Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
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.
Toxicol In Vitro ; 67: 104920, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32590029

ABSTRACT

Gut microbiota can impact liver disease development via the gut-liver axis. Liver inflammation is a shared pathological event in various liver diseases and gut microbiota might influence this pathological process. In this study, we studied the influence of gut microbiota on the inflammatory response of the liver to lipopolysaccharide (LPS). The inflammatory response to LPS (1-10 µg/ml) of livers of specific-pathogen-free (SPF) or germ-free (GF) mice was evaluated ex vivo, using precision-cut liver slices (PCLS). LPS induced a more pronounced inflammatory response in GF PCLS than in SPF PCLS. Baseline TNF-α gene expression was significantly higher in GF slices as compared to SPF slices. LPS treatment induced TNF-α, IL-1ß, IL-6 and iNOS expression in both SPF and GF PCLS, but the increase was more intense in GF slices. The anti-inflammatory markers SOCS3 and IRAK-M gene expression was significantly higher in GF PCLS than SPF PCLS at 24h with 1 µg/ml LPS treatment, and IL-10 was not differently expressed in GF PCLS than SPF PCLS. In addition, TLR-4 mRNA, but not protein, at basal level was higher in GF slices than in SPF slices. Taken together, this study shows that, in mice, the host microbiota attenuates the pro-inflammatory impact of LPS in the liver, indicating a positive role of the gut microbiota on the immune homeostasis of the liver.


Subject(s)
Lipopolysaccharides/pharmacology , Liver/drug effects , Microbiota , Animals , Cytokines/genetics , Cytokines/immunology , Germ-Free Life , Inflammation/genetics , Inflammation/immunology , Interleukin-1 Receptor-Associated Kinases/genetics , Liver/immunology , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
3.
Chem Commun (Camb) ; 56(44): 5941-5944, 2020 Jun 04.
Article in English | MEDLINE | ID: mdl-32347235

ABSTRACT

Targeted epitope-based mass spectrometry imaging (MSI) utilizes laser cleavable mass-tags bound to targeting moieties for detecting proteins in tissue sections. Our work constitutes the first proof-of-concept of a novel laser desorption ionization (LDI)-MSI strategy using photocleavable Ru(ii) polypyridine complexes as mass-tags for imaging of integrins αvß3 in human cancer tissues.


Subject(s)
Head and Neck Neoplasms/metabolism , Integrin alphaVbeta3/metabolism , Peptides, Cyclic/pharmacology , Pyridines/pharmacology , Ruthenium/pharmacology , Humans , Mass Spectrometry/methods , Peptides, Cyclic/chemistry , Pyridines/chemistry , Ruthenium/chemistry
4.
Toxicol Res (Camb) ; 8(6): 885-895, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-32190293

ABSTRACT

Gold-based compounds are of great interest in the field of medicinal chemistry as novel therapeutic (anticancer) agents due to their peculiar reactivity and mechanisms of action with respect to organic drugs. Despite their promising pharmacological properties, the possible toxic effects of gold compounds need to be carefully evaluated in order to optimize their design and applicability. This study reports on the potential toxicity of three experimental gold-based anticancer compounds featuring lansoprazole ligands (1-3) studied in an ex vivo model, using rat precision cut kidney and liver slices (PCKS and PCLS, respectively). The results showed a different toxicity profile for the tested compounds, with the neutral complex 2 being the least toxic, even less toxic than cisplatin, followed by the cationic complex 1. The dinuclear cationic gold complex 3 was the most toxic in both liver and kidney slices. This result correlated with the metal uptake of the different compounds assessed by ICP-MS, where complex 3 showed the highest accumulation of gold in liver and kidney slices. Interestingly compound 1 showed the highest selectivity towards cancer cells compared to the healthy tissues. Histomorphology evaluation showed a similar pattern for all three Au(i) complexes, where the distal tubular cells suffered the most extensive damage, in contrast to the damage in the proximal tubules induced by cisplatin. The binding of representative gold compounds with the model ubiquitin was also studied by ESI-MS, showing that after 24 h incubation only 'naked' Au ions were bound to the protein following ligands' loss. The mRNA expression of stress response genes appeared to be similar for both evaluated organs, suggesting oxidative stress as the possible mechanism of toxicity. The obtained results open new perspectives towards the design and testing of bifunctional gold complexes with chemotherapeutic applications.

5.
Bioconjug Chem ; 29(11): 3856-3865, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30380298

ABSTRACT

Cisplatin occupies a crucial role in the treatment of various malignant tumors. However, its efficacy and applicability are heavily restricted by severe systemic toxicities and drug resistance. Our study exploits the active targeting of supramolecular metallacages to enhance the activity of cisplatin in cancer cells while reducing its toxicity. Thus, Pd2L4 cages (L = ligand) have been conjugated to four integrin ligands with different binding affinity and selectivity. Cage formation and encapsulation of cisplatin was proven by NMR spectroscopy. Upon encapsulation, cisplatin showed increased cytotoxicity in vitro, in melanoma A375 cells overexpressing αvß3 integrins. Moreover, ex vivo studies in tissue slices indicated reduced toxicity toward healthy liver and kidney tissues for cage-encapsulated cisplatin. Analysis of metal content by ICP-MS demonstrated that the encapsulated drug is less accumulated in these organs compared to the "free" cisplatin.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Drug Carriers/metabolism , Integrin alphaVbeta3/metabolism , Melanoma/drug therapy , Metal-Organic Frameworks/metabolism , Palladium/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Drug Carriers/chemistry , Drug Delivery Systems , Humans , Ligands , Male , Melanoma/metabolism , Metal-Organic Frameworks/chemistry , Palladium/chemistry , Rats, Wistar
7.
Rev Physiol Biochem Pharmacol ; 175: 71-102, 2018.
Article in English | MEDLINE | ID: mdl-29728869

ABSTRACT

Oxidative stress is a reflection of the imbalance between the production of reactive oxygen species (ROS) and the scavenging capacity of the antioxidant system. Excessive ROS, generated from various endogenous oxidative biochemical enzymes, interferes with the normal function of liver-specific cells and presumably plays a role in the pathogenesis of liver fibrosis. Once exposed to harmful stimuli, Kupffer cells (KC) are the main effectors responsible for the generation of ROS, which consequently affect hepatic stellate cells (HSC) and hepatocytes. ROS-activated HSC undergo a phenotypic switch and deposit an excessive amount of extracellular matrix that alters the normal liver architecture and negatively affects liver function. Additionally, ROS stimulate necrosis and apoptosis of hepatocytes, which causes liver injury and leads to the progression of end-stage liver disease. In this review, we overview the role of ROS in liver fibrosis and discuss the promising therapeutic interventions related to oxidative stress. Most importantly, novel drugs that directly target the molecular pathways responsible for ROS generation, namely, mitochondrial dysfunction inhibitors, endoplasmic reticulum stress inhibitors, NADPH oxidase (NOX) inhibitors, and Toll-like receptor (TLR)-affecting agents, are reviewed in detail. In addition, challenges for targeting oxidative stress in the management of liver fibrosis are discussed.


Subject(s)
Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/therapy , Oxidative Stress , Reactive Oxygen Species/metabolism , Endoplasmic Reticulum Stress/drug effects , Humans , Mitochondria/drug effects , NADPH Oxidases/antagonists & inhibitors , Toll-Like Receptors/antagonists & inhibitors
9.
Eur J Pharm Sci ; 115: 175-184, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29309877

ABSTRACT

Drug-induced liver injury (DILI) is a common reason for drug withdrawal from the market. An important cause of DILI is drug-induced cholestasis. One of the major players involved in drug-induced cholestasis is the bile salt efflux pump (BSEP; ABCB11). Inhibition of BSEP by drugs potentially leads to cholestasis due to increased (toxic) intrahepatic concentrations of bile acids with subsequent cell injury. In order to investigate the possibilities for in silico prediction of cholestatic effects of drugs, we developed a mechanistic biokinetic model for human liver bile acid handling populated with human in vitro data. For this purpose we considered nine groups of bile acids in the human bile acid pool, i.e. chenodeoxycholic acid, deoxycholic acid, the remaining unconjugated bile acids and the glycine and taurine conjugates of each of the three groups. Michaelis-Menten kinetics of the human uptake transporter Na+-taurocholate cotransporting polypeptide (NTCP; SLC10A1) and BSEP were measured using NTCP-transduced HEK293 cells and membrane vesicles from BSEP-overexpressing HEK293 cells. For in vitro-in vivo scaling, transporter abundance was determined by LC-MS/MS in these HEK293 cells and vesicles as well as in human liver tissue. Other relevant human kinetic parameters were collected from literature, such as portal bile acid levels and composition, bile acid synthesis and amidation rate. Additional empirical scaling was applied by increasing the excretion rate with a factor 2.4 to reach near physiological steady-state intracellular bile acid concentrations (80µM) after exposure to portal vein bile acid levels. Simulations showed that intracellular bile acid concentrations increase 1.7 fold in the presence of the BSEP inhibitors and cholestatic drugs cyclosporin A or glibenclamide, at intrahepatic concentrations of 6.6 and 20µM, respectively. This simplified model provides a tool for a first indication whether drugs at therapeutic concentrations might cause cholestasis by inhibiting BSEP.


Subject(s)
Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Cholestasis/chemically induced , Cholestasis/metabolism , Liver/metabolism , Pharmaceutical Preparations/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , Biological Transport/drug effects , Cell Line , HEK293 Cells , Humans , Kinetics , Membrane Transport Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism
10.
Chembiochem ; 19(7): 736-743, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29356267

ABSTRACT

Formaldehyde fixation is widely used for long-term maintenance of tissue. However, due to formaldehyde-induced crosslinks, fixed tissue proteins are difficult to extract, which hampers mass spectrometry (MS) proteomic analyses. Recent years have seen the use of different combinations of high temperature and solubilizing agents (usually derived from antigen retrieval techniques) to unravel formaldehyde-fixed paraffin-embedded tissue proteomes. However, to achieve protein extraction yields similar to those of fresh-frozen tissue, high-temperature heating is necessary. Such harsh extraction conditions can affect sensitive amino acids and post-translational modifications, resulting in the loss of important information, while still not resulting in protein yields comparable to those of fresh-frozen tissue. Herein, the objective is to evaluate cleavable protein crosslinkers as fixatives that allow tissue preservation and efficient protein extraction from fixed tissue for MS proteomics under mild conditions. With this goal in mind, disuccinimidyl tartrate (DST) and dithiobis(succinimidylpropionate) (DSP) are investigated as cleavable fixating reagents. These compounds crosslink proteins by reacting with amino groups, leading to amide bond formation, and can be cleaved with sodium metaperiodate (cis-diols, DST) or reducing agents (disulfide bonds, DSP), respectively. Results show that cleavable protein crosslinking with DST and DSP allows tissue fixation with morphology preservation comparable to that of formaldehyde. In addition, cleavage of DSP improves protein recovery from fixed tissue by a factor of 18 and increases the number of identified proteins by approximately 20 % under mild extraction conditions compared with those of formaldehyde-fixed paraffin-embedded tissue. A major advantage of DSP is the introduction of well-defined protein modifications that can be taken into account during database searching. In contrast to DSP fixation, DST fixation followed by cleavage with sodium metaperiodate, although effective, results in side reactions that prevent effective protein extraction and interfere with protein identification. Protein crosslinkers that can be cleaved under mild conditions and result in defined modifications, such as DSP, are thus viable alternatives to formaldehyde as tissue fixatives to facilitate protein analysis from paraffin-embedded, fixed tissue.


Subject(s)
Cross-Linking Reagents/chemistry , Fixatives/chemistry , Proteome/analysis , Succinimides/chemistry , Tissue Fixation/methods , Animals , Chromatography, Liquid , Liver/cytology , Periodic Acid/chemistry , Proteome/chemistry , Proteomics/methods , Rats , Tandem Mass Spectrometry
11.
J Tissue Eng Regen Med ; 12(1): e227-e236, 2018 01.
Article in English | MEDLINE | ID: mdl-27943623

ABSTRACT

Over recent years, the progress in the development of a bioartificial liver (BAL) as an extracorporeal device or as a tissue engineered transplantable organ has been immense. However, many important BAL characteristics that are necessary to meet clinical demands have not been sufficiently addressed. This review describes the existing challenges in the development of a BAL for clinical applications, highlighting multicellularity and sinusoidal microarchitecture as crucial BAL characteristics to fulfil various liver functions. Currently available sources of nonparenchymal liver cells, such as endothelial cells, cholangiocytes and macrophages, used in BAL development are defined. Also, we discuss the recent studies on the reconstruction of the complex liver sinusoid microarchitecture using various liver cell types. Moreover, we highlight some other aspects of a BAL, such as liver zonation and formation of a vascular as well as biliary network for an adequate delivery, biotransformation and removal of substrates and waste products. Finally, the benefits of a multicellular BAL for the pharmaceutical industry are briefly described. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Liver, Artificial , Acute-On-Chronic Liver Failure/therapy , Animals , Bile/metabolism , Coculture Techniques , Humans , Liver Failure, Acute/therapy , Pharmaceutical Preparations
12.
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
13.
Metallomics ; 9(12): 1786-1795, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29104982

ABSTRACT

Mechanisms of toxicity and cellular transport of anticancer metallodrugs, including platinum-based agents, have not yet been fully elucidated. Here, we studied the toxic effects and accumulation mechanisms of cisplatin in healthy rat kidneys ex vivo, using the Precision Cut Tissue Slices (PCTS) method. In addition, for the first time, we investigated the nephrotoxic effects of an experimental anticancer cyclometallated complex [Au(pyb-H)(PTA)Cl]PF6 (PTA = 1,3,5-triazaphosphaadamantane). The viability of the kidney slices after metallodrug treatment was evaluated by ATP content determination and histomorphology analysis. A concentration dependent decrease in viability of PCKS was observed after exposure to cisplatin or the Au(iii) complex, which correlated with the increase in slice content of Pt and Au, respectively. Metal accumulation in kidney slices was analysed by ICP-MS. The involvement of OCTs and MATE transporters in the accumulation of both metal compounds in kidneys was evaluated co-incubating the tissues with cimitedine, inhibitor of OCT and MATE. Studies of mRNA expression of the markers KIM-1, villin, p53 and Bax showed that cisplatin damages proximal tubules, whereas the Au(iii) complex preferentially affects the distal tubules. However, no effect of cimetidine on the toxicity or accumulation of cisplatin and the Au(iii) complex was observed. The effect of temperature on metallodrug accumulation in kidneys suggests the involvement of a carrier-mediated uptake process, other than OCT2, for cisplatin; while carrier-mediated excretion was suggested in the cases of the Au(iii) complex.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cytotoxins/toxicity , Gold/toxicity , Kidney/metabolism , Adenosine Triphosphate/metabolism , Animals , Antiporters/metabolism , Kidney/drug effects , Kidney/pathology , Male , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism , Rats , Rats, Wistar
14.
ChemMedChem ; 12(17): 1429-1435, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28741878

ABSTRACT

A series of organometallic AuI N-heterocyclic carbene (NHC) complexes was synthesized and characterized for anticancer activity in four human cancer cell lines. The compounds' toxicity in healthy tissue was determined using precision-cut kidney slices (PCKS) as a tool to determine the potential selectivity of the gold complexes ex vivo. All evaluated compounds presented cytotoxic activity toward the cancer cells in the nano- or low micromolar range. The mixed AuI NHC complex, (tert-butylethynyl)-1,3-bis-(2,6-diisopropylphenyl)imidazol-2-ylidene gold(I), bearing an alkynyl moiety as ancillary ligand, showed high cytotoxicity in cancer cells in vitro, while being barely toxic in healthy rat kidney tissues. The obtained results open new perspectives toward the design of mixed NHC-alkynyl gold complexes for cancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Organogold Compounds/chemistry , Organogold Compounds/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Line, Tumor , Heterocyclic Compounds/toxicity , Humans , Kidney/drug effects , Male , Methane/analogs & derivatives , Methane/chemistry , Methane/pharmacology , Methane/toxicity , Neoplasms/drug therapy , Organogold Compounds/toxicity , Rats, Wistar
15.
Arch Toxicol ; 91(10): 3403-3413, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28391356

ABSTRACT

Drug-induced cholestasis (DIC) is one of the leading manifestations of drug-induced liver injury (DILI). As the underlying mechanisms for DIC are not fully known and specific and predictive biomarkers and pre-clinical models are lacking, the occurrence of DIC is often only reported when the drug has been approved for registration. Therefore, appropriate models that predict the cholestatic potential of drug candidates and/or provide insight into the mechanism of DIC are highly needed. We investigated the application of rat precision-cut liver slices (PCLS) to predict DIC, using several biomarkers of cholestasis: hepatocyte viability, intracellular accumulation of total as well as individual bile acids and changes in the expression of genes known to play a role in cholestasis. Rat PCLS exposed to the cholestatic drugs chlorpromazine, cyclosporine A and glibenclamide for 48 h in the presence of a 60 µM physiological bile acid (BA) mix reflected various changes associated with cholestasis, such as decrease in hepatocyte viability, accumulation and changes in the composition of BA and changes in the gene expression of Fxr, Bsep and Ntcp. The toxicity of the drugs was correlated with the accumulation of BA, and especially DCA and CDCA and their conjugates, but to a different extent for different drugs, indicating that BA toxicity is not the only cause for the toxicity of cholestatic drugs. Moreover, our study supports the use of several biomarkers to test drugs for DIC. In conclusion, our results indicate that PCLS may represent a physiological and valuable model to identify cholestatic drugs and provide insight into the mechanisms underlying DIC.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Organ Culture Techniques/methods , Toxicity Tests/methods , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , Animals , Bile Acids and Salts/metabolism , Cholestasis/chemically induced , Gene Expression Regulation/drug effects , Liver/metabolism , Liver/pathology , Male , Organic Anion Transporters, Sodium-Dependent/genetics , Rats, Wistar , Symporters/genetics
16.
Arch Toxicol ; 91(3): 1401-1412, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27344345

ABSTRACT

Hepatotoxicity is one of the major reasons for withdrawal of drugs from the market. Therefore, there is a need to screen new drugs for hepatotoxicity in humans at an earlier stage. The aim of this study was to validate human precision-cut liver slices (PCLS) as an ex vivo model to predict drug-induced cholestasis and identify the possible mechanisms of cholestasis-induced toxicity using gene expression profiles. Five hepatotoxicants, which are known to induce cholestasis (alpha-naphthyl isothiocyanate, chlorpromazine, cyclosporine, ethinyl estradiol and methyl testosterone) were used at concentrations inducing low (<30 %) and medium (30-50 %) toxicity, based on ATP content. Human PCLS were incubated with the drugs in the presence of a non-toxic concentration (60 µM) of a bile acid mixture (portal vein concentration and composition) as model for bile acid-induced cholestasis. Regulated genes include bile acid transporters and cholesterol transporters. Pathway analysis revealed that hepatic cholestasis was among the top ten regulated pathways, and signaling pathways such as farnesoid X receptor- and liver X receptor-mediated responses, which are known to play a role in cholestasis, were significantly affected by all cholestatic compounds. Other significantly affected pathways include unfolded protein response and protein ubiquitination implicating the role of endoplasmic reticulum stress. This study shows that human PCLS incubated in the presence of a physiological bile acid mixture correctly reflect the pathways affected in drug-induced cholestasis in the human liver. In the future, this human PCLS model can be used to identify cholestatic adverse drug reactions of new chemical entities.


Subject(s)
Cholestasis/chemically induced , Liver/drug effects , 1-Naphthylisothiocyanate/toxicity , Aged , Chlorpromazine/adverse effects , Cholestasis/genetics , Cyclosporine/adverse effects , Dose-Response Relationship, Drug , Ethinyl Estradiol/adverse effects , Female , Gene Expression Profiling/methods , Humans , Male , Methyltestosterone/adverse effects , Middle Aged , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Young Adult
18.
Biopharm Drug Dispos ; 38(2): 127-138, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27757966

ABSTRACT

Although intestinal P-glycoprotein (P-gp) has been extensively studied in vitro and in animals, its activity and the consequences of P-gp inhibition for drug disposition and toxicity in humans are still difficult to accurately extrapolate from these studies. Moreover, existing in vitro models do not take into consideration that the intestine is heterogeneous with respect to P-gp expression. Recently, we reported rat precision-cut intestinal slices (PCIS) as a physiological ex vivo model to study the regional gradient of P-gp activity and inhibition. Here we extended the application of PCIS to the human intestine. For this purpose rhodamine 123 (R123) accumulation in the presence or absence of the P-gp inhibitors verapamil, cyclosporine A, quinidine, ketoconazole, PSC833 and CP100356 was measured in PCIS of human duodenum, jejunum, ileum and colon. R123 accumulation in the presence of the P-gp inhibitors appeared to be most enhanced in the ileum compared to the other regions. Moreover, the regional differences in accumulation are in line with published differences in abundance of P-gp. The rank order of the potency of the P-gp inhibitors, reflected by their IC50 , was comparable to that in rat PCIS. However, the increase in accumulation of the P-gp substrate R123 by the inhibitors was larger in human ileum PCIS than in rat PCIS, indicating species difference in P-gp abundance. These data show that human PCIS are an appropriate ex vivo model to study the activity of intestinal P-gp and predict the inhibitory effect of drugs and of transporter-mediated drug-drug interactions in the human intestine. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Colon/drug effects , Colon/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Membrane Transport Modulators/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Aged , Biological Transport , Dose-Response Relationship, Drug , Duodenum/drug effects , Duodenum/metabolism , Female , Fluorescent Dyes/metabolism , Humans , Ileum/drug effects , Ileum/metabolism , In Vitro Techniques , Jejunum/drug effects , Jejunum/metabolism , Kinetics , Male , Middle Aged , Rhodamine 123/metabolism
19.
Toxicol In Vitro ; 40: 26-33, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27939799

ABSTRACT

Intestinal P-gp and CYP3A4 work coordinately to reduce the intracellular concentration of drugs, and drug-drug interactions (DDIs) based on this interplay are of clinical importance and require pre-clinical investigation. Using precision-cut intestinal slices (PCIS) of human jejunum, ileum and colon, we investigated the P-gp/CYP3A4 interplay and related DDIs with P-gp inhibitors at the different regions of the human intestine with quinidine (Qi), dual substrate of P-gp and CYP3A4, as probe. All the P-gp inhibitors increased the intracellular concentrations of Qi by 2.1-2.6 fold in jejunum, 2.6-3.8 fold in ileum but only 1.2-1.3 fold in colon, in line with the different P-gp expression in these intestinal regions. The selective P-gp inhibitors (CP100356 and PSC833) enhanced 3-hydroxy-quinidine (3OH-Qi) in jejunum and ileum, while dual inhibitors of P-gp and CYP3A4 (verapamil and ketoconazole) decreased the 3OH-Qi production, despite of the increased intracellular Qi concentration, due to inhibition of CYP3A4. The outcome of DDIs based on P-gp/CYP3A4 interplay, shown as remarkable changes in the intracellular concentration of both the parent drug and the metabolite, varied among the intestinal regions, probably due to the different expression of P-gp and CYP3A4, and were different from those found in rat PCIS, which may have important implications for the disposition and toxicity of drugs and their metabolites.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Colon/metabolism , Cytochrome P-450 CYP3A/metabolism , Ileum/metabolism , Jejunum/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Cyclosporins/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions , Female , Humans , In Vitro Techniques , Isoquinolines/pharmacology , Ketoconazole/pharmacology , Male , Middle Aged , Quinazolines/pharmacology , Quinidine/analogs & derivatives , Quinidine/metabolism , Quinidine/pharmacokinetics , Verapamil/pharmacology
20.
Arch Toxicol ; 91(5): 2079-2092, 2017 May.
Article in English | MEDLINE | ID: mdl-27717970

ABSTRACT

Human precision-cut liver slices (hPCLS) are a valuable ex vivo model that can be used in acute toxicity studies. However, a rapid decline in metabolic enzyme activity limits their use in studies that require a prolonged xenobiotic exposure. The aim of the study was to extend the viability and function of hPCLS to 5 days of incubation. hPCLS were incubated in two media developed for long-term culture of hepatocytes, RegeneMed®, and Cellartis®, and in the standard medium WME. Maintenance of phase I and II metabolism was studied both on gene expression as well as functional level using a mixture of CYP isoform-specific substrates. Albumin synthesis, morphological integrity, and glycogen storage was assessed, and gene expression was studied by transcriptomic analysis using microarrays with a focus on genes involved in drug metabolism, transport and toxicity. The data show that hPCLS retain their viability and functionality during 5 days of incubation in Cellartis® medium. Albumin synthesis as well as the activity and gene expression of phase I and II metabolic enzymes did not decline during 120-h incubation in Cellartis® medium, with CYP2C9 activity as the only exception. Glycogen storage and morphological integrity were maintained. Moreover, gene expression changes in hPCLS during incubation were limited and mostly related to cytoskeleton remodeling, fibrosis, and moderate oxidative stress. The expression of genes involved in drug transport, which is an important factor in determining the intracellular xenobiotic exposure, was also unchanged. Therefore, we conclude that hPCLS cultured in Cellartis® medium are a valuable human ex vivo model for toxicological and pharmacological studies that require prolonged xenobiotic exposure.


Subject(s)
Enzymes/metabolism , Liver/metabolism , Organ Culture Techniques/methods , Adenosine Triphosphate/metabolism , Albumins/biosynthesis , Carrier Proteins/metabolism , Culture Media , Fibrosis/genetics , Gene Expression Regulation , Humans , Inactivation, Metabolic , Liver/drug effects , Liver/pathology , Oxidative Stress/genetics , Xenobiotics/metabolism , Xenobiotics/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...