Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jul 1.
Published in final edited form as: J Steroid Biochem Mol Biol. 2012 Oct 8;136:252–257. doi: 10.1016/j.jsbmb.2012.09.031

24-Hydroxylase in Cancer: Impact on Vitamin D-based Anticancer Therapeutics

Wei Luo a, Pamela A Hershberger a, Donald L Trump b, Candace S Johnson a,*
PMCID: PMC3686893  NIHMSID: NIHMS422350  PMID: 23059474

Abstract

The active vitamin D hormone 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) plays a major role in regulating calcium homeostasis and bone mineralization. 1,25(OH)2D3 also modulates cellular proliferation and differentiation in a variety of cell types. 24-hydroxylase, encoded by the CYP24A1 gene, is the key enzyme which converts 1,25(OH)2D3 to less active calcitroic acid. Nearly all cell types express 24-hydroxylase, the highest activity being observed in the kidney. There is increasing evidence linking the incidence and prognosis of certain cancers to low serum 25 (OH)D3 levels and high expression of vitamin D 24-hydroxylase supporting the idea that elevated CYP24A1 expression may stimulate degradation of vitamin D metabolites including 25-(OH)D3 and 1,25(OH)2D3. The over expression of CYP24A1 in cancer cells may be a factor affecting 1,25(OH)2D3 bioavailability and anti-proliferative activity pre-clinically and clinically. The combination of 1,25(OH)2D3 with CYP24A1 inhibitors enhances 1,25(OH)2D3 mediated signaling and anti-proliferative effects and may be useful in overcoming effects of aberrant CYP24 expression.

Keywords: CYP24, 24-hydroxylase, calcitriol and anti-tumor

1. Introduction

Vitamin D is a fat-soluble secosteroid which is synthesized in the body and has broad biological effects. 1,25(OH)2D3 is the most potent biologically active form of vitamin D3 and plays a central role in regulating calcium and phosphorus homeostasis[1, 2]. Recent studies have shown that 1,25(OH)2D3 has a broad range of functions including modulating the immune system and regulating cellular differentiation and proliferation[15]. 1,25(OH)2D3 inhibits cancer cell proliferation, cell migration and invasion and induces apoptosis in a wide variety of cancer cell types[612], including leukemia, prostate, colon, breast, ovarian, pancreas and liver.

Vitamin D3 (cholecalciferol), a precursor of 1,25(OH)2D3, is available in the diet, but is primarily supplied through synthesis from 7-dehydrocholesterol in the skin after exposure to ultraviolet B-light. Once produced, vitamin D3 is carried as a complex with the plasma vitamin D binding protein (DBP) to the liver[13], where it is hydroxylated by 25-hydroxylase to yield 25(OH)D3, the major circulating form of vitamin D. 25(OH)D3 is 1!-hydroxylated to 1,25(OH)2D3, also known as calcitriol, by 1α-hydroxylase (CYP27B1) mainly in kidney, and this is subject to tight control to avoid hypercalcemia. 1,25(OH)2D3 is also synthesized in many extrarenal sites that express CYP27B1[1417].

24-hydroxylase encoded by CYP24A1 is the key enzyme to inactivate 1,25(OH)2D3 [18, 19]. The blood 1,25(OH)2D3 level is tightly controlled through feedback regulation of its biosynthesis and catabolism by CYP27B1 and CYP24A1, respectively. Both CYP27B1 and CYP24A1 are highly regulated enzymes and respond to modulating agents such as parathyroid hormone (PTH), calcitonin, calcium, phosphorus as well as 1,25(OH)2D3. CYP24A1 is constitutively expressed in kidney and gastrointestinal mucosa. In most other tissues, CYP24 is transcriptionally induced by 1,25(OH)2D3 [1820]. This limits 1,25(OH)2D3 signaling and potential to cause hypercalcemia. Increased expression of CYP24A1 has been found in several human tumors[21, 22]. By stimulating 1,25(OH)2D3 degradation, over expression of CYP24A1 limits 1,25(OH)2D3 biologic activity and may diminish the effect of 1,25(OH)2D3 in modulating proliferation and motility of cancer cells. The understanding of CYP24A1 dysregulation in and cancers is likely to uncover potential ways to inhibit CYP24A1 in an effort to improve the anti-tumor efficacy of 1,25(OH)2D3.

2. Regulation of CYP24A1 expression

2.1 VDR/RXR binding to VDREs

The human CYP24A1 gene, located on chromosome 20q13.2 is highly conserved across all phyla[18, 23]. CYP24A1 is a mitochondrial protein and a member of the cytochrome class I P450 superfamily of enzymes. CYP24A1 expression is controlled by a 1,25(OH)2D3- vitamin D receptor (VDR) dependent process[24]. In VDR null mice, there are lower levels of CYP24A1 and 24-hydroxylated metabolites demonstrating that CYP24A1 expression is dependent on VDR[25]. In most normal tissues, CYP24A1 is expressed at very low basal levels and is induced by 1,25(OH)2D3 via positive transcriptional regulation.

CYP24A1 is regulated through binding of 1,25(OH)2D3 to the VDR and the subsequent interaction of the VDR-1,25(OH)2D3 complex with its heterodimeric partner, retinoid-X-receptor (RXR)[26]. Unliganded VDR/RXR binds to the vitamin D response elements (VDRE) to recruit corepressors, such as NCoR or Alien[27]. This results in recruitment of enzymes with HDAC activity and leads to a closed chromatin structure and gene repression. The binding of 1,25-(OH)2D3 to VDR leads to replacement of the repressor by a coactivator complex, such as nuclear coactivators (NCoAs)[28, 29]. These coactivators link the ligand-activated VDR to enzymes displaying histone acetyltransferase activity that cause chromatin relaxation and activate target gene transcription[30, 31]. A cluster of VDREs has been reported in the proximal promoter of both the human and rat CYP24 genes[32, 33]. Single nucleotide polymorphisms (SNPs) in the VDRE within the human CYP24A1 promoter have been identified and are associated with lower affinity for the VDR-RXR heterodimer, decreased transactivation, and reduced expression of CYP24A1[34].

2.2 Stimulation of intracellular pathways

1,25(OH)2D3 activation of protein kinase signaling pathways may also be involved in the regulation of CYP24A1 expression[18, 35, 36]. This rapid (minutes), non-genomic action involves intracellular signaling pathways such as protein kinase C (PKC) and mitogen activated protein kinase (MAPK), through a plasma-membrane-associated VDR. The signaling mechanisms that regulate transcriptional activation of the CYP24A1 promoter are poorly defined and further investigation is needed. Some transcription factors that interact with VDR and enhance 1,25(OH)2D3-mediated induction of the CYP24A1 promoter through intracellular signaling pathways have been identified. For example, Ets-1 has been shown to induce CYP24A1 promoter activity[37, 38]. Ets-1 can be activated by the Ras/Raf signaling pathway through phosphorylation of threonine residue 38[39]. Furthermore, 1,25(OH)2D3 stimulates MAP kinase and ERK5 to activate Ets-1 through phosphorylation in COS-1 monkey kidney fibroblast cells[37]. Dwivedi et al.[40] reported that the interaction of VDR and Ets-1 caused a significant induction of CYP24A1 promoter activity in COS-1 cells. The direct interaction between VDR and Ets-1 protein was demonstrated by an in vitro GST-protein pull-down study[41]. Signal transduction-regulated CYP24A1 expression could be cell type specific. COS-1 cells treated with inhibitors for ERK1/2 and ERK5 abolished 1,25(OH)2D3 induction of the CYP24A1 promoter[40]. However, this was not found to be the case in human embryonic kidney 293T (HEK-293T) cells[37]. Cellular modulators, such as glucocorticoids also regulate CYP24A1 expression. The cooperation between CCAAT/enhancer-binding protein (C/EBP) and glucocorticoid receptor (GR) results in enhancement of 1,25(OH)2D3-induced CYP24A1 expression[42]. Glucocorticoids may also indirectly increase CYP24A1 transcription by increasing VDR gene expression[43].

2.3 Epigenetics

Epigenetic changes could operate on a more intermediate time scale to serve as an additional mechanism controlling CYP24 expression[44, 45]. Epigenetic regulation of steroid signaling is an emerging field of research[46]. However, at present, little is known about the consequences of epigenetic regulation of the vitamin D metabolism pathway. Novakovic et al. reported tissue-specific CYP24A1 promoter methylation in human placenta, purified cytotrophoblasts, and primary cultures of chorionic villus tissue.[45]. They demonstrated that promoter methylation directly down-regulated basal promoter activity and abolished vitamin D-mediated gene activation. No methylation was detected in most normal human tissues including kidney, skeletal muscle, skin fibroblasts, brain (prefrontal cortex), sperm, whole blood, buccal mucosa, endometrial stroma, decidualized stroma, bone marrow, umbilical cord tissue[45] and peripheral blood lymphocytes[47].

3. CYP24A1 in Cancer

CYP24A1, which is a candidate oncogene[22, 48], is aberrantly expressed in numerous human tumor types[21, 22, 48]. High CYP24A1 levels seem to be a common feature of several solid tumors. Elevated tumor CYP24A1 expression is associated with a poorer prognosis[22, 49]. The increased intra-tumoral levels of CYP24A1 may lead to rapid degradation of 1,25(OH)2D3 and abrogation of its antiproliferative effects.

The mechanisms underlying the aberrant expression of CYP24A1 in tumors are not well defined. CYP24A1 overexpression does not correlate with VDR expression level in several types of tumors, since the latter are actually reduced or unchanged. This suggests that high CYP24A1 levels are not the result of the normal physiological transcriptional process observed when VDR-bound 1,25(OH)2D3 activates CYP24A1. There is evidence that multiple factors might be involved in dysregulation of CYP24 expression in cancer[14, 5052].

One possibility is amplification at the CYP24A1 locus. Albertson et al.[53] found that CYP24A1 levels in breast tumors are higher in specimens with amplification at the CYP24A1 locus. Kallioniemi et al.[54] showed that a significant increase of regional copy number and amplification at the CYP24A1 locus was observed in approximately 18% of primary breast tumors and 40% of breast cell lines. Alternatively, CYP24A1 regulation in tumors may be related to miRNAs that target the CYP24 gene. Komagata et al.[50] identified miR- 125b, which binds at the 3′ UTR of CYP24A1 mRNA to post-transcriptionally repress CYP24 translation. In these cells, CYP24A1 protein levels were increased by the inhibition of miR-125b and were decreased by miR-125b over-expression. Furthermore, CYP24A1 levels in breast cancer tissues were inversely associated with miR-125b levels. Additionally, the number of functional vitamin D receptor binding sites could be responsible for a difference in CYP24 gene expression level induced by vitamin D in malignant and normal mammary cells[52]. The number of functional VDREs is higher in malignant breast MCF-7 cells with high CYP24A1 expression than in normal breast MCF-10A cells. Lastly, CYP24 expression in cancer cells may be modified via epigenetic processes. CYP24 DNA promoter hypermethylation was observed in human choriocarcinoma cell lines[45], human prostate cancer cell lines[51, 55] and human prostate cancerous lesions. The promoter DNA methylation in cancer was inversely correlated with CYP24A1 expression[51, 55]. In addition, studies indicate that repression of CYP24A1 gene expression in human prostate cancer cells was mediated in part by promoter DNA methylation and repressive histone modifications. The tumor microenvironment also affects the methylation status of the CYP24 promoter. Differential methylation of the CYP24A1 gene promoter was also observed in endothelium from benign and malignant human prostate[56].

4. CYP24A1 inhibitors

As a key enzyme responsible for vitamin D catabolism, the aberrant expression of CYP24A1 in tumors may negatively impact vitamin D therapy. Furthermore, the induction of CYP24A1 expression by administration of 1,25(OH)2D3 through the negative feedback mechanism also limits the amount of 1,25(OH)2D3 available systemically and locally in tumor cells. In vitro studies demonstrated that differences in 1,25(OH)2D3-mediated growth inhibition among various cancer cell lines correlated inversely to levels of CYP24A1 expression in these cells[21, 57]. Data obtained using CYP24 inhibitors suggest that increased CYP24A1 expression in tumors restricts 1,25(OH)2D3 anti-tumor activity[58]. Inhibition of CYP24A1 slows the loss of 1,25(OH)2D3 and extends the half-life of 1,25(OH)2D3, thereby increasing 1,25(OH)2D3 exposure and anti-proliferative effects[58, 59].

Different types of CYP24A1 inhibitors have been developed. These include azole and non-azole inhibitors and vitamin D analogues. Azole inhibitors have a nitrogen heterocyclic ring, commonly an imidazole or triazole, and they bind with the Fe3+ heme of CYP24. Non-azole inhibitors lack this nitrogen heterocycle and instead bind within the CYP24 active site via hydrogen bonding and hydrophobic interactions.

Ketoconazole and liarozole[60, 61], are non-selective azole CYP24A1 inhibitors, which target the active sites of P450 enzymes[62]. Combination of 1,25(OH)2D3 with liarozole or ketoconazole increases the half-life of 1,25(OH)2D3 and potentiates its antiproliferative effects in human cancer cells exhibiting CYP24A1 activity.

The tetralone derivative (2-(4-hydroxybenzyl)-6-methoxy-3,4-dihydro-2H-naphthalen-1-one), a non-azole CYP24 inhibitor, enhanced 1,25(OH)2D3 antiproliferative activity in DU145 cells and enhanced the upregulation of vitamin D target genes, p21waf1/cip1 and GADD45a[63]. Isoflavones are non-specific inhibitors of the CYP enzymes. Genistein is one of the isoflavones found in a number of plants[64]. Genistein inhibits the expression of CYP24, leading to an increase in the half-life of 1,25(OH)2D3 [65]. Combination of 1,25(OH)2D3 with genistein resulted in synergistic growth inhibition of prostate cancer cells by activating vitamin D signaling[66]and enhanced 1,25(OH)2D3 mediated functional responses including upregulation of mRNA levels of p21/WAF1 and IGFBP-3[67].

The broad inhibitory effects of non-selective inhibitors such as ketoconazole on other P450 enzymes, particularly CYP27B1, limit their potential as CYP24A1 inhibitors. This has resulted in a decade long effort to develop CYP24 selective inhibitors. Schuster et al.[68] synthesized and screened a large number of azole compounds using primary human keratinocyte cultures as a source of CYP24 and CYP27B1, with 25(OH)D3as substrate to analyze the complex metabolite profiles. Among them, VID400 ((R)-N-(2-(1H-Imidazol-1-yl)-2-Phenylethyl)-4-Chlorobiphenyl-4-Carboxamide), exhibited strong and selective CYP24A1 inhibitory activity (IC50: 15 nM), with little effect on CYP27B1 activity (IC50: 616nM). VID400 occupies the substrate site in the CYP24A1 enzyme. Indicative of its potential utility, the antiproliferative effect of 1,25(OH)2D3 was enhanced 100-fold in human keratinocytes by VID400[69].

Posner et al have developed CYP24 inhibitors that are 1,25(OH)2D3 analogs[70]. These include the sulfone and sulfoximine derivatives designated CTA018 and CTA091, respectively. CTA018, which also binds the VDR and functions as a VDR agonist, is currently in Phase II clinical trials for patients with end stage renal disease and secondary hyperparathyroidism. CTA019 significantly increases 1,25(OH)2D3-mediated gene expression and anti-proliferative activity in tumor cells and also significantly and specifically increases tumor exposure to co-administered 1,25(OH)2D3 in lung tumor xenograft models[71, 72].

5. Summary

The anti-proliferative and pro-apoptotic effects of 1,25(OH)2D3 have been demonstrated in various tumor model systems in vitro and in vivo. However, limited anti-tumor effects of 1,25(OH)2D3 have been observed in clinical trials. This may be attributed to a variety of factors including the over-expression of CYP24A1 in tumors, which likely leads to the rapid local inactivation of 1,25(OH)2D3. Efforts to dissect the mechanisms responsible for CYP24 over-expression in tumors are underway. Insights gained from these studies are expected to yield novel strategies to suppress CYP24 expression and improve the efficacy of 1,25(OH)2D3treatment. An alternative strategy for improving 1,25(OH)2D3 activity involves combining it with a selective CYP24 inhibitor. The validity of this approach is supported by numerous pre-clinical investigations, which demonstrate that CYP24 inhibitors suppress 1,25(OH)2D3 catabolism by tumor cells and increase the effects of 1,25(OH)2D3 on gene expression and cell growth. Studies are now required to determine whether selective CYP24A1 inhibitors can be used safely and efficaciously in the oncology clinic and how best to combine them with a vitamin D supplementation regimen.

Highlights.

  • An overview is presented on the role of 24-hydroxylase or CYP24 in cancer

  • CYP24 is overexpressed in cancer

  • CYP24 could influence the effectiveness of vitamin D therapeutics

  • The role of CYP24 inhibitors in cancer has not been explored

Acknowledgments

The work is supported by NIH/NCI CA67267, CA85142, CA95045 and CA132844.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Bikle DD, Pillai S. Vitamin D, calcium, and epidermal differentiation. Endocr Rev. 1993;14(1):3–19. doi: 10.1210/edrv-14-1-3. [DOI] [PubMed] [Google Scholar]
  • 2.DeLuca HF. Overview of general physiologic features and functions of vitamin D. Am J Clin Nutr. 2004;80(6 Suppl):1689S–96S. doi: 10.1093/ajcn/80.6.1689S. [DOI] [PubMed] [Google Scholar]
  • 3.Muller K, Bendtzen K. 1,25-Dihydroxyvitamin D3 as a natural regulator of human immune functions. J Investig Dermatol Symp Proc. 1996;1(1):68–71. [PubMed] [Google Scholar]
  • 4.Peehl DM, et al. Antiproliferative effects of 1,25-dihydroxyvitamin D3 on primary cultures of human prostatic cells. Cancer Res. 1994;54(3):805–10. [PubMed] [Google Scholar]
  • 5.Masuda S, Jones G. Promise of vitamin D analogues in the treatment of hyperproliferative conditions. Mol Cancer Ther. 2006;5(4):797–808. doi: 10.1158/1535-7163.MCT-05-0539. [DOI] [PubMed] [Google Scholar]
  • 6.Sung V, Feldman D. 1,25-Dihydroxyvitamin D3 decreases human prostate cancer cell adhesion and migration. Mol Cell Endocrinol. 2000;164(1–2):133–43. doi: 10.1016/s0303-7207(00)00226-4. [DOI] [PubMed] [Google Scholar]
  • 7.Tanaka H, et al. 1 alpha,25-Dihydroxycholecalciferol and a human myeloid leukaemia cell line (HL-60) Biochem J. 1982;204(3):713–9. doi: 10.1042/bj2040713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Gonzalez-Sancho JM, et al. Effects of 1alpha,25-dihydroxyvitamin D3 in human colon cancer cells. Anticancer Res. 2006;26(4A):2669–81. [PubMed] [Google Scholar]
  • 9.Colston KW, et al. Vitamin D status and breast cancer risk. Anticancer Res. 2006;26(4A):2573–80. [PubMed] [Google Scholar]
  • 10.Zhang X, Nicosia SV, Bai W. Vitamin D receptor is a novel drug target for ovarian cancer treatment. Curr Cancer Drug Targets. 2006;6(3):229–44. doi: 10.2174/156800906776842939. [DOI] [PubMed] [Google Scholar]
  • 11.Chiang KC, Chen TC. Vitamin D for the prevention and treatment of pancreatic cancer. World J Gastroenterol. 2009;15(27):3349–54. doi: 10.3748/wjg.15.3349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ghous Z, et al. Inhibition of hepatocellular cancer by EB1089: in vitro and in vive study. Anticancer Res. 2008;28(6A):3757–61. [PubMed] [Google Scholar]
  • 13.White P, Cooke N. The multifunctional properties and characteristics of vitamin D-binding protein. Trends Endocrinol Metab. 2000;11(8):320–7. doi: 10.1016/s1043-2760(00)00317-9. [DOI] [PubMed] [Google Scholar]
  • 14.Cross HS. Extrarenal vitamin D hydroxylase expression and activity in normal and malignant cells: modification of expression by epigenetic mechanisms and dietary substances. Nutr Rev. 2007;65(8 Pt 2):S108–12. doi: 10.1111/j.1753-4887.2007.tb00334.x. [DOI] [PubMed] [Google Scholar]
  • 15.Schwartz GG, et al. Human prostate cells synthesize 1,25-dihydroxyvitamin D3 from 25-hydroxyvitamin D3. Cancer Epidemiol Biomarkers Prev. 1998;7(5):391–5. [PubMed] [Google Scholar]
  • 16.Chen TC, et al. Prostatic 25-hydroxyvitamin D-1alpha-hydroxylase and its implication in prostate cancer. J Cell Biochem. 2003;88(2):315–22. doi: 10.1002/jcb.10342. [DOI] [PubMed] [Google Scholar]
  • 17.Townsend K, et al. Biological actions of extra-renal 25-hydroxyvitamin D-1alpha-hydroxylase and implications for chemoprevention and treatment. J Steroid Biochem Mol Biol. 2005;97(1–2):103–9. doi: 10.1016/j.jsbmb.2005.06.004. [DOI] [PubMed] [Google Scholar]
  • 18.Prosser DE, Jones G. Enzymes involved in the activation and inactivation of vitamin D. Trends Biochem Sci. 2004;29(12):664–73. doi: 10.1016/j.tibs.2004.10.005. [DOI] [PubMed] [Google Scholar]
  • 19.Omdahl JaMBK. The 25-hydroxyvitamin D 24-hydroxylase. In: FJPJaGFH, editor. Vitamin D. 2. 2005. pp. 85–104. [Google Scholar]
  • 20.Omdahl JL, et al. Overview of regulatory cytochrome P450 enzymes of the vitamin D pathway. Steroids. 2001;66(3–5):381–9. doi: 10.1016/s0039-128x(00)00157-4. [DOI] [PubMed] [Google Scholar]
  • 21.Anderson MG, et al. Expression of VDR and CYP24A1 mRNA in human tumors. Cancer Chemother Pharmacol. 2006;57(2):234–40. doi: 10.1007/s00280-005-0059-7. [DOI] [PubMed] [Google Scholar]
  • 22.Mimori K, et al. Clinical significance of the overexpression of the candidate oncogene CYP24 in esophageal cancer. Ann Oncol. 2004;15(2):236–41. doi: 10.1093/annonc/mdh056. [DOI] [PubMed] [Google Scholar]
  • 23.Nelson DR, et al. Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants. Pharmacogenetics. 2004;14(1):1–18. doi: 10.1097/00008571-200401000-00001. [DOI] [PubMed] [Google Scholar]
  • 24.Haussler MR, et al. The nuclear vitamin D receptor: biological and molecular regulatory properties revealed. J Bone Miner Res. 1998;13(3):325–49. doi: 10.1359/jbmr.1998.13.3.325. [DOI] [PubMed] [Google Scholar]
  • 25.Endres B, Kato S, DeLuca HF. Metabolism of 1alpha,25-dihydroxyvitamin D(3) in vitamin Dreceptor-ablated mice in vivo. Biochemistry. 2000;39(8):2123–9. doi: 10.1021/bi9923757. [DOI] [PubMed] [Google Scholar]
  • 26.Pike JW, Meyer MB. The vitamin D receptor: new paradigms for the regulation of gene expression by 1,25-dihydroxyvitamin D(3) Endocrinol Metab Clin North Am. 2010;39(2):255–69. doi: 10.1016/j.ecl.2010.02.007. table of contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Burke LJ, Baniahmad A. Co-repressors 2000. FASEB J. 2000;14(13):1876–88. doi: 10.1096/fj.99-0943rev. [DOI] [PubMed] [Google Scholar]
  • 28.Leo C, Chen JD. The SRC family of nuclear receptor coactivators. Gene. 2000;245(1):1–11. doi: 10.1016/s0378-1119(00)00024-x. [DOI] [PubMed] [Google Scholar]
  • 29.Rachez C, Freedman LP. Mechanisms of gene regulation by vitamin D(3) receptor: a network of coactivator interactions. Gene. 2000;246(1–2):9–21. doi: 10.1016/s0378-1119(00)00052-4. [DOI] [PubMed] [Google Scholar]
  • 30.Rachez C, et al. Ligand-dependent transcription activation by nuclear receptors requires the DRIP complex. Nature. 1999;398(6730):824–8. doi: 10.1038/19783. [DOI] [PubMed] [Google Scholar]
  • 31.Carlberg C, Polly P. Gene regulation by vitamin D3. Crit Rev Eukaryot Gene Expr. 1998;8(1):19–42. doi: 10.1615/critreveukargeneexpr.v8.i1.20. [DOI] [PubMed] [Google Scholar]
  • 32.Chen KS, DeLuca HF. Cloning of the human 1 alpha,25-dihydroxyvitamin D-3 24-hydroxylase gene promoter and identification of two vitamin D-responsive elements. Biochim Biophys Acta. 1995;1263(1):1–9. doi: 10.1016/0167-4781(95)00060-t. [DOI] [PubMed] [Google Scholar]
  • 33.Zierold C, Darwish HM, DeLuca HF. Two vitamin D response elements function in the rat 1,25-dihydroxyvitamin D 24-hydroxylase promoter. J Biol Chem. 1995;270(4):1675–8. doi: 10.1074/jbc.270.4.1675. [DOI] [PubMed] [Google Scholar]
  • 34.Roff A, Wilson RT. A novel SNP in a vitamin D response element of the CYP24A1 promoter reduces protein binding, transactivation, and gene expression. J Steroid Biochem Mol Biol. 2008;112(1–3):47–54. doi: 10.1016/j.jsbmb.2008.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Norman AW, et al. Rapid and genomic biological responses are mediated by different shapes of the agonist steroid hormone, 1alpha,25(OH)2vitamin D3. Steroids. 1999;64(1–2):120–8. doi: 10.1016/s0039-128x(98)00091-9. [DOI] [PubMed] [Google Scholar]
  • 36.Cui M, et al. Effects of MAPK signaling on 1,25-dihydroxyvitamin D-mediated CYP24 gene expression in the enterocyte-like cell line, Caco-2. J Cell Physiol. 2009;219(1):132–42. doi: 10.1002/jcp.21657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Nutchey BK, et al. Molecular action of 1,25-dihydroxyvitamin D3 and phorbol ester on the activation of the rat cytochrome P450C24 (CYP24) promoter: role of MAP kinase activities and identification of an important transcription factor binding site. Biochem J. 2005;389(Pt 3):753–62. doi: 10.1042/BJ20041947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Cui M, et al. The effect of differentiation on 1,25 dihydroxyvitamin D-mediated gene expression in the enterocyte-like cell line, Caco-2. J Cell Physiol. 2009;218(1):113–21. doi: 10.1002/jcp.21574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yang BS, et al. Ras-mediated phosphorylation of a conserved threonine residue enhances the transactivation activities of c-Ets1 and c-Ets2. Mol Cell Biol. 1996;16(2):538–47. doi: 10.1128/mcb.16.2.538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dwivedi PP, et al. Role of MAP kinases in the 1,25-dihydroxyvitamin D3-induced transactivation of the rat cytochrome P450C24 (CYP24) promoter. Specific functions for ERK1/ERK2 and ERK5. J Biol Chem. 2002;277(33):29643–53. doi: 10.1074/jbc.M204561200. [DOI] [PubMed] [Google Scholar]
  • 41.Tolon RM, et al. Association with Ets-1 causes ligand- and AF2-independent activation of nuclear receptors. Mol Cell Biol. 2000;20(23):8793–802. doi: 10.1128/mcb.20.23.8793-8802.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dhawan P, Christakos S. Novel regulation of 25-hydroxyvitamin D3 24-hydroxylase (24(OH)ase) transcription by glucocorticoids: cooperative effects of the glucocorticoid receptor, C/EBP beta, and the Vitamin D receptor in 24(OH)ase transcription. J Cell Biochem. 2010;110(6):1314–23. doi: 10.1002/jcb.22645. [DOI] [PubMed] [Google Scholar]
  • 43.Hidalgo AA, et al. Dexamethasone enhances 1{alpha},25-dihydroxyvitamin D3 effects by increasing vitamin D receptor transcription. J Biol Chem. 2011 doi: 10.1074/jbc.M111.244061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Leader JE, et al. Epigenetic regulation of nuclear steroid receptors. Biochem Pharmacol. 2006;72(11):1589–96. doi: 10.1016/j.bcp.2006.05.024. [DOI] [PubMed] [Google Scholar]
  • 45.Novakovic B, et al. Placenta-specific methylation of the vitamin D 24-hydroxylase gene: implications for feedback autoregulation of active vitamin D levels at the fetomaternal interface. J Biol Chem. 2009;284(22):14838–48. doi: 10.1074/jbc.M809542200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Vieth R. How to optimize vitamin D supplementation to prevent cancer, based on cellular adaptation and hydroxylase enzymology. Anticancer Res. 2009;29(9):3675–84. [PubMed] [Google Scholar]
  • 47.Wjst M, et al. Epigenetic regulation of vitamin D converting enzymes. J Steroid Biochem Mol Biol. 2010;121(1–2):80–3. doi: 10.1016/j.jsbmb.2010.03.056. [DOI] [PubMed] [Google Scholar]
  • 48.Horvath HC, et al. The candidate oncogene CYP24A1: A potential biomarker for colorectal tumorigenesis. J Histochem Cytochem. 2010;58(3):277–85. doi: 10.1369/jhc.2009.954339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chen G, et al. CYP24A1 is an independent prognostic marker of survival in patients with lung adenocarcinoma. Clin Cancer Res. 2011;17(4):817–26. doi: 10.1158/1078-0432.CCR-10-1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Komagata S, et al. Human CYP24 catalyzing the inactivation of calcitriol is post-transcriptionally regulated by miR-125b. Mol Pharmacol. 2009;76(4):702–9. doi: 10.1124/mol.109.056986. [DOI] [PubMed] [Google Scholar]
  • 51.Khorchide M, Lechner D, Cross HS. Epigenetic regulation of vitamin D hydroxylase expression and activity in normal and malignant human prostate cells. J Steroid Biochem Mol Biol. 2005;93(2–5):167–72. doi: 10.1016/j.jsbmb.2004.12.022. [DOI] [PubMed] [Google Scholar]
  • 52.Matilainen JM, et al. The number of vitamin D receptor binding sites defines the different vitamin D responsiveness of the CYP24 gene in malignant and normal mammary cells. J Biol Chem. 2010;285(31):24174–83. doi: 10.1074/jbc.M110.124073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Albertson DG, et al. Quantitative mapping of amplicon structure by array CGH identifies CYP24 as a candidate oncogene. Nat Genet. 2000;25(2):144–6. doi: 10.1038/75985. [DOI] [PubMed] [Google Scholar]
  • 54.Kallioniemi A, et al. Detection and mapping of amplified DNA sequences in breast cancer by comparative genomic hybridization. Proc Natl Acad Sci U S A. 1994;91(6):2156–60. doi: 10.1073/pnas.91.6.2156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Luo W, et al. Epigenetic regulation of vitamin D 24-hydroxylase/CYP24A1 in human prostate cancer. Cancer Res. 2010;70(14):5953–62. doi: 10.1158/0008-5472.CAN-10-0617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Deeb KK, et al. Differential vitamin D 24-hydroxylase/CYP24A1 gene promoter methylation in endothelium from benign and malignant human prostate. Epigenetics. 2011;6(8) doi: 10.4161/epi.6.8.16536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Miller GJ, et al. Vitamin D receptor expression, 24-hydroxylase activity, and inhibition of growth by 1alpha,25-dihydroxyvitamin D3 in seven human prostatic carcinoma cell lines. Clin Cancer Res. 1995;1(9):997–1003. [PubMed] [Google Scholar]
  • 58.Parise RA, et al. CYP24, the enzyme that catabolizes the antiproliferative agent vitamin D, is increased in lung cancer. Int J Cancer. 2006;119(8):1819–28. doi: 10.1002/ijc.22058. [DOI] [PubMed] [Google Scholar]
  • 59.Schuster I, et al. Selective inhibitors of vitamin D metabolism--new concepts and perspectives. Anticancer Res. 2006;26(4A):2653–68. [PubMed] [Google Scholar]
  • 60.Reinhardt TA, Horst RL. Ketoconazole inhibits self-induced metabolism of 1,25-dihydroxyvitamin D3 and amplifies 1,25-dihydroxyvitamin D3 receptor up-regulation in rat osteosarcoma cells. Arch Biochem Biophys. 1989;272(2):459–65. doi: 10.1016/0003-9861(89)90240-3. [DOI] [PubMed] [Google Scholar]
  • 61.Ly LH, et al. Liarozole acts synergistically with 1alpha,25-dihydroxyvitamin D3 to inhibit growth of DU 145 human prostate cancer cells by blocking 24-hydroxylase activity. Endocrinology. 1999;140(5):2071–6. doi: 10.1210/endo.140.5.6698. [DOI] [PubMed] [Google Scholar]
  • 62.Yee SW, Simons C. Synthesis and CYP24 inhibitory activity of 2-substituted-3,4-dihydro-2H-naphthalen-1-one (tetralone) derivatives. Bioorg Med Chem Lett. 2004;14(22):5651–4. doi: 10.1016/j.bmcl.2004.08.040. [DOI] [PubMed] [Google Scholar]
  • 63.Yee SW, Campbell MJ, Simons C. Inhibition of Vitamin D3 metabolism enhances VDR signalling in androgen-independent prostate cancer cells. J Steroid Biochem Mol Biol. 2006;98(4–5):228–35. doi: 10.1016/j.jsbmb.2005.11.004. [DOI] [PubMed] [Google Scholar]
  • 64.Shanmugam MK, Kannaiyan R, Sethi G. Targeting cell signaling and apoptotic pathways by dietary agents: role in the prevention and treatment of cancer. Nutr Cancer. 2011;63(2):161–73. doi: 10.1080/01635581.2011.523502. [DOI] [PubMed] [Google Scholar]
  • 65.Farhan H, Cross HS. Transcriptional inhibition of CYP24 by genistein. Ann N Y Acad Sci. 2002;973:459–62. doi: 10.1111/j.1749-6632.2002.tb04683.x. [DOI] [PubMed] [Google Scholar]
  • 66.Rao A, et al. Vitamin D receptor and p21/WAF1 are targets of genistein and 1,25-dihydroxyvitamin D3 in human prostate cancer cells. Cancer Res. 2004;64(6):2143–7. doi: 10.1158/0008-5472.can-03-3480. [DOI] [PubMed] [Google Scholar]
  • 67.Swami S, et al. Genistein potentiates the growth inhibitory effects of 1,25-dihydroxyvitamin D3 in DU145 human prostate cancer cells: role of the direct inhibition of CYP24 enzyme activity. Mol Cell Endocrinol. 2005;241(1–2):49–61. doi: 10.1016/j.mce.2005.05.001. [DOI] [PubMed] [Google Scholar]
  • 68.Schuster I, et al. Inhibitors of vitamin D hydroxylases: structure-activity relationships. J Cell Biochem. 2003;88(2):372–80. doi: 10.1002/jcb.10365. [DOI] [PubMed] [Google Scholar]
  • 69.Schuster I, et al. Combination of vitamin D metabolites with selective inhibitors of vitamin D metabolism. Recent Results Cancer Res. 2003;164:169–88. doi: 10.1007/978-3-642-55580-0_13. [DOI] [PubMed] [Google Scholar]
  • 70.Posner GH, et al. Vitamin D analogues targeting CYP24 in chronic kidney disease. J Steroid Biochem Mol Biol. 2010;121(1–2):13–9. doi: 10.1016/j.jsbmb.2010.03.065. [DOI] [PubMed] [Google Scholar]
  • 71.Zhang Q, et al. CYP24 inhibition preserves 1alpha,25-dihydroxyvitamin D(3) anti-proliferative signaling in lung cancer cells. Mol Cell Endocrinol. 2012;355(1):153–61. doi: 10.1016/j.mce.2012.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Beumer JH, et al. A local effect of CYP24 inhibition on lung tumor xenograft exposure to 1,25-dihydroxyvitamin D(3) is revealed using a novel LC-MS/MS assay. Steroids. 2012;77(5):477–83. doi: 10.1016/j.steroids.2012.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES