Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
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.
Clin Transl Sci ; 14(5): 1659-1680, 2021 09.
Article in English | MEDLINE | ID: mdl-33982436

ABSTRACT

Nonclinical testing has served as a foundation for evaluating potential risks and effectiveness of investigational new drugs in humans. However, the current two-dimensional (2D) in vitro cell culture systems cannot accurately depict and simulate the rich environment and complex processes observed in vivo, whereas animal studies present significant drawbacks with inherited species-specific differences and low throughput for increased demands. To improve the nonclinical prediction of drug safety and efficacy, researchers continue to develop novel models to evaluate and promote the use of improved cell- and organ-based assays for more accurate representation of human susceptibility to drug response. Among others, the three-dimensional (3D) cell culture models present physiologically relevant cellular microenvironment and offer great promise for assessing drug disposition and pharmacokinetics (PKs) that influence drug safety and efficacy from an early stage of drug development. Currently, there are numerous different types of 3D culture systems, from simple spheroids to more complicated organoids and organs-on-chips, and from single-cell type static 3D models to cell co-culture 3D models equipped with microfluidic flow control as well as hybrid 3D systems that combine 2D culture with biomedical microelectromechanical systems. This article reviews the current application and challenges of 3D culture systems in drug PKs, safety, and efficacy assessment, and provides a focused discussion and regulatory perspectives on the liver-, intestine-, kidney-, and neuron-based 3D cellular models.


Subject(s)
Animal Use Alternatives/methods , Cell Culture Techniques, Three Dimensional , Drug Evaluation, Preclinical/methods , Animal Use Alternatives/standards , Cells, Cultured , Coculture Techniques , Drug Evaluation, Preclinical/standards , Humans , Intestines/cytology , Kidney/cytology , Liver/cytology , Neurons , Spheroids, Cellular , Toxicity Tests/methods , Toxicity Tests/standards , United States , United States Food and Drug Administration/standards
3.
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
4.
Clin Cancer Res ; 24(18): 4365-4370, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29743182

ABSTRACT

Passage of the Biologics Price Competition and Innovation Act of 2009 created an abbreviated licensure pathway for biosimilar products. The FDA approved ABP215 (MVASI, bevacizumab-awwb; Amgen) as a biosimilar to U.S.-licensed Avastin (bevacizumab; Genentech) based on an extensive comparative analytic characterization, data obtained in a pharmacokinetic similarity study in healthy subjects, and a comparative clinical study in patients with non-small cell lung cancer. The totality of the evidence for biosimilarity supported extrapolation of the data to support licensure as a biosimilar for other approved indications of U.S.-licensed Avastin, without the need of additional clinical studies. Clin Cancer Res; 24(18); 4365-70. ©2018 AACR.


Subject(s)
Bevacizumab/therapeutic use , Biosimilar Pharmaceuticals/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug-Related Side Effects and Adverse Reactions/epidemiology , Bevacizumab/pharmacokinetics , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/genetics , Drug Approval , Drug-Related Side Effects and Adverse Reactions/classification , Drug-Related Side Effects and Adverse Reactions/pathology , Humans , Randomized Controlled Trials as Topic , United States , Vascular Endothelial Growth Factor A/genetics
5.
J Pharm Sci ; 107(5): 1330-1341, 2018 05.
Article in English | MEDLINE | ID: mdl-29289674

ABSTRACT

Delivering a drug in amorphous form in a formulated product is a strategy used to enhance the apparent solubility of a drug substance and its oral bioavailability. Drug crystallization in such products may occur during the manufacturing process or on storage, reducing the solubility advantage of the amorphous drug. However, the impact of partial drug crystallization in the drug product on the resulting bioavailability and pharmacokinetics is unknown. In this study, dissolution testing of commercial tacrolimus capsules (which are formulated to contain amorphous drug), both fresh and those containing different amounts of crystalline drug, was conducted using both United States Pharmacopeia and noncompendial dissolution tests with different dissolution media and volumes. A physiologically based pharmacokinetic (PBPK) absorption model was developed to predict the impact of crystallinity extent on the oral absorption of the products and to evaluate the discriminatory ability of the different dissolution methods. Virtual bioequivalence simulations between partially crystallized tacrolimus capsules versus fresh Prograf or generic tacrolimus capsules were performed using the PBPK model and in vitro dissolution data of the various fresh and partially crystallized capsules under United States Pharmacopeia and noncompendial dissolution conditions. The results suggest that compendial dissolution tests may not be sufficiently discriminatory with respect to the presence of crystallinity in an amorphous formulation. Nonsink dissolution tests using lower dissolution volumes generate more discriminatory profiles that predict different pharmacokinetics of tacrolimus capsules containing different extents of drug crystallinity. In conclusion, the PBPK modeling approach can be used to assess the impact of partial drug crystallinity in the formulated product and to guide the development of appropriate dissolution methods.


Subject(s)
Immunosuppressive Agents/pharmacokinetics , Tacrolimus/pharmacokinetics , Capsules , Computer Simulation , Crystallization , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Models, Biological , Powder Diffraction , Solubility , Tacrolimus/administration & dosage , Tacrolimus/chemistry , Tacrolimus/metabolism , Therapeutic Equivalency , X-Ray Diffraction
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Drug Metab Dispos ; 43(4): 631-45, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25650380

ABSTRACT

We applied physiologically based pharmacokinetic (PBPK) modeling to study the dose-dependent metabolism and excretion of verapamil and its preformed metabolite, norverapamil, to unravel the kinetics of norverapamil formation via N-demethylation. Various initial verapamil (1, 50, and 100 µM) and preformed norverapamil (1.5 and 5 µM) concentrations, perfused at 12 ml/min, were investigated in the perfused rat liver preparation. Perfusate and bile were collected over 90 minutes, and livers were harvested at the end of perfusion for high-performance liquid chromatography analysis. After correction for the adsorption of 10%-25% dose verapamil and norverapamil onto Tygon tubing and binding to albumin and red blood cell, fitting of verapamil and formed and preformed norverapamil data with ADAPT5 revealed nonlinearity for protein binding, N-demethylation (V(max,met1)(VER --> NOR) = 96.6 ± 33.4 nmol/min; K(m,met1)(VER --> NOR) = 10.4 ± 4.1 µM), formation of other metabolites (V(max,met2(VER -->others) 288 ± 51 nmol/min; K(m.met2)(VER -->others )= 14.1 ± 4.9 µM), as well as biliary excretion (V(max,sec)(VER)= 0.911 ± 0.505 nmol/min; K(m,sec)(VER) = 4.75 ± 2.29 µM). The hepatic clearance of verapamil (CL(L)(VER) decreased with the dose (8.16-10.2 ml/min), with values remaining high relative to perfusate blood flow rate among the doses. The hepatic clearance of preformed norverapamil (11 ml/min) remained unchanged for the concentrations studied and approximated perfusate blood flow rate, suggesting a high norverapamil extraction ratio. The fractional formation of norverapamil and biliary excretion of verapamil based on fitted constants were 31.1% and 0.64% of CL(L)(VER), respectively. Enantiomeric disposition and auto-inhibition of verapamil failed to perturb these estimaties according to PBPK modeling, due to the low values of the Michaelis-Menten constant, Km, and inhibition parameter, kI.


Subject(s)
Erythrocytes/metabolism , Liver/metabolism , Models, Biological , Verapamil/analogs & derivatives , Animals , Blood Proteins/metabolism , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Male , Metabolic Clearance Rate , Nonlinear Dynamics , Perfusion , Protein Binding , Rats, Sprague-Dawley , Stereoisomerism , Time Factors , Tissue Distribution , Verapamil/blood , Verapamil/chemistry , Verapamil/metabolism , Verapamil/pharmacokinetics
12.
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
13.
J Neurosci ; 34(21): 7091-101, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24849345

ABSTRACT

We demonstrate a role of the vitamin D receptor (VDR) in reducing cerebral soluble and insoluble amyloid-ß (Aß) peptides. Short-term treatment of two human amyloid precursor protein-expressing models, Tg2576 and TgCRND8 mice, with 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], the endogenous active ligand of VDR, resulted in higher brain P-glycoprotein (P-gp) and lower soluble Aß levels, effects negated with coadministration of elacridar, a P-gp inhibitor. Long-term treatment of TgCRND8 mice with 1,25(OH)2D3 during the period of plaque formation reduced soluble and insoluble plaque-associated Aß, particularly in the hippocampus in which the VDR is abundant and P-gp induction is greatest after 1,25(OH)2D3 treatment, and this led to improved conditioned fear memory. In mice fed a vitamin D-deficient diet, lower cerebral P-gp expression was observed, but levels were restored on replenishment with VDR ligands. The composite data suggest that the VDR is an important therapeutic target in the prevention and treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Cerebral Cortex/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Vitamin D/analogs & derivatives , Vitamins/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Cerebral Cortex/drug effects , Conditioning, Psychological/drug effects , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Vitamin D/blood , Vitamin D/therapeutic use
14.
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
15.
Biopharm Drug Dispos ; 34(7): 402-16, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23897575

ABSTRACT

Previous studies have shown that 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ] treatment in mice resulted in induction of intestinal and renal Cyp24a1 and Trpv6 expression, increased hepatic Cyp7a1 expression and activity, as well as higher renal Mdr1/P-gp expression. The present study compared the equimolar efficacies of 1α-hydroxyvitamin D3 [1α(OH)D3 ] (6 nmol/kg i.p. q2d × 4), a lipophilic precursor with a longer plasma half-life that is converted to 1,25(OH)2 D3 , and 1,25(OH)2 D3 on vitamin D receptor (VDR) target genes. To clarify whether changes in VDR genes was due to VDR and not secondary, farnesoid X receptor (FXR)-directed effects, namely, lower Cyp7a1 expression in rat liver due to increased bile acid absorption, wildtype [fxr(+/+)] and FXR knockout [fxr(-/-)] mice were used to distinguish between VDR and FXR effects. With the exception that hepatic Sult2a1 mRNA was increased equally well by 1α(OH)D3 and 1,25(OH)2 D3 , 1α(OH)D3 treatment led to higher increases in hepatic Cyp7a1, renal Cyp24a1, VDR, Mdr1 and Mrp4, and intestinal Cyp24a1 and Trpv6 mRNA expression in both fxr(+/+) and fxr(-/-) mice compared to 1,25(OH)2 D3 treatment. A similar induction in protein expression and microsomal activity of hepatic Cyp7a1 and renal P-gp and Mrp4 protein expression was noted for both compounds. A higher intestinal induction of Trpv6 was observed, resulting in greater hypercalcemic effect following 1α(OH)D3 treatment. The higher activity of 1α(OH)D3 was explained by its rapid conversion to 1,25(OH)2 D3 in tissue sites, furnishing higher plasma and tissue 1,25(OH)2 D3 levels compared to following 1,25(OH)2 D3 -treatment. In conclusion, 1α(OH)D3 exerts a greater effect on VDR gene induction than equimolar doses of 1,25(OH)2 D3 in mice.


Subject(s)
Calcitriol/pharmacology , Hydroxycholecalciferols/pharmacology , Receptors, Calcitriol/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Calcitriol/blood , Calcitriol/pharmacokinetics , Calcium/blood , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Hydroxycholecalciferols/pharmacokinetics , Ileum/drug effects , Ileum/metabolism , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Phosphorus/blood , Sulfotransferases/genetics
16.
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
17.
Curr Drug Metab ; 14(1): 57-79, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22935069

ABSTRACT

Intestinal transporters and enzymes are factors that can influence the absorption of orally administrated drugs. Compartmental models are no longer adequate to describe the sequential handling of drugs and metabolites by the intestine and liver during oral drug absorption, especially when intestinal removal is substantial relative to the liver, and when induction/inhibition elicits different extents of change for identical intestinal and hepatic enzymes or transporters. In this review, we described PBPK models for the intestine (with differential flow patterns: traditional model, TM, and segregated flow model, SFM, and QGut model) as well as semi- or whole bodyphysiological- based pharmacokinetic (PBPK) models to describe the impact of the flow pattern, and the intestinal transporters and enzymes and their attendant heterogeneities on intestinal (FI or FG) and oral (Fsys) bioavailability. The modeling efforts have led to a refinement in providing mechanistic insight on the accurate prediction of drug and metabolite profiles for DDI, pharmacogenomics, age factors and disease conditions.


Subject(s)
Intestinal Absorption , Models, Biological , Pharmaceutical Preparations/metabolism , Administration, Oral , Age Factors , Animals , Drug Interactions , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Pharmaceutical Preparations/administration & dosage , Pharmacogenetics , Pharmacokinetics
18.
J Neurochem ; 123(6): 944-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035695

ABSTRACT

Induction of the multidrug resistance protein 1 (MDR1)/P-glycoprotein (P-gp) by the vitamin D receptor (VDR) was investigated in isolated rat brain capillaries and rat (RBE4) and human (hCMEC/D3) brain microvessel endothelial cell lines. Incubation of isolated rat brain capillaries with 10 nM of the VDR ligand, 1α,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] for 4 h increased P-gp protein expression fourfold. Incubation with 1,25(OH)(2)D(3) for 4 or 24 h increased P-gp transport activity (specific luminal accumulation of NBD-CSA, the fluorescent P-gp substrate) by 25-30%. In RBE4 cells, Mdr1b mRNA was induced in a concentration-dependent manner by exposure to 1,25(OH)(2)D(3). Concomitantly, P-gp protein expression increased 2.5-fold and was accompanied by a 20-35% reduction in cellular accumulation of the P-gp substrates, rhodamine 6G (R6G), and HiLyte Fluor 488-labeled human amyloid beta 1-42 (hAß(42)). In hCMEC/D3 cells, a 3 day exposure to 100 nM 1,25(OH)(2)D(3) increased MDR1 mRNA expression (40%) and P-gp protein (threefold); cellular accumulation of R6G and hAß(42) was reduced by 30%. Thus, VDR activation up-regulates Mdr1/MDR1 and P-gp protein in isolated rat brain capillaries and rodent and human brain microvascular endothelia, implicating a role for VDR in increasing the brain clearance of P-gp substrates, including hAß(42), a plaque-forming precursor in Alzheimer's disease.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/cytology , Brain/blood supply , Calcitriol/metabolism , Calcitriol/physiology , Endothelial Cells/metabolism , Receptors, Calcitriol/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiology , Brain/cytology , Cell Line , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Ligands , Male , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptors, Calcitriol/metabolism , Up-Regulation/physiology
19.
Drug Metab Dispos ; 40(10): 1869-77, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22745334

ABSTRACT

Physiologically based pharmacokinetic (PBPK) models for the intestine, comprising of different flow rates perfusing the enterocyte region, were revisited for appraisal of flow affects on the intestinal availability (F(I)) and, in turn, the systemic availability (F(sys)) and intestinal versus liver contribution to the first-pass effect during oral drug absorption. The traditional model (TM), segregated flow model (SFM), and effective flow (Q(Gut)) model stipulate that 1.0, ∼0.05 to 0.3, and ≤0.484× of the total intestinal flow, respectively, reach the enterocyte region that houses metabolically active and transporter-enriched enterocytes. The fractional flow rate to the enterocyte region (f(Q)), when examined under varying experimental conditions, was found to range from 0.024 to 0.2 for the SFM and 0.065 to 0.43 for the Q(Gut) model. Appraisal of these flow intestinal models, when used in combination with whole-body PBPK models, showed the ranking as SFM < Q(Gut) model < TM in the description of F(I), and the same ranking existed for the contribution of the intestine to first-pass removal. However, the ranking for the predicted contribution of hepatic metabolism, when present, to first-pass removal was the opposite: SFM > Q(Gut) model > TM. The findings suggest that the f(Q) value strongly influences the rate of intestinal metabolism (F(I) and F(sys)) and indirectly affects the rate of liver metabolism due to substrate sparing effect. Thus, the f(Q) value in the intestinal flow models pose serious implications on the interpretation of data on the first-pass effect and oral absorption of drugs.


Subject(s)
Enterohepatic Circulation , Intestinal Absorption , Intestinal Mucosa/metabolism , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Administration, Oral , Animals , Biological Availability , Computer Simulation , Enterocytes/metabolism , Humans , Intestines/cytology , Liver/metabolism , Pharmaceutical Preparations/administration & dosage
20.
J Pharmacol Exp Ther ; 337(3): 846-59, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21421739

ABSTRACT

Secondary farnesoid X receptor (FXR) effects, in addition to vitamin D receptor (VDR) effects, were observed in the rat liver after treatment with 1α,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)], the natural ligand of VDR, caused by increased bile acid absorption as a consequence of apical sodium-dependent bile acid transporter induction. To investigate whether the increased multidrug resistance protein 1 (Mdr1)/P-glycoprotein (P-gp) expression in the rat liver and kidney was caused by the VDR and not the FXR, we examined changes in Mdr1/P-gp expression in fxr(+/+) and fxr(-/-) mice after intraperitoneal dosing of vehicle versus 1,25(OH)(2)D(3) (0 or 2.5 µg/kg every other day for 8 days). Renal and brain levels of Mdr1 mRNA and P-gp protein were significantly increased in both fxr(+/+) and fxr(-/-) mice treated with 1,25(OH)(2)D(3), confirming that Mdr1/P-gp induction occurred independently of the FXR. Increased P-gp function was evident in 1,25(OH)(2)D(3)-treated fxr(+/+) mice given intravenous bolus doses of the P-gp probe, [(3)H]digoxin (0.1 mg/kg). Decreased blood (24%) and brain (29%) exposure, estimated as reduced areas under the curve, caused by increased renal (74%) and total body (34%) clearances of digoxin, were observed in treated mice. These events were predicted by physiologically based pharmacokinetic modeling that showed increased renal secretory intrinsic clearance (3.45-fold) and brain efflux intrinsic clearance (1.47-fold) in the 1,25(OH)(2)D(3)-treated mouse, trends that correlated well with increases in P-gp protein expression in tissues. The clearance changes were less apparent because of the high degree of renal reabsorption of digoxin. The observations suggest an important role of the VDR in the regulation of P-gp in the renal and brain disposition of P-gp substrates.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/drug effects , Cardiotonic Agents/pharmacology , Digoxin/pharmacokinetics , Kidney/drug effects , Receptors, Calcitriol/metabolism , Vitamin D/analogs & derivatives , Vitamins/pharmacology , Animals , Area Under Curve , Bile Acids and Salts/metabolism , Brain/metabolism , Cardiotonic Agents/pharmacokinetics , Digoxin/metabolism , Digoxin/pharmacology , Female , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Symporters/metabolism , Up-Regulation/drug effects , Vitamin D/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...