Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • Log out
  • My Cart
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleMETABOLISM, TRANSPORT, AND PHARMACOGENOMICS

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

Jianghong Fan, Shanjun Liu, Yimin Du, Jodi Morrison, Robert Shipman and K. Sandy Pang
Journal of Pharmacology and Experimental Therapeutics August 2009, 330 (2) 389-402; DOI: https://doi.org/10.1124/jpet.108.149815
Jianghong Fan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shanjun Liu
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yimin Du
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jodi Morrison
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Robert Shipman
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
K. Sandy Pang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

The effects of 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3] on gene expression and function were studied in Caco-2 cells. Microarray analyses, real-time quantitative polymerase chain reactions, and Western blotting were used to determine the mRNA and protein expression of transporters and enzymes after 1,25(OH)2D3 or vehicle (0.1% ethanol) treatment for 1, 3, 6, and 10 days. The mRNA and protein expressions of the apical sodium-dependent bile acid transporter, oligopeptide transporter 1, multidrug resistance-associated protein (MRP) 3, and sulfotransferase 1E1 remained unchanged with 1,25(OH)2D3 treatment, whereas those for CYP3A4, multidrug resistance protein 1, and MRP2 were significantly increased (P < 0.05). 1,25(OH)2D3 treatment significantly enhanced MRP4 protein expression by increasing protein stability without affecting mRNA expression, as confirmed in cycloheximide experiments. Marked increase in 6β-hydroxylation of testosterone by CYP3A4 was also observed in the 6-day 1,25(OH)2D3-treated (100 nM) cell lysate. The transport of [3H]digoxin, the P-glycoprotein (P-gp) substrate, after treatment with 100 nM 1,25(OH)2D3 for 3 days revealed a higher apparent permeability (Papp) value in the basal (B)-to-apical (A) direction over that of vehicle treatment (15.1 ± 0.53 × 10-6 versus 11.8 ± 0.58 × 10-6 cm/s; P < 0.05), whereas the Papp in the A-to-B direction was unchanged; the efflux ratio was increased (from 5.8 to 8.0). Reduced cellular retention of 5-(and-6)-carboxy-2′,7′-dichlorofluorescein, suggestive of higher MRP2 activity, was observed in the 3-day 100 nM 1,25(OH)2D3-treated cells over controls. Higher protein expression of CYP3A4, MRP2, P-gp, and MRP4 was also observed after a 6-day treatment with other vitamin D analogs (100 nM 1α-hydroxyvitamin D3,1α-hydroxyvitamin D2 or Hectorol, and 25-hydroxyvitamin D3) in Caco-2 cells, suggesting a role of 1,25(OH)2D3 and analogs in the activation of enzymes and transporters via the vitamin D receptor.

The intestine plays an important role in the absorption of orally administered drugs. The expression and proximity of metabolic enzymes and efflux transporters in the enterocyte contribute to intestinal first-pass removal and delimit the tissue accumulation of endo- and xenobiotics. In the small intestine, cytochrome P450 3A4 (CYP3A4) accounts for approximately 70% of total cytochrome P450 content and is responsible for the metabolism of approximately 50% of drugs currently in use (Pelkonen et al., 2008). The ATP-dependent drug efflux P-glycoprotein (P-gp), encoded by the multidrug resistance 1 gene (MDR1) located in the apical membrane of the enterocyte, is involved in the active excretion of a variety of lipophilic, cationic drugs from the intestine (Artursson et al., 2001). The multidrug resistance-associated proteins (MRPs), another important subfamily of ATP-binding cassette transporters, are involved in the transport of unconjugated amphiphilic anions and glutathione, glucuronide, and sulfate conjugates. MRP2 is an efflux transporter localized in the apical membrane, whereas MRP1, MRP3, and MRP5 are basolateral efflux transporters. The localization of MRP4 in enterocytes has been inferred to exist at the basolateral membrane for efflux (Prime-Chapman et al., 2004).

The expression and function of drug metabolic enzymes and transporters are under regulation of nuclear receptors. In addition to the pregnane X receptor (PXR), the constitutive androstane receptor (CAR), farnesoid X receptor, and glucocorticoid receptor (for review, see Urquhart et al., 2007), the vitamin D receptor (VDR) is known to induce enzyme and transporter genes in the intestine. These include the human CYP3A4 (Thummel et al., 2001), SULT2A1 (Song et al., 2006), murine Mrp3 (McCarthy et al., 2005), and the rat apical sodium-dependent bile acid transporter (Asbt) (Chen et al., 2006).

1α,25-Dihydroxyvitamin D3 [1,25(OH)2D3], the biologically active form of vitamin D, is the natural ligand of the VDR. Vitamin D is synthesized in the skin from its precursor, 7-dehydrocholesterol, in response to ultraviolet light, and it is converted to 25-hydroxyvitamin D3 [25(OH)D3] in the liver and then to 1,25(OH)2D3 by 1α-hydroxylase in the kidney (Darwish and DeLuca, 1996). Upon activation, the ligand-receptor complex recruits coactivators and heterodimerizes with the retinoid X receptor and then binds to the vitamin D response element (VDRE) and regulates target gene expression. During the past three decades, evidence has accumulated that 1,25(OH)2D3 mediates its biological activities through specific binding to the nuclear VDR. The therapeutic potential of 1,25(OH)2D3 is limited by its tendency to induce hypercalcemia (Prudencio et al., 2001). Vitamin D analogs that have lower hypercalcemic effects are potential therapeutic agents that can be used for treatment of human diseases and disorders (Stein and Wark, 2003). 1α-Hydroxyvitamin D3 [1α(OH)D3] is a prodrug of the active form of vitamin D3 and undergoes metabolic conversion by 25-hydroxylase in the liver or intestine to 1,25(OH)2D3 before exerting its effect (Brickman et al., 1976). 1α-Hydroxyvitamin D2 [1α(OH)D2] or Hectorol is activated by 25-hydroxylase to produce 1,25(OH)2D2, a biologically active form of vitamin D2, and it is reported to display significantly reduced hypercalcemia at therapeutic dosages for the treatment of secondary hyperparathyroidism (Brown, 2001).

The Caco-2 cell monolayer is a well established human carcinoma cell line that differentiates spontaneously in culture upon reaching confluence to become enterocyte-like cells that contain transporters and enzymes (Artursson et al., 2001). Effects of 1,25(OH)2D3 and the vitamin D analogs on the expression and activity of enzymes and transporters in the human intestine may thus be conveniently ascertained in Caco-2 cells due to high expression level of the VDR. Most transporters, with the exception of breast cancer resistance protein, are present at reasonably high levels (Taipalensuu et al., 2001). It had been demonstrated that CYP3A4 expression was up-regulated by 1,25(OH)2D3 via binding to the VDRE (Schmiedlin-Ren et al., 1997). More recently, a functional VDRE has been identified in the human MDR1 gene (Saeki et al., 2008). However, the role of the VDR on other target genes is unknown. In this study, we investigated the effects of 1,25(OH)2D3 and the vitamin D analogs on the expression and function of major metabolic enzymes and transporters in Caco-2 cells. To confirm that CYP3A4 function was catalytically induced by 1,25(OH)2D3 in Caco-2 cells, the activity of testosterone 6β-hydroxylase, a marker of induction of CYP3A4 (Chan et al., 2004), was measured. Digoxin, a prototypic substrate of P-gp, was used to evaluate P-gp function in Caco-2 cells (Cavet et al., 1996), whereas the fluorescent MRP2 substrate, 5-(and-6)-carboxy-2′,7′-dichlorofluorescein (CDF), whose diacetate prodrug 5-(and-6)-carboxy-2′,7′-dichlorofluorescein diacetate (CDF-DA) readily diffuses across the cell membrane for hydrolysis by intracellular esterases to form CDF, was used to evaluate MRP2 function (Tian et al., 2004).

Materials and Methods

Materials

1α,25(OH)2D3, lα(OH)D3, 25(OH)D3, cycloheximide, and glucose were purchased from Sigma-Aldrich Canada (Mississauga, ON, Canada). lα(OH)D2 was kindly provided by Dr. Peter Bonate (Genzyme, Cambridge, MA). Testosterone and 6β-hydroxytestosterone were obtained from Sigma-Aldrich (St. Louis, MO). CDF-DA, Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum, 0.05% trypsin-EDTA, penicillin-streptomycin, and nonessential amino acids were all obtained from Invitrogen (Carlsbad, CA). A 12-well Transwell plate was purchased from Corning Life Sciences (Lowell, MA). [3H]Digoxin (specific activity, 40 mCi/μmol), whose purity exceeded 98% as verified by HPLC, and [14C]mannitol (specific activity, 51 mCi/mmol) were procured from PerkinElmer Life and Analytical Sciences (Boston, MA). The materials and reagents for real-time quantitative polymerase chain reaction (RT-qPCR) were purchased from Applied Biosystems (Foster City, CA). Reagents for mRNA sample preparation were from Sigma-Aldrich and Ambion (Austin, TX). Anti-CYP3A4, MRP2, and P-gp antibodies were purchased from Abcam (Cambridge, MA). Anti-villin antibody was obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). Anti-VDR and anti-Cyp3A4 antibody were from Thermo Fisher Scientific (Waltham, MA) and BD Biosciences (Mississauga, ON, Canada), respectively. Donkey anti-rabbit IgG, sheep anti-mouse IgG, and the enhanced chemiluminescence were purchased from GE Healthcare (Baie d'Urfe, QC, Canada). Other antibodies were kindly provided by the following investigators: anti-PEPT1 (Dr. Wolfgang Sadee, The Ohio State University, Columbus, OH), anti-ASBT (Dr. Paul A. Dawson, Wake Forest University School of Medicine, Winston-Salem, NC), anti-MRP3 (Dr. Yuichi Sugiyama, University of Tokyo, Tokyo, Japan), and anti-MRP4 (Dr. John D. Schuetz, St. Jude Children's Research Hospital, Memphis, TN).

Cell Culture

Caco-2 cells were obtained from the American Type Culture Collection (Manassas, VA). Cells were allowed to undergo two passages before use. Cells then were cultured in DMEM supplemented with 10% fetal bovine serum, 1% nonessential amino acids, 100 U/ml penicillin, and 100 μg/ml streptomycin under an atmosphere of 5% CO2 and 95% relative humidity at 37°C. Cells used in the studies were between passages 30 and 50. Caco-2 cells were seeded in a density of 2.5 × 104 cells/cm2 in 60-mm dishes; the medium was changed every other day, with the exception of treatment days. For the treatment with 1,25(OH)2D3 and vitamin D analogs (10 or 100 nM), cells were treated with control medium containing 0.1% ethanol or 1,25(OH)2D3 and vitamin D analogs in 0.1% ethanol daily on day 15 for six consecutive days, on day 18 for three consecutive days, or on day 20 for 1 day; cells were then harvested on day 21. For studies investigating the effect of 1,25(OH)2D3 on MRP4 protein stability, Caco-2 cells were first treated with 100 nM 1,25(OH)2D3 or ethanol (0.1%) for 6 days and then with cycloheximide (10 μg/ml) to arrest protein synthesis, followed by protein isolation for MRP4 determination at 0, 8, 24, and 48 h after the cycloheximide treatment. For studies determining the effect of 25(OH)D3 on gene expression in the presence or absence of FBS, Caco-2 cells were treated with 100 nM 1,25(OH)2D3, 25(OH)D3, or ethanol (0.1%) for 6 days in the presence of 10% FBS or in the serum-free condition, wherein a 1% mixture of insulin-transferrin-selenium (ITS; Invitrogen) was used instead.

DTEx Microarray Analysis

Total RNA (∼2 μg) was converted to aRNA using the MessageAmp II aRNA amplification kit (ABI, Chicago IL; and Ambion) according to the manufacturers' instructions. Labeled cDNA was prepared from purified aRNA using Cy5-labeled random nonamers and SuperScript II reverse transcriptase (Invitrogen, Burlington, ON, Canada) according to the manufacturer's instructions. Cy5-labeled cDNA was purified on QIAquik PCR purification columns (QIAGEN, Mississauga ON, Canada). Purified Cy5-labeled cDNA was quantified using a spectrophotometer (NanoDrop Inc., Wilmington, DE). Cy5-labeled cDNA (∼3 μg) was denatured at 95°C for 10 min and hybridized to a DTEx microarray slide [145 absorption, distribution, metabolism, and excretion-associated genes printed on Nexterion E slides (Schott, Louisville, KY)] in Nexterion 1× Hyb solution for 18 h at 45°C. Before hybridization, each DTEx microarray slide was denatured in boiled water for 60 s and blocked in Nexterion Block E for 15 min at 45°C. After hybridization, the DTEx microarray slides were washed once for 15 min at 45°C in each of the following solutions: 2× SSC/0.2% SDS, 2× SSC, and 0.2× SSC. The DTEx microarray slides were “spin-dried” at 1200 rpm for 2 min at room temperature and then scanned in a ProScanArray HT (PerkinElmer, Woodbridge, ON, Canada). After image acquisition, fluorescent signals from the DTEx microarray were assessed using QuantArray. Results were exported to GeneLinker Gold (Improved Outcomes Software, Kingston, ON, Canada) for normalization, hierarchical cluster analysis, and matrix plot images. Four independent DTEx microarray experiments were performed for each RNA sample in this study. Each gene on the DTEx microarray was printed in quadruplicate, resulting in n = 16 for each gene interrogated in the DTEx microarray analysis.

Real-Time Quantitative Polymerase Chain Reactions

RNA levels of the drug transporters and enzymes were analyzed by RT-qPCR. Total RNA was isolated from cells using TRI Reagent (Sigma Chemical Co., Oakville, ON, Canada). The first-strand cDNA was synthesized by using the First Strand cDNA synthesis kit (Fermentas, Burlington, ON, Canada). cDNA samples then were subjected to qPCR assays with the fluorescent dye SYBR Green methodology and an ABI 7500 detector (Applied Biosystems, Foster City, CA). Gene-specific primers, designed with the Primer3 software (Whitehead Institute for Biomedical Research, Cambridge, MA), are summarized in Table 1. Primers were optimized and validated with the comparative CT method, as described in the manufacturer's manual. Reactions were performed using the following protocol: 95°C for 5 min, followed by 40 cycles of 95°C for 15 s and 60°C for 1 min. A dissociation curve protocol was linked to every PCR assay to ensure the specificity of the PCR product. The expression of specific genes was normalized to the value of villin mRNA of the same sample. Relative quantification was obtained by the comparative threshold cycle (ΔΔCT) method (RQ software, version 1.2.3; Applied Biosystems).

View this table:
  • View inline
  • View popup
TABLE 1

Primer sets for quantitative real-time PCR

Western Blot Analysis

Cellular proteins were extracted by dissolving cells in the CelLytic cell lysis reagent (Sigma-Aldrich). Protein concentration was measured by the method of Lowry et al. (1951). Sixty micrograms of whole cell protein was mixed with 4× gel loading buffer (250 mM Tris-HCl, pH 6.8, 8% SDS, 20% glycerol, 0.2% bromphenol blue, and 0.4 M dithiothreitol), separated on a 7.5% SDS-polyacrylamide gel under reducing conditions, and transferred to nitrocellulose membranes (Hybond ECL; GE Healthcare). Nonspecific antibody binding was blocked by preincubation of the membranes with 5% skim milk in 1× Tris-buffered saline for 1 h at room temperature. Membranes were then incubated overnight at 4°C with the appropriate primary antibodies at different dilutions with 2% skim milk in 1× Tris-buffered saline containing 0.1% Tween 20. After washing, they were incubated with donkey anti-rabbit IgG or sheep anti-mouse IgG at 1:1000 dilutions for 1 h at room temperature. Bands were visualized according to the enhanced chemiluminescence kit per the manufacturer's instructions. Villin served as loading control. Band intensity was analyzed with the ImageJ 1.32 software (National Institutes of Health, Bethesda, MD), and protein expression was presented as the ratio of target proteins band intensity to villin band intensity in the same blot.

Functional Studies in Caco-2 Cells

CYP3A4-Mediated Testosterone 6β-Hydroxylation after 6 Days of Treatment with 100 nM 1,25(OH)2D3. Cell lysate (4 mg), testosterone (200 μM), and Tris-HCl buffer (50 mM; pH 7.4) in a volume of 470 μl were preincubated for 5 min at 37°C. Thereafter, an NADPH-regenerating system composed of 25 μl of solution A and 5 μl of solution B (BD Biosciences) was added to initiate the reaction. The reaction was terminated at 60 min upon addition of 1 ml of ice-cold acetonitrile that contained 500 ng/ml desmethyldiazepam (internal standard for HPLC). The solution was added to 25 μl of 1 N NaOH and extracted against 5 ml of ethyl acetate after mixing and centrifugation at 5000g (for 5 min). The supernatant was removed and dried under nitrogen gas and then reconstituted in 100 μl of methanol. The 6β-hydroxytestosterone and testosterone in the reconstituted sample were assayed by a validated HPLC method with the following conditions (Brimer et al., 2000): the HPLC system (Shimadzu, Kyoto, Japan) consisted of an SCL-10A system controller, LC-10AT pump, and SIL-10A XL autoinjector; and an Altima C18 reverse-phase column (4.6 × 250 mm; particle size, 10 μm; Alltech Associates, Deerfield, IL) and precolumn [2.2 × 0.34 cm i.d. filled with μBondapak C18/Corasil 37–55 μm (Waters, Milford, MA)]. The mobile phase, consisting of methanol and water (58:42, v/v), was used at the flow rate of 1.0 ml/min. The effluent was monitored at 254 nm with an SPD-10A UV detector (Shimadzu). Data acquisition and integration were performed with the Star-Chrom LITE HPLC data system software (D-Star Instruments, Manassas, VA). The retention times of 6β-hydroxytestosterone and testosterone were 9.2 and 22 min, respectively. The enzymatic activity of CYP3A4 was expressed as picomoles of 6β-hydroxytestosterone formed per gram of protein per minute.

    Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

DTEx microarray analysis of enzyme and transporter gene expression after ethanol (control; 0.1% as vehicle) and 1,25(OH)2d3 (0–100 nM) treatment for 0, 1, 3, 6, or 10 days (D1, D3, D6, or D10). Expression levels of genes that were regulated by 1,25(OH)2d3 (A) and genes displaying the highest differential levels of expression in response to 1,25(OH)2d3 treatment (10 and 100 nM, B) in Caco-2 cells for 1, 3, 6, or 10 consecutive days; ethanol (0.1%) was used for treatment in the control group. Cells were then harvested on day 21, and the expression levels of 1,25(OH)2d3-regulated genes were determined by DTEx microarray analysis and normalized to that of the house-keeping gene glyceraldehyde-3-phosphate dehydrogenase.

Transport Studies for [3H]Digoxin (5 μM) in Transwell Plates after 3 Days of Treatment with 10 and 100 nM 1,25(OH)2D3. For transport studies, Caco-2 cells were used at passages between 39 and 50 and seeded in a density of 8 × 104 cells/cm2 in Transwell plates with a pore size of 0.4 μm and a growth area of 1.13 cm2 (Corning Inc., Corning, NY). The medium was changed every other day up to day 10 and then daily thereafter. Before and after the transport experiment was performed, the integrity of the monolayer was checked by measurement of the transepithelial electrical resistance (TEER) values with a millicell-ERS volt/ohm meter (Millipore, Billerica, MA). Monolayers that demonstrated a resistance value between 600 and 1000 Ω · cm2 were used for the transport experiments. Routine analysis of TEER values showed normal development of the tight junctions (>600 Ω · cm2). [3H]Digoxin (5960 ± 3.69 × 103 dpm/ml with 5 μM unlabeled digoxin) or CDF-DA (10 μM) was dissolved in dimethyl sulfoxide (0.1%). [14C]Mannitol (1.90 ± 0.52 × 103 dpm/ml), a marker of paracellular transport, was used for each set of studies to assess membrane integrity.

The cell monolayer at 21 to 24 days of culture was used for transport studies. The procedure was similar to that described previously (Artursson et al., 2001). Before the experiment, the incubation medium was gently suctioned off and replaced by prewarmed transport HBSS buffer and incubated for 60 min at 37°C thereafter. The transport experiment for the apical-to-basolateral (A→B) or basolateral-to-apical (B→A) direction was initiated upon gentle removal of the transport buffer in the donor compartment and placement of 0.55 or 1.55 ml of [3H]digoxin into the donor compartment; the receiver compartment was filled with 1.5 or 0.5 ml of blank transport buffer. An aliquot of 0.05 ml of sample was quickly withdrawn from the donor compartment to define the loading concentration, C0, to result in starting volumes of 0.5 ml in the apical compartment and 1.5 ml in the basolateral compartment. TEER values were measured at various stages to ensure that the integrity of the monolayer was retained with the buffer changes. At 20, 40, 60, and 90 min, 750 and 250 μl were removed from the receiver compartment for the A→B and B→A transport studies, respectively. The volume removed was replenished by prewarmed blank transport buffer to ensure the sink conditions. At the end of the experiment, 250 and 750 μl of sample were collected from the apical side and the basolateral side, respectively. Cells collected at the end of the transport study were washed three times with 2 ml of ice-cold HBSS; 750 μl of NaOH (0.2 N) was added to digest the cells, and the sample was back-neutralized by the addition of 150 μl of HCl (1 N).

    Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Effects of 1,25(OH)2d3 on VDR mRNA and protein expression in Caco-2 cells. Caco-2 cells were treated with 1,25(OH)2d3 (10 or 100 nM) on day 20, 18, 15, or 11 for 1, 3, 6, or 10 consecutive days, whereas the control group was treated with ethanol (0.1%). Cells were harvested on day 21, and the expression of VDR mRNA and protein was determined by using qPCR and Western blotting and normalized to that of the house-keeping gene villin. A, data are the mean ± S.D. of qPCR quantifications from three independent cultures. B, data are the mean ± S.D. of the density histograms from three separate Western blot analyses. *, P < 0.05 between the 1,25(OH)2d3-treated and ethanol-treated groups.

    Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Effects of 1,25(OH)2d3 on CYP3A4 expression in Caco-2 cells. Caco-2 cells were treated with 1,25(OH)2d3 (10 or 100 nM) on day 20, 18, 15, or 11 for 1, 3, 6, or 10 consecutive days, whereas the control group was treated with ethanol (0.1%). Cells were harvested on day 21 and the expression of mRNA and protein was determined by using qPCR and Western blotting and normalized to that of the house-keeping gene villin. A, data are the mean ± S.D. of qPCR quantifications from three independent cultures. B, data are the mean ± S.D. of the density histograms from three separate Western blot analyses. *, P < 0.05 between the 1,25(OH)2d3-treated and ethanol-treated groups.

The radioactivities of samples were measured by liquid scintillation spectrometry (model 5801 beta counter; Beckman Coulter Canada, Inc., Mississauga, ON, Canada) upon addition of 9.5 ml of scintillation fluor (Ready Safe; Beckman Coulter Canada, Inc.). The cellular contents and amounts in the donor and receiver compartments were used for the mass balance considerations or the estimation of recovery. The apparent permeability coefficient (Papp, centimeter per second) of [3H]digoxin across Caco-2 monolayers was expressed in centimeter per second and calculated as shown in eq. 1: Embedded Image where ΔAR is the amount accumulated in the receiver side during the interval time (Δt), area is the surface area of the filter/membrane of the Transwell (square centimeters), and C0 is the initial drug concentration in the donor chamber. The efflux ratio, EfR is given by the ratio of Papp in the (B→A) to that in the (A→B) direction: Math

CDF Transport Studies in Transwell Plates after 3 Days of Treatment with 10 and 100 nM 1,25(OH)2D3 in Caco-2 Cells. For the definition of MRP2 function, 10 μM CDF-DA, the nonfluorescent prodrug that was dissolved in transport buffer, was added into either the apical or basolateral compartment of the Transwell plates. At 10, 20, and 40 min, 750 and 250 μl were removed from the receiver compartment for the A→B and B→A transport studies, respectively. The volume removed was replenished by prewarmed, blank transport buffer to ensure the sink conditions. At the end of the experiment, cells were lysed with 500 μl of 1% Triton X-100 in PBS for 20 min at room temperature. All retrieved samples were stored at -20°C until analyses. The extent of transport (transport efficiency) of the fluorescent CDF metabolite in the A-to-B or B-to-A direction was assessed as shown in eq. 3: Math CDF fluorescence in the receiver compartment and in cell lysate were analyzed by a microplate fluorescence reader (λex = 485 nm; λem = 590 nm) (SpectraMax Gemini XS; Molecular Devices, Sunnyvale CA).

    Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Effects of 1,25(OH)2d3 on ASBT and PEPT1 expression in Caco-2 cells. Caco-2 cells were treated with 1,25(OH)2d3 (10 or 100 nM) on day 20, 18, 15, or 11 for 1, 3, 6, or 10 consecutive days, whereas the control group was treated with ethanol (0.1%). Cells were harvested on day 21, and the expression of mRNA and protein was determined by using qPCR and Western blotting and normalized to that of the house-keeping gene villin. A and C, data are the mean ± S.D. of qPCR quantifications from three independent cultures. B and D, data are the mean ± S.D. of the density histograms from three separate Western blot analyses. *, P < 0.05 between the 1,25(OH)2d3-treated and ethanol-treated groups.

CDF Accumulation in Caco-2 Cells in Culture Dish. The extent of accumulation of CDF, an index of MRP2 function, in Caco-2 cells (Tian et al., 2004) was also assessed in Caco-2 cells grown on a culture dish. Caco-2 cells were seeded with a density of 2.5 × 104 cells/cm2 in a six-well plate and treated with control medium containing 0.1% ethanol or 1,25(OH)2D3 (100 nM) daily on day 18 for three consecutive days. On day 21, triplicate plates were washed three times with HBSS and incubated with 10 μM CDF-DA in transport buffer for 30 min at 37°C. At the end of incubation, CDF fluorescence in the lysed cells was assayed.

Data Analysis

Student's t test and analysis of variance were used to determine statistical difference between control and treatment groups. Results were expressed as mean ± S.D. A P value of <0.05 was considered as statistically significant.

Results

DTEx Microarray Analysis of Enzymes and Transporters in 1,25(OH)2D3-Treated Caco-2 Cells

DTEx microarray analysis of Caco-2 cells treated with 1,25(OH)2D3 (0, 10, and 100 nM for 1, 3, 6, and 10 days) showed that maximal differential expression occurred after 6 days of treatment with 10 nM 1,25(OH)2D3. At this point, 99 genes were up-regulated at least 2-fold (range, 2–20-fold) compared with vehicle-treated controls. This group of differentially expressed, up-regulated genes included 37 ABC transporters (range, 2–16-fold), 13 cytochromes P450 (range, 2–20-fold), 15 nuclear xenobiotic receptors (range, 2–10-fold), 23 solute ligand carrier transporters (range, 2–15-fold), 5 sulfotransferases (range, 2–15-fold), and 6 UDP-glucuronosyltransferases (range, 2–11-fold). Genes that were regulated by 1,25(OH)2D3 treatment are shown in Fig. 1A. The 10 most up-regulated genes are shown in Fig. 1B. A close alignment between 1,25(OH)2D3 concentration and days of induction with increased DTEx signal was not achieved, an observation that probably can be attributed to the toxic effects of 1,25(OH)2D3 in the system.

    Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Effect of 1,25(OH)2d3 on P-gp and MRP2 expression in Caco-2 cells. Caco-2 cells were treated with 1,25(OH)2d3 (10 or 100 nM) on day 20, 18, 15, or 11 for 1, 3, 6, or 10 consecutive days, and the control group was treated with ethanol (0.1%). Cells were harvested on day 21, and the expression of mRNA and protein were determined by using qPCR and Western blotting and normalized to that of the house-keeping gene villin. A and C, data are the mean ± S.D. of qPCR quantifications from three independent cultures. B and D, data are the mean ± S.D. of the density histograms from three separate Western blot analyses. *, P < 0.05 between the 1,25(OH)2d3-treated and ethanol-treated groups.

Effects of 1,25(OH)2D3 on Gene Expression in Caco-2 Cells

The mRNA and protein levels of the house-keeping gene villin were unchanged and independent of the duration and concentration of 1,25(OH)2D3 used in the treatments. There was a decrease in VDR mRNA level in response to 1,25(OH)2D3; a decreasing trend of VDR protein expression, although statistically insignificant, was observed (Fig. 2, A and B). 1,25(OH)2D3 (10 and 100 nM) resulted in a dramatic increase in CYP3A4 mRNA and protein expression in a dose-dependent manner in Caco-2 cells, with the maximal response being achieved at the concentration of 100 nM after 6 or 10 days of treatment (Fig. 3, A and B). The changes for CYP3A4 mRNA and protein levels were consistent with findings of Schmiedlin-Ren et al. (1997). No significant change was observed for the mRNA expression of other phase I and phase II enzymes, such as CYP1A2, SULT1E1, SULT2A1, and SULT1A1, or the protein level of CYP1A2 (data not shown).

No change was observed for the absorptive transporters PEPT1 and ASBT (Fig. 4), with the exception of a small decrease in PEPT1 mRNA at 100 nM 1,25(OH)2D3 after the 6-day treatment (Fig. 4C). For apical efflux transporters, MDR1 mRNA levels in the treated groups were significantly up-regulated upon exposure to 1,25(OH)2D3 for 1 or 6 days at the designated concentrations, and P-gp protein levels were increased significantly for the 10-day treatment group compared with controls (Fig. 5, A and B). Increased MRP2 mRNA and protein levels were observed after treatment for 1 or 3 days, being higher after the 3-day treatment for the protein level (Fig. 5, C and D).

1,25(OH)2D3 failed to elicit any change in MRP3 mRNA and protein levels (Fig. 6, A and B), but it exhibited a significant inductive effect on MRP4 protein expression of 1.8-, 1.6-, or 1.9-fold after 3-, 6-, or 10-day treatment with 100 nM 1,25(OH)2D3, respectively; however, mRNA levels of MRP4 remained unaltered (Fig. 6, C and D), suggesting a post-transcriptional regulation of 1,25(OH)2D3 on MRP4 in the Caco-2 cells.

Post-Transcriptional Modulation Instead of de Novo Protein Synthesis for 1,25(OH)2D3-Mediated Increase in MRP4 Protein

To determine the underlying mechanism for the up-regulation of MRP4 protein by 1,25(OH)2D3, Caco-2 cells were incubated with the protein synthesis inhibitor cycloheximide (10 μg/ml) after induction with 1,25(OH)2D3 (100 nM for 6 days). At 0, 8, 24, and 48 h after the addition of cycloheximide, samples were retrieved and subjected to Western blot analysis. As shown in Fig. 7, levels of MRP4 fell to less than 62% of the initial value by 8 h after cycloheximide treatment and 44% by 48 h in the vehicle-treated group. In the 1,25(OH)2D3-treated group, however, degradation of MRP4 was arrested, and MRP4 levels showed a 1.3-fold increase for the 8-h sample relative to that at time 0, followed by a gradual decrease of MRP4 protein expression for the next 16 h. These data suggest that 1,25(OH)2D3 had probably increased the stability of MRP4 protein. However, whether 1,25(OH)2D3 directly affects MRP4 protein or other related proteinase activities needs to be further investigated.

    Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

Effects of 1,25(OH)2d3 on MRP3 and MRP4 expression in Caco-2 cells. Caco-2 cells were treated with 1,25(OH)2d3 (10 or 100 nM) on day 20, 18, 15, or 11 for 1, 3, 6, or 10 consecutive days, whereas the control group was treated with ethanol (0.1%). Cells were harvested on day 21, and the expression of mRNA and protein was determined by using qPCR and Western blotting and normalized to that of the house-keeping gene villin. A and C, data are the mean ± S.D. of qPCR quantifications from three independent cultures. B and D, data are the mean ± S.D. of the density histograms from three separate Western blot analyses. *, P < 0.05 between the 1,25(OH)2d3-treated and ethanol-treated groups.

    Fig. 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 7.

1,25(OH)2d3 improved MRP4 protein stability in Caco-2 cells. Caco-2 cells were treated with 100 nM 1,25(OH)2d3 on day 15 for six consecutive days, whereas the control group was treated with ethanol (0.1%). The protein synthesis inhibitor cycloheximide (10 μg/ml) was added and incubated with the cells on day 21. A, cell extracts were collected at the indicated times for determination of protein expression using Western blotting and normalized to the expression of the house-keeping gene villin. B, density histograms from three separate Western blot analyses (mean ± S.D.). *, P < 0.05 between the 1,25(OH)2d3-treated and ethanol-treated groups.

    Fig. 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 8.

Cumulative amounts of [3H]digoxin transported for the A→B and B→A direction at various times in the receiver compartment of the Caco-2 cell monolayer. Caco-2 cells were treated with ethanol (0.1%) and 1,25(OH)2d3 (10 and 100 nM) on day 18 for three consecutive days. The transport study was performed on day 21. Data are presented as mean ± S.D. (n = 3). *, P < 0.05 between the 1,25(OH)2d3-treated and ethanol-treated groups.

Functional Studies of 1,25(OH)2D3 on CYP3A4 and Transporters

Effect of 1,25(OH)2D3 on CYP3A4 Catalytic Activity. To determine the correlation between the higher CYP3A4 protein levels observed with 1,25(OH)2D3 treatment in Caco-2 cells lysate and catalytic activity, the rate of formation of 6β-hydroxytestosterone from 200 μM testosterone by CYP3A4 was assessed. With a 6-day treatment of 100 nM 1,25(OH)2D3 in Caco-2 cells, the rate of 6β-hydroxytestosterone formation was dramatically increased from undetectable levels in vehicle-treated and PBS control samples to 202 ± 41.2 pmol/min/g cellular protein.

Effects of 1,25(OH)2D3 on P-gp Function: [3H]Digoxin Transport across the Caco-2 Monolayer. The effects of the 3-day treatment of 1,25(OH)2D3 on P-gp function were evaluated by measuring the transport of [3H]digoxin across the Caco-2 cell monolayer. The TEER values for different treatment groups at the end of the 3-day treatment, measured before and after transport studies, remained high (>600 Ω · cm2) (data not shown). [3H]Digoxin transport across the PBS control Caco-2 monolayer showed marked asymmetry, with the Papp value for B→A (11.5 ± 1.8 × 10-6 cm/s, with the rate or ΔAR/Δt in eq. 1 being estimated as the slope of cumulative amount in receiver side versus time plot) exceeding that for the A→B direction (1.7 ± 0.4 × 10-6 cm/s); the efflux ratio was 6.8 (Table 2). Values of the vehicle (0.1% ethanol)-treated cells were similar to those of the PBS controls. 1,25(OH)2D3 (100 nM in 0.1% ethanol) treatment had no significant effect on the A-to-B permeability (Table 2; Fig. 8) but increased the Papp value of [3H]digoxin significantly in the B-to-A direction from 11.8 to 15.1 × 10-6 cm/s (P < 0.05), resulting in an increased efflux ratio of 8.0. In instances where 1,25(OH)2D3 was used for treatment, the apparent permeability of mannitol had remained unchanged. These data suggest the presence of functionally active P-gp protein in the 100 nM 1,25(OH)2D3-treated Caco-2 cells.

View this table:
  • View inline
  • View popup
TABLE 2

Apparent permeability coefficients (Papp × 10–6 cm/s) and net efflux ratio of bidirectional [3H]digoxin and [14C]mannitol transport for 60 min across the Caco-2 cell monolayer Caco-2 cells were treated with 1,25(OH)2D3 (10 and 100 nM) on day 18 for three consecutive days. The transport experiment was performed on day 21 for 60 min. Results are mean ± S.D. in triplicates.

Effect of 1,25(OH)2D3 after 3 Days of Treatment on MRP2 Function. The effective transport of the fluorescent CDF after application of CDF-DA into either the apical or basolateral, donor side, is shown. There was asymmetry in the accumulation of CDF in the receiver side and retention in the cell from the B-to-A (Fig. 9A) and A-to-B direction (Fig. 9B); CDF preferably accumulated in the apical side and not the basolateral side. With basolateral administration, the retention of CDF in the cell was significantly lower with 100 nM 1,25(OH)2D3 treatment, and there was no change in CDF appearance in the receiver, apical side (Fig. 9A). With apical administration, the appearance of CDF in the basolateral, receiver side was lower with 1,25(OH)2D3 treatment, but there was no change in CDF fluorescence in the cellular compartment (Fig. 9B). The plot of the transport efficiency (eq. 3) revealed the trend of transmembrane transport, showing a significantly higher transport at the apical membrane compared with the basolateral membrane, a decreasing trend of net basolateral efflux, and an increasing net apical efflux with 1,25(OH)2D3 treatment (Fig. 9C). These trends were, however, not statistically significant, suggesting that changes in MRP2 function were minor.

Results on CDF accumulation (administered as CDF-DA) in control and 1,25(OH)2D3-treated Caco-2 cells cultured on culture dishes are summarized in Fig. 9D. Consistent with increased MRP2 protein expression in 1,25(OH)2D3-treated Caco-2 cells, the fluorescence associated with the MRP2 substrate CDF inside the cells was similar between the control and ethanol (0.1%)-treated cells, but the fluorescence was significantly decreased after 1,25(OH)2D3 (100 nM) treatment for 3 days (44.0 ± 2.5 versus 29.2 ± 1.5 fluorescence/mg protein/min), suggesting appreciable excretion of CDF with 1,25(OH)2D3 treatment.

    Fig. 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 9.

1,25(OH)2d3 on MRP2 function in Caco-2 cells. CDF accumulation, measured as fluorescence, in the receiver side and in cells for ethanol (0.1%) and 1,25(OH)2d3 (10 and 100 nM) treated Caco-2 cells after 40 min of incubation after basolateral (A) and apical (B) dosing of the precursor 10 μM CDF-DA, and transport efficiency (C) (amount of CDF in receiver side/amounts of CDF in receiver and cell) at the apical or basolateral membrane over 40 min in the Caco-2 cell monolayer in Transwell plates. Data are presented as mean ± S.D. (n = 3). *, P < 0.05 between the 1,25(OH)2d3-treated and ethanol-treated groups. D, CDF retention/accumulation in cultured Caco-2 cells grown on culture dishes, measured as fluorescence, for the ethanol-(0.1%) and 1,25(OH)2d3-treated Caco-2 cells after 30 min of incubation.

    Fig. 10.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 10.

Effects of vitamin D analogs on genes expression in Caco-2 cells. Caco-2 cells were treated with 100 nM 1α,25(OH)2d3, lα(OH)D3, 1α(OH)D2, and 25(OH)D3 on day 15 for six consecutive days, whereas the control group was treated with ethanol (0.1%) in the course of culture. Cells were harvested on day 21, and protein expression was determined using Western blot and normalized to that of the house-keeping gene villin.

Effects of Vitamin D Analogs on Gene Expression in Caco-2 Cells Cultured in FBS

Effects of 1α(OH)D3 and 1α(OH)D2 on P-gp, MRP2, MRP4, and CYP3A4 protein expression were similar to those of 1,25(OH)2D3 (Fig. 10; Table 3). Because all of the vitamin D analogs and 1,25(OH)2D3 were tested at the 100 nM concentration, the relative potencies could not be directly compared because the analogs require activation to furnish the active forms. 1α(OH)D3, 1,25(OH)2D3, and 1α(OH)D2 were all capable of significantly inducing P-gp, MRP2, MRP4, and CYP3A4 protein expression in Caco-2 cells after 3 or 6 days of treatment. MRP2 and P-gp protein expressions were highest after 3 days of treatment, and a higher induction on MRP4 and CYP3A4 protein expression was achieved after six consecutive days of treatment with the vitamin D analogs. However, 25(OH)D3 failed to exert any effect.

View this table:
  • View inline
  • View popup
TABLE 3

Relative protein expression in Caco-2 cells after treatment with vitamin D analogs for 3 and 6 days Results are means ± S.D. from triplicate experiments; the mean value for the ethanol (0.1%) group was expressed as 1.0.

Effects of the Vitamin D Analog 25(OH)D3 on Protein Expression in Caco-2 Cells Cultured in Absence of FBS

The role of the vitamin D binding protein (DBP) in FBS on the availability of free 25(OH)D3 was further investigated by culturing the cells during the six consecutive treatment days in absence of 10% FBS; for this, an ITS-containing medium was used. 1,25(OH)2D3 was again used as a standard for comparison with 25(OH)D3 (Fig. 11). In the absence of FBS (replaced by ITS containing medium), significantly higher inductive activities of 25(OH)D3 toward P-gp and MRP2 protein were observed compared with those with FBS in the culture medium (Fig. 11, C and D). Approximately 1.4- and 2-fold increases in P-gp and MRP2 protein expression over the vehicle-treated groups were achieved in response to 25(OH)D3 in the absence of FBS, and the activities were less than that for 1,25(OH)2D3. Unexpectedly, protein expressions of CYP3A4 and MRP4 in Caco-2 cells cultured with 25(OH)D3 in absence of FBS were similar to those cultured in FBS and were slightly but insignificantly higher than those of controls (Fig. 11, B and E). An amelioration of inductive effects for CYP3A4 and MRP4 in the absence of FBS was further observed for 1,25(OH)2D3 (Fig. 11, B and E), results that were consistent with observations of Schmiedlin-Ren et al. (1997), who speculated that FBS was essential to prevent the toxicity of 1,25(OH)2D3 by limiting its free concentration. The same reasoning was adopted here for the observations on CYP3A4 and MRP4, genes that may be sensitive to toxic effects of 1,25(OH)2D3 (see lane 4 of Fig. 11A) and 25(OH)D3 (Fig. 11A, lane 3).

Discussion

Vitamin D is used for increasing calcium absorption and regulating bone mineral metabolism, and it is increasingly being used for the prevention and treatment of cancer and cardiovascular and autoimmune diseases (Holick, 2004; Schwartz and Skinner, 2007). Normally, 1,25(OH)2D3 exists at low levels in blood after vitamin D intake (DeLuca, 1988). However, upon administration of the commercially available 1,25(OH)2D3 via injection (calcitriol injection, Calcijex) or capsule (calcitriol, Rocaltrol) for the treatment of hypocalcemia, osteoporosis, hypoparathyroidism, hypoparathyroidism, osteomalacia, rickets, renal osteodystrophy (Bailie and Johnson, 2002), or cancer (Beer and Myrthue, 2006), high systemic levels of 1,25(OH)2D3 are attained. Consequently, due to the marked toxicity associated with 1,25(OH)2D3, vitamin D analogs that possess less hypercalcemic effects and toxicity are being developed as new drug entities (Masuda and Jones, 2006). The contribution of this class of 1,25(OH)2D3 products/vitamin D analogs to the regulation of genes that modulate drug disposition via the VDR has not been systematically examined.

Inductive effects of 1,25(OH)2D3 on transporters and enzymes have been observed in Caco-2 cells because of high levels of VDR and transporter and enzyme genes, rendering the system well suited for investigation (Taipalensuu et al., 2001). Our results demonstrated that there was a decrease in VDR mRNA level in 1,25(OH)2D3-treated cells compared with vehicle-treated cells (Fig. 2). Different responses of the VDR to 1,25(OH)2D3 with respect to VDR expression or VDR binding activity have been noted (Halline et al., 1994). 1,25(OH)2D3 could result in the up-regulation of VDR in undifferentiated Caco-2 cells, or would lose the inducing capability, or even down-regulate VDR expression and VDR binding activity in fully differentiated cells (Halline et al., 1994), suggesting factors other than VDR levels are important in the VDR response to 1,25(OH)2D3.

Our results showed that the 1,25(OH)2D3 treatment of Caco-2 cells significantly induced the mRNA and protein expression of various genes in a dose- and time-dependent manner. Results on the overall induction of CYP3A4 expression from microarray analyses (Fig. 1), RT-qPCR, Western blotting (Fig. 3, A and B), and the rate of 6β-hydroxylation of testosterone in 1,25(OH)2D3-treated Caco-2 cells in the present study were consistent with those of previous reports (Schmiedlin-Ren et al., 1997; Chan et al., 2004). Our results on the up-regulation of MDR1 mRNA and increased P-gp protein (Fig. 5, A and B) were validated functionally by the higher apical efflux of [3H]digoxin across the 1,25(OH)2D3-treated Caco-2 cell monolayer (Fig. 8; Table 2). The parallel changes in MDR1 mRNA and P-gp protein expression, similar to those for CYP3A4, suggest coordinate, transcriptional modulation of CYP3A4 and MDR1 by the VDR. The activation of both CYP3A4 and MDR1 via the PXR is unlikely because low levels of PXR exist in Caco-2 cells, and the VDR/vitamin D pathway had previously failed to stimulate PXR expression (Schmiedlin-Ren et al., 2001). Due to the inability of 1,25(OH)2D3 to activate PXR even in the mucosal villus of the small intestine (Thummel et al., 2001), it was concluded that it was VDR not PXR that mediated the 1,25(OH)2D3-induced gene expression of CYP3A4 and MDR1 in Caco-2 cells. In vivo, both PXR and VDR could mediate the regulation of MDR1 and CYP3A4 (Lemaire et al., 2004; Satsu et al., 2008), rendering it difficult to resolve the VDR versus the PXR effects when vitamin D analogs and PXR-activating ligands such as rifampin are administered concomitantly.

Song et al. (2006) reported that SULT2A1 is a target for transcriptional activation by vitamin D mediated through VDR and CCAAT/enhancer-binding protein α in Caco-2 cells. However, we did not detect a significant change in mRNA of SULT2A1, SULT1A1, or SULT1E1 from qPCR assay, a discrepancy that could be explained by the different Caco-2 cells among laboratories and the differing cell culture conditions. We used Caco-2 cells that were fully differentiated and grown in DMEM with 10% FBS for 21 days, whereas cells used by Song et al. (2006) were grown in DMEM supplemented with 5% charcoal-dextran-stripped FBS and cultured only for 3 days. We were not able to detect SULT2A1 protein because these may surface only when cells are kept in a confluent stage for an extended period beyond 21 days, as suggested by Meinl et al. (2008).

No change in protein and mRNA levels of ASBT, PEPT1, and MRP3 was detected using Western blot and qPCR after 1,25(OH)2D3 treatment (Figs. 4 and 6, A and B). By contrast, changes in mRNA levels of the apical efflux transporter MRP2 were commensurate with increased protein (Fig. 5, C and D). The increased MRP2 expression correlated with the lower cellular retention of CDF, which was formed through the rapid hydrolysis after entry of the precursor CDF-DA (Fig. 9D). Due to the lack of involvement of MRP4 (Pratt et al., 2006) and lack of change in mRNA or protein expression of MRP3 (Fig. 6, A and B), the alternate route of efflux of CDF (Tian et al., 2004), namely, the reduction in cellular accumulation of CDF in Caco-2 cells, was found to correlate well with increased MRP2 function after 1,25(OH)2D3 treatment. A similar reduction was noted in the cells in Transwell transport studies in the B-to-A direction (Fig. 9A).

    Fig. 11.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 11.

Effects of 25(OH)2d3 on P-gp, MRP2, MRP4, and CYP3A4 genes expression in Caco-2 cells. Cells were treated with 100 nM 1α,25(OH)2d3 and 25(OH)D3 on day 15 for six consecutive days in the absence (-FBS) or presence (+FBS) of 10% FBS in cell culture medium. Control group was treated with ethanol (0.1%). Cells were harvested on day 21. Protein expression was determined using Western blot and normalized to that of the housekeeping gene villin (A). The relative intensity of protein expression from Western blot analyses, and the summarized data (density histograms) were shown. B to E, mean ± S.D. (n = 3) for CYP3A4 (B), P-gp (C), MRP2 (D), and MRP4 (E). *, P < 0.05 between the ethanol- and 1,25(OH)2d3-treated groups.

For MRP4, an increase in protein and not mRNA, probably the result of post-transcriptional regulation, was observed (Fig. 6, C and D). This post-transcriptional regulation of MRP4 expression was also observed in primary biliary cirrhosis (Zollner et al., 2007). The regulation mechanism, confirmed by cycloheximide treatment of Caco-2 cells, is probably due to 1,25(OH)2D3 being able to enhance MRP4 protein stability (Fig. 7). We attempted to assess the transport of [3H]PMEA ([3H]adefovir) as the substrate of MRP4 (Reid et al., 2003) after the addition of [3H]bis(POM)-PMEA (or [3H]adefovir dipivoxil) into donor (apical or basolateral) compartments after 1,25(OH)2D3 pretreatment in Transwell plates. The complexity of the multiple transporters: P-gp for the handling of [3H]bis(POM)-PMEA; involvement of MRP4 and other MRP proteins for active transport of [3H]bis(POM)-PMEA metabolites, mono(POM)-PMEA and PMEA, at both the apical and basolateral membranes (Annaert et al., 1998); and the varying extents of induction of P-gp, MRP2, and MRP4 by 1,25(OH)2D3 drastically compromised data interpretation. A thorough investigation is underway to examine the involvement of MRP4 with use of specific inhibitors.

The inert vitamin D analogs that require activation to the respective active species on induction of expression of major drug metabolic enzymes and transporters were also investigated. Our studies demonstrated, for the first time, the inductive effects of vitamin D analogs lα(OH)D3, 25(OH)D3, and 1α(OH)D2 on CYP3A4, MDR1, MRP2, and MRP4 protein expression in Caco-2 cells (Fig. 10). The potencies of these vitamin D analogs to induce target genes expression depend on the binding affinities of the analogs toward the DBP and the binding affinity of the activated ligand(s) for the VDR. It is known that some nonrenal cells, including those of the prostate (Lou et al., 2004), colon, and pancreas, also express 1α-hydroxylase and 25-hydroxylase, which may lead to the local conversion of 25(OH)D3,1α(OH)D3, and 1α(OH)D2 to the active forms 1,25(OH)2D3 and 1,25(OH)2D2, establishing the VDR signaling pathway among these cells. It has been noted that 25(OH)D3 binds more tightly to DBP than 1,25(OH)2D3, whereas 1,25(OH)2D3 binds more tightly to the VDR than 25(OH)D3 (Norman et al., 2001). Due to its high binding affinity for the DBP, the effect of 25(OH)D3 on gene expression becomes limited due to reduced cellular availability of free 25(OH)D3. Our observation in this study showed that the effectiveness of 25(OH)D3 on gene expression in Caco-2 cells was lowered in the presence of FBS, a source of DBP, whereas much higher activity was re-established in the absence of FBS (Fig. 11). However, 25(OH)D3, at a high concentration (1 μM), was reported to stimulate gene expression in the presence of FBS in Caco-2 cells (Taparia et al., 2006), implying that the interactions related to 25(OH)D3 might be possible because the circulating concentration of 25(OH)D3 is 1000-fold higher than that of 1,25(OH)2D3 in vivo (Taparia et al., 2006). All of the analogs—lα(OH)D3, 25(OH)D3, and 1α(OH)D2—activated CYP3A4, MDR1, MRP2, mRNA, and protein expression and increased MRP4 protein, although absent for MRP3 (Fig. 10; Table 3). The activity of 25(OH)D3 was masked when FBS was present in the culture medium (Fig. 11). Harada et al. (1993) had demonstrated previously that 1,25(OH)2D3 was more potent than lα(OH)D3 and 25(OH)D3 in bone calcium mobilization.

This study on Caco-2 cells suggests statistically significant changes in CYP3A4, MDR1, MRP2, and MRP4 protein with 1,25(OH)2D3. Several other genes were stimulated by 1,25(OH)2D3, as revealed from the positive and significant changes with microarray analyses (Fig. 1) that directed us to areas for exploration. Similar changes were observed with the inert vitamin D analogs, albeit with slightly lower inducing activities, suggesting that the Caco-2 cells possess enzymes to form the active 1,25(OH)2D3 and 1,25(OH)2D2. These changes on transporters and enzymes in the Caco-2 cells after 1,25(OH)2D3 treatment attest to the importance of VDR in the regulation of transporters and enzymes. In addition to PXR and CAR, the VDR is another important nuclear receptor that influences drug disposition.

Footnotes

  • This study was supported by the Canadian Institutes for Health Research [Grant MOP89850].

  • Article, publication date, and citation information can be found at http://jpet.aspetjournals.org.

  • doi:10.1124/jpet.108.149815.

  • ABBREVIATIONS: P-gp, P-glycoprotein; MDR1, multidrug resistance protein 1; MRP, multidrug resistance-associated protein; PXR, pregnane X receptor; CAR, constitutive androstane receptor; VDR, vitamin D receptor; VDRE, vitamin D response element; SULT, sulfotransferase; ASBT, apical sodium dependent bile acid transporter; PEPT1, oligopeptide transporter 1; 1,25(OH)2D3,1α,25-dihydroxyvitamin D3; 25(OH)D3, 25-hydroxyvitamin D3;lα(OH)D3,1α-hydroxyvitamin D3;lα(OH)D2,1α-hydroxyvitamin D2 or Hectorol; CDF, 5-(and-6)-carboxy-2′,7′-dichlorofluorescein; CDF-DA, 5-(and-6)-carboxy-2′,7′-dichlorofluorescein diacetate; DMEM, Dulbecco's modified Eagle's medium; HPLC, high-performance liquid chromatography; RT-qPCR, real-time quantitative polymerase chain reaction; aRNA, aberrant RNA; FBS, fetal bovine serum; EfR, efflux ratio; ITS, insulin-transferrin-selenium; SSC, standard saline citrate; TEER, transepithelial electrical resistance; A, apical; B, basolateral; HBSS, Hanks' balanced salt solution; Papp, apparent permeability; PBS, phosphate-buffered saline; DBP, vitamin D binding protein; PMEA, 9-(2-phosphonylmethoxyethyl)adenine; POM, pivaloxymethyl.

    • Received December 15, 2008.
    • Accepted May 1, 2009.
  • The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    Annaert P, Van Gelder J, Naesens L, De Clercq E, Van den Mooter G, Kinget R, and Augustijns P (1998) Carrier mechanisms involved in the transepithelial transport of bis(POM)-PMEA and its metabolites across Caco-2 monolayers. Pharm Res 15: 1168-1173.
    OpenUrlCrossRefPubMed
  2. ↵
    Artursson P, Palm K, and Luthman K (2001) Caco-2 monolayers in experimental and theoretical predictions of drug transport. Adv Drug Deliv Rev 46: 27-43.
    OpenUrlCrossRefPubMed
  3. ↵
    Bailie GR and Johnson CA (2002) Comparative review of the pharmacokinetics of vitamin D analogues. Semin Dial 15: 352-357.
    OpenUrlCrossRefPubMed
  4. ↵
    Beer TM and Myrthue A (2006) Calcitriol in the treatment of prostate cancer. Anticancer Res 26: 2647-2651.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Brickman AS, Coburn JW, Friedman GR, Okamura WH, Massry SG, and Norman AW (1976) Comparison of effects of 1α-hydroxy-vitamin D3 and 1,25-dihydroxyvitamin D3 in man. J Clin Invest 57: 1540-1547.
    OpenUrlCrossRefPubMed
  6. ↵
    Brimer C, Dalton JT, Zhu Z, Schuetz J, Yasuda K, Vanin E, Relling MV, Lu Y, and Schuetz EG (2000) Creation of polarized cells coexpressing CYP3A4, NADPH cytochrome P450 reductase and MDR1/P-glycoprotein. Pharm Res 17: 803-810.
    OpenUrlCrossRefPubMed
  7. ↵
    Brown AJ (2001) Therapeutic uses of vitamin D analogues. Am J Kidney Dis 38: S3-S19.
    OpenUrlPubMed
  8. ↵
    Cavet ME, West M, and Simmons NL (1996) Transport and epithelial secretion of the cardiac glycoside, digoxin, by human intestinal epithelial (Caco-2) cells. Br J Pharmacol 118: 1389-1396.
    OpenUrlCrossRefPubMed
  9. ↵
    Chan LM, Cooper AE, Dudley AL, Ford D, and Hirst BH (2004) P-glycoprotein potentiates CYP3A4-mediated drug disappearance during Caco-2 intestinal secretory detoxification. J Drug Target 12: 405-413.
    OpenUrlCrossRefPubMed
  10. ↵
    Chen X, Chen F, Liu S, Glaeser H, Dawson PA, Hofmann AF, Kim RB, Shneider BL, and Pang KS (2006) Transactivation of rat apical sodium-dependent bile acid transporter and increased bile acid transport by 1α,25-dihydroxyvitamin D3 via the vitamin D receptor. Mol Pharmacol 69: 1913-1923.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Darwish HM and DeLuca HF (1996) Recent advances in the molecular biology of vitamin D action. Prog Nucleic Acid Res Mol Biol 53: 321-344.
    OpenUrlCrossRefPubMed
  12. ↵
    DeLuca HF (1988) The vitamin D story: a collaborative effort of basic science and clinical medicine. FASEB J 2: 224-236.
    OpenUrlAbstract
  13. ↵
    Halline AG, Davidson NO, Skarosi SF, Sitrin MD, Tietze C, Alpers DH, and Brasitus TA (1994) Effects of 1,25-dihydroxyvitamin D3 on proliferation and differentiation of Caco-2 cells. Endocrinology 134: 1710-1717.
    OpenUrlCrossRefPubMed
  14. ↵
    Harada M, Miyahara T, Miyata M, Komiyama H, Sugure A, Ikemoto Y, Kondo S, Takamura T, Higuchi S, and Otomo S (1993) Calcium regulating activity of 24a-homo-24,24-difluoro-1α,25-dihydroxyvitamin D3 and 26,27-dimethyl-24,24-difluoro-1α,25-dihydroxyvitamin D3. Calcif Tissue Int 53: 318-323.
    OpenUrlCrossRefPubMed
  15. ↵
    Holick MF (2004) Vitamin D: importance in the prevention of cancers, type 1 diabetes, heart disease, and osteoporosis. Am J Clin Nutr 79: 362-371.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Lemaire G, de Sousa G, and Rahmani R (2004) A PXR reporter gene assay in a stable cell culture system: CYP3A4 and CYP2B6 induction by pesticides. Biochem Pharmacol 68: 2347-2358.
    OpenUrlCrossRefPubMed
  17. ↵
    Lou YR, Laaksi I, Syvälä H, Bläuer M, Tammela TL, Ylikomi T, and Tuohimaa P (2004) 25-hydroxyvitamin D3 is an active hormone in human primary prostatic stromal cells. FASEB J 18: 332-334.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Lowry OH, Rosebrough NJ, Farr AL, and Randall RJ (1951) Protein measurement with the Folin phenol reagent. J Biol Chem 193: 265-275.
    OpenUrlFREE Full Text
  19. ↵
    Masuda S and Jones G (2006) Promise of vitamin D analogues in the treatment of hyperproliferative conditions. Mol Cancer Ther 5: 797-808.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    McCarthy TC, Li X, and Sinal CJ (2005) Vitamin D receptor-dependent regulation of colon multidrug resistance-associated protein 3 gene expression by bile acids. J Biol Chem 280: 23232-23242.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Meinl W, Ebert B, Glatt H, and Lampen A (2008) Sulfotransferase forms expressed in human intestinal Caco-2 and TC7 cells at varying stages of differentiation and role in benzo[a]pyrene metabolism. Drug Metab Dispos 36: 276-283.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Norman AW, Ishizuka S, and Okamura WH (2001) Ligands for the vitamin D endocrine system: different shapes function as agonists and antagonists for genomic and rapid response receptors or as a ligand for the plasma vitamin D binding protein. J Steroid Biochem Mol Biol 76: 49-59.
    OpenUrlCrossRefPubMed
  23. ↵
    Pelkonen O, Turpeinen M, Hakkola J, Honkakoski P, Hukkanen J, and Raunio H (2008) Inhibition and induction of human cytochrome P450 enzymes: current status. Arch Toxicol 82: 667-715.
    OpenUrlCrossRefPubMed
  24. ↵
    Pratt S, Chen V, Perry WI 3rd, Starling JJ, and Dantzig AH (2006) Kinetic validation of the use of carboxydichlorofluorescein as a drug surrogate for MRP5-mediated transport. Eur J Pharm Sci 27: 524-532.
    OpenUrlCrossRefPubMed
  25. ↵
    Prime-Chapman HM, Fearn RA, Cooper AE, Moore V, and Hirst BH (2004) Differential multidrug resistance-associated protein 1 through 6 isoform expression and function in human intestinal epithelial Caco-2 cells. J Pharmacol Exp Ther 311: 476-484.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Prudencio J, Akutsu N, Benlimame N, Wang T, Bastien Y, Lin R, Black MJ, Alaoui-Jamali MA, and White JH (2001) Action of low calcemic 1α,25-dihydroxyvitamin D3 analogue EB1089 in head and neck squamous cell carcinoma. J Natl Cancer Inst 93: 745-753.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Reid G, Wielinga P, Zelcer N, De Haas M, Van Deemter L, Wijnholds J, Balzarini J, and Borst P (2003) Characterization of the transport of nucleoside analog drugs by the human multidrug resistance proteins MRP4 and MRP5. Mol Pharmacol 63: 1094-1103.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Saeki M, Kurose K, Tohkin M, and Hasegawa R (2008) Identification of the functional vitamin D response elements in the human MDR1 gene. Biochem Pharmacol 76: 531-542.
    OpenUrlCrossRefPubMed
  29. ↵
    Satsu H, Hiura Y, Mochizuki K, Hamada M, and Shimizu M (2008) Activation of pregnane X receptor and induction of MDR1 by dietary phytochemicals. J Agric Food Chem 56: 5366-5373.
    OpenUrlCrossRefPubMed
  30. ↵
    Schmiedlin-Ren P, Thummel KE, Fisher JM, Paine MF, Lown KS, and Watkins PB (1997) Expression of enzymatically active CYP3A4 by Caco-2 cells grown on extracellular matrix-coated permeable supports in the presence of 1α,25-dihydroxyvitamin D3. Mol Pharmacol 51: 741-754.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Schmiedlin-Ren P, Thummel KE, Fisher JM, Paine MF, and Watkins PB (2001) Induction of CYP3A4 by 1α,25-dihydroxyvitamin D3 is human cell line-specific and is unlikely to involve pregnane X receptor. Drug Metab Dispos 29: 1446-1453.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Schwartz GG and Skinner HG (2007) Vitamin D status and cancer: new insights. Curr Opin Clin Nutr Metab Care 10: 6-11.
    OpenUrlPubMed
  33. ↵
    Song CS, Echchgadda I, Seo YK, Oh T, Kim S, Kim SA, Cho S, Shi L, and Chatterjee B (2006) An essential role of the CAAT/enhancer binding protein-alpha in the vitamin D-induced expression of the human steroid/bile acid-sulfotransferase (SULT2A1). Mol Endocrinol 20: 795-808.
    OpenUrlCrossRefPubMed
  34. ↵
    Stein MS and Wark JD (2003) An update on the therapeutic potential of vitamin D analogues. Expert Opin Investig Drugs 12: 825-840.
    OpenUrlCrossRefPubMed
  35. ↵
    Taipalensuu J, Törnblom H, Lindberg G, Einarsson C, Sjöqvist F, Melhus H, Garberg P, Sjöström B, Lundgren B, and Artursson P (2001) Correlation of gene expression of ten drug efflux proteins of the ATP-binding cassette transporter family in normal human jejunum and in human intestinal epithelial Caco-2 cell monolayers. J Pharmacol Exp Ther 299: 164-170.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Taparia S, Fleet JC, Peng JB, Wang XD, and Wood RJ (2006) 1,25-Dihydroxyvitamin D and 25-hydroxyvitamin D-mediated regulation of TRPV6 (a putative epithelial calcium channel) mRNA expression in Caco-2 cells. Eur J Nutr 45: 196-204.
    OpenUrlCrossRefPubMed
  37. ↵
    Thummel KE, Brimer C, Yasuda K, Thottassery J, Senn T, Lin Y, Ishizuka H, Kharasch E, Schuetz J, and Schuetz E (2001) Transcriptional control of intestinal cytochrome P-4503A by 1α,25-dihydroxy vitamin D3. Mol Pharmacol 60: 1399-1406.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Tian X, Zamek-Gliszczynski MJ, Zhang P, and Brouwer KL (2004) Modulation of multidrug resistance-associated protein 2 (Mrp2) and Mrp3 expression and function with small interfering RNA in sandwich-cultured rat hepatocytes. Mol Pharmacol 66: 1004-1010.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Urquhart BL, Tirona RG, and Kim RB (2007) Nuclear receptors and the regulation of drug-metabolizing enzymes and drug transporters: implications for interindividual variability in response to drugs. J Clin Pharmacol 47: 566-578.
    OpenUrlCrossRefPubMed
  40. ↵
    Zollner G, Wagner M, Fickert P, Silbert D, Gumhold J, Zatloukal K, Denk H, and Trauner M (2007) Expression of bile acid synthesis and detoxification enzymes and the alternative bile acid efflux pump MRP4 in patients with primary biliary cirrhosis. Liver Int 27: 920-929.
    OpenUrlCrossRefPubMed
View Abstract
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 376 (2)
Journal of Pharmacology and Experimental Therapeutics
Vol. 376, Issue 2
1 Feb 2021
  • Table of Contents
  • About the Cover
  • Index by author
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
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
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleMETABOLISM, TRANSPORT, AND PHARMACOGENOMICS

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

Jianghong Fan, Shanjun Liu, Yimin Du, Jodi Morrison, Robert Shipman and K. Sandy Pang
Journal of Pharmacology and Experimental Therapeutics August 1, 2009, 330 (2) 389-402; DOI: https://doi.org/10.1124/jpet.108.149815

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Research ArticleMETABOLISM, TRANSPORT, AND PHARMACOGENOMICS

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

Jianghong Fan, Shanjun Liu, Yimin Du, Jodi Morrison, Robert Shipman and K. Sandy Pang
Journal of Pharmacology and Experimental Therapeutics August 1, 2009, 330 (2) 389-402; DOI: https://doi.org/10.1124/jpet.108.149815
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • HDL Mimetic 4F Modulates Aβ Distribution in Brain and Plasma
  • AOX1 Inhibition by Gefitinib, Erlotinib, and Metabolites
  • Catalytic Activity of CYP2C9 Variants
Show more Metabolism, Transport, and Pharmacogenomics

Similar Articles

  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2021 by the American Society for Pharmacology and Experimental Therapeutics