Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Basic Res Cardiol ; 119(2): 309-327, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38305903

ABSTRACT

Statins are effective drugs in reducing cardiovascular morbidity and mortality by inhibiting cholesterol synthesis. These effects are primarily beneficial for the patient's vascular system. A significant number of statin users suffer from muscle complaints probably due to mitochondrial dysfunction, a mechanism that has recently been elucidated. This has raised our interest in exploring the effects of statins on cardiac muscle cells in an era where the elderly and patients with poorer functioning hearts and less metabolic spare capacity start dominating our patient population. Here, we investigated the effects of statins on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-derived CMs). hiPSC-derived CMs were exposed to simvastatin, atorvastatin, rosuvastatin, and cerivastatin at increasing concentrations. Metabolic assays and fluorescent microscopy were employed to evaluate cellular viability, metabolic capacity, respiration, intracellular acidity, and mitochondrial membrane potential and morphology. Over a concentration range of 0.3-100 µM, simvastatin lactone and atorvastatin acid showed a significant reduction in cellular viability by 42-64%. Simvastatin lactone was the most potent inhibitor of basal and maximal respiration by 56% and 73%, respectively, whereas simvastatin acid and cerivastatin acid only reduced maximal respiration by 50% and 42%, respectively. Simvastatin acid and lactone and atorvastatin acid significantly decreased mitochondrial membrane potential by 20%, 6% and 3%, respectively. The more hydrophilic atorvastatin acid did not seem to affect cardiomyocyte metabolism. This calls for further research on the translatability to the clinical setting, in which a more conscientious approach to statin prescribing might be considered, especially regarding the current shift in population toward older patients with poor cardiac function.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Induced Pluripotent Stem Cells , Simvastatin/analogs & derivatives , Humans , Aged , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocytes, Cardiac/metabolism , Atorvastatin/pharmacology , Simvastatin/pharmacology , Mitochondria/metabolism , Lactones/metabolism , Lactones/pharmacology , Hydrogen-Ion Concentration
2.
Curr Res Toxicol ; 6: 100149, 2024.
Article in English | MEDLINE | ID: mdl-38292667

ABSTRACT

Tofacitinib is a small molecule Janus kinase (JAK) inhibitor, introduced to the European market in 2017, for the treatment of rheumatoid arthritis, psoriatic arthritis and ulcerative colitis. In the treatment of women with autoimmune diseases, pregnancy is a relevant issue, as such diseases typically affect women in their reproductive years. Currently, there is limited data on the use of tofacitinib during pregnancy. To estimate the extent of placental transfer in the absence of clinical data, we conducted ex vivo dual-side perfused human placental cotyledon perfusions. Term placentas were perfused for 180 min with tofacitinib (100 nM, added to the maternal circuit) in a closed-closed configuration. At the end of the perfusions, drug concentrations in the maternal and fetal reservoirs were near equilibrium, at 35.6 ± 5.5 and 24.8 ± 4.7 nM, respectively. Transfer of tofacitinib was similar to that observed for the passive diffusion marker antipyrine (100 µg/mL, added to the maternal reservoir). Final antipyrine maternal and fetal concentrations amounted to 36.9 ± 3.0 and 36.7 ± 1.3 µg/mL, respectively. In conclusion, in the ex vivo perfused placenta tofacitinib traverses the placental barrier rapidly and extensively. This suggests that substantial fetal tofacitinib exposure will take place after maternal drug dosing.

3.
Antibiotics (Basel) ; 12(4)2023 Apr 03.
Article in English | MEDLINE | ID: mdl-37107064

ABSTRACT

Linezolid is used off-label for treatment of central nervous system infections. However, its pharmacokinetics and target attainment in cranial cerebrospinal fluid (CSF) in tuberculous meningitis patients is unknown. This study aimed to predict linezolid cranial CSF concentrations and assess attainment of pharmacodynamic (PD) thresholds (AUC:MIC of >119) in plasma and cranial CSF of adults and children with tuberculous meningitis. A physiologically based pharmacokinetic (PBPK) model was developed to predict linezolid cranial CSF profiles based on reported plasma concentrations. Simulated steady-state PK curves in plasma and cranial CSF after linezolid doses of 300 mg BID, 600 mg BID, and 1200 mg QD in adults resulted in geometric mean AUC:MIC ratios in plasma of 118, 281, and 262 and mean cranial CSF AUC:MIC ratios of 74, 181, and 166, respectively. In children using ~10 mg/kg BID linezolid, AUC:MIC values at steady-state in plasma and cranial CSF were 202 and 135, respectively. Our model predicts that 1200 mg per day in adults, either 600 mg BID or 1200 mg QD, results in reasonable (87%) target attainment in cranial CSF. Target attainment in our simulated paediatric population was moderate (56% in cranial CSF). Our PBPK model can support linezolid dose optimization efforts by simulating target attainment close to the site of TBM disease.

4.
Toxicol In Vitro ; 80: 105327, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35134484

ABSTRACT

Tumor necrosis factor (TNF) regulates trophoblast turnover during the formation of the placental syncytium and can be a potentially relevant target for adverse effects of xenobiotics. We mimicked syncytialization in vitro by stimulating BeWo cells with 50 µM forskolin. Undifferentiated and syncytialized BeWo cells were exposed to TNF (10 pg/mL-10 ng/mL) for 48 h after which cell viability, progesterone release and gene expression of a selected set of markers representative for placental function were assessed. In undifferentiated BeWo cells, high TNF levels (1-10 ng/mL) increased gene expression of TNF, NF-κB, and TNFRSF1B to maximally 99 ± 17, 2.2 ± 0.2, and 3.0 ± 0.4 of control values, respectively (p < 0.001). These effects were also found in syncytialized BeWo cells but less pronounced. Additionally, TNF may induce syncytialization in BeWo cells as it upregulated ERVW-1 expression by 1.55 ± 0.14-fold (p < 0.05). On the contrary, TNF levels of 10 and 100 pg/mL did not affect gene expression in both undifferentiated and syncytialized BeWo cells, but did enhance cell viability in syncytialised BeWo cells (p < 0.001). In conclusion, we found that high TNF levels (1-10 ng/mL) increased gene expression of TNF, NF-κB, and TNFRSF1B especially in undifferentiated BeWo cells, while physiological TNF concentrations positively affected cell viability and while there was no effect on any of the investigated functional markers.


Subject(s)
Trophoblasts/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Cell Line , Cell Survival/drug effects , Colforsin/pharmacology , Female , Gene Expression , Humans , Pregnancy , Progesterone/metabolism , Trophoblasts/metabolism
5.
J Clin Pharmacol ; 62(3): 385-396, 2022 03.
Article in English | MEDLINE | ID: mdl-34554580

ABSTRACT

Moxifloxacin has an important role in the treatment of tuberculosis (TB). Unfortunately, coadministration with the cornerstone TB drug rifampicin results in suboptimal plasma exposure. We aimed to gain insight into the moxifloxacin pharmacokinetics and the interaction with rifampicin. Moreover, we provided a mechanistic framework to understand moxifloxacin pharmacokinetics. We developed a physiologically based pharmacokinetic model in Simcyp version 19, with available and newly generated in vitro and in vivo data, to estimate pharmacokinetic parameters of moxifloxacin alone and when administered with rifampicin. By combining these strategies, we illustrate that the role of P-glycoprotein in moxifloxacin transport is limited and implicate MRP2 as transporter of moxifloxacin-glucuronide followed by rapid hydrolysis in the gut. Simulations of multiple dose area under the plasma concentration-time curve (AUC) of moxifloxacin (400 mg once daily) with and without rifampicin (600 mg once daily) were in accordance with clinically observed data (predicted/observed [P/O] ratio of 0.87 and 0.80, respectively). Importantly, increasing the moxifloxacin dose to 600 mg restored the plasma exposure both in actual patients with TB as well as in our simulations. Furthermore, we extrapolated the single dose model to pediatric populations (P/O AUC ratios, 1.04-1.52) and the multiple dose model to children with TB (P/O AUC ratio, 1.51). In conclusion, our combined approach resulted in new insights into moxifloxacin pharmacokinetics and accurate simulations of moxifloxacin exposure with and without rifampicin. Finally, various knowledge gaps were identified, which may be considered as avenues for further physiologically based pharmacokinetic refinement.


Subject(s)
Antitubercular Agents/pharmacology , Moxifloxacin/pharmacokinetics , Rifampin/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Adult , Antitubercular Agents/pharmacokinetics , Area Under Curve , Child , Drug Therapy, Combination , Glucuronosyltransferase/metabolism , HEK293 Cells , Humans , Models, Biological , Multidrug Resistance-Associated Protein 2/metabolism
6.
Br J Clin Pharmacol ; 87(4): 2128-2131, 2021 04.
Article in English | MEDLINE | ID: mdl-32986871

ABSTRACT

Eculizumab is known to cross the placenta to a limited degree, but recently therapeutic drug levels in cord blood were found in a single case. We report maternal, cord and placental levels of unbound eculizumab, C5 and C5-eculizumab in two pregnancies of a paroxysmal nocturnal haemoglobinuria patient who received 900 mg eculizumab every 2 weeks. In both pregnancies, cord blood concentrations of unbound eculizumab were below 4 µg/mL, while C5-eculizumab levels were 22 and 26 µg/mL, suggesting that a considerable fraction of C5 was blocked in the newborn. Concentrations in each placenta of unbound eculizumab were 41 ± 3 and 45 ± 4 µg/g tissue, of C5-eculizumab 19 ± 2 and 32 ± 3 µg/g, and of C5 20 ± 3 and 30 ± 2 µg/g (mean ± SD, in three tissue samples per placenta). Placental levels of unbound eculizumab were higher than those of C5-eculizumab complexes, while maternal concentrations were approximately equal, suggesting selective transport of unbound eculizumab across the placenta.


Subject(s)
Hemoglobinuria, Paroxysmal , Antibodies, Monoclonal, Humanized , Female , Hemoglobinuria, Paroxysmal/drug therapy , Humans , Infant, Newborn , Placenta , Pregnancy
7.
Arch Toxicol ; 95(2): 557-571, 2021 02.
Article in English | MEDLINE | ID: mdl-33083868

ABSTRACT

The application of anticancer drugs during pregnancy is associated with placenta-related adverse pregnancy outcomes. Therefore, it is important to study placental toxicity of anticancer drugs. The aim of this study was to compare effects on viability and steroidogenesis in placental tissue explants and trophoblast cell lines. Third trimester placental tissue explants were exposed for 72 h (culture day 4-7) to a concentration range of doxorubicin, paclitaxel, cisplatin, carboplatin, crizotinib, gefitinib, imatinib, or sunitinib. JEG-3, undifferentiated BeWo, and syncytialised BeWo cells were exposed for 48 h to the same drugs and concentrations. After exposure, tissue and cell viability were assessed and progesterone and estrone levels were quantified in culture medium. Apart from paclitaxel, all compounds affected both cell and tissue viability at clinically relevant concentrations. Paclitaxel affected explant viability moderately, while it reduced cell viability by 50% or more in all cell lines, at 3-10 nM. Doxorubicin (1 µM) reduced viability in explants to 83 ± 7% of control values, whereas it fully inhibited viability in all cell types. Interference with steroid release in explants was difficult to study due to large variability in measurements, but syncytialised BeWo cells proved suitable for this purpose. We found that 1 µM sunitinib reduced progesterone release to 76 ± 6% of control values, without affecting cell viability. While we observed differences between the models for paclitaxel and doxorubicin, most anticancer drugs affected viability significantly in both placental explants and trophoblast cell lines. Taken together, the placenta should be recognized as a potential target organ for toxicity of anticancer drugs.


Subject(s)
Antineoplastic Agents/toxicity , Estrone/analysis , Placenta/drug effects , Progesterone/analysis , Trophoblasts/drug effects , Cell Line, Tumor , Cell Survival , Cells, Cultured , Cytostatic Agents/toxicity , Female , Humans , Pregnancy , Pregnancy Trimester, Third/drug effects
8.
Biochim Biophys Acta Mol Basis Dis ; 1866(6): 165727, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32070771

ABSTRACT

Mitochondrial complex I (CI), the first multiprotein enzyme complex of the OXPHOS system, executes a major role in cellular ATP generation. Consequently, dysfunction of this complex has been linked to inherited metabolic disorders, including Leigh disease (LD), an often fatal disease in early life. Development of clinical effective treatments for LD remains challenging due to the complex pathophysiological nature. Treatment with the peroxisome proliferation-activated receptor (PPAR) agonist bezafibrate improved disease phenotype in several mitochondrial disease mouse models mediated via enhanced mitochondrial biogenesis and fatty acid ß-oxidation. However, the therapeutic potential of this mixed PPAR (α, δ/ß, γ) agonist is severely hampered by hepatotoxicity, which is possibly caused by activation of PPARγ. Here, we aimed to investigate the effects of the PPARα-specific fibrate clofibrate in mitochondrial CI-deficient (Ndufs4-/-) mice. Clofibrate increased lifespan and motor function of Ndufs4-/- mice, while only marginal hepatotoxic effects were observed. Due to the complex clinical and cellular phenotype of CI-deficiency, we also aimed to investigate the therapeutic potential of clofibrate combined with the redox modulator KH176. As described previously, single treatment with KH176 was beneficial, however, combining clofibrate with KH176 did not result in an additive effect on disease phenotype in Ndufs4-/- mice. Overall, both drugs have promising, but independent and nonadditive, properties for the pharmacological treatment of CI-deficiency-related mitochondrial diseases.


Subject(s)
Chromans/pharmacology , Clofibrate/pharmacology , Electron Transport Complex I/deficiency , Longevity/drug effects , Mitochondrial Diseases/drug therapy , Adenosine Triphosphate/metabolism , Animals , Bezafibrate/pharmacology , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Fatty Acids/metabolism , Humans , Leigh Disease/drug therapy , Leigh Disease/metabolism , Leigh Disease/pathology , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Motor Activity/drug effects , Oxidation-Reduction/drug effects , Peroxisome Proliferator-Activated Receptors/agonists , Peroxisome Proliferator-Activated Receptors/genetics
10.
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
11.
Toxicol In Vitro ; 29(7): 1868-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26216510

ABSTRACT

The uremic solutes p-cresyl sulfate (pCS) and p-cresyl glucuronide (pCG) accumulate in patients with chronic kidney disease (CKD), and might contribute to disease progression. Moreover, retention of these solutes may directly be related to renal tubular function. Here, we investigated the role of the efflux transporters Multidrug Resistance Protein 4 (MRP4) and Breast Cancer Resistance Protein (BCRP) in pCS and pCG excretion, and studied the impact of both solutes on the phenotype of human conditionally immortalized renal proximal tubule epithelial cells (ciPTEC). Our results show that p-cresol metabolites accumulate during CKD, with a shift from sulfation to glucuronidation upon progression. Moreover, pCS inhibited the activity of MRP4 by 40% and BCRP by 25%, whereas pCG only reduced MRP4 activity by 75%. Moreover, BCRP-mediated transport of both solutes was demonstrated. Exposure of ciPTEC to pCG caused epithelial-to-mesenchymal transition, indicated by increased expression of vimentin and Bcl-2, and diminished E-cadherin. This was associated with altered expression of key tubular transporters. In conclusion, BCRP is likely involved in the renal excretion of both solutes, and pCG promotes phenotypical changes in ciPTEC, supporting the notion that uremic toxins may be involved in CKD progression by negatively affecting renal tubule cell phenotype and functionality.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cresols/metabolism , Glucuronides/metabolism , Kidney Tubules, Proximal/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Sulfuric Acid Esters/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Adult , Aged , Baculoviridae/genetics , Cell Line , Female , HEK293 Cells , Humans , Male , Middle Aged , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/genetics , Renal Elimination , Renal Insufficiency, Chronic/metabolism , Transduction, Genetic
12.
J Hypertens ; 33(10): 2075-82, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26203967

ABSTRACT

BACKGROUND: Tyrosine kinase inhibitors targeting angiogenesis have become an important part of the treatment of patients with several types of cancer. One of the most reported side effects of vascular endothelial growth factor receptor (VEGFR)-targeted therapies is hypertension. In this study, we hypothesized that the development of hypertension in patients treated with sunitinib, a multitargeted tyrosine kinase inhibitor, is preceded by reduced endothelium-dependent vasodilation. Moreover, we hypothesized that this endothelial dysfunction is a result of impaired nitric oxide release. METHOD: In a placebo-controlled experiment, we determined vascular responses in isolated mesenteric arteries of rats (n = 26) after 7 days of sunitinib treatment. RESULTS: Sunitinib reduced endothelium-dependent vasodilation, but not endothelium-independent vasodilation. Moreover, we observed that the difference in endothelium-dependent vasodilation between controls and sunitinib-treated animals disappeared in the presence of N-nitro-L-arginine methyl ester (L-NAME), a nitric oxide antagonist. In patients with metastatic renal cell carcinoma, before and 1 week after start of sunitinib, the endothelium-dependent vasodilator response to intra-arterial acetycholine and the endothelium-independent vasodilator response to intra-arterial sodium nitroprusside was assessed with venous occlusion plethysmography. No changes in forearm blood flow ratios were observed. Mean arterial pressure did significantly increase from 101.9 ±â€Š3.8 to 106.1 ±â€Š2.6 mmHg after 1 week and further to 115.8 (±4.9) mmHg after 2 weeks of treatment. CONCLUSION: In animals, this study confirms that exposure to high concentrations of sunitinib reduces endothelium-dependent vasodilation by reducing endothelial release of nitric oxide. In humans, however, reduced endothelium-dependent vasodilation does not precede the development of hypertension in patients treated with sunitinib.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Renal Cell/drug therapy , Endothelium, Vascular/drug effects , Hypertension/physiopathology , Indoles/pharmacology , Kidney Neoplasms/drug therapy , Pyrroles/pharmacology , Vasodilation/drug effects , Acetylcholine/pharmacology , Adult , Aged , Angiogenesis Inhibitors/adverse effects , Animals , Endothelium, Vascular/physiopathology , Forearm/blood supply , Humans , Hypertension/chemically induced , Indoles/adverse effects , Male , Mesenteric Arteries/drug effects , Middle Aged , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Pyrroles/adverse effects , Rats , Regional Blood Flow/drug effects , Sunitinib , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vasodilator Agents/pharmacology
13.
Transl Res ; 164(1): 46-56, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24486136

ABSTRACT

Mycophenolic acid (MPA) is an immunosuppressant used in transplant rejection, often in combination with cyclosporine (CsA) and tacrolimus (Tac). The drug is cleared predominantly via the kidneys, and 95% of the administered dose appears in urine as 7-hydroxy mycophenolic acid glucuronide (MPAG). The current study was designed to unravel the renal excretory pathway of MPA and MPAG, and their potential drug-drug interactions. The role of multidrug resistance protein (MRP) 2 and MRP4 in MPA disposition was studied using human embryonic kidney 293 (HEK293) cells overexpressing the human transporters, and in isolated, perfused kidneys of Mrp2-deficient rats and Mrp4-deficient mice. Using these models, we identified MPA as substrate of MRP2 and MRP4, whereas its MPAG appeared to be a substrate of MRP2 only. CsA inhibited MPAG transport via MRP2 for 50% at 8 µM (P < 0.05), whereas Tac had no effect. This was confirmed by cell survival assays, showing a 10-fold increase in MPA cytotoxicity (50% reduction in cell survival changed from 12.2 ± 0.3 µM to 1.33 ± 0.01 µM by MPA + CsA; P < 0.001) and in perfused kidneys, showing a 50% reduction in MPAG excretion (P < 0.05). The latter effect was observed in Mrp2-deficient animals as well, supporting the importance of Mrp2 in MPAG excretion. CsA, but not Tac, inhibited MPA glucuronidation by rat kidney homogenate and human uridine 5'-diphospho-glucuronosyltransferase-glucuronosyltransferase 1A9 (P < 0.05 and P < 0.01, respectively). We conclude that MPA is a substrate of both MRP2 and MRP4, but MRP2 is the main transporter involved in renal MPAG excretion. In conclusion, CsA, but not Tac, influences MPA clearance by inhibiting renal MPA glucuronidation and MRP2-mediated MPAG secretion.


Subject(s)
Cyclosporine/pharmacokinetics , Kidney/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Mycophenolic Acid/pharmacokinetics , Tacrolimus/pharmacokinetics , Animals , Cyclosporine/metabolism , Drug Interactions , Gene Expression Regulation/drug effects , Glucuronides/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Mycophenolic Acid/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Tacrolimus/metabolism
14.
Drug Metab Dispos ; 40(6): 1076-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22415933

ABSTRACT

Coumarin (1,2-benzopyrone) is a natural compound that has been used as a fragrance in the food and perfume industry and could have therapeutic usefulness in the treatment of lymphedema and different types of cancer. Several previous pharmacokinetic studies of coumarin have been performed in humans, which revealed extensive first-pass metabolism of the compound. 7-Hydroxycoumarin (7-HC) and its glucuronide (7-HC-G) are the main metabolites formed in humans, and via this route, 80 to 90% of the absorbed coumarin is excreted into urine, mainly as 7-HC-G. Active transport processes play a role in the urinary excretion of 7-HC-G; however, until now, the transporters involved remained to be elucidated. In this study, we investigated whether the efflux transporters multidrug resistance-associated proteins (MRP)1-4, breast cancer resistance protein, or P-glycoprotein play a role in 7-HC and 7-HC-G transport. For this purpose, we measured uptake of the metabolites into membrane vesicles overexpressing these transporters. Our results showed that 7-HC is not transported by any of the efflux transporters tested, whereas 7-HC-G was a substrate of MRP3 and MRP4. These results are in line with the pharmacokinetic profile of coumarin and suggest that MRP3 and MRP4 are the main transporters involved in the excretion of the coumarin metabolite 7-HC-G from liver and kidney.


Subject(s)
Coumarins/metabolism , Multidrug Resistance-Associated Proteins/physiology , Umbelliferones/metabolism , Biological Transport, Active/physiology , HEK293 Cells , Humans
15.
Mol Pharm ; 9(5): 1351-60, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22428727

ABSTRACT

Although the CB1 receptor antagonist/inverse agonist rimonabant has positive effects on weight loss and cardiometabolic risk factors, neuropsychiatric side effects have prompted researchers to develop peripherally acting derivatives. Here, we investigated for a series of 3,4-diarylpyrazoline CB1 receptor antagonists if transport by the brain efflux transporter P-gp could be used as a selection criterion in the development of such drugs. All 3,4-diarylpyrazolines and rimonabant inhibited P-gp transport activity in membrane vesicles isolated from HEK293 cells overexpressing the transporter, but only the 1,1-dioxo-thiomorpholino analogue 23 exhibited a reduced accumulation (-38 ± 2%) in these cells, which could be completely reversed by the P-gp/BCRP inhibitor elacridar. In addition, 23 appeared to be a BCRP substrate, whereas rimonabant was not. In rats, the in vivo brain/plasma concentration ratio of 23 was significantly lower than for rimonabant (0.4 ± 0.1 vs 6.2 ± 1.6, p < 0.001). Coadministration of elacridar resulted in an 11-fold increase of the brain/plasma ratio for 23 (p < 0.01) and only 1.4-fold for rimonabant (p < 0.05), confirming the involvement of P-gp and possibly BCRP in limiting the brain entrance of 23 in vivo. In conclusion, these data support the conception that efflux via transporters such as P-gp and BCRP can limit the brain penetration of CB1 receptor antagonists, and that this property could be used in the development of peripheral antagonists.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cannabinoid Receptor Antagonists/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Acridines/pharmacology , Animals , Biological Transport/drug effects , Blood-Brain Barrier , Blotting, Western , Cannabinoid Receptor Antagonists/metabolism , Cell Line , Humans , Kinetics , Male , Neoplasm Proteins/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Quinidine/pharmacology , Rats , Rats, Wistar , Rimonabant , Tandem Mass Spectrometry , Tetrahydroisoquinolines/pharmacology
16.
Drug Metab Dispos ; 39(7): 1294-302, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21511945

ABSTRACT

Cannabinoid type 1 (CB1) receptor antagonists have been developed for the treatment of obesity, but a major disadvantage is that they cause unwanted psychiatric effects. Selective targeting of peripheral CB1 receptors might be an option to circumvent these side effects. Multidrug resistance-associated proteins (MRPs) can influence the pharmacokinetics of drugs and thereby affect their disposition in the body. In this study, we investigated the interaction of the prototypic CB1 receptor antagonist rimonabant and a series of 3,4-diarylpyrazoline CB1 receptor antagonists with MRP1, MRP2, MRP3, and MRP4 in vitro. Their effect on ATP-dependent transport of estradiol 17-ß-D-glucuronide (E(2)17ßG) was measured in inside-out membrane vesicles isolated from transporter-overexpressing human embryonic kidney 293 cells. Rimonabant inhibited MRP1 transport activity more potently than MRP4 (K(i) of 1.4 and 4 µM, respectively), whereas the 3,4-diarylpyrazolines were stronger inhibitors of MRP4- than MRP1-mediated transport. A number of CB1 receptor antagonists, including rimonabant, stimulated MRP2 and MRP3 transport activity at low substrate concentrations but inhibited E(2)17ßG transport at high substrate concentrations. The interaction of 3,4-diarylpyrazolines and rimonabant with MRP1-4 indicates their potential for drug-drug interactions. Preliminary in vivo data suggested that for some 3,4-diarylpyrazolines the relatively lower brain efficacy may be related to their inhibitory potency against MRP4 activity. Furthermore, this study shows that the modulatory effects of the 3,4-diarylpyrazolines were influenced by their chemical properties and that small variations in structure can determine the affinity of these compounds for efflux transporters and thereby affect their pharmacokinetic behavior.


Subject(s)
Multidrug Resistance-Associated Proteins/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Blotting, Western , Cell Line , Chromatography, Liquid , Humans , Protein Transport , Tandem Mass Spectrometry
17.
Hypertension ; 56(4): 722-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20679180

ABSTRACT

3-Hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors (statins) are effective in the primary and secondary prevention of cardiovascular events. Although originally developed to improve lipid profile, statins have demonstrated a surplus of beneficial pleiotropic effects, including improved endothelial function, reduced inflammation, and increased tolerance to ischemia-reperfusion injury. In preclinical studies, increased ecto-5'-nucleotidase activity, the key enzyme in extracellular adenosine formation, plays an important role in these effects. Because human data are absent, we explored the effects of rosuvastatin on ecto-5'-nucleotidase activity and the clinical relevance of increased extracellular adenosine during ischemia in humans in vivo. The forearm vasodilator responses to 3 increasing periods of forearm ischemia (2, 5, and 13 minutes) were determined during placebo and caffeine (an adenosine receptor antagonist) infusion into the brachial artery. At the end of an 8-day treatment period with rosuvastatin (20 mg per day), this whole procedure was repeated. During both experiments, ecto-5'-nucleotidase activity was determined. Vasodilator responses are expressed as the percentage increase in forearm blood flow ratio from baseline. Rosuvastatin increased ecto-5'-nucleotidase activity by 49±17% and enhanced the vasodilator response after 2, 5, and 13 minutes of ischemia in the absence (146±19, 330±26, and 987±133 to 312±77, 566±107, and 1533±267) but not in the presence of caffeine (98±25, 264±54, and 727±111 versus 95±19, 205±34, and 530±62). Rosuvastatin increases extracellular formation of adenosine in humans in vivo probably by enhancing ecto-5'-nucleotidase activity. This action results in the improvement of reactive hyperemia and may further enhance the clinical benefit of statins, in particular in conditions of ischemia.


Subject(s)
5'-Nucleotidase/metabolism , Fluorobenzenes/pharmacology , Ischemia/physiopathology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Vasodilator Agents/pharmacology , Adenosine/metabolism , Blood Flow Velocity/drug effects , Brachial Artery/drug effects , Brachial Artery/physiopathology , Caffeine/administration & dosage , Caffeine/pharmacology , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/pharmacology , Female , Fluorobenzenes/administration & dosage , Forearm/blood supply , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Infusions, Intra-Arterial , Male , Pyrimidines/administration & dosage , Rosuvastatin Calcium , Sulfonamides/administration & dosage , Time Factors , Up-Regulation/drug effects , Young Adult
18.
Am J Physiol Endocrinol Metab ; 297(3): E759-66, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19602584

ABSTRACT

In animal models, administration of the adenosine analog AICA-riboside has shown beneficial effects on ischemia-reperfusion injury and glucose homeostasis. The vascular and/or metabolic effects of AICA-riboside administration in humans remain to be established. AICA-riboside was infused intra-arterially in four different dosages up to 8 mg x min(-1) x dl(-1) in 24 healthy subjects. Forearm blood flow (FBF) and glucose uptake and plasma glucose, free fatty acid, and AICA-riboside concentrations were assessed. We also combined AICA-riboside infusion (2 mg x min(-1) x dl(-1)) with the intra-arterial administration of the adenosine receptor antagonist caffeine (90 microg x min(-1) x dl(-1); n = 6) and with the endothelial NO synthase inhibitor l-NMMA (0.4 mg x min(-1) x dl(-1); n = 6). Additional in vitro experiments were performed to explain our in vivo effects of AICA-riboside in humans. AICA-riboside increased FBF dose dependently from 2.0 +/- 0.2 to 13.2 +/- 1.9 ml x min(-1) x dl(-1) maximally (P < 0.05 for all dosages). The latter was not reduced by caffeine administration but was significantly attenuated by l-NMMA infusion. Despite high plasma AICA-riboside concentrations, forearm glucose uptake did not change. In vitro experiments showed rapid uptake of AICA-riboside by the equilibrative nucleoside transporter in erythrocytes and subsequent phosphorylation to AICA-ribotide. We conclude that AICA-riboside induces a potent vasodilator response in humans that is mediated by NO. Despite high local plasma concentrations, AICA-riboside does not increase skeletal muscle glucose uptake.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Muscle, Skeletal/blood supply , Nitric Oxide/pharmacology , Ribonucleosides/pharmacology , Vasodilation/drug effects , Adult , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/pharmacology , Brachial Artery/drug effects , Brachial Artery/physiology , Caffeine/administration & dosage , Cells, Cultured , Enzyme Inhibitors/administration & dosage , Female , Forearm/blood supply , Hemodynamics/drug effects , Humans , Injections, Intra-Arterial , Male , Muscle, Skeletal/drug effects , NG-Nitroarginine Methyl Ester/administration & dosage , Regional Blood Flow/drug effects , Ribonucleosides/administration & dosage , Young Adult
19.
Arterioscler Thromb Vasc Biol ; 29(6): 963-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19359665

ABSTRACT

OBJECTIVE: Statins may increase extracellular adenosine formation from adenosine monophosphate by enhancing ecto-5'-nucleotidase activity. This theory was tested in humans using dipyridamole-induced vasodilation as a read-out for local adenosine formation. Dipyridamole inhibits the transport of extracellular adenosine into the cytosol resulting in increased extracellular adenosine and subsequent vasodilation. In addition, we studied the effect of statin therapy in a forearm model of ischemia-reperfusion injury. METHODS AND RESULTS: Volunteers randomly received rosuvastatin or placebo in a double-blind parallel design (n=21). The forearm vasodilator response to intraarterial dipyridamole was determined in the absence and presence of the adenosine antagonist caffeine. During a separate visit the vasodilator response to nitroprusside and adenosine was established. In addition, healthy men were randomly divided in 3 groups to receive either placebo (n=10), rosuvastatin (n=22), or rosuvastatin combined with intravenous caffeine (n=12). Subsequently, volunteers performed forearm ischemic exercise. At reperfusion, Tc-99m-labeled annexin A5 was infused intravenously and scintigraphic images were acquired, providing an early marker of cell injury. Rosuvastatin treatment significantly increased the vasodilator response to dipyridamole, which was prevented by caffeine. Rosuvastatin did not influence the response to either sodium nitroprusside or adenosine indicating a specific interaction between rosuvastatin and dipyridamole, which does not result from an effect of rosuvastatin on adenosine clearance nor adenosine-receptor affinity or efficacy. Rosuvastatin increased tolerance to ischemia-reperfusion injury, which was attenuated by caffeine. CONCLUSIONS: Rosuvastatin increases extracellular adenosine formation, which provides protection against ischemia-reperfusion injury in humans in vivo. Therefore, statins and dipyridamole may interact synergistically.


Subject(s)
Adenosine/metabolism , Fluorobenzenes/therapeutic use , Forearm/blood supply , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Reperfusion Injury/prevention & control , Sulfonamides/therapeutic use , Vasodilation/drug effects , Adenosine/administration & dosage , Administration, Oral , Annexin A5/metabolism , Caffeine/administration & dosage , Dipyridamole/administration & dosage , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fluorobenzenes/administration & dosage , Fluorobenzenes/adverse effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Infusions, Intra-Arterial , Male , Nitroprusside/administration & dosage , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Radionuclide Imaging , Regional Blood Flow/drug effects , Reperfusion Injury/diagnostic imaging , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Rosuvastatin Calcium , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Time Factors , Up-Regulation , Vasodilator Agents/administration & dosage
20.
Clin Pharmacol Ther ; 78(1): 52-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16003293

ABSTRACT

BACKGROUND: Adenosine receptor stimulation induces several effects that could limit ischemia-reperfusion injury. We hypothesize that treatment with the nucleoside uptake inhibitor dipyridamole increases endogenous adenosine and limits ischemia-reperfusion injury in humans. METHODS: Ischemia-reperfusion injury was studied in forearm skeletal muscle by technetium Tc 99m-labeled annexin A5 scintigraphy. Ischemia-reperfusion injury was induced by unilateral forearm ischemic exercise. Immediately on reperfusion, annexin A5 labeled with technetium Tc 99m was administered intravenously, and ischemia-reperfusion injury was expressed as the percentage difference in radioactivity between the experimental arm and the control arm 1 and 4 hours after reperfusion. Targeting was quantified in the region of the thenar muscle and forearm flexor muscles. This approach was used in 9 healthy male volunteers after a 1-week treatment with dipyridamole (200 mg, slow release, twice daily) and in 23 control subjects. RESULTS: Dipyridamole treatment significantly reduced annexin A5 targeting in skeletal muscle compared with the control group (thenar region, 13% +/- 7% versus 22% +/- 15% at 1 hour after reperfusion and 9% +/- 6% versus 27% +/- 13% at 4 hours for dipyridamole and control groups, respectively [P = .01]; flexor region, 4% +/- 8% versus 7% +/- 6% at 1 hour after reperfusion and 1% +/- 4% versus 10% +/- 9% at 4 hours for dipyridamole and control groups, respectively [P = .01]). CONCLUSIONS: One week of oral treatment with the nucleoside uptake inhibitor dipyridamole (200 mg, slow release, twice daily) significantly limits ischemia-reperfusion injury in humans in vivo, as assessed by technetium Tc 99m-labeled annexin A5 scintigraphy of forearm skeletal muscle.


Subject(s)
Dipyridamole/administration & dosage , Dipyridamole/pharmacokinetics , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Administration, Oral , Adult , Annexin A5/administration & dosage , Blood Pressure/drug effects , Caffeine/blood , Drug Administration Schedule , Exercise , Exercise Tolerance , Forearm/blood supply , Forearm/diagnostic imaging , Heart Rate/drug effects , Humans , Ischemic Preconditioning/methods , Male , Muscle, Skeletal/blood supply , Muscle, Skeletal/diagnostic imaging , Radionuclide Imaging , Technetium/administration & dosage , Uridine/antagonists & inhibitors , Uridine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...