. 1998).
VDR down-regulation causes unresponsiveness to 1,25(OH)2D3 and failure of the therapy with 1,25(OH)2D3 or its analogues. For this reason, our group has searched for mechanisms to explain the loss of VDR expression in advanced CRC. We found that SNAIL1, a repressor of E-cadherin and a strong EMT inducer, represses the expression of VDR and blocks 1,25(OH)2D3 anti-tumoural actions in CRC cells in culture and such cells xenografted in mice (Pálmer et al. 2004, Larriba et al. 2007). Moreover, SNAIL1 is up-regulated in ∼60% of colorectal tumours and is associated with VDR down-regulation (Pálmer et al. 2004, Peña et al. 2005, 2006). In addition to SNAIL1, its family member SNAIL2 (but not other EMT inducers such as ZEB1, ZEB2, TWIST1 or E47) also represses VDR expression and blocks 1,25(OH)2D3 action in CRC cells (Larriba et al. 2009). Moreover, SNAIL1 and SNAIL2 show an additive repressive effect on VDR expression in cultured cells and in colorectal tumours (Larriba et al. 2009, 2010). Thus, it is probable that efficient VDR repression during CRC progression requires the expression of both SNAIL1 and SNAIL2, and that their expression in colorectal tumours could be used to identify patients who are likely to be resistant to the therapy with 1,25(OH)2D3 or its analogues.
Post-transcriptional down-regulation of VDR by miRNAs may be another mechanism responsible for the low VDR levels found in advanced CRC. Thus, it has been reported that miR-27b (MIR27B) and miR-298 (MIR298) reduce VDR levels in LS-180 colon cancer cells (Pan et al. 2009). In addition, Mohri et al. (2009) have shown that miR-125b (MIR125B) down-regulates VDR expression and reduces 1,25(OH)2D3 action in breast cancer cells. Interestingly, miR-125b has been found to be over-expressed in colon cancer metastases (Baffa et al. 2009). Therefore, the over-expression of miRNAs that target VDR may constitute a novel strategy used by colon cancer cells to escape from the anti-tumoural action of 1,25(OH)2D3.
Extensive work by Campbell's group has shown that lack of 1,25(OH)2D3 sensitivity in prostate and breast cancer cells is associated with elevated expression of the VDR corepressors SMRT and NCoR respectively (Khanim et al. 2004, Banwell et al. 2006). Accordingly, cotreatment with inhibitors of HDACs (trichostatin A, sodium butyrate) or DNA methyltransferases (5-aza-2′-deoxycytidine) reverses the resistance to the anti-proliferative effects of 1,25(OH)2D3 in different types of cancer cells, including CRC cells (Abedin et al. 2006). These results suggest that altered expression patterns of corepressors inappropriately sustain epigenetic modification at 1,25(OH)2D3 target genes and generate resistance to 1,25(OH)2D3 action. However, the effect of sodium butyrate on the sensitivity of CRC cells to 1,25(OH)2D3 seems to be mainly mediated by butyrate-induced VDR over-expression (Gaschott et al. 2001a,b, Gaschott & Stein 2003). Notably, increased levels of VDR corepressors have not been reported in colon cancer, instead, NCoR and SMRT have been found aberrantly located in the cytoplasm of cells from human CRC biopsies (Fernández-Majada et al. 2007a,b, Tzelepi et al. 2009). Thus, the possible contribution of this epigenetic mechanism to the development of 1,25(OH)2D3 resistance in CRC is unclear.
Anti-tumoural action of vitamin D in animal models of CRC
Results obtained in several types of studies using experimental animals support a protective and therapeutic action of vitamin D against CRC. Administration of a western-style diet (high in fat and low in vitamin D and calcium) generates colonic crypt hyperplasia and colon dysplasia in wild-type mice, and increases the incidence of pre-neoplastic lesions in various pre-clinical mouse models of intestinal tumourigenesis. These effects are suppressed upon diet supplementation with calcium and vitamin D (reviewed in Lamprecht & Lipkin (2003) and Ordóñez-Morán et al. (2005)).
Human tumour cells implanted s.c. into immunosuppressed mice (xenografts) are commonly used as an in vivo approach in pre-clinical anti-cancer drug development. Numerous studies have shown that 1,25(OH)2D3 and several analogues clearly reduce the growth of colorectal xenografts (reviewed in Ordóñez-Morán et al. (2005), Deeb et al. (2007) and Kang et al. (2011)). Similar results were obtained using different chemical carcinogens (N-methyl-N-nitrosourea, azoxymethane and others) to generate colorectal tumours in mice or rats. The number of tumours generated after chronic treatment with the carcinogens was reduced by the administration of 1,25(OH)2D3 or several of its analogues (reviewed in Ordóñez-Morán et al. (2005)).
Both 1,25(OH)2D3 and its less calcaemic derivative 1,25(OH)2-16-ene-19-nor-24-oxo-D3 reduce tumour load (the sum of all polyp areas) in Apcmin/+ mice (Huerta et al. 2002), a mouse model of intestinal carcinogenesis that carries a mutated allele of the tumour suppressor gene Apc and spontaneously develops multiple neoplasias throughout the intestinal tract (Su et al. 1992). Xu et al. (2010) confirmed these results and also found that treatment with 1,25(OH)2D3 or two of its analogues increases the expression of E-cadherin, inhibits that of Myc and reduces β-catenin nuclear levels in the small intestine and colon of Apcmin/+ mice. However, a recent study by Irving et al. (2011) has shown that treatment with 25(OH)D3 or two 1,25(OH)2D3 analogues did not affect intestinal tumour development in the ApcPirc/+ rat or in the Apcmin/+ mouse models in the absence or presence of the colonic tumour inducer dextran sodium sulphate. As the authors comment, it is however possible that the length of treatment with the vitamin D compounds or the putative loss of Vdr expression in the tumours might have influenced the results of the study (Irving et al. 2011).
Genetically modified mice have also been used to study the effects of the disruption of VDR expression on colorectal tumourigenesis. Vdr-deficient mice do not show a spontaneous increase in cancer but are more prone to oncogene- or carcinogen-induced tumours (reviewed in Bouillon et al. (2008)). These animals display colonic crypt hyper-proliferation, and increased levels of DNA damage and oxidative stress in the intestine (Kállay et al. 2001, 2002). Recently, two teams have bred Vdr−/+ and Apcmin/+ mice in order to evaluate the consequences of VDR deficiency on the initiation and development of intestinal cancer (Larriba et al. 2011, Zheng et al. 2012). These studies described increased tumour burden in Vdr−/−Apcmin/+ mice as compared to Vdr+/+ Apcmin/+. Importantly, Vdr−/−Apcmin/+ mice also show a higher activation of the WNT/β-catenin pathway in the lesions and an increase in the number of colonic aberrant crypt foci (Larriba et al. 2011).
Human studies: clinical and molecular data
Few large-scale cancer intervention trials on vitamin D treatment in human CRC patients have been reported. The first of these was a randomised, double-blind, placebo-controlled trial of 2686 individuals (65–85 years of age) receiving 100 000 IU vitamin D3 or placebo every 4 months (∼833 IU/day). This study did not find any significant effect of vitamin D3 supplementation on CRC incidence or mortality after 5 years (Trivedi et al. 2003). Accordingly, another randomised, double-blinded, placebo-controlled trial performed by the US Women's Health Initiative, including 36 282 post-menopausal women (50–79 years of age) who were treated with 400 IU/day vitamin D3 plus 1 g/day calcium vs placebo, failed to find any effect of the treatment on CRC incidence (Wactawski-Wende et al. 2006). However, the study had serious drawbacks that challenge its validity: the low amount of vitamin D3 used did not increase the circulating 25(OH)D3 levels, the degree of patient adherence to the treatment was low and the follow-up period was short (7 years) (Wactawski-Wende et al. 2006). Still, re-analysis of the data showed that concurrent estrogenic therapy increased CRC risk and that the treatment with vitamin D3 plus calcium was beneficial in its absence (relative risk (RR) 0.71; 95% confidence interval (CI) 0.46–1.09) (Ding et al. 2008). The third study with published results was a smaller trial performed in Nebraska (USA) with 1179 post-menopausal women (mean age 67 years) who were treated with 1100 IU/day vitamin D3 plus 1.4–1.5 g/day calcium vs calcium alone for 4 years (Lappe et al. 2007). Treatment with vitamin D3 plus calcium reduced total cancer incidence (RR 0.40; 95% CI 0.20–0.82), including CRC, particularly if the first-year cases were not considered. However, the low number of CRC cases reduces the validity of the study (Lappe et al. 2007).
At the molecular level, Holt et al. (2006) showed that combined daily supplementation of 11 patients with vitamin D3 (400 IU) and calcium (1.5 g) for 6 months reduced the proliferation of colonic flat mucosa and polyps, and increased the expression of BAK1 and diminished that of the CRC-associated mucin MUC5AC in the polyps when compared with the placebo group (8 patients). In a series of recent papers, Fedirko et al. reported that 800 IU/day vitamin D3 and/or 2 g/day calcium (in 92 patients with at least one adenoma, 6 months treatment) increased BAX and p21CIP1 expression in colorectal crypts (Fedirko et al. 2009a,b). These authors also observed a decreased labelling of the DNA oxidation marker 8-OH-dG (only by vitamin D3 or calcium alone, but not by the combination) and a reduced expression of the marker of long-term proliferation hTERT in the upper part of the crypt (only in the vitamin D3 plus calcium group) (Fedirko et al. 2009b, 2010). In addition, the group treated with vitamin D3 alone showed a reduction in the expression of the proinflammatory cytokines TNF-α, IL6, IL1β and IL8, and the proinflammatory marker C-reactive protein (Hopkins et al. 2011).
Clearly, there is a need for large and well-designed randomised, double-blind, placebo-controlled clinical studies. Up to now, clinical trials have included mostly elderly people, frequently post-menopausal women and so on, they may not fully reveal the potential action of vitamin D. Additional problems are the lack of an optimal vitamin D3 dose to be used, its possible combination with other agents (calcium), and the long follow-up period that seems recommendable in the case of a disease such as CRC, which probably develops over one or two decades. One promising ongoing trial is the VITamin D and OmegA-3 TriaL (VITAL), a study designed to investigate the role of high doses of vitamin D3 (2000 IU/day), omega-3 fatty acids (1 g/day) and their combination on cancer and cardiovascular diseases in 20 000 initially healthy men and women aged 60 and 65 years and older respectively (Nicholas 2011, Okereke & Manson 2011). In addition, a recent look at the US National Institutes of Health clinical trials website (http://clinicaltrials.gov) showed that more than 1000 trials are currently underway to assess the effects of vitamin D compounds on cancer and other diseases.
Conclusions and perspectives
In summary, the vast majority of molecular and genetic data suggests a protective role of vitamin D against CRC, and epidemiological studies also indicate that vitamin D deficiency is linked to high risk of this neoplasia. Globally, the results available on the loss of VDR expression and on the alteration of CYP27B1 and CYP24A1 levels during CRC progression support a role for vitamin D in the prevention and/or in the therapy of early stages rather than in the treatment of advanced cases of this neoplasia.
Declaration of interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.
Funding
The work in the authors' laboratory is supported by Ministerio de Ciencia e Innovación of Spain (grant number SAF2010-18302), Fondo Europeo de Desarrollo Regional-Instituto de Salud Carlos III (grant number RD06/0020/0009) and Comunidad de Madrid (grant number S2011/BMD-2344, Colomics2).
Acknowledgements
We thank Prof. Carsten Carlberg for critical reading of part of this review and Robin Rycroft for his valuable assistance in the preparation of the English manuscript.
References
Abe E, Miyaura C, Sakagami H, Takeda M, Konno K, Yamazaki T, Yoshiki S & Suda T 1981 Differentiation of mouse myeloid leukemia cells induced by 1α,25-dihydroxyvitamin D3. PNAS 78 4990–4994. doi:10.1073/pnas.78.8.4990.
Abedin SA, Banwell CM, Colston KW, Carlberg C & Campbell MJ 2006 Epigenetic corruption of VDR signalling in malignancy. Anticancer Research 26 2557–2566.
Agger K, Cloos PA, Rudkjaer L, Williams K, Andersen G, Christensen J & Helin K 2009 The H3K27me3 demethylase JMJD3 contributes to the activation of the INK4A-ARF locus in response to oncogene- and stress-induced senescence. Genes and Development 23 1171–1176. doi:10.1101/gad.510809.
Aguilera O, Peña C, García JM, Larriba MJ, Ordóñez-Morán P, Navarro D, Barbáchano A, López de Silanes I, Ballestar E & Fraga MF et al. 2007 The Wnt antagonist DICKKOPF-1 gene is induced by 1α,25-dihydroxyvitamin D3 associated to the differentiation of human colon cancer cells. Carcinogenesis 28 1877–1884. doi:10.1093/carcin/bgm094.
Aiba T, Susa M, Fukumori S & Hashimoto Y 2005 The effects of culture conditions on CYP3A4 and MDR1 mRNA induction by 1α,25-dihydroxyvitamin D3 in human intestinal cell lines, Caco-2 and LS180. Drug Metabolism and Pharmacokinetics 20 268–274. doi:10.2133/dmpk.20.268.
Ajibade D, Benn BS & Christakos S 2010 Mechanism of action of 1,25-dihydroxyvitamin D3 on intestinal calcium absorption and renal calcium transport. In Vitamin D: Physiology, Molecular Biology, and Clinical Applications, 2nd edn, pp 175–187. Ed MF Holick. Boston, MA: Humana Press
Álvarez-Díaz S, Valle N, García JM, Peña C, Freije JM, Quesada V, Astudillo A, Bonilla F, López-Otín C & Muñoz A 2009 Cystatin D is a candidate tumor suppressor gene induced by vitamin D in human colon cancer cells. Journal of Clinical Investigation 119 2343–2358. doi:10.1172/JCI37205.
Álvarez-Díaz S, Larriba MJ, López-Otín C & Muñoz A 2010 Vitamin D: proteases, protease inhibitors and cancer. Cell Cycle 9 32–37. doi:10.4161/cc.9.1.10266.
Álvarez-Díaz S, Valle N, Ferrer-Mayorga G, Lombardía L, Herrera M, Domínguez O, Segura MF, Bonilla F, Hernando E & Muñoz A 2012 MicroRNA-22 is induced by vitamin D and contributes to its antiproliferative, antimigratory and gene regulatory effects in colon cancer cells. Human Molecular Genetics [in press]. doi:10.1093/hmg/dds031.
Anderson MG, Nakane M, Ruan X, Kroeger PE & Wu-Wong JR 2006 Expression of VDR and CYP24A1 mRNA in human tumors. Cancer Chemotherapy and Pharmacology 57 234–240. doi:10.1007/s00280-005-0059-7.
Andl CD & Rustgi AK 2005 No one-way street: cross-talk between E-cadherin and receptor tyrosine kinase (RTK) signaling: a mechanism to regulate RTK activity. Cancer Biology & Therapy 4 28–31. doi:10.4161/cbt.4.1.1431.
Baffa R, Fassan M, Volinia S, O'Hara B, Liu CG, Palazzo JP, Gardiman M, Rugge M, Gomella LG & Croce CM et al. 2009 MicroRNA expression profiling of human metastatic cancers identifies cancer gene targets. Journal of Pathology 219 214–221. doi:10.1002/path.2586.
Banwell CM, MacCartney DP, Guy M, Miles AE, Uskokovic MR, Mansi J, Stewart PM, O'Neill LP, Turner BM & Colston KW et al. 2006 Altered nuclear receptor corepressor expression attenuates vitamin D receptor signaling in breast cancer cells. Clinical Cancer Research 12 2004–2013. doi:10.1158/1078-0432.CCR-05-1218.
Barbáchano A, Ordóñez-Morán P, García JM, Sánchez A, Pereira F, Larriba MJ, Martínez N, Hernández J, Landolfi S & Bonilla F et al. 2010 SPROUTY-2 and E-cadherin regulate reciprocally and dictate colon cancer cell tumourigenicity. Oncogene 29 4800–4813. doi:10.1038/onc.2010.225.
Bareis P, Bises G, Bischof MG, Cross HS & Peterlik M 2001 25-Hydroxyvitamin D metabolism in human colon cancer cells during tumor progression. Biochemical and Biophysical Research Communications 285 1012–1017. doi:10.1006/bbrc.2001.5289.
Barnes JD, Arhel NJ, Lee SS, Sharp A, Al-Okail M, Packham G, Hague A, Paraskeva C & Williams AC 2005 Nuclear BAG-1 expression inhibits apoptosis in colorectal adenoma-derived epithelial cells. Apoptosis 10 301–311. doi:10.1007/s10495-005-0804-8.
Ben-Shoshan M, Amir S, Dang DT, Dang LH, Weisman Y & Mabjeesh NJ 2007 1α,25-Dihydroxyvitamin D3 (calcitriol) inhibits hypoxia-inducible factor-1/vascular endothelial growth factor pathway in human cancer cells. Molecular Cancer Therapeutics 6 1433–1439. doi:10.1158/1535-7163.MCT-06-0677.
Bises G, Kállay E, Weiland T, Wrba F, Wenzl E, Bonner E, Kriwanek S, Obrist P & Cross HS 2004 25-Hydroxyvitamin D3-1α-hydroxylase expression in normal and malignant human colon. Journal of Histochemistry and Cytochemistry 52 985–989. doi:10.1369/jhc.4B6271.2004.
Bouillon R, Carmeliet G, Verlinden L, van Etten E, Verstuyf A, Luderer HF, Lieben L, Mathieu C & Demay M 2008 Vitamin D and human health: lessons from vitamin D receptor null mice. Endocrine Reviews 29 726–776. doi:10.1210/er.2008-0004.
Brehier A & Thomasset M 1988 Human colon cell line HT-29: characterisation of 1,25-dihydroxyvitamin D3 receptor and induction of differentiation by the hormone. Journal of Steroid Biochemistry 29 265–270. doi:10.1016/0022-4731(88)90275-0.
Cabrita MA & Christofori G 2008 Sprouty proteins, masterminds of receptor tyrosine kinase signaling. Angiogenesis 11 53–62. doi:10.1007/s10456-008-9089-1.
Cantorna MT, Munsick C, Bemiss C & Mahon BD 2000 1,25-Dihydroxycholecalciferol prevents and ameliorates symptoms of experimental murine inflammatory bowel disease. Journal of Nutrition 130 2648–2652.
Cantorna MT, Zhu Y, Froicu M & Wittke A 2004 Vitamin D status, 1,25-dihydroxyvitamin D3, and the immune system. American Journal of Clinical Nutrition 80 1717S–1720S.
Carlberg C & Dunlop TW 2006 An integrated biological approach to nuclear receptor signaling in physiological control and disease. Critical Reviews in Eukaryotic Gene Expression 16 1–22.
Carlberg C & Seuter S 2009 A genomic perspective on vitamin D signaling. Anticancer Research 29 3485–3493.
Carlberg C, Seuter S & Heikkinen S 2012 The first genome-wide view of vitamin D receptor locations and their mechanistic implications. Anticancer Research 32 271–282.
Chaffer CL & Weinberg RA 2011 A perspective on cancer cell metastasis. Science 331 1559–1564. doi:10.1126/science.1203543.
Chakrabarty S, Wang H, Canaff L, Hendy GN, Appelman H & Varani J 2005 Calcium sensing receptor in human colon carcinoma: interaction with Ca2+ and 1,25-dihydroxyvitamin D3. Cancer Research 65 493–498.
Chen Y & Goldstein JA 2009 The transcriptional regulation of the human CYP2C genes. Current Drug Metabolism 10 567–578.
Chen A, Davis BH, Sitrin MD, Brasitus TA & Bissonnette M 2002 Transforming growth factor-β 1 signaling contributes to Caco-2 cell growth inhibition induced by 1,25(OH)2D3. American Journal of Physiology. Gastrointestinal and Liver Physiology 283 G864–G874.
Clevers H 2006 Wnt/β-catenin signaling in development and disease. Cell 127 469–480. doi:10.1016/j.cell.2006.10.018.
Colston K, Colston MJ & Feldman D 1981 1,25-Dihydroxyvitamin D3 and malignant melanoma: the presence of receptors and inhibition of cell growth in culture. Endocrinology 108 1083–1086. doi:10.1210/endo-108-3-1083.
Cristobo I, Larriba MJ, de los Ríos V, García F, Muñoz A & Casal JI 2011 Proteomic analysis of 1α,25-dihydroxyvitamin D3 action on human colon cancer cells reveals a link to splicing regulation. Journal of Proteomics 75 384–397. doi:10.1016/j.jprot.2011.08.003.
Cross HS, Bajna E, Bises G, Genser D, Kállay E, Pötzi R, Wenzl E, Wrba F, Roka R & Peterlik M 1996 Vitamin D receptor and cytokeratin expression may be progression indicators in human colon cancer. Anticancer Research 16 2333–2337.
Cross HS, Bareis P, Hofer H, Bischof MG, Bajna E, Kriwanek S, Bonner E & Peterlik M 2001 25-Hydroxyvitamin D3-1α-hydroxylase and vitamin D receptor gene expression in human colonic mucosa is elevated during early cancerogenesis. Steroids 66 287–292. doi:10.1016/S0039-128X(00)00153-7.
Cross HS, Nittke T & Kallay E 2011 Colonic vitamin D metabolism: implications for the pathogenesis of inflammatory bowel disease and colorectal cancer. Molecular and Cellular Endocrinology 347 70–79. doi:10.1016/j.mce.2011.07.022.
David CJ & Manley JL 2011 Alternative pre-mRNA splicing regulation in cancer: pathways and programs unhinged. Genes and Development 24 2343–2364. doi:10.1101/gad.1973010.
Deeb KK, Trump DL & Johnson CS 2007 Vitamin D signalling pathways in cancer: potential for anticancer therapeutics. Nature Reviews. Cancer 7 684–700. doi:10.1038/nrc2196.
De Santa F, Totaro MG, Prosperini E, Notarbartolo S, Testa G & Natoli G 2007 The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell 130 1083–1094. doi:10.1016/j.cell.2007.08.019.
De Smet K & Contreras R 2005 Human antimicrobial peptides: defensins, cathelicidins and histatins. Biotechnology Letters 27 1337–1347. doi:10.1007/s10529-005-0936-5.
Díaz GD, Paraskeva C, Thomas MG, Binderup L & Hague A 2000 Apoptosis is induced by the active metabolite of vitamin D3 and its analogue EB1089 in colorectal adenoma and carcinoma cells: possible implications for prevention and therapy. Cancer Research 60 2304–2312.
Ding EL, Mehta S, Fawzi WW & Giovannucci EL 2008 Interaction of estrogen therapy with calcium and vitamin D supplementation on colorectal cancer risk: reanalysis of Women's Health Initiative randomized trial. International Journal of Cancer 122 1690–1694. doi:10.1002/ijc.23311.
Drocourt L, Ourlin JC, Pascussi JM, Maurel P & Vilarem MJ 2002 Expression of CYP3A4, CYP2B6, and CYP2C9 is regulated by the vitamin D receptor pathway in primary human hepatocytes. Journal of Biological Chemistry 277 25125–25132. doi:10.1074/jbc.M201323200.
Echchgadda I, Song CS, Roy AK & Chatterjee B 2004 Dehydroepiandrosterone sulfotransferase is a target for transcriptional induction by the vitamin D receptor. Molecular Pharmacology 65 720–729. doi:10.1124/mol.65.3.720.
Egan JB, Thompson PA, Vitanov MV, Bartik L, Jacobs ET, Haussler MR, Gerner EW & Jurutka PW 2010 Vitamin D receptor ligands, adenomatous polyposis coli, and the vitamin D receptor FokI polymorphism collectively modulate β-catenin activity in colon cancer cells. Molecular Carcinogenesis 49 337–352.
Evans SRT, Nolla J, Hanfelt J, Shabahang M, Nauta RJ & Shchepotin IB 1998 Vitamin D receptor expression as a predictive marker of biological behavior in human colorectal cancer. Clinical Cancer Research 4 1591–1595.
Evans SRT, Soldatenkov V, Shchepotin EB, Bogrash E & Shchepotin IB 1999 Novel 19-nor-hexafluoride vitamin D3 analog (Ro 25-6760) inhibits human colon cancer in vitro via apoptosis. International Journal of Oncology 14 979–985.
Fabian MR, Sonenberg N & Filipowicz W 2010 Regulation of mRNA translation and stability by microRNAs. Annual Review of Biochemistry 79 351–379. doi:10.1146/annurev-biochem-060308-103103.
Fan J, Liu S, Du Y, Morrison J, Shipman R & Pang KS 2009 Up-regulation of transporters and enzymes by the vitamin D receptor ligands, 1α,25-dihydroxyvitamin D3 and vitamin D analogs, in the Caco-2 cell monolayer. Journal of Pharmacology and Experimental Therapeutics 330 389–402. doi:10.1124/jpet.108.149815.
Fedirko V, Bostick RM, Flanders WD, Long Q, Shaukat A, Rutherford RE, Daniel CR, Cohen V & Dash C 2009a Effects of vitamin D and calcium supplementation on markers of apoptosis in normal colon mucosa: a randomized, double-blind, placebo-controlled clinical trial. Cancer Prevention Research 2 213–223. doi:10.1158/1940-6207.CAPR-08-0157.
Fedirko V, Bostick RM, Flanders WD, Long Q, Sidelnikov E, Shaukat A, Daniel CR, Rutherford RE & Woodard JJ 2009b Effects of vitamin D and calcium on proliferation and differentiation in normal colon mucosa: a randomized clinical trial. Cancer Epidemiology, Biomarkers & Prevention 18 2933–2941. doi:10.1158/1055-9965.EPI-09-0239.
Fedirko V, Bostick RM, Long Q, Flanders WD, McCullough ML, Sidelnikov E, Daniel CR, Rutherford RE & Shaukat A 2010 Effects of supplemental vitamin D and calcium on oxidative DNA damage marker in normal colorectal mucosa: a randomized clinical trial. Cancer Epidemiology, Biomarkers & Prevention 19 280–291. doi:10.1158/1055-9965.EPI-09-0448.
Ferlay J, Shin HR, Bray F, Forman D, Mathers C & Parkin DM 2010 Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. International Journal of Cancer 127 2893–2917. doi:10.1002/ijc.25516.
Fernández-García NI, Pálmer HG, García M, González-Martín A, del Río M, Barettino D, Volpert O, Muñoz A & Jiménez B 2005 1α,25-Dihydroxyvitamin D3 regulates the expression of Id1 and Id2 genes and the angiogenic phenotype of human colon carcinoma cells. Oncogene 24 6533–6544.
Fernández-Majada V, Aguilera C, Villanueva A, Vilardell F, Robert-Moreno A, Aytés A, Real FX, Capella G, Mayo MW & Espinosa L et al. 2007a Nuclear IKK activity leads to dysregulated notch-dependent gene expression in colorectal cancer. PNAS 104 276–281. doi:10.1073/pnas.0606476104.
Fernández-Majada V, Pujadas J, Vilardell F, Capella G, Mayo MW, Bigas A & Espinosa L 2007b Aberrant cytoplasmic localization of N-CoR in colorectal tumors. Cell Cycle 6 1748–1752. doi:10.4161/cc.6.14.4429.
Freedman DM, Looker AC, Chang SC & Graubard BI 2007 Prospective study of serum vitamin D and cancer mortality in the United States. Journal of the National Cancer Institute 99 1594–1602. doi:10.1093/jnci/djm204.
Froicu M, Zhu Y & Cantorna MT 2006 Vitamin D receptor is required to control gastrointestinal immunity in IL-10 knockout mice. Immunology 117 310–318. doi:10.1111/j.1365-2567.2005.02290.x.
Fujita H, Sugimoto K, Inatomi S, Maeda T, Osanai M, Uchiyama Y, Yamamoto Y, Wada T, Kojima T & Yokozaki H et al. 2008 Tight junction proteins claudin-2 and -12 are critical for vitamin D-dependent Ca2+ absorption between enterocytes. Molecular Biology of the Cell 19 1912–1921. doi:10.1091/mbc.E07-09-0973.
Gandini S, Boniol M, Haukka J, Byrnes G, Cox B, Sneyd MJ, Mullie P & Autier P 2011 Meta-analysis of observational studies of serum 25-hydroxyvitamin D levels and colorectal, breast and prostate cancer and colorectal adenoma. International Journal of Cancer 128 1414–1424. doi:10.1002/ijc.25439.
Garland CF & Garland FC 1980 Do sunlight and vitamin D reduce the likelihood of colon cancer? International Journal of Epidemiology 9 65–71. doi:10.1093/ije/9.3.227.
Garland CF, Comstock GW, Garland FC, Helsing KJ, Shaw EK & Gorham ED 1989 Serum 25-hydroxyvitamin D and colon cancer: eight-year prospective study. Lancet 2 1176–1178. doi:10.1016/S0140-6736(89)91789-3.
Gaschott T & Stein J 2003 Short-chain fatty acids and colon cancer cells: the vitamin D receptor–butyrate connection. Recent Results in Cancer Research 164 247–257.
Gaschott T, Steinhilber D, Milovic V & Stein J 2001a Tributyrin, a stable and rapidly absorbed prodrug of butyric acid, enhances antiproliferative effects of dihydroxycholecalciferol in human colon cancer cells. Journal of Nutrition 131 1839–1843.
Gaschott T, Werz O, Steinmeyer A, Steinhilber D & Stein J 2001b Butyrate-induced differentiation of Caco-2 cells is mediated by vitamin D receptor. Biochemical and Biophysical Research Communications 288 690–696. doi:10.1006/bbrc.2001.5832.
Gaschott T, Steinmeyer A, Steinhilber D & Stein J 2002 ZK 156718, a low calcemic, antiproliferative, and prodifferentiating vitamin D analog. Biochemical and Biophysical Research Communications 290 504–509. doi:10.1006/bbrc.2001.6213.
Gay O, Gilquin B, Nakamura F, Jenkins ZA, McCartney R, Krakow D, Deshiere A, Assard N, Hartwig JH & Robertson SP et al. 2011 RefilinB (FAM101B) targets filamin A to organize perinuclear actin networks and regulates nuclear shape. PNAS 108 11464–11469. doi:10.1073/pnas.1104211108.
Giovannucci E 2011 The epidemiology of vitamin D and cancer risk. In Vitamin D, 3rd edn, pp 227–234. Eds D Feldman, JW Pike & JS Adams. San Diego, CA: Academic Press
Giuliano AR, Franceschi RT & Wood RJ 1991 Characterization of the vitamin D receptor from the Caco-2 human colon carcinoma cell line: effect of cellular differentiation. Archives of Biochemistry and Biophysics 285 261–269. doi:10.1016/0003-9861(91)90358-P.
Gocek E, Wang X, Liu X, Liu CG & Studzinski GP 2011 MicroRNA-32 upregulation by 1,25-dihydroxyvitamin D3 in human myeloid leukemia cells leads to Bim targeting and inhibition of AraC-induced apoptosis. Cancer Research 71 6230–6239. doi:10.1158/0008-5472.CAN-11-1717.
Gombart AF, Borregaard N & Koeffler HP 2005 Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3. FASEB Journal 19 1067–1077. doi:10.1096/fj.04-3284com.
González-Sancho JM, Larriba MJ, Ordóñez-Morán P, Pálmer HG & Muñoz A 2006 Effects of 1α,25-dihydroxyvitamin D3 in human colon cancer cells. Anticancer Research 26 2669–2681.
González-Sancho JM, Larriba MJ & Muñoz A 2011 Vitamin D and Wnt/β-catenin signaling. In Vitamin D, 3rd edn, pp 235–250. Eds D Feldman, JW Pike & JS Adams. San Diego, CA: Academic Press
Halline AG, Davidson NO, Skarosi SF, Sitrin MD, Tietze C, Alpers DH & Brasitus TA 1994 Effects of 1,25-dihydroxyvitamin D3 on proliferation and differentiation of Caco-2 cells. Endocrinology 134 1710–1717. doi:10.1210/en.134.4.1710.
Hansen CM, Binderup L, Hamberg KJ & Carlberg C 2001 Vitamin D and cancer: effects of 1,25(OH)2D3 and its analogs on growth control and tumorigenesis. Frontiers in Bioscience 6 D820–D848. doi:10.2741/Hansen.
Haussler MR, Whitfield GK, Haussler CA, Hsieh J-C, Thompson PD, Selznick SH, Dominguez CE & Jurutka PW 1998 The nuclear vitamin D receptor: biological and molecular regulatory properties revealed. Journal of Bone and Mineral Research 13 325–349. doi:10.1359/jbmr.1998.13.3.325.
Haussler MR, Jurutka PW, Mizwicki M & Norman AW 2011 Vitamin D receptor (VDR)-mediated actions of 1α,25(OH)2vitamin D3: genomic and non-genomic mechanisms. Best Practice & Research. Clinical Endocrinology & Metabolism 25 543–559. doi:10.1016/j.beem.2011.05.010.
Heikkinen S, Väisänen S, Pehkonen P, Seuter S, Benes V & Carlberg C 2011 Nuclear hormone 1α,25-dihydroxyvitamin D3 elicits a genome-wide shift in the locations of VDR chromatin occupancy. Nucleic Acids Research 39 9181–9193. doi:10.1093/nar/gkr654.
Heyman RA, Mangelsdorf DJ, Dyck JA, Stein RB, Eichele G, Evans RM & Thaller C 1992 9-cis retinoic acid is a high affinity ligand for the retinoid X receptor. Cell 68 397–406. doi:10.1016/0092-8674(92)90479-V.
Holick MF 2005 Photobiology of vitamin D. In Vitamin D, 2nd edn, pp 37–43. Eds D Feldman, AN Pike & FH Glorieux. San Diego, CA: Elsevier
Holick MF 2007 Vitamin D deficiency. New England Journal of Medicine 357 266–281. doi:10.1056/NEJMra070553.
Holt PR, Bresalier RS, Ma CK, Liu KF, Lipkin M, Byrd JC & Yang K 2006 Calcium plus vitamin D alters preneoplastic features of colorectal adenomas and rectal mucosa. Cancer 106 287–296. doi:10.1002/cncr.21618.
Hopkins MH, Owen J, Ahearn T, Fedirko V, Flanders WD, Jones DP & Bostick RM 2011 Effects of supplemental vitamin D and calcium on biomarkers of inflammation in colorectal adenoma patients: a randomized, controlled clinical trial. Cancer Prevention Research 4 1645–1654. doi:10.1158/1940-6207.CAPR-11-0105.
Horst RL, Reinhardt TA & Reddy GS 2005 Vitamin D metabolism. In Vitamin D, 2nd edn, pp 15–29. Eds D Feldman, AN Pike & FH Glorieux. San Diego, CA: Elsevier
Huerta S, Irwin RW, Heber D, Go VLW, Koeffler HP, Uskokovic MR & Harris DM 2002 1α,25-(OH)2D3 and its synthetic analogue decrease tumor load in the Apcmin mouse. Cancer Research 62 741–746.
Iacopetta B 2003 TP53 mutation in colorectal cancer. Human Mutation 21 271–276. doi:10.1002/humu.10175.
IARC 2008 Vitamin D and cancer. In IARC Working Group Reports Vol 5. International Agency for Research on Cancer: Lyon, France
Irving AA, Halberg RB, Albrecht DM, Plum LA, Krentz KJ, Clipson L, Drinkwater N, Amos-Landgraf JM, Dove WF & DeLuca HF 2011 Supplementation by vitamin D compounds does not affect colonic tumor development in vitamin D sufficient murine models. Archives of Biochemistry and Biophysics 515 64–71. doi:10.1016/j.abb.2011.08.011.
Jenab M, Bueno-de-Mesquita HB, Ferrari P, van Duijnhoven FJ, Norat T, Pischon T, Jansen EH, Slimani N, Byrnes G & Rinaldi S et al. 2010 Association between pre-diagnostic circulating vitamin D concentration and risk of colorectal cancer in European populations:a nested case–control study. BMJ 340 b5500 doi:10.1136/bmj.b5500.
Kaler P, Augenlicht L & Klampfer L 2009 Macrophage-derived IL-1β stimulates Wnt signaling and growth of colon cancer cells: a crosstalk interrupted by vitamin D3. Oncogene 28 3892–3902. doi:10.1038/onc.2009.247.
Kaler P, Galea V, Augenlicht L & Klampfer L 2010 Tumor associated macrophages protect colon cancer cells from TRAIL-induced apoptosis through IL-1β-dependent stabilization of Snail in tumor cells. PLoS ONE 5 e11700 doi:10.1371/journal.pone.0011700.
Kállay E, Pietschmann P, Toyokuni S, Bajna E, Hahn P, Mazzucco K, Bieglmayer C, Kato S & Cross HS 2001 Characterization of a vitamin D receptor knockout mouse as a model of colorectal hyperproliferation and DNA damage. Carcinogenesis 22 1429–1435. doi:10.1093/carcin/22.9.1429.
Kállay E, Bareis P, Bajna E, Kriwanek S, Bonner E, Toyokuni S & Cross HS 2002 Vitamin D receptor activity and prevention of colonic hyperproliferation and oxidative stress. Food and Chemical Toxicology 40 1191–1196. doi:10.1016/S0278-6915(02)00030-3.
Kaminsky LS & Zhang Q-Y 2003 The small intestine as a xenobiotic-metabolizing organ. Drug Metabolism and Disposition 31 1520–1525. doi:10.1124/dmd.31.12.1520.
Kamysz W, Okrój M & Łukasiak J 2003 Novel properties of antimicrobial peptides. Acta Biochimica Polonica 50 461–469.
Kang W, Lee S, Jeon E, Yun YR, Kim KH & Jang JH 2011 Emerging role of vitamin D in colorectal cancer. World Journal of Gastrointestinal Oncology 3 123–127. doi:10.4251/wjgo.v3.i8.123.
Kato S, Kim MS, Yamaoka K & Fujiki R 2007 Mechanisms of transcriptional repression by 1,25(OH)2 vitamin D. Current Opinion in Nephrology and Hypertension 16 297–304. doi:10.1097/MNH.0b013e3281c55f16.
Khanim FL, Gommersall LM, Wood VH, Smith KL, Montalvo L, O'Neill LP, Xu Y, Peehl DM, Stewart PM & Turner BM et al. 2004 Altered SMRT levels disrupt vitamin D3 receptor signalling in prostate cancer cells. Oncogene 23 6712–6725. doi:10.1038/sj.onc.1207772.
Klaus A & Birchmeier W 2008 Wnt signalling and its impact on development and cancer. Nature Reviews. Cancer 8 387–398. doi:10.1038/nrc2389.
Klose RJ & Zhang Y 2007 Regulation of histone methylation by demethylimination and demethylation. Nature Reviews. Molecular Cell Biology 8 307–318. doi:10.1038/nrm2143.
Kouzmenko A, Ohtake F, Fujiki R & Kato S 2011 Epigenetic modifications in vitamin D receptor-mediated transrepression. In Vitamin D, 3rd edn, pp 227–234. Eds D Feldman, JW Pike & JS Adams. San Diego, CA: Academic Press
Kriebitzsch C, Verlinden L, Eelen G, Tan BK, Van Camp M, Bouillon R & Verstuyf A 2009 The impact of 1,25(OH)2D3 and its structural analogs on gene expression in cancer cells – a microarray approach. Anticancer Research 29 3471–3483.
Krishnan AV & Feldman D 2011 Mechanisms of the anti-cancer and anti-inflammatory actions of vitamin D. Annual Review of Pharmacology and Toxicology 51 311–336. doi:10.1146/annurev-pharmtox-010510-100611.
Lagishetty V, Misharin AV, Liu NQ, Lisse TS, Chun RF, Ouyang Y, McLachlan SM, Adams JS & Hewison M 2010 Vitamin D deficiency in mice impairs colonic antibacterial activity and predisposes to colitis. Endocrinology 151 2423–2432. doi:10.1210/en.2010-0089.
Lamprecht SA & Lipkin M 2003 Chemoprevention of colon cancer by calcium, vitamin D and folate: molecular mechanisms. Nature Reviews. Cancer 3 601–614. doi:10.1038/nrc1144.
Lappe JM, Travers-Gustafson D, Davies KM, Recker RR & Heaney RP 2007 Vitamin D and calcium supplementation reduces cancer risk: results of a randomized trial. American Journal of Clinical Nutrition 85 1586–1591.
Larriba MJ & Muñoz A 2005 SNAIL vs vitamin D receptor expression in colon cancer: therapeutics implications. British Journal of Cancer 92 985–989. doi:10.1038/sj.bjc.6602484.
Larriba MJ, Valle N, Pálmer HG, Ordóñez-Morán P, Álvarez-Díaz S, Becker KF, Gamallo C, García de Herreros A, González-Sancho JM & Muñoz A 2007 The inhibition of Wnt/β-catenin signalling by 1α,25-dihydroxyvitamin D3 is abrogated by Snail1 in human colon cancer cells. Endocrine-Related Cancer 14 141–151. doi:10.1677/ERC-06-0028.
Larriba MJ, Valle N, Álvarez S & Muñoz A 2008 Vitamin D3 and colorectal cancer. Advances in Experimental Medicine and Biology 617 271–280.
Larriba MJ, Martín-Villar E, García JM, Pereira F, Peña C, García de Herreros A, Bonilla F & Muñoz A 2009 Snail2 cooperates with Snail1 in the repression of vitamin D receptor in colon cancer. Carcinogenesis 30 1459–1468. doi:10.1093/carcin/bgp140.
Larriba MJ, Bonilla F & Muñoz A 2010 The transcription factors Snail1 and Snail2 repress vitamin D receptor during colon cancer progression. Journal of Steroid Biochemistry and Molecular Biology 121 106–109. doi:10.1016/j.jsbmb.2010.01.014.
Larriba MJ, Ordóñez-Morán P, Chicote I, Martín-Fernández G, Puig I, Muñoz A & Pálmer HG 2011 Vitamin D receptor deficiency enhances Wnt/β-catenin signaling and tumor burden in colon cancer. PLoS ONE 6 e23524 doi:10.1371/journal.pone.0023524.
Lechner D, Kállay E & Cross HS 2005 Phytoestrogens and colorectal cancer prevention. Vitamins and Hormones 70 169–198. doi:10.1016/S0083-6729(05)70006-6.
Levin AA, Sturzenbecker LJ, Kazmer S, Bosakowski T, Huselton C, Allenby G, Speck J, Kratzeisen C, Rosenberger M & Lovey A et al. 1992 9-cis retinoic acid stereoisomer binds and activates the nuclear receptor RXRα. Nature 355 359–361. doi:10.1038/355359a0.
Liu N, Nguyen L, Chun RF, Lagishetty V, Ren S, Wu S, Hollis B, DeLuca HF, Adams JS & Hewison M 2008 Altered endocrine and autocrine metabolism of vitamin D in a mouse model of gastrointestinal inflammation. Endocrinology 149 4799–4808. doi:10.1210/en.2008-0060.
Liu G, Hu X & Chakrabarty S 2010 Vitamin D mediates its action in human colon carcinoma cells in a calcium-sensing receptor-dependent manner: downregulates malignant cell behavior and the expression of thymidylate synthase and survivin and promotes cellular sensitivity to 5-FU. International Journal of Cancer 126 631–639. doi:10.1002/ijc.24762.
Ma Y, Zhang P, Wang F, Yang J, Liu Z & Qin H 2011 Association between vitamin D and risk of colorectal cancer: a systematic review of prospective studies. Journal of Clinical Oncology 29 3775–3782. doi:10.1200/JCO.2011.35.7566.
Markowitz SD & Bertagnolli MM 2009 Molecular origins of cancer: molecular basis of colorectal cancer. New England Journal of Medicine 361 2449–2460. doi:10.1056/NEJMra0804588.
Matusiak D, Murillo G, Carroll RE, Mehta RG & Benya RV 2005 Expression of vitamin D receptor and 25-hydroxyvitamin D3-1α-hydroxylase in normal and malignant human colon. Cancer Epidemiology, Biomarkers & Prevention 14 2370–2376. doi:10.1158/1055-9965.EPI-05-0257.
McCarthy TC, Li X & Sinal CJ 2005 Vitamin D receptor-dependent regulation of colon multidrug resistance-associated protein 3 gene expression by bile acids. Journal of Biological Chemistry 280 23232–23242. doi:10.1074/jbc.M411520200.
Meyer MB, Goetsch PD & Pike JW 2012 VDR/RXR and TCF4/β-catenin cistromes in colonic cells of colorectal tumor origin: impact on c-FOS and c-MYC gene expression. Molecular Endocrinology 26 37–51. doi:10.1210/me.2011-1109.
Miura K, Fujibuchi W & Sasaki I 2011 Alternative pre-mRNA splicing in digestive tract malignancy. Cancer Science 102 309–316. doi:10.1111/j.1349-7006.2010.01797.x.
Modica S, Gofflot F, Murzilli S, D'Orazio A, Salvatore L, Pellegrini F, Nicolucci A, Tognoni G, Copetti M & Valanzano R et al. 2010 The intestinal nuclear receptor signature with epithelial localization patterns and expression modulation in tumors. Gastroenterology 138 636–648. doi:10.1053/j.gastro.2009.09.060.
Mohri T, Nakajima M, Takagi S, Komagata S & Yokoi T 2009 MicroRNA regulates human vitamin D receptor. International Journal of Cancer 125 1328–1333. doi:10.1002/ijc.24459.
Murayama A, Kim MS, Yanagisawa J, Takeyama K & Kato S 2004 Transrepression by a liganded nuclear receptor via a bHLH activator through co-regulator switching. EMBO Journal 23 1598–1608. doi:10.1038/sj.emboj.7600157.
Nakamura F, Stossel TP & Hartwig JH 2011 The filamins: organizers of cell structure and function. Cell Adhesion & Migration 5 160–169. doi:10.4161/cam.5.2.14401.
Nicholas J 2011 Vitamin D and cancer: uncertainty persists; research continues. Journal of the National Cancer Institute 103 851–852. doi:10.1093/jnci/djr215.
Norman AW 2008 From vitamin D to hormone D: fundamentals of the vitamin D endocrine system essential for good health. American Journal of Clinical Nutrition 88 491S–499S. doi:10.3945/ajcn.2008.27049.
Norman AW & Bouillon R 2010 Vitamin D nutritional policy needs a vision for the future. Experimental Biology and Medicine 235 1034–1045. doi:10.1258/ebm.2010.010014.
Norman AW, Mizwicki MT & Norman DP 2004 Steroid-hormone rapid actions, membrane receptors and a conformational ensemble model. Nature Reviews. Drug Discovery 3 27–41. doi:10.1038/nrd1283.
Oh YS, Kim EJ, Schaffer BS, Kang YH, Binderup L, MacDonald RG & Park JHY 2001 Synthetic low-calcaemic vitamin D3 analogues inhibit secretion of insulin-like growth factor II and stimulate production of insulin-like growth factor-binding protein-6 in conjunction with growth suppression of HT-29 colon cancer cells. Molecular and Cellular Endocrinology 183 141–149. doi:10.1016/S0303-7207(01)00598-6.
Okereke OI & Manson JE 2011 The VITamin D and OmegA-3 TriaL (VITAL): rationale and design of a large-scale randomized controlled trial. In Vitamin D, 3rd edn, pp 2043–2055. Eds D Feldman, JW Pike & JS Adams. San Diego, CA: Academic Press
Okumura K, Itoh A, Isogai E, Hirose K, Hosokawa Y, Abiko Y, Shibata T, Hirata M & Isogai H 2004 C-terminal domain of human CAP18 antimicrobial peptide induces apoptosis in oral squamous cell carcinoma SAS-H1 cells. Cancer Letters 212 185–194. doi:10.1016/j.canlet.2004.04.006.
Oppenheim JJ, Biragyn A, Kwak LW & Yang D 2003 Roles of antimicrobial peptides such as defensins in innate and adaptive immunity. Annals of Rheumatic Disease 62 17–21. doi:10.1136/ard.62.suppl_2.ii17.
Ordóñez-Morán P & Muñoz A 2009 Nuclear receptors: genomic and non-genomic effects converge. Cell Cycle 8 1675–1680. doi:10.4161/cc.8.11.8579.
Ordóñez-Morán P, Larriba MJ, Pendás-Franco N, Aguilera O, González-Sancho JM & Muñoz A 2005 Vitamin D and cancer: an update of in vitro and in vivo data. Frontiers in Bioscience 10 2723–2749. doi:10.2741/1731.
Ordóñez-Morán P, Larriba MJ, Pálmer HG, Valero RA, Barbáchano A, Duñach M, García de Herreros A, Villalobos C, Berciano MT & Lafarga M et al. 2008 RhoA-ROCK and p38MAPK-MSK1 mediate vitamin D effects on gene expression, phenotype, and Wnt pathway in colon cancer cells. Journal of Cell Biology 183 697–710. doi:10.1083/jcb.200803020.
Ordóñez-Morán P, Álvarez-Díaz S, Valle N, Larriba MJ, Bonilla F & Muñoz A 2010 The effects of 1,25-dihydroxyvitamin D3 on colon cancer cells depend on RhoA-ROCK-p38MAPK-MSK signaling. Journal of Steroid Biochemistry and Molecular Biology 121 355–361. doi:10.1016/j.jsbmb.2010.02.031.
Pálmer HG, González-Sancho JM, Espada J, Berciano MT, Puig I, Baulida J, Quintanilla M, Cano A, García de Herreros A & Lafarga M et al. 2001 Vitamin D3 promotes the differentiation of colon carcinoma cells by the induction of E-cadherin and the inhibition of β-catenin signaling. Journal of Cell Biology 154 369–387. doi:10.1083/jcb.200102028.
Pálmer HG, Sánchez-Carbayo M, Ordóñez-Morán P, Larriba MJ, Cordón-Cardó C & Muñoz A 2003 Genetic signatures of differentiation induced by 1α,25-dihydroxyvitamin D3 in human colon cancer cells. Cancer Research 63 7799–7806.
Pálmer HG, Larriba MJ, García JM, Ordóñez-Morán P, Peña C, Peiró S, Puig I, Rodríguez R, de la Fuente R & Bernad A et al. 2004 The transcription factor SNAIL represses vitamin D receptor expression and responsiveness in human colon cancer. Nature Medicine 10 917–919. doi:10.1038/nm1095.
Pan Q & Simpson RU 1999 c-myc intron element-binding proteins are required for 1, 25-dihydroxyvitamin D3 regulation of c-myc during HL-60 cell differentiation and the involvement of HOXB4. Journal of Biological Chemistry 274 8437–8444. doi:10.1074/jbc.274.13.8437.
Pan Q, Martell RE, O'Connell TD & Simpson RU 1996 1,25-Dihydroxyvitamin D3-regulated binding of nuclear proteins to a c-myc intron element. Endocrinology 137 4154–4160. doi:10.1210/en.137.10.4154.
Pan YZ, Gao W & Yu AM 2009 MicroRNAs regulate CYP3A4 expression via direct and indirect targeting. Drug Metabolism and Disposition 37 2112–2117. doi:10.1124/dmd.109.027680.
Peña C, García JM, Silva J, García V, Rodríguez R, Alonso I, Millán I, Salas C, García de Herreros A & Muñoz A et al. 2005 E-cadherin and vitamin D receptor regulation by SNAIL and ZEB1 in colon cancer: clinicopathological correlations. Human Molecular Genetics 14 3361–3370. doi:10.1093/hmg/ddi366.
Peña C, García JM, García V, Silva J, Domínguez G, Rodríguez R, Maximiano C, García de Herreros A, Muñoz A & Bonilla F 2006 The expression levels of the transcriptional regulators p300 and CtBP modulate the correlations between SNAIL, ZEB1, E-cadherin and vitamin D receptor in human colon carcinomas. International Journal of Cancer 119 2098–2104. doi:10.1002/ijc.22083.
Pendás-Franco N, García JM, Peña C, Valle N, Pálmer HG, Heinäniemi M, Carlberg C, Jiménez B, Bonilla F & Muñoz A et al. 2008 DICKKOPF-4 is induced by TCF/β-catenin and upregulated in human colon cancer, promotes tumour cell invasion and angiogenesis and is repressed by 1α,25-dihydroxyvitamin D3. Oncogene 27 4467–4477. doi:10.1038/onc.2008.88.
Pereira F, Barbáchano A, Silva J, Bonilla F, Campbell MJ, Muñoz A & Larriba MJ 2011 KDM6B/JMJD3 histone demethylase is induced by vitamin D and modulates its effects in colon cancer cells. Human Molecular Genetics 20 4655–4665. doi:10.1093/hmg/ddr399.
Pereira F, Barbáchano A, Singh PK, Campbell MJ, Muñoz A & Larriba MJ 2012 Vitamin D has wide regulatory effects on histone demethylase genes. Cell Cycle [in press].
Pfrunder A, Gutmann H, Beglinger C & Drewe J 2003 Gene expression of CYP3A4, ABC-transporters (MDR1 and MRP1-MRP5) and hPXR in three different human colon carcinoma cell lines. Journal of Pharmacy and Pharmacology 55 59–66. doi:10.1111/j.2042-7158.2003.tb02434.x.
Pike JW & Meyer MB 2010 The vitamin D receptor: new paradigms for the regulation of gene expression by 1,25-dihydroxyvitamin D3. Endocrinology and Metabolism Clinics of North America 39 255–269. doi:10.1016/j.ecl.2010.02.007.
Platz EA, Hankinson SE, Hollis BW, Colditz GA, Hunter DJ, Speizer FE & Giovannucci E 2000 Plasma 1,25-dihydroxy- and 25-hydroxyvitamin D and adenomatous polyps of the distal colorectum. Cancer Epidemiology, Biomarkers & Prevention 9 1059–1065.
Ramagopalan SV, Heger A, Berlanga AJ, Maugeri NJ, Lincoln MR, Burrell A, Handunnetthi L, Handel AE, Disanto G & Orton SM et al. 2010 A ChIP-seq defined genome-wide map of vitamin D receptor binding: associations with disease and evolution. Genome Research 20 1352–1360. doi:10.1101/gr.107920.110.
Schuster I, Egger H, Reddy GS & Vorisek G 2003 Combination of vitamin D metabolites with selective inhibitors of vitamin D metabolism. Recent Results in Cancer Research 164 169–188.
Schwab M, Reynders V, Loitsch S, Steinhilber D, Schröder O & Stein J 2008 The dietary histone deacetylase inhibitor sulforaphane induces human β-defensin-2 in intestinal epithelial cells. Immunology 125 241–251. doi:10.1111/j.1365-2567.2008.02834.x.
Shabahang M, Buras RR, Davoodi F, Schumaker LM, Nauta RJ & Evans SRT 1993 1,25-Dihydroxyvitamin D3 receptor as a marker of human colon carcinoma cell line differentiation and growth inhibition. Cancer Research 53 3712–3718.
Shah S, Islam MN, Dakshanamurthy S, Rizvi I, Rao M, Herrell R, Zinser G, Valrance M, Aranda A & Moras D et al. 2006 The molecular basis of vitamin D receptor and β-catenin crossregulation. Molecular Cell 21 799–809. doi:10.1016/j.molcel.2006.01.037.
Sheinin Y, Kaserer K, Wrba F, Wenzl E, Kriwanek S, Peterlik M & Cross HS 2000 In situ mRNA hybridization analysis and immunolocalization of the vitamin D receptor in normal and carcinomatous human colonic mucosa: relation to epidermal growth factor receptor expression. Virchows Archiv 437 501–507. doi:10.1007/s004280000275.
Stambolsky P, Tabach Y, Fontemaggi G, Weisz L, Maor-Aloni R, Siegfried Z, Shiff I, Kogan I, Shay M & Kalo E et al. 2010 Modulation of the vitamin D3 response by cancer-associated mutant p53. Cancer Cell 17 273–285. doi:10.1016/j.ccr.2009.11.025.
Su LK, Kinzler KW, Vogelstein B, Preisinger AC, Moser AR, Luongo C, Gould KA & Dove WF 1992 Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science 256 668–670. doi:10.1126/science.1350108.
Sun J, Mustafi R, Cerda S, Chumsangsri A, Xia YR, Li YC & Bissonnette M 2008 Lithocholic acid down-regulation of NF-kappaB activity through vitamin D receptor in colonic cancer cells. Journal of Steroid Biochemistry and Molecular Biology 111 37–40. doi:10.1016/j.jsbmb.2008.01.003.
Thiery JP, Acloque H, Huang RYJ & Nieto MA 2009 Epithelial–mesenchymal transitions in development and disease. Cell 139 871–890. doi:10.1016/j.cell.2009.11.007.
Thompson PD, Jurutka PW, Whitfield GK, Myskowski SM, Eichhorst KR, Dominguez CE, Haussler CA & Haussler MR 2002 Liganded VDR induces CYP3A4 in small intestinal and colon cancer cells via DR3 and ER6 vitamin D responsive elements. Biochemical and Biophysical Research Communications 299 730–738. doi:10.1016/S0006-291X(02)02742-0.
Thorne J & Campbell MJ 2008 The vitamin D receptor in cancer. Proceedings of the Nutrition Society 67 115–127. doi:10.1017/S0029665108006964.
Thorne JL, Maguire O, Doig CL, Battaglia S, Fehr L, Sucheston LE, Heinaniemi M, O'Neill LP, McCabe CJ & Turner BM et al. 2011 Epigenetic control of a VDR-governed feed-forward loop that regulates p21(waf1/cip1) expression and function in non-malignant prostate cells. Nucleic Acids Research 39 2045–2056. doi:10.1093/nar/gkq875.
Thummel KE, Brimer C, Yasuda K, Thottassery J, Senn T, Lin Y, Ishizuka H, Kharasch E, Schuetz J & Schuetz E 2001 Transcriptional control of intestinal cytochrome P-4503A by 1α,25-dihydroxy vitamin D3. Molecular Pharmacology 60 1399–1406.
Tong W-M, Hofer H, Ellinger A, Peterlik M & Cross HS 1999 Mechanism of antimitogenic action of vitamin D in human colon carcinoma cells: relevance for suppression of epidermal growth factor-stimulated cell growth. Oncology Research 11 77–84.
Toropainen S, Väisänen S, Heikkinen S & Carlberg C 2010 The down-regulation of the human MYC gene by the nuclear hormone 1α,25-dihydroxyvitamin D3 is associated with cycling of corepressors and histone deacetylases. Journal of Molecular Biology 400 284–294. doi:10.1016/j.jmb.2010.05.031.
Townsend K, Evans KN, Campbell MJ, Colston KW, Adams JS & Hewison M 2005 Biological actions of extra-renal 25-hydroxyvitamin D-1α-hydroxylase and implications for chemoprevention and treatment. Journal of Steroid Biochemistry and Molecular Biology 97 103–109. doi:10.1016/j.jsbmb.2005.06.004.
Trivedi DP, Doll R & Khaw KT 2003 Effect of four monthly oral vitamin D3 (cholecalciferol) supplementation on fractures and mortality in men and women living in the community: randomised double blind controlled trial. BMJ 326 469 doi:10.1136/bmj.326.7387.469.
Tzelepi V, Grivas P, Kefalopoulou Z, Kalofonos H, Varakis JN & Sotiropoulou-Bonikou G 2009 Expression of estrogen receptor co-regulators NCoR and PELP1 in epithelial cells and myofibroblasts of colorectal carcinomas: cytoplasmic translocation of NCoR in epithelial cells correlates with better prognosis. Virchows Archiv 454 41–53. doi:10.1007/s00428-008-0708-4.
de Urquiza AM, Liu S, Sjoberg M, Zetterstrom RH, Griffiths W, Sjovall J & Perlmann T 2000 Docosahexaenoic acid, a ligand for the retinoid X receptor in mouse brain. Science 290 2140–2144. doi:10.1126/science.290.5499.2140.
Verstuyf A, Carmeliet G, Bouillon R & Mathieu C 2010 Vitamin D: a pleiotropic hormone. Kidney International 78 140–145. doi:10.1038/ki.2010.17.
Wactawski-Wende J, Kotchen JM, Anderson GL, Assaf AR, Brunner RL, O'Sullivan MJ, Margolis KL, Ockene JK, Phillips L & Pottern L et al. 2006 Calcium plus vitamin D supplementation and the risk of colorectal cancer. New England Journal of Medicine 354 684–696. doi:10.1056/NEJMoa055222.
Welch C, Santra MK, El-Assaad W, Zhu X, Huber WE, Keys RA, Teodoro JG & Green MR 2009 Identification of a protein, G0S2, that lacks Bcl-2 homology domains and interacts with and antagonizes Bcl-2. Cancer Research 69 6782–6789. doi:10.1158/0008-5472.CAN-09-0128.
Wood RJ, Tchack L, Angelo G, Pratt RE & Sonna LA 2004 DNA microarray analysis of vitamin D-induced gene expression in a human colon carcinoma cell line. Physiological Genomics 17 122–129. doi:10.1152/physiolgenomics.00002.2003.
Woolcott CG, Wilkens LR, Nomura AM, Horst RL, Goodman MT, Murphy SP, Henderson BE, Kolonel LN & Le Marchand L 2010 Plasma 25-hydroxyvitamin D levels and the risk of colorectal cancer: the multiethnic cohort study. Cancer Epidemiology, Biomarkers & Prevention 19 130–134. doi:10.1158/1055-9965.EPI-09-0475.
Wu K, Feskanich D, Fuchs CS, Willett WC, Hollis BW & Giovannucci EL 2007 A nested case control study of plasma 25-hydroxyvitamin D concentrations and risk of colorectal cancer. Journal of the National Cancer Institute 99 1120–1129. doi:10.1093/jnci/djm038.
Xu H, Posner GH, Stevenson M & Campbell FC 2010 ApcMIN modulation of vitamin D secosteroid growth control. Carcinogenesis 31 1434–1441. doi:10.1093/carcin/bgq098.
Yue J, Wang Q, Lu H, Brenneman M, Fan F & Shen Z 2009 The cytoskeleton protein filamin-A is required for an efficient recombinational DNA double strand break repair. Cancer Research 69 7978–7985. doi:10.1158/0008-5472.CAN-09-2177.
Zhao X & Feldman D 1993 Regulation of vitamin D receptor abundance and responsiveness during differentiation of HT29 human colon cancer cells. Endocrinology 132 1808–1814. doi:10.1210/en.132.4.1808.
Zheng W, Wong KE, Zhang Z, Dougherty U, Mustafi R, Kong J, Deb DK, Zheng H, Bissonnette M & Li YC 2012 Inactivation of the vitamin D receptor in APCmin/+ mice reveals a critical role for the vitamin D receptor in intestinal tumor growth. International Journal of Cancer 130 10–19. doi:10.1002/ijc.25992.
Zhu Y, Mahon BD, Froicu M & Cantorna MT 2005 Calcium and 1α,25-dihydroxyvitamin D3 target the TNF-α pathway to suppress experimental inflammatory bowel disease. European Journal of Immunology 35 217–224. doi:10.1002/eji.200425491.