Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
AAPS J ; 24(4): 71, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35650371

ABSTRACT

The vitamin D receptor (VDR), in addition to other nuclear receptors, the pregnane X receptor (PXR) and constitutive androstane receptor (CAR), is involved in the regulation of enzymes, transporters and receptors, and therefore intimately affects drug disposition, tissue health, and the handling of endogenous and exogenous compounds. This review examines the role of 1α,25-dihydroxyvitamin D3 or calcitriol, the natural VDR ligand, on activation of the VDR and its crosstalk with other nuclear receptors towards the regulation of enzymes and transporters, notably many of the cytochrome P450s including CYP3A4 and sulfotransferase 2A1 (SULT2A1) as well as cholesterol 7α-hydroxylase (CYP7A1). Moreover, the VDR upregulates the intestinal channel, TRPV6, for calcium absorption, LDL receptor-related protein 1 (LRP1) and receptor for advanced glycation end products (RAGE) in brain for ß-amyloid peptide efflux and influx, the sodium phosphate transporters (NaPi), the apical sodium-dependent bile acid transporter (ASBT) and organic solute transporters (OSTα-OSTß) for bile acid absorption and efflux, respectively, the renal organic anion transporter 3 (OAT3) and several of the ATP-binding cassette protein transporters-the multidrug resistance protein 1 (MDR1) and the multidrug resistance-associated proteins (MRPs). Hence, the role of the VDR is increasingly being recognized for its therapeutic potential and pharmacologic activity, giving rise to drug-drug interactions (DDI). Therapeutically, ligand-activated VDR shows anti-inflammatory effects towards the suppression of inflammatory mediators, improves cognition by upregulating amyloid-beta (Aß) peptide clearance in brain, and maintains phosphate, calcium, and parathyroid hormone (PTH) balance and kidney function and bone health, demonstrating the crucial roles of the VDR in disease progression and treatment of diseases.


Subject(s)
Calcium , Receptors, Calcitriol , Calcium/metabolism , Ligands , Membrane Transport Proteins , Receptors, Calcitriol/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
2.
Biopharm Drug Dispos ; 41(3): 126-148, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32319119

ABSTRACT

Calcitriol or 1,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ] is the active ligand of the vitamin D receptor (VDR) that plays a vital role in health and disease. Vitamin D is converted to the relatively inactive metabolite, 25-hydroxyvitamin D3 [25(OH)D3 ], by CYP27A1 and CYP2R1 in the liver, then to 1,25(OH)2 D3 by a specific, mitochondrial enzyme, CYP27B1 (1α-hydroxylase) that is present primarily in the kidney. The degradation of both metabolites is mostly carried out by the more ubiquitous mitochondrial enzyme, CYP24A1. Despite the fact that calcitriol inhibits its formation and degradation, allometric scaling revealed strong interspecies correlation of the net calcitriol clearance (CL estimated from dose/AUC∞ ), production rate (PR), and basal, plasma calcitriol concentration with body weight (BW). PBPK-PD (physiologically based pharmacokinetic-pharmacodynamic) modeling confirmed the dynamic interactions between calcitriol and Cyp27b1/Cyp24a1 on the decrease in the PR and increase in CL in mice. Close scrutiny of the literature revealed that basal levels of calcitriol had not been taken into consideration for estimating the correct AUC∞ and CL after exogenous calcitriol dosing in both animals and humans, leading to an overestimation of AUC∞ and underestimation of the plasma CL. In humans, CL was decreased in chronic kidney disease but increased in cancer. Collectively, careful pharmacokinetic data analysis and improved definition are achieved with PBPK-PD modeling, which embellishes the complexity of dose, enzyme regulation, and disease conditions. Allometric scaling and PBPK-PD modeling were applied successfully to extend the PBPK model to predict calcitriol kinetics in cancer patients.


Subject(s)
Vitamin D/analogs & derivatives , Animals , Cytochrome P-450 Enzyme System/metabolism , Humans , Kinetics , Mice , Models, Biological , Receptors, Calcitriol/metabolism , Vitamin D/metabolism , Vitamin D/pharmacokinetics
3.
Biopharm Drug Dispos ; 39(4): 196-204, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29488238

ABSTRACT

Vitamin D3 and the synthetic vitamin D analogs, 1α-hydroxyvitamin D3 [1α(OH)D3 ], 1α-hydroxyvitamin D2 [1α(OH)D2 ] and 25-hydroxyvitamin D3 [25(OH)D3 ] were appraised for their vitamin D receptor (VDR) associated-potencies as cholesterol lowering agents in mice in vivo. These precursors are activated in vivo: 1α(OH)D3 and 1α(OH)D2 are transformed by liver CYP2R1 and CYP27A1 to active VDR ligands, 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ] and 1α,25-dihydroxyvitamin D2 [1,25(OH)2 D2 ], respectively. 1α(OH)D2 may also be activated by CYP24A1 to 1α,24-dihydroxyvitamin D2 [1,24(OH)2 D2 ], another active VDR ligand. 25(OH)D3 , the metabolite formed via CYP2R1 and or CYP27A1 in liver from vitamin D3 , is activated by CYP27B1 in the kidney to 1,25(OH)2 D3 . In C57BL/6 mice fed the high fat/high cholesterol Western diet for 3 weeks, vitamin D analogs were administered every other day intraperitoneally during the last week of the diet. The rank order for cholesterol lowering, achieved via mouse liver small heterodimer partner (Shp) inhibition and increased cholesterol 7α-hydroxylase (Cyp7a1) expression, was: 1.75 nmol/kg 1α(OH)D3  > 1248 nmol/kg 25(OH)D3 (dose ratio of 0.0014) > > 1625 nmol/kg vitamin D3 . Except for 1.21 nmol/kg 1α(OH)D2 that failed to lower liver and plasma cholesterol contents, a significant negative correlation was observed between the liver concentration of 1,25(OH)2 D3 formed from the precursors and liver cholesterol levels. The composite results show that vitamin D analogs 1α(OH)D3 and 25(OH)D3 exhibit cholesterol lowering properties upon activation to 1,25(OH)2 D3 : 1α(OH)D3 is rapidly activated by liver enzymes and 25(OH)D3 is slowly activated by renal Cyp27b1 in mouse.


Subject(s)
Anticholesteremic Agents/therapeutic use , Hypercholesterolemia/drug therapy , Vitamin D/analogs & derivatives , Vitamin D/therapeutic use , Animals , Anticholesteremic Agents/blood , Cholesterol/blood , Cholesterol/metabolism , Diet, High-Fat , Hypercholesterolemia/blood , Hypercholesterolemia/metabolism , Ileum/drug effects , Ileum/metabolism , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Membrane Transport Proteins/genetics , Mice, Inbred C57BL , Receptors, Cytoplasmic and Nuclear/genetics , Vitamin D/blood
4.
Drug Metab Dispos ; 46(1): 75-87, 2018 01.
Article in English | MEDLINE | ID: mdl-29084783

ABSTRACT

We expanded our published physiologically based pharmacokinetic model (PBPK) on 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], ligand of the vitamin D receptor (VDR), to appraise VDR-mediated pharmacodynamics in mice. Since 1,25(OH)2D3 kinetics was best described by a segregated-flow intestinal model (SFM) that described a low/partial intestinal (blood/plasma) flow to enterocytes, with feedback regulation of its synthesis (Cyp27b1) and degradation (Cyp24a1) enzymes, this PBPK(SFM) model was expanded to describe the VDR-mediated changes (altered/basal mRNA expression) of target genes/responses with the indirect response model. We examined data on 1) renal Trpv5 (transient receptor potential cation channel, subfamily V member 5) and Trpv6 and intestinal Trpv6 (calcium channels) for calcium absorption; 2) liver 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (Hmgcr) and cytochrome 7α-hydroxylase (Cyp7a1) for cholesterol synthesis and degradation, respectively; and 3) renal and brain Mdr1 (multidrug-resistance protein that encodes the P-glycoprotein) for digoxin disposition after repetitive intraperitoneal doses of 120 pmol 1,25(OH)2D3 Fitting, performed with modeling software, yielded reasonable prediction of a dominant role of intestinal Trpv6 in calcium absorption, circadian rhythm that is characterized by simple cosine models for Hmgcr and Cyp7a1 on liver cholesterol, and brain and renal Mdr1 on tissue efflux of digoxin. Fitted parameters on the Emax, EC50, and turnover rate constants of VDR-target genes [zero-order production (kin) and first-order degradation (kout) rate constants] showed low coefficients of variation and acceptable median prediction errors (4.5%-40.6%). Sensitivity analyses showed that the Emax and EC50 values are key parameters that could influence the pharmacodynamic responses. In conclusion, the PBPK(SFM)-pharmacodynamic model successfully characterized VDR gene activation and serves as a useful tool to predict the therapeutic effects of 1,25(OH)2D3.


Subject(s)
Calcitriol/pharmacology , Models, Biological , Receptors, Calcitriol/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Calcitriol/therapeutic use , Calcium/metabolism , Cholesterol/analysis , Cholesterol/metabolism , Digoxin/pharmacology , Hydroxymethylglutaryl CoA Reductases/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Animal , RNA, Messenger/metabolism , Sensitivity and Specificity , TRPV Cation Channels/metabolism
5.
Biopharm Drug Dispos ; 39(2): 99-115, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29243851

ABSTRACT

The vitamin D-deficient model, established in the C57BL/6 mouse after 8 weeks of feeding vitamin D-deficient diets in the absence or presence of added calcium, was found associated with elevated levels of plasma parathyroid hormone (PTH) and plasma and liver cholesterol, and a reduction in cholesterol 7α-hydroxylase (Cyp7a1, rate-limiting enzyme for cholesterol metabolism) and renal Oat3 mRNA/protein expression levels. However, there was no change in plasma calcium and phosphate levels. Appraisal of the liver revealed an up-regulation of mRNA expressions of the small heterodimer partner (Shp) and attenuation of Cyp7a1, which contributed to hypercholesterolemia in vitamin D-deficiency. When vitamin D-sufficient or D-deficient mice were further rendered hypercholesterolemic with 3 weeks of feeding the respective, high fat/high cholesterol (HF/HC) diets, treatment with 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ], active vitamin D receptor (VDR) ligand, or vitamin D (cholecalciferol) to HF/HC vitamin D-deficient mice lowered the cholesterol back to baseline levels. Cholecalciferol treatment partially restored renal Oat3 mRNA/protein expression back to that of vitamin D-sufficient mice. When the protein expression of protein kinase C (PKC), a known, negative regulator of Oat3, was examined in murine kidney, no difference in PKC expression was observed for any of the diets with/without 1,25(OH)2 D3 /cholecalciferol treatment, inferring that VDR regulation of renal Oat3 did not involve PKC in mice. As expected, plasma calcium levels were not elevated by cholecalciferol treatment of vitamin D-deficient mice, while 1,25(OH)2 D3 treatment led to hypercalcemia. In conclusion, vitamin D-deficiency resulted in down-regulation of liver Cyp7a1 and renal Oat3, conditions that are alleviated upon replenishment of cholecalciferol.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/biosynthesis , Down-Regulation , Gene Expression Regulation, Enzymologic , Kidney/metabolism , Liver/metabolism , Organic Anion Transporters, Sodium-Independent/biosynthesis , Vitamin D Deficiency/enzymology , Vitamin D Deficiency/genetics , Animals , Bile Acids and Salts/metabolism , Calcifediol/blood , Calcium/blood , Calcium/pharmacology , Cholecalciferol/pharmacology , Cholesterol/blood , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/blood , Cholesterol 7-alpha-Hydroxylase/genetics , Diet/methods , Gallbladder/metabolism , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Organic Anion Transporters, Sodium-Independent/genetics , Parathyroid Hormone/blood , Vitamin D/analogs & derivatives , Vitamin D/blood
6.
J Pharmacol Exp Ther ; 360(1): 174-191, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27789682

ABSTRACT

The humanized liver mouse model is being exploited increasingly for human drug metabolism studies. However, its model stability, intercommunication between human hepatocytes and mouse nonparenchymal cells in liver and murine intestine, and changes in extrahepatic transporter and enzyme expressions have not been investigated. We examined these issues in FRGN [fumarylacetoacetate hydrolase (Fah-/-), recombination activating gene 2 (Rag2-/-), and interleukin 2 receptor subunit gamma (IL-2rg -/-) triple knockout] on nonobese diabetic (NOD) background] and chimeric mice: mFRGN and hFRGN (repopulated with mouse or human hepatocytes, respectively). hFRGN mice showed markedly higher levels of liver cholesterol, biliary bilirubin, and bile acids (liver, bile, and plasma; mainly human forms, but also murine bile acids) but lower transforming growth factor beta receptor 2 (TGFBR2) mRNA expression levels (10%) in human hepatocytes and other proliferative markers in mouse nonparenchymal cells (Tgf-ß1) and cholangiocytes [plasma membrane-bound, G protein-coupled receptor for bile acids (Tgr5)], suggestive of irregular regeneration processes in hFRGN livers. Changes in gene expression in murine intestine, kidney, and brain of hFRGN mice, in particular, induction of intestinal farnesoid X receptor (Fxr) genes: fibroblast growth factor 15 (Fgf15), mouse ileal bile acid binding protein (Ibabp), small heterodimer partner (Shp), and the organic solute transporter alpha (Ostα), were observed. Proteomics revealed persistence of remnant murine proteins (cyotchrome P450 7α-hydroxylase (Cyp7a1) and other enzymes and transporters) in hFRGN livers and suggest the likelihood of mouse activity. When compared with normal human liver tissue, hFRGN livers showed lower SHP mRNA and higher CYP7A1 (300%) protein expression, consequences of tß- and tα-muricholic acid-mediated inhibition of the FXR-SHP cascade and miscommunication between intestinal Fgf15 and human liver fibroblast growth factor receptor 4 (FGFR4), as confirmed by the unchanged hepatic pERK/total ERK ratio. Dysregulation of hepatocyte proliferation and bile acid homeostasis in hFRGN livers led to hepatotoxicity, gallbladder distension, liver deformity, and other extrahepatic changes, making questionable the use of the preparation for drug metabolism studies.


Subject(s)
Bile Acids and Salts/metabolism , Homeostasis , Intestines/cytology , Liver/cytology , Liver/metabolism , Signal Transduction , Adolescent , Adult , Animals , Bile Acids and Salts/blood , Child , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation , Gene Knockout Techniques , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hydrolases/deficiency , Hydrolases/genetics , Male , Mice , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics
7.
Drug Metab Dispos ; 44(9): 1524-35, 2016 09.
Article in English | MEDLINE | ID: mdl-27342868

ABSTRACT

Chimeric mouse liver models are useful in vivo tools for human drug metabolism studies; however, liver integrity and the microcirculation remain largely uninvestigated. Hence, we conducted liver perfusion studies to examine these attributes in FRGN [Fah(-/-), Rag2(-/-), and Il2rg(-/-), NOD strain] livers (control) and chimeric livers repopulated with mouse (mFRGN) or human (hFRGN) hepatocytes. In single-pass perfusion studies (2.5 ml/min), outflow dilution profiles of noneliminated reference indicators ((51)Cr-RBC, (125)I-albumin, (14)C-sucrose, and (3)H-water) revealed preservation of flow-limited distribution and reduced water and albumin spaces in hFRGN livers compared with FRGN livers, a view supported microscopically by tightly packed sinusoids. With prograde and retrograde perfusion of harmol (50 µM) in FRGN livers, an anterior sulfation (Sult1a1) over the posterior distribution of glucuronidation (Ugt1a1) activity was preserved, evidenced by the 42% lower sulfation-to-glucuronidation ratio (HS/HG) and 14% higher harmol extraction ratio (E) upon switching from prograde to retrograde flow. By contrast, zonation was lost in mFRGN and hFRGN livers, with HS/HG and E for both flows remaining unchanged. Remnant mouse genes persisted in hFRGN livers (10%-300% those of FRGN). When hFRGN livers were compared with human liver tissue, higher UGT1A1 and MRP2, lower MRP3, and unchanged SULT1A1 and MRP4 mRNA expression were observed. Total Sult1a1/SULT1A1 protein expression in hFRGN livers was higher than that of FRGN livers, consistent with higher harmol sulfate formation. The composite data on humanized livers suggest a loss of zonation, lack of complete liver humanization, and persistence of murine hepatocyte activities leading to higher sulfation.


Subject(s)
Chimera , Enzymes/metabolism , Liver/enzymology , Animals , Antibodies, Monoclonal, Humanized/immunology , Arylsulfotransferase/metabolism , Glucuronosyltransferase/metabolism , Humans , Liver/blood supply , Mice , Microcirculation
8.
Drug Metab Dispos ; 44(2): 189-208, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26586377

ABSTRACT

1α,25-Dihydroxyvitamin D3 [1,25(OH)2D3] concentrations are regulated by renal CYP27B1 for synthesis and CYP24A1 for degradation. Published plasma and tissue 1,25(OH)2D3 concentrations and mRNA fold change expression of Cyp24a1 and Cyp27b1 following repetitive i.p. injections to C57BL/6 mice (2.5 µg × kg(-1) every 2 days for 4 doses) were fitted with a minimal and full physiologically-based pharmacokinetic-pharmacodynamic models (PBPK-PD). The minimal physiologically-based pharmacokinetic-pharmacodynamic linked model (mPBPK-PD) related Cyp24a1 mRNA fold changes to linear changes in tissue/tissue baseline 1,25(OH)2D3 concentration ratios, whereas the full physiologically-based pharmacokinetic-pharmacodynamic model (PBPK-PD) related measured tissue Cyp24a1 and Cyp27b1 fold changes to tissue 1,25(OH)2D3 concentrations with indirect response, sigmoidal maximal stimulatory effect/maximal inhibitory effect functions. Moreover, the intestinal segregated flow model (SFM) that describes a low and partial intestinal (blood/plasma) flow to enterocytes was nested within both models for comparison with the traditional model for intestine (TM) where the entire flow perfuses the intestine. Both the mPBPK(SFM)-PD and full PBPK(SFM)-PD models described the i.p. plasma and tissue 1,25(OH)2D3 concentrations and fold changes in mRNA expression significantly better than the TM counterparts with F test comparisons. The full PBPK(SFM)-PD fits showed estimates with good precision (lower percentage of coefficient of variation), and the model was more robust in predicting data from escalating i.v. doses (2, 60, and 120 pmol) and the rebound in 1,25(OH)2D3 tissue concentrations after dosing termination. The full PBPK(SFM)-PD model performed the best among the tested models for describing the complex pharmacokinetic-pharmacodynamic interplay among Cyp27b1, Cyp24a1, and 1,25(OH)2D3.


Subject(s)
Calcitriol/metabolism , Vitamin D/analogs & derivatives , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Animals , Enterocytes/metabolism , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Calcitriol/metabolism , Vitamin D/metabolism
9.
Br J Pharmacol ; 172(14): 3611-26, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25829051

ABSTRACT

BACKGROUND AND PURPOSE: Concentrations of 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ], the active ligand of the vitamin D receptor, are tightly regulated by CYP27B1 for synthesis and CYP24A1 for degradation. However, the dose-dependent pharmacokinetic (PK)-pharmacodynamic (PD) relationship between these enzymes and 1,25(OH)2 D3 concentrations has not been characterized. EXPERIMENTAL APPROACH: The pharmacokinetics of 1,25(OH)2 D3 were evaluated after administration of single (2, 60 and 120 pmol) and repeated (2 and 120 pmol q2d ×3) i.v. doses to male C57BL/6 mice. mRNA expression of CYP27B1 and CYP24A1 was examined by quantitative PCR and 1,25(OH)2 D3 concentrations were determined by enzyme immunoassay. KEY RESULTS: CYP27B1 and CYP24A1 changes were absent for the 2 pmol dose and biexponential decay profiles showed progressively shorter terminal half-lives with increasing doses. Fitting with a two-compartment model revealed decreasing net synthesis rates and increasing total clearances with dose, consistent with a dose-dependent down-regulation of renal CYP27B1 and the induction of renal/intestinal CYP24A1 mRNA expression. Upon incorporation of PD parameters for inhibition of CYP27B1 and induction of CYP24A1 to the simple two-compartment model, fitting was significantly improved. Moreover, fitted estimates for the 2 pmol dose, together with the PD parameters as modifiers, were able to predict profiles reasonably well for the higher (60 and 120 pmol) doses. Lastly, an indirect response model, which considered the synthesis and degradation of enzymes, adequately described the PK and PD profiles. CONCLUSIONS AND IMPLICATIONS: The unique PK of exogenously administered 1,25(OH)2 D3 led to changes in PD of CYP27B1 and CYP24A1, which hastened the clearance of 1,25(OH)2 D3 .


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/biosynthesis , Vitamin D3 24-Hydroxylase/metabolism , Vitamin D/analogs & derivatives , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Animals , Dose-Response Relationship, Drug , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Vitamin D/administration & dosage , Vitamin D/pharmacokinetics , Vitamin D3 24-Hydroxylase/genetics
10.
J Pharm Sci ; 103(11): 3793-3805, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25266751

ABSTRACT

Evidence in the literature suggests that 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ], the vitamin D receptor ligand, down-regulated the expression of the rat renal organic anion (renal organic anion transporter, rOAT) and oligopeptide (rPEPT) transporters, but increased intestinal rPEPT1 expression. We investigated, in rats, the intravenous and oral pharmacokinetics of 2 mg/kg cefdinir and cefadroxil, two cephalosporins that are eliminated via renal OAT1/OAT3 and are substrates of PEPT1/PEPT2, with and without 1,25(OH)2 D3 treatment. The area under the plasma concentration-time curve (AUC) of cefdinir or cefadroxil after 1,25(OH)2 D3 treatment was increased significantly because of decreased clearance (CL). Both kidney uptake and cumulative urinary recovery were significantly decreased, whereas liver uptake and fecal recovery remained unchanged in 1,25(OH)2 D3 -treated rats. Similar changes in AUC and CL were observed for both drugs upon coadministration of probenecid, the OAT inhibitor. Oral availability of cefdinir and cefadroxil remained unchanged with 1,25(OH)2 D3 treatment, suggesting lack of a role for intestinal rPEPT1. Rather, reduction of rOAT1/rOAT3 mRNA expression in kidney with 1,25(OH)2 D3 -treatment was observed, confirmed by decreased function in MDCKII cells overexpressing human OAT1 and OAT3. These composite results suggest that 1,25(OH)2 D3 treatment reduces cefdinir and cefadroxil clearances by diminution of renal OAT1/OAT3 expression, implicating a role for 1,25(OH)2 D3 in eliciting transporter-based drug interactions.


Subject(s)
Calcitriol/administration & dosage , Cefadroxil/pharmacokinetics , Cephalosporins/pharmacokinetics , Kidney/drug effects , Organic Anion Transport Protein 1/drug effects , Organic Anion Transporters, Sodium-Independent/drug effects , Receptors, Calcitriol/agonists , Administration, Oral , Animals , Area Under Curve , Biological Availability , Cefadroxil/administration & dosage , Cefadroxil/urine , Cefdinir , Cephalosporins/administration & dosage , Cephalosporins/urine , Dogs , Down-Regulation , Drug Interactions , Humans , Injections, Intravenous , Kidney/metabolism , Ligands , Liver/drug effects , Liver/metabolism , Madin Darby Canine Kidney Cells , Male , Metabolic Clearance Rate , Models, Biological , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Peptide Transporter 1 , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Renal Elimination/drug effects , Symporters/metabolism , Tissue Distribution , Transfection
11.
Gastroenterology ; 146(4): 1048-59, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24365583

ABSTRACT

BACKGROUND & AIMS: Little is known about the effects of the vitamin D receptor (VDR) on hepatic activity of human cholesterol 7α-hydroxylase (CYP7A1) and cholesterol metabolism. We studied these processes in mice in vivo and mouse and human hepatocytes. METHODS: Farnesoid X receptor (Fxr)(-/-), small heterodimer partner (Shp)(-/-), and C57BL/6 (wild-type control) mice were fed normal or Western diets for 3 weeks and were then given intraperitoneal injections of vehicle (corn oil) or 1α,25-dihydroxyvitamin D3 (1,25[OH]2D3; 4 doses, 2.5 µg/kg, every other day). Plasma and tissue samples were collected and levels of Vdr, Shp, Cyp7a1, Cyp24a1, and rodent fibroblast growth factor (Fgf) 15 expression, as well as levels of cholesterol, were measured. We studied the regulation of Shp by Vdr using reporter and mobility shift assays in transfected human embryonic kidney 293 cells, quantitative polymerase chain reaction with mouse tissues and mouse and human hepatocytes, and chromatin immunoprecipitation assays with mouse liver. RESULTS: We first confirmed the presence of Vdr mRNA and protein expression in livers of mice. In mice fed normal diets and given injections of 1,25(OH)2D3, liver and plasma concentrations of 1,25(OH)2D3 increased and decreased in unison. Changes in hepatic Cyp7a1 messenger RNA (mRNA) correlated with those of Cyp24a1 (a Vdr target gene) and inversely with Shp mRNA, but not ileal Fgf15 mRNA. Similarly, incubation with 1,25(OH)2D3 increased levels of Cyp24a1/CYP24A1 and Cyp7a1/CYP7A1 mRNA in mouse and human hepatocytes, and reduced levels of Shp mRNA in mouse hepatocytes. In Fxr(-/-) and wild-type mice with hypercholesterolemia, injection of 1,25(OH)2D3 consistently reduced levels of plasma and liver cholesterol and Shp mRNA, and increased hepatic Cyp7a1 mRNA and protein; these changes were not observed in Shp(-/-) mice given 1,25(OH)2D3 and fed Western diets. Truncation of the human small heterodimer partner (SHP) promoter and deletion analyses revealed VDR-dependent inhibition of SHP, and mobility shift assays showed direct binding of VDR to enhancer regions of SHP. In addition, chromatin immunoprecipitation analysis of livers from mice showed that injection of 1,25(OH)2D3 increased recruitment of Vdr and rodent retinoid X receptor to the Shp promoter. CONCLUSIONS: Activation of the VDR represses hepatic SHP to increase levels of mouse and human CYP7A1 and reduce cholesterol.


Subject(s)
Calcitriol/pharmacology , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholesterol/metabolism , Hepatocytes/drug effects , Liver/drug effects , Receptors, Calcitriol/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Binding Sites , Disease Models, Animal , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Enzymologic , HEK293 Cells , Hepatocytes/enzymology , Humans , Hypercholesterolemia/drug therapy , Hypercholesterolemia/enzymology , Hypercholesterolemia/genetics , Ileum/drug effects , Ileum/enzymology , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Calcitriol/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Steroid Hydroxylases/metabolism , Time Factors , Transfection , Vitamin D3 24-Hydroxylase
12.
Am J Physiol Endocrinol Metab ; 304(9): E977-89, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23482451

ABSTRACT

The vitamin D receptor (VDR) maintains a balance of plasma calcium and 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], its natural active ligand, by directly regulating the calcium ion channel (TRPV6) and degradation enzyme (CYP24A1), and indirectly regulating the parathyroid hormone (PTH) for feedback regulation of the synthetic enzyme CYP27B1. Studies that examined the intricate relationships between plasma and tissue 1,25(OH)2D3 levels and changes in VDR target genes and plasma calcium and PTH are virtually nonexistent. In this study, we investigated temporal correlations between tissue 1,25(OH)2D3 concentrations and VDR target genes in ileum and kidney and plasma calcium and PTH concentrations in response to 1,25(OH)2D3 treatment in mice (2.5 µg/kg ip, singly or q2d × 4). After a single ip dose, plasma 1,25(OH)2D3 peaked at ∼0.5 h and then decayed biexponentially, falling below basal levels after 24 h and then returning to baseline after 8 days. Upon repetitive ip dosing, plasma, ileal, renal, and bone 1,25(OH)2D3 concentrations rose and decayed in unison. Temporal profiles showed increased expressions of ileal Cyp24a1 and renal Cyp24a1, Mdr1/P-gp, and VDR but decreased renal Cyp27b1 mRNA after a time delay in VDR activation. Increased plasma calcium and attenuated PTH levels and increased ileal and renal Trpv6 expression paralleled the changes in tissue 1,25(OH)2D3 concentrations. Gene changes in the kidney were more sustained than those in intestine, but the magnitudes of change for Cyp24a1 and Trpv6 were lower than those in intestine. The data revealed that 1,25(OH)2D3 equilibrates with tissues rapidly, and VDR target genes respond quickly to exogenously administered 1,25(OH)2D3.


Subject(s)
Calcitriol/metabolism , Calcitriol/pharmacology , Calcium/metabolism , Parathyroid Hormone/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Vitamins/metabolism , Vitamins/pharmacology , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/biosynthesis , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Animals , Blotting, Western , Calcitriol/pharmacokinetics , Calcium/blood , Calcium Channels/biosynthesis , Calcium Channels/genetics , Feedback, Physiological/physiology , Intestinal Mucosa/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorus/blood , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Steroid Hydroxylases/biosynthesis , Steroid Hydroxylases/genetics , TRPV Cation Channels/biosynthesis , TRPV Cation Channels/genetics , Vitamin D3 24-Hydroxylase
13.
PLoS One ; 7(12): e51739, 2012.
Article in English | MEDLINE | ID: mdl-23251616

ABSTRACT

The colonic epithelium continuously regenerates with transitions through various cellular phases including proliferation, differentiation and cell death via apoptosis. Human colonic adenocarcinoma (Caco-2) cells in culture undergo spontaneous differentiation into mature enterocytes in association with progressive increases in expression of glutathione S-transferase alpha-1 (GSTA1). We hypothesize that GSTA1 plays a functional role in controlling proliferation, differentiation and apoptosis in Caco-2 cells. We demonstrate increased GSTA1 levels associated with decreased proliferation and increased expression of differentiation markers alkaline phosphatase, villin, dipeptidyl peptidase-4 and E-cadherin in postconfluent Caco-2 cells. Results of MTS assays, BrdU incorporation and flow cytometry indicate that forced expression of GSTA1 significantly reduces cellular proliferation and siRNA-mediated down-regulation of GSTA1 significantly increases cells in S-phase and associated cell proliferation. Sodium butyrate (NaB) at a concentration of 1 mM reduces Caco-2 cell proliferation, increases differentiation and increases GSTA1 activity 4-fold by 72 hours. In contrast, 10 mM NaB causes significant toxicity in preconfluent cells via apoptosis through caspase-3 activation with reduced GSTA1 activity. However, GSTA1 down-regulation by siRNA does not alter NaB-induced differentiation or apoptosis in Caco-2 cells. While 10 mM NaB causes GSTA1-JNK complex dissociation, phosphorylation of JNK is not altered. These findings suggest that GSTA1 levels may play a role in modulating enterocyte proliferation but do not influence differentiation or apoptosis.


Subject(s)
Glutathione Transferase/pharmacology , Alkaline Phosphatase/metabolism , Apoptosis/drug effects , Butyrates/pharmacology , Caco-2 Cells , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Enzyme Activation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , S Phase/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...