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










Publication year range
1.
J Med Chem ; 66(6): 4179-4196, 2023 03 23.
Article in English | MEDLINE | ID: mdl-36883854

ABSTRACT

The CXCR3 chemokine receptor is a G protein-coupled receptor mainly expressed on immune cells from the lymphoid lineage, including activated T cells. Binding of its inducible chemokine ligands CXCL9, CXCL10, and CXCL11 leads to downstream signaling events and the migration of activated T cells to sites of inflammation. Herein, we report the third part of our CXCR3 antagonist program in the field of autoimmunity, culminating in the discovery of the clinical compound ACT-777991 (8a). A previously disclosed advanced molecule was exclusively metabolized by the CYP2D6 enzyme, and options to address the issue are described. ACT-777991 is a highly potent, insurmountable, and selective CXCR3 antagonist that showed dose-dependent efficacy and target engagement in a mouse model of acute lung inflammation. The excellent properties and safety profile warranted progress in the clinics.


Subject(s)
Chemokine CXCL10 , Receptors, Chemokine , Animals , Mice , Chemokine CXCL10/metabolism , Chemokine CXCL9 , Receptors, Chemokine/metabolism , Ligands , Signal Transduction , Receptors, CXCR3/metabolism
2.
J Med Chem ; 65(17): 11533-11549, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35969159

ABSTRACT

The chemokine receptor CXCR3 allows the selective recruitment of innate and adaptive inflammatory immune cells into inflamed tissue. CXCR3 ligands are secreted after exposure to pro-inflammatory cytokines. Upon binding to CXCR3 ligands, CXCR3 expressing T-lymphocytes migrate toward sites of inflammation and can promote tissue damage. Therefore, antagonizing this receptor may provide clinical benefits for patients suffering from autoimmune diseases characterized by high concentrations of CXCR3 ligands. Herein, we report the second part of our CXCR3 discovery program where we explored the benzimidazolo-thiazole core scaffold. The optimization of potency and the mitigation of an hERG liability are described. Further pharmacokinetic considerations led to the identification of the potent CXCR3 antagonist ACT-672125 (29). The compound showed good physicochemical properties and safety profile. In a proof-of-mechanism model of lung inflammation, ACT-672125 inhibited the recruitment of CXCR3 expressing T cells into the inflamed lung in a dose-dependent manner.


Subject(s)
Autoimmune Diseases , Thiazoles , Autoimmune Diseases/drug therapy , Cytokines , Humans , Ligands , Receptors, CXCR3/metabolism , T-Lymphocytes/metabolism , Thiazoles/pharmacology , Thiazoles/therapeutic use
3.
J Med Chem ; 65(17): 11513-11532, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35947786

ABSTRACT

The chemokine receptor CXCR3 is a seven-transmembrane G-protein-coupled receptor (GPCR) involved in various pathologies, in particular autoimmune diseases. It is activated by the three chemokine ligands CXCL9, CXCL10, and CXCL11 and enables the recruitment of immune cell subsets leading to damage of inflamed tissues. Starting from a high-throughput screening hit, we describe the iterative optimization of a chemical series culminating in the discovery of the selective CXCR3 antagonist ACT-660602 (9j). The careful structural modifications during the lead optimization phase led to a compound with high biological potency in inhibiting cell migration together with improvements of the metabolic stability and hERG issue. In a LPS-induced lung inflammation model in mice, ACT-660602 led to significantly reduced recruitment of the CXCR3+ CD8+ T cell in the bronchoalveolar lavage compartment when administered orally at a dose of 30 mg/kg.


Subject(s)
Autoimmune Diseases , Chemokine CXCL10 , Animals , Autoimmune Diseases/drug therapy , CD8-Positive T-Lymphocytes/metabolism , Chemokine CXCL9/metabolism , Ligands , Mice , Receptors, CXCR3/metabolism
4.
Xenobiotica ; 48(7): 704-719, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28737453

ABSTRACT

1. The metabolism of selexipag has been studied in vivo in man and the main excreted metabolites were identified. Also, metabolites circulating in human plasma have been structurally identified and quantified. 2. The main metabolic pathway of selexipag in man is the formation of the active metabolite ACT-333679. Other metabolic pathways include oxidation and dealkylation reactions. All primary metabolites undergo subsequent hydrolysis of the sulphonamide moiety to their corresponding acids. ACT-333679 undergoes conjugation with glucuronic acid and aromatic hydroxylation to P10, the main metabolite detected in human faeces. 3. The formation of the active metabolite ACT-333679 is catalysed by carboxylesterases, while the oxidation and dealkylation reactions are metabolized by CYP2C8 and CYP3A4. CYP2C8 is the only P450 isoform catalysing the aromatic hydroxylation to P10. CYP2C8 together with CYP3A4 are also involved in the formation of several minor ACT-333679 metabolites. UGT1A3 and UGT2B7 catalyse the glucuronidation of ACT-333679. 4. The potential of selexipag to inhibit or induce cytochrome P450 enzymes or drug transport proteins was studied in vitro. Selexipag is an inhibitor of CYP2C8 and CYP2C9 and induces CYP3A4 and CYP2C9 in vitro. Also, selexipag inhibits the transporters OATP1B1, OATP1B3, OAT1, OAT3, and BCRP. However, due to its low dose and relatively low unbound exposure, selexipag has a low potential for causing drug-drug interactions.


Subject(s)
Acetamides/metabolism , Acetamides/pharmacology , Pyrazines/metabolism , Pyrazines/pharmacology , Receptors, Epoprostenol/agonists , Acetamides/blood , Acetamides/chemistry , Acetates/pharmacology , Drug Interactions , Enzyme Inhibitors/pharmacology , Esterases/antagonists & inhibitors , Esterases/metabolism , Hepatocytes/metabolism , Humans , Membrane Transport Proteins/metabolism , Metabolic Networks and Pathways/drug effects , Metabolome , Metabolomics , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , NADP/metabolism , Pyrazines/blood , Pyrazines/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Epoprostenol/metabolism , Recombinant Proteins/metabolism
5.
Br J Clin Pharmacol ; 80(4): 670-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25851691

ABSTRACT

AIMS: This study investigated the effect of a fixed dose combination of lopinavir/ritonavir on the pharmacokinetics (PK) of selexipag and its active metabolite ACT-333679. METHODS: This was an open label, randomized, single centre, two way, crossover study. Twenty healthy male subjects were treated with a single dose of 400 µg selexipag alone and in combination with multiple doses of lopinavir/ritonavir (400/100 mg) twice daily. RESULTS: The results showed that lopinavir/ritonavir approximately doubled the exposure to selexipag. The area under the plasma concentration-time curve from time zero to infinity (AUC(0,∞) and the maximum plasma concentration (Cmax) of selexipag were 2.2- and 2.1-fold higher, respectively, than under selexipag alone, with a 90% confidence interval (CI) of the geometric mean ratio (GMR) of 1.9, 2.7 and 1.7, 2.6, respectively. For ACT-333679, the clinically more relevant component of selexipag, systemic exposure was increased by 8% (GMR of AUC(0,∞) 1.1, 90% CI 0.9, 1.3), when lopinavir/ritonavir was co-administered with selexipag. The most frequently reported adverse event (AE) was headache. A single dose of selexipag, administered either alone or together with multiple doses of lopinavir/ritonavir, was safe and well tolerated. CONCLUSIONS: Lopinavir/ritonavir does not affect the PK parameters of selexipag and ACT-333679 to a clinically relevant extent. Therefore, adaptation of the selexipag dose is not required when co-administered with inhibitors of the organic anion-transporting polypeptide (OATP) 1B1/ 1B3, P-glycoprotein (P-gp) and/or CYP3A4.


Subject(s)
Acetamides/metabolism , Acetamides/pharmacokinetics , Acetates/pharmacokinetics , Lopinavir/pharmacology , Pyrazines/metabolism , Pyrazines/pharmacokinetics , Ritonavir/pharmacology , Acetamides/adverse effects , Acetamides/blood , Acetates/blood , Adult , Antihypertensive Agents/adverse effects , Antihypertensive Agents/blood , Antihypertensive Agents/pharmacokinetics , Cross-Over Studies , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Combinations , Drug Interactions , Healthy Volunteers , Humans , Male , Middle Aged , Pyrazines/adverse effects , Pyrazines/blood , Young Adult
6.
J Pharmacol Exp Ther ; 350(1): 130-43, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24769543

ABSTRACT

Treatment of pulmonary arterial hypertension with the endothelin receptor antagonist bosentan has been associated with transient increases in liver transaminases. Mechanistically, bosentan inhibits the bile salt export pump (BSEP) leading to an intrahepatic accumulation of cytotoxic bile salts, which eventually results in hepatocellular damage. BSEP inhibition by bosentan is amplified by its accumulation in the liver as bosentan is a substrate of organic anion-transporting polypeptide (OATP) transport proteins. The novel endothelin receptor antagonist macitentan shows a superior liver safety profile. Introduction of the less acidic sulfamide moiety and increased lipophilicity yield a hepatic disposition profile different from other endothelin receptor antagonists. Passive diffusion rather than OATP-mediated uptake is the driving force for macitentan uptake into the liver. Interaction with the sodium taurocholate cotransporting polypeptide and BSEP transport proteins involved in hepatic bile salt homeostasis is therefore limited due to the low intrahepatic drug concentrations. Evidence for this conclusion is provided by in vitro experiments in drug transporter-expressing cell lines, acute and long-term studies in rats and dogs, absence of plasma bile salt changes in healthy human volunteers after multiple dosing, and finally the liver safety profile of macitentan in the completed phase III morbidity/mortality SERAPHIN (Study with an Endothelin Receptor Antagonist in Pulmonary Arterial Hypertension to Improve Clinical Outcome) trial.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Bile Acids and Salts/blood , Liver/metabolism , Organic Anion Transporters, Sodium-Dependent/drug effects , Pyrimidines/pharmacokinetics , Pyrimidines/toxicity , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity , Symporters/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Animals , Bosentan , Cell Line , Cricetinae , Dogs , Dose-Response Relationship, Drug , Endothelin Receptor Antagonists , Hepatocytes , Humans , Male , Organic Anion Transporters/drug effects , Pyrimidines/adverse effects , Rats , Sulfonamides/adverse effects
7.
Biochemistry ; 52(19): 3297-309, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23600489

ABSTRACT

The ATP-binding cassette exporters Sav1866 from Staphylococcus aureus and P-glycoprotein are known to share a certain sequence similarity and disposition for cationic allocrites. Conversely, the two ATPases react very differently to neutral detergents that have previously been shown to be inhibitory allocrites for P-glycoprotein. To gain insight into the functional differences of the two proteins, we compared their basal and detergent-stimulated ATPase activity. P-Glycoprotein was investigated in NIH-MDR1-G185 plasma membrane vesicles and Sav1866 in lipid vesicles exhibiting a membrane packing density and a surface potential similar to those of the plasma membrane vesicles. Under basal conditions, Sav1866 revealed a lower catalytic efficiency and concomitantly a more pronounced sodium chloride and pH dependence than P-glycoprotein. As expected, the cationic allocrites (alkyltrimethylammonium chlorides) induced similar bell-shaped activity curves as a function of concentration for both exporters, suggesting stimulation upon binding of the first and inhibition upon binding of the second allocrite molecule. However, the neutral allocrites (n-alkyl-ß-d-maltosides and n-ethylene glycol monododecyl ethers) reduced P-glycoprotein's ATPase activity at concentrations well below their critical micelle concentration (CMC) but strongly enhanced Sav1866's ATPase activity even at concentrations above their CMC. The lack of ATPase inhibition at high concentrations of neutral of detergents could be explained by their comparatively low binding affinity for the transmembrane domains of Sav1866, which seems to prevent binding of a second inhibitory molecule. The high ATPase activity in the presence of hydrophobic, long chain detergents moreover revealed that Sav1866, despite its lower basal catalytic efficiency, is a more efficient floppase for lipidlike amphiphiles than P-glycoprotein.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Staphylococcus aureus/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Bacterial Proteins/genetics , Circular Dichroism , Detergents , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Lipids/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Salinity , Sequence Homology, Amino Acid , Staphylococcus aureus/genetics , Thermodynamics , Vanadates/pharmacology
8.
AAPS J ; 14(1): 68-78, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22189899

ABSTRACT

Macitentan is a dual endothelin receptor antagonist under phase 3 investigation in pulmonary arterial hypertension. We investigated the effect of cyclosporine (Cs) and rifampin on the pharmacokinetics of macitentan and its metabolites ACT-132577 and ACT-373898 in healthy male subjects. In addition, in vitro studies were performed to investigate interactions between macitentan and its active metabolite ACT-132577 with human organic anion-transporting polypeptides (OATPs). The clinical study (AC-055-111) was conducted as a two-part, one-sequence, crossover study. Ten subjects in each part received multiple-dose macitentan followed by multiple-dose co-administration of Cs (part A) or rifampin (part B). In the presence of Cs, steady-state area under the plasma concentration-time profiles during a dose interval (AUC(τ)) for macitentan and ACT-373898 increased 10% and 7%, respectively, and decreased 3% for ACT-132577. Steady-state AUC(τ) of macitentan and ACT-373898 in the presence of rifampin decreased 79% and 64%, respectively. For ACT-132577, no relevant difference in AUC(τ) between the two treatments was observed. Macitentan co-administered with Cs or rifampin was well tolerated. The complementary in vitro studies demonstrated no marked differences in uptake rates of macitentan and ACT-132577 between the wild-type and OATP over-expressing cells over the concentration range tested. Concomitant treatment with Cs did not have any clinically relevant effect on the exposure to macitentan or its metabolites, at steady-state. Concomitant treatment with rifampin reduced significantly the exposure to macitentan and its metabolite ACT-373898 at steady-state but did not affect the exposure to the active metabolite ACT-132577 to a clinically relevant extent.


Subject(s)
Cyclosporine/pharmacology , Pyrimidines/pharmacokinetics , Rifampin/pharmacology , Sulfonamides/pharmacokinetics , Adult , Animals , Area Under Curve , CHO Cells , Cricetinae , Cricetulus , Cross-Over Studies , Cyclosporine/administration & dosage , Drug Interactions , Endothelin A Receptor Antagonists , Endothelin B Receptor Antagonists , Humans , Male , Organic Anion Transporters/metabolism , Pyrimidines/administration & dosage , Rifampin/administration & dosage , Sulfonamides/administration & dosage , Young Adult
9.
Biochim Biophys Acta ; 1798(3): 515-25, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20004641

ABSTRACT

We compared the P-glycoprotein ATPase activity in inside-out plasma membrane vesicles and living NIH-MDR1-G185 cells with the aim to detect substrate transport. To this purpose we used six substrates which differ significantly in their passive influx through the plasma membrane. In cells, the cytosolic membrane leaflet harboring the substrate binding site of P-glycoprotein has to be approached by passive diffusion through the lipid membrane, whereas in inside-out plasma membrane vesicles, it is accessible directly from the aqueous phase. Compounds exhibiting fast passive influx compared to active efflux by P-glycoprotein induced similar ATPase activity profiles in cells and inside-out plasma membrane vesicles, because their concentrations in the cytosolic leaflets were similar. Compounds exhibiting similar influx as efflux induced in contrast different ATPase activity profiles in cells and inside-out vesicles. Their concentration was significantly lower in the cytosolic leaflet of cells than in the cytosolic leaflet of inside-out membrane vesicles, indicating that P-glycoprotein could cope with passive influx. P-glycoprotein thus transported all compounds at a rate proportional to ATP hydrolysis (i.e. all compounds were substrates). However, it prevented substrate entry into the cytosol only if passive influx of substrates across the lipid bilayer was in a similar range as active efflux.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphatases/metabolism , Transport Vesicles/enzymology , Acids , Air , Animals , Biological Transport/drug effects , Cell Survival/drug effects , Cyclosporine/pharmacology , Detergents/pharmacology , Extracellular Space/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Mice , Micelles , NIH 3T3 Cells , Permeability/drug effects , Substrate Specificity/drug effects , Time Factors , Transfection , Water
10.
Biochemistry ; 47(38): 10197-207, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18759452

ABSTRACT

P-glycoprotein (ABCB1) prevents absorption (e.g., blood-brain barrier) or enhances excretion (e.g., kidney) by moving substrates from the cytosolic to the extracellular membrane leaflet at the expense of ATP hydrolysis. It translocates various drugs and functions in membranes exhibiting different lateral packing densities. To gain more functional insight, we measured the temperature dependence of the P-glycoprotein ATPase activity in NIH-MDR1-G185 cell membranes in the absence and presence of three drugs (promazine, verapamil, and PSC833), exhibiting significantly different transporter affinities. Activation enthalpies (Delta H(++)) and entropies ( TDelta S(++)) were derived from Eyring plots. In the absence of drugs, the activation enthalpy and the free energy of activation for P-glycoprotein ATPase activity was determined as Delta H(++) = 92.6 +/- 4.2 kJ/mol and Delta G(++) = 73.1 +/- 7.2 kJ/mol, respectively. Increasing the drug concentration reduced the activation enthalpy, whereby the drug with the highest transporter affinity had the strongest effect (DeltaDelta H(++) = -21%). The free energy of activation decreased for activating (DeltaDelta G(++) = approximately -3.8%) and increased for inhibitory compounds (DeltaDelta G(++) = approximately +0.7%). The drug-specific changes of the free energy of activation are thus barely above thermal energy. A comparison with literature data revealed that a decrease of the lateral membrane packing density reduces the enthalpic and the entropic contribution to the free energy of activation. Although the P-glycoprotein ATPase activity increases only slightly with decreasing lateral membrane packing density, the mode of action changes from strongly entropy-driven at high, to essentially enthalpy-driven at low packing densities. This suggests that the transporter and the membrane form a functional entity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Membrane/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Animals , Catalysis , Cell Membrane/chemistry , Drug Delivery Systems , Enzyme Activation/physiology , Mice , NIH 3T3 Cells , Substrate Specificity/physiology , Thermodynamics , Transport Vesicles/chemistry , Transport Vesicles/metabolism
11.
Biochemistry ; 46(11): 3394-404, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17302433

ABSTRACT

P-glycoprotein (MDR1, ABCB1) is an ATP-dependent efflux transporter of a large variety of compounds. To understand P-glycoprotein in more detail, it is important to elucidate its activity in the cellular ensemble as well as in plasma membrane vesicles (under conditions where other ATP dependent proteins are blocked). We measured P-glycoprotein activity in inside-out vesicles formed from plasma membranes of MDR1-transfected mouse embryo fibroblasts (NIH-MDR1-G185) for comparison with previous measurements of P-glycoprotein activity in living NIH-MDR1-G185 cells. In plasma membrane vesicles activity was measured by monitoring phosphate release upon ATP hydrolysis and in living cells by monitoring the extracellular acidification rate upon ATP synthesis via glycolysis. P-glycoprotein was stimulated as a function of the concentration with 19 structurally different drugs, including local anesthetics, cyclic peptides, and cytotoxic drugs. The concentrations of half-maximum P-glycoprotein activation, K1, were identical in inside-out plasma membrane vesicles and in living cells and covered a broad range of concentrations (K1 approximately (10(-8)-10(-3)) M). The influence of the pH, drug association, and vesicle aggregation on the concentration of half-maximum P-glycoprotein activation was investigated. The turnover numbers in plasma membrane vesicles and in living cells were also approximately identical if the latter were measured in the presence of pyruvate. However, in the absence of pyruvate they were higher in living cells. The rate of ATP hydrolysis/ATP synthesis decreased exponentially with decreasing free energy of drug binding from water to the transporter, DeltaG0(tw)(1) (or increasing binding affinity). This suggests that drug release from the transmembrane domains has to occur before ATP is hydrolyzed for resetting the transporter.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Membrane/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Transport/drug effects , Cell Membrane/drug effects , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Hydrogen-Ion Concentration , Kinetics , Mice , NIH 3T3 Cells , Vanadates/pharmacology
12.
Biochemistry ; 45(9): 3020-32, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16503657

ABSTRACT

It is generally accepted that P-glycoprotein binds its substrates in the lipid phase of the membrane. Quantification and characterization of the lipid-transporter binding step are, however, still a matter of debate. We therefore selected 15 structurally diverse drugs and measured the binding constants from water to the activating (inhibitory) binding region of P-glycoprotein, K(tw(1)) (K(tw(2))), as well as the lipid-water partition coefficients, K(lw). The former were obtained by measuring the concentrations of half-maximum activation (inhibition), K(1) (K(2)), in living NIH-MDR-G185 mouse embryo fibroblasts using a Cytosensor microphysiometer, and the latter were derived from surface activity measurements. This allowed determination of the membrane concentration of drugs at half-maximum P-glycoprotein activation (C(b(1)) = (0.02 to 67) mmol/L lipid), which is much higher than the corresponding aqueous concentration (K(1) = (0.02 to 376) microM). Moreover we determined the free energy of drug binding from water to the activating binding region of the transporter (DeltaG degrees (tw(1)) = (-30 to -54) kJ/mol), the free energy of drug partitioning into the lipid membrane (DeltaG degrees (lw) = (-23 to -34) kJ/mol), and, as the difference of the two, the free energy of drug binding from the lipid membrane to the activating binding region of the transporter (DeltaG degrees (tl(1)) = (-7 to -27) kJ/mol). For the compounds tested DeltaG degrees (tl(1)) was less negative than DeltaG degrees (lw) but varied more strongly. The free energies of substrate binding to the transporter within the lipid phase, DeltaG degrees (tl(1)), are consistent with a modular binding concept, where the energetically most efficient binding module comprises two hydrogen bond acceptor groups.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Membrane Lipids/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Genes, MDR , Humans , Hydrogen Bonding/drug effects , Immunosuppressive Agents/metabolism , Kinetics , Mice , NIH 3T3 Cells , Protein Binding/drug effects , Structure-Activity Relationship , Thermodynamics , Time Factors , Transfection , Water/chemistry , Water/metabolism
13.
Biochemistry ; 43(46): 14840-51, 2004 Nov 23.
Article in English | MEDLINE | ID: mdl-15544355

ABSTRACT

P-glycoprotein ATPase activity has been studied almost exclusively by measuring inorganic phosphate release from inside-out cellular vesicles. We have recently proposed a new method based on measurements of the extracellular acidification rate (ECAR) of living cells with a Cytosensor microphysiometer. This method allows for systematic investigation of the various factors influencing P-glycoprotein activation in living cells. Basal metabolic rates or ECARs of different MDR1-transfected cell lines were compared with those of the Mdr1a(-/-)1b(-/-) knockout, MRP1-transfected, and corresponding wild-type cell lines. Basal ECARs of all cells were on the order of 10(7) protons/cell/s, whereby those of genetically modified cells were on average (over all cell lines) slightly lower than those of wild-type cells. The expression level of P-glycoprotein in MDR1-transfected cells had no influence on basal ECARs. Verapamil-induced ECARs were specific for MDR1-transfected cells and increased with the expression level of P-glycoprotein. Moreover, ECARs were dependent on the metabolic state of the cell and were (2.8 +/- 1.2) x 10(6) and (8.0 +/- 1.5) x 10(6) protons/cell/s in glucose-deficient and glucose-fed NIH-MDR-G185 cells, respectively, after verapamil (10 muM) stimulation. The ECARs were practically identical to the rates of lactate extrusion and thus reflect the rates of ATP synthesis via glycolysis. Taking into account the number of P-glycoprotein molecules per cell, the rate of ATP hydrolysis in inside-out vesicles of the same cells was determined as (9.2 +/- 1.5) x 10(6) phosphates/cell/s, in good agreement with the rate of ATP synthesized in glucose-fed cells. The energy required for P-glycoprotein activation relative to the basal metabolic energy was twice as large in glucose-deficient as in glucose-fed cells, suggesting cellular protection by P-glycoprotein even under conditions of starvation.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Extracellular Space/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adenosine Triphosphatases/metabolism , Animals , Biosensing Techniques , Cell Culture Techniques , Cell Membrane/enzymology , Cell Membrane/metabolism , Culture Media, Conditioned/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Extracellular Space/drug effects , Extracellular Space/enzymology , Extracellular Space/genetics , Humans , Hydrogen-Ion Concentration , LLC-PK1 Cells , Lactic Acid/metabolism , Mice , Mice, Knockout , NIH 3T3 Cells , Swine , Transfection , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...