1
|
Ågren R, Zeberg H, Stępniewski TM, Free RB, Reilly SW, Luedtke RR, Århem P, Ciruela F, Sibley DR, Mach RH, Selent J, Nilsson J, Sahlholm K. Ligand with Two Modes of Interaction with the Dopamine D 2 Receptor-An Induced-Fit Mechanism of Insurmountable Antagonism. ACS Chem Neurosci 2020; 11:3130-3143. [PMID: 32865974 PMCID: PMC7553383 DOI: 10.1021/acschemneuro.0c00477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
![]()
A solid
understanding of the mechanisms governing ligand binding
is crucial for rational design of therapeutics targeting the dopamine
D2 receptor (D2R). Here, we use G protein-coupled
inward rectifier potassium (GIRK) channel activation in Xenopus oocytes to measure the kinetics of D2R antagonism by
a series of aripiprazole analogues, as well as the recovery of dopamine
(DA) responsivity upon washout. The aripiprazole analogues comprise
an orthosteric and a secondary pharmacophore and differ by the length
of the saturated carbon linker joining these two pharmacophores. Two
compounds containing 3- and 5-carbon linkers allowed for a similar
extent of recovery from antagonism in the presence of 1 or 100 μM
DA (>25 and >90% of control, respectively), whereas recovery
was less
prominent (∼20%) upon washout of the 4-carbon linker compound,
SV-III-130, both with 1 and 100 μM DA. Prolonging the coincubation
time with SV-III-130 further diminished recovery. Curve-shift experiments
were consistent with competition between SV-III-130 and DA. Two mutations
in the secondary binding pocket (V91A and E95A) of D2R
decreased antagonistic potency and increased recovery from SV-III-130
antagonism, whereas a third mutation (L94A) only increased recovery.
Our results suggest that the secondary binding pocket influences recovery
from inhibition by the studied aripiprazole analogues. We propose
a mechanism, supported by in silico modeling, whereby
SV-III-130 initially binds reversibly to the D2R, after
which the drug-receptor complex undergoes a slow transition to a second
ligand-bound state, which is dependent on secondary binding pocket
integrity and irreversible during the time frame of our experiments.
Collapse
Affiliation(s)
- Richard Ågren
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Hugo Zeberg
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Tomasz Maciej Stępniewski
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences of Pompeu Fabra University (UPF)-Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- InterAx Biotech AG, PARK innovAARE, 5234 Villigen, Switzerland
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw 02-089, Poland
| | - R. Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland 20892-3723, United States
| | - Sean W. Reilly
- Department of Radiology, Division of Nuclear Medicine and Clinical Molecular Imaging, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Robert R. Luedtke
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107, United States
| | - Peter Århem
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat 08907, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat 08907, Spain
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland 20892-3723, United States
| | - Robert H. Mach
- Department of Radiology, Division of Nuclear Medicine and Clinical Molecular Imaging, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences of Pompeu Fabra University (UPF)-Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Johanna Nilsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
- Department of Integrative Medical Biology, Umeå University, Umeå 901 87, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| |
Collapse
|
2
|
Guo D, Peletier LA, Bridge L, Keur W, de Vries H, Zweemer A, Heitman LH, IJzerman AP. A two-state model for the kinetics of competitive radioligand binding. Br J Pharmacol 2018; 175:1719-1730. [PMID: 29486053 PMCID: PMC5913406 DOI: 10.1111/bph.14184] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 12/11/2017] [Accepted: 02/19/2018] [Indexed: 01/09/2023] Open
Abstract
Background and Purpose Ligand–receptor binding kinetics is receiving increasing attention in the drug research community. The Motulsky and Mahan model, a one‐state model, offers a method for measuring the binding kinetics of an unlabelled ligand, with the assumption that the labelled ligand has no preference while binding to distinct states or conformations of a drug target. As such, the one‐state model is not applicable if the radioligand displays biphasic binding kinetics to the receptor. Experimental Approach We extended the Motulsky and Mahan model to a two‐state model, in which the kinetics of the unlabelled competitor binding to different receptor states (R1 and R2) can be measured. With this extended model, we determined the binding kinetics of unlabelled N‐5′‐ethylcarboxamidoadenosine (NECA), a representative agonist for the adenosine A1 receptor. Subsequently, an application of the model was exemplified by measuring the binding kinetics of other A1 receptor ligands. In addition, limitations of the model were investigated as well. Key Results The kinetic rate constants of unlabelled NECA were comparable with the results of kinetic radioligand binding assays in which [3H]‐NECA was used. The model was further validated by good correlation between simulated results and the experimental data. Conclusion The two‐state model is sufficient to analyse the binding kinetics of an unlabelled ligand, when a radioligand shows biphasic association characteristics. We expect this two‐state model to have general applicability for other targets as well.
Collapse
Affiliation(s)
- Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | | | - Lloyd Bridge
- Department of Mathematics, Swansea University, Swansea, UK.,Department of Engineering Design and Mathematics, University of the West of England, Bristol, UK
| | - Wesley Keur
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Henk de Vries
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Annelien Zweemer
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| |
Collapse
|
3
|
Hoare SR. Receptor binding kinetics equations: Derivation using the Laplace transform method. J Pharmacol Toxicol Methods 2018; 89:26-38. [DOI: 10.1016/j.vascn.2017.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/02/2017] [Accepted: 08/08/2017] [Indexed: 01/29/2023]
|
4
|
Vlot AHC, de Witte WEA, Danhof M, van der Graaf PH, van Westen GJP, de Lange ECM. Target and Tissue Selectivity Prediction by Integrated Mechanistic Pharmacokinetic-Target Binding and Quantitative Structure Activity Modeling. AAPS JOURNAL 2017; 20:11. [PMID: 29204742 DOI: 10.1208/s12248-017-0172-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/20/2017] [Indexed: 12/31/2022]
Abstract
Selectivity is an important attribute of effective and safe drugs, and prediction of in vivo target and tissue selectivity would likely improve drug development success rates. However, a lack of understanding of the underlying (pharmacological) mechanisms and availability of directly applicable predictive methods complicates the prediction of selectivity. We explore the value of combining physiologically based pharmacokinetic (PBPK) modeling with quantitative structure-activity relationship (QSAR) modeling to predict the influence of the target dissociation constant (K D) and the target dissociation rate constant on target and tissue selectivity. The K D values of CB1 ligands in the ChEMBL database are predicted by QSAR random forest (RF) modeling for the CB1 receptor and known off-targets (TRPV1, mGlu5, 5-HT1a). Of these CB1 ligands, rimonabant, CP-55940, and Δ8-tetrahydrocanabinol, one of the active ingredients of cannabis, were selected for simulations of target occupancy for CB1, TRPV1, mGlu5, and 5-HT1a in three brain regions, to illustrate the principles of the combined PBPK-QSAR modeling. Our combined PBPK and target binding modeling demonstrated that the optimal values of the K D and k off for target and tissue selectivity were dependent on target concentration and tissue distribution kinetics. Interestingly, if the target concentration is high and the perfusion of the target site is low, the optimal K D value is often not the lowest K D value, suggesting that optimization towards high drug-target affinity can decrease the benefit-risk ratio. The presented integrative structure-pharmacokinetic-pharmacodynamic modeling provides an improved understanding of tissue and target selectivity.
Collapse
Affiliation(s)
- Anna H C Vlot
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands
| | - Wilhelmus E A de Witte
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands
| | - Meindert Danhof
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands
| | - Piet H van der Graaf
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands.,Certara Quantitative Systems Pharmacology, Canterbury Innovation Centre, Canterbury, CT2 7FG, UK
| | - Gerard J P van Westen
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands.
| |
Collapse
|
5
|
Xia L, de Vries H, Lenselink EB, Louvel J, Waring MJ, Cheng L, Pahlén S, Petersson MJ, Schell P, Olsson RI, Heitman LH, Sheppard RJ, IJzerman AP. Structure-Affinity Relationships and Structure-Kinetic Relationships of 1,2-Diarylimidazol-4-carboxamide Derivatives as Human Cannabinoid 1 Receptor Antagonists. J Med Chem 2017; 60:9545-9564. [PMID: 29111736 PMCID: PMC5734604 DOI: 10.1021/acs.jmedchem.7b00861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
![]()
We
report on the synthesis and biological evaluation of a series of 1,2-diarylimidazol-4-carboxamide
derivatives developed as CB1 receptor antagonists. These
were evaluated in a radioligand displacement binding assay, a [35S]GTPγS binding assay, and in a competition association
assay that enables the relatively fast kinetic screening of multiple
compounds. The compounds show high affinities and a diverse range
of kinetic profiles at the CB1 receptor and their structure–kinetic
relationships (SKRs) were established. Using the recently resolved
hCB1 receptor crystal structures, we also performed a modeling
study that sheds light on the crucial interactions for both the affinity
and dissociation kinetics of this family of ligands. We provide evidence
that, next to affinity, additional knowledge of binding kinetics is
useful for selecting new hCB1 receptor antagonists in the
early phases of drug discovery.
Collapse
Affiliation(s)
- Lizi Xia
- Division of Medicinal Chemistry, LACDR, Leiden University , 2300RA Leiden, The Netherlands
| | - Henk de Vries
- Division of Medicinal Chemistry, LACDR, Leiden University , 2300RA Leiden, The Netherlands
| | - Eelke B Lenselink
- Division of Medicinal Chemistry, LACDR, Leiden University , 2300RA Leiden, The Netherlands
| | - Julien Louvel
- Division of Medicinal Chemistry, LACDR, Leiden University , 2300RA Leiden, The Netherlands
| | | | | | - Sara Pahlén
- Medicinal Chemistry, Cardiovascular and Metabolic Diseases, IMED Biotech Unit, AstraZeneca , Gothenburg SE-431 83, Sweden
| | - Maria J Petersson
- Medicinal Chemistry, Cardiovascular and Metabolic Diseases, IMED Biotech Unit, AstraZeneca , Gothenburg SE-431 83, Sweden
| | | | | | - Laura H Heitman
- Division of Medicinal Chemistry, LACDR, Leiden University , 2300RA Leiden, The Netherlands
| | - Robert J Sheppard
- Medicinal Chemistry, Oncology, IMED Biotech Unit, AstraZeneca , Cambridge SK10 2NA, United Kingdom
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, LACDR, Leiden University , 2300RA Leiden, The Netherlands
| |
Collapse
|
6
|
Wong YC, Ilkova T, van Wijk RC, Hartman R, de Lange ECM. Development of a population pharmacokinetic model to predict brain distribution and dopamine D2 receptor occupancy of raclopride in non-anesthetized rat. Eur J Pharm Sci 2017; 111:514-525. [PMID: 29106979 DOI: 10.1016/j.ejps.2017.10.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/13/2017] [Accepted: 10/22/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Raclopride is a selective antagonist of the dopamine D2 receptor. It is one of the most frequently used in vivo D2 tracers (at low doses) for assessing drug-induced receptor occupancy (RO) in animals and humans. It is also commonly used as a pharmacological blocker (at high doses) to occupy the available D2 receptors and antagonize the action of dopamine or drugs on D2 in preclinical studies. The aims of this study were to comprehensively evaluate its pharmacokinetic (PK) profiles in different brain compartments and to establish a PK-RO model that could predict the brain distribution and RO of raclopride in the freely moving rat using a LC-MS based approach. METHODS Rats (n=24) received a 10-min IV infusion of non-radiolabeled raclopride (1.61μmol/kg, i.e. 0.56mg/kg). Plasma and the brain tissues of striatum (with high density of D2 receptors) and cerebellum (with negligible amount of D2 receptors) were collected. Additional microdialysis experiments were performed in some rats (n=7) to measure the free drug concentration in the extracellular fluid of the striatum and cerebellum. Raclopride concentrations in all samples were analyzed by LC-MS. A population PK-RO model was constructed in NONMEM to describe the concentration-time profiles in the unbound plasma, brain extracellular fluid and brain tissue compartments and to estimate the RO based on raclopride-D2 receptor binding kinetics. RESULTS In plasma raclopride showed a rapid distribution phase followed by a slower elimination phase. The striatum tissue concentrations were consistently higher than that of cerebellum tissue throughout the whole experimental period (10-h) due to higher non-specific tissue binding and D2 receptor binding in the striatum. Model-based simulations accurately predicted the literature data on rat plasma PK, brain tissue PK and D2 RO at different time points after intravenous or subcutaneous administration of raclopride at tracer dose (RO <10%), sub-pharmacological dose (RO 10%-30%) and pharmacological dose (RO >30%). CONCLUSION For the first time a predictive model that could describe the quantitative in vivo relationship between dose, PK and D2 RO of raclopride in non-anesthetized rat was established. The PK-RO model could facilitate the selection of optimal dose and dosing time when raclopride is used as tracer or as pharmacological blocker in various rat studies. The LC-MS based approach, which doses and quantifies a non-radiolabeled tracer, could be useful in evaluating the systemic disposition and brain kinetics of tracers.
Collapse
Affiliation(s)
- Yin Cheong Wong
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Trayana Ilkova
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Rob C van Wijk
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Robin Hartman
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
7
|
Zeilinger M, Pichler F, Nics L, Wadsak W, Spreitzer H, Hacker M, Mitterhauser M. New approaches for the reliable in vitro assessment of binding affinity based on high-resolution real-time data acquisition of radioligand-receptor binding kinetics. EJNMMI Res 2017; 7:22. [PMID: 28271461 PMCID: PMC5340791 DOI: 10.1186/s13550-016-0249-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/15/2016] [Indexed: 12/02/2022] Open
Abstract
Background Resolving the kinetic mechanisms of biomolecular interactions have become increasingly important in early-phase drug development. Since traditional in vitro methods belong to dose-dependent assessments, binding kinetics is usually overlooked. The present study aimed at the establishment of two novel experimental approaches for the assessment of binding affinity of both, radiolabelled and non-labelled compounds targeting the A3R, based on high-resolution real-time data acquisition of radioligand-receptor binding kinetics. A novel time-resolved competition assay was developed and applied to determine the Ki of eight different A3R antagonists, using CHO-K1 cells stably expressing the hA3R. In addition, a new kinetic real-time cell-binding approach was established to quantify the rate constants kon and koff, as well as the dedicated Kd of the A3R agonist [125I]-AB-MECA. Furthermore, lipophilicity measurements were conducted to control influences due to physicochemical properties of the used compounds. Results Two novel real-time cell-binding approaches were successfully developed and established. Both experimental procedures were found to visualize the kinetic binding characteristics with high spatial and temporal resolution, resulting in reliable affinity values, which are in good agreement with values previously reported with traditional methods. Taking into account the lipophilicity of the A3R antagonists, no influences on the experimental performance and the resulting affinity were investigated. Conclusions Both kinetic binding approaches comprise tracer administration and subsequent binding to living cells, expressing the dedicated target protein. Therefore, the experiments resemble better the true in vivo physiological conditions and provide important markers of cellular feedback and biological response.
Collapse
Affiliation(s)
- Markus Zeilinger
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Radiopharmacy and Experimental Nuclear Medicine, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Faculty of Engineering, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Florian Pichler
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Radiopharmacy and Experimental Nuclear Medicine, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Faculty of Engineering, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Lukas Nics
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Radiopharmacy and Experimental Nuclear Medicine, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Radiopharmacy and Experimental Nuclear Medicine, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Department of Inorganic Chemistry, University of Vienna, Vienna, Austria
| | - Helmut Spreitzer
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Radiopharmacy and Experimental Nuclear Medicine, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Radiopharmacy and Experimental Nuclear Medicine, Waehringer Guertel 18-20, 1090, Vienna, Austria. .,Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Vienna, Austria. .,Ludwig Boltzmann Institute for Applied Diagnostics, Vienna, Austria.
| |
Collapse
|
8
|
A new, simple and robust radioligand binding method used to determine kinetic off-rate constants for unlabeled ligands. Application at α 2A - and α 2C -adrenoceptors. Eur J Pharmacol 2016; 788:113-121. [DOI: 10.1016/j.ejphar.2016.06.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 10/21/2022]
|
9
|
Vauquelin G. Cell membranes… and how long drugs may exert beneficial pharmacological activity in vivo. Br J Clin Pharmacol 2016; 82:673-82. [PMID: 27135195 PMCID: PMC5338106 DOI: 10.1111/bcp.12996] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022] Open
Abstract
The time course of the beneficial pharmacological effect of a drug has long been considered to depend merely on the temporal fluctuation of its free concentration. Only in the last decade has it become widely accepted that target-binding kinetics can also affect in vivo pharmacological activity. Although current reviews still essentially focus on genuine dissociation rates, evidence is accumulating that additional micro-pharmacokinetic (PK) and -pharmacodynamic (PD) mechanisms, in which the cell membrane plays a central role, may also increase the residence time of a drug on its target. The present review provides a compilation of otherwise widely dispersed information on this topic. The cell membrane can intervene in drug binding via the following three major mechanisms: (i) by acting as a sink/repository for the drug; (ii) by modulating the conformation of the drug and even by participating in the binding process; and (iii) by facilitating the approach (and rebinding) of the drug to the target. To highlight these mechanisms, we focus on drugs that are currently used in clinical therapy, such as the antihypertensive angiotensin II type 1 receptor antagonist candesartan, the atypical antipsychotic agent clozapine and the bronchodilator salmeterol. Although the role of cell membranes in PK-PD modelling is gaining increasing interest, many issues remain unresolved. It is likely that novel biophysical and computational approaches will provide improved insights in the near future.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department Molecular and Biochemical PharmacologyVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
10
|
Shimizu Y, Ogawa K, Nakayama M. Characterization of Kinetic Binding Properties of Unlabeled Ligands via a Preincubation Endpoint Binding Approach. ACTA ACUST UNITED AC 2016; 21:729-37. [PMID: 27270099 DOI: 10.1177/1087057116652065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/06/2016] [Indexed: 11/16/2022]
Abstract
The dissociation rates of unlabeled drugs have been well studied by kinetic binding analyses. Since kinetic assays are laborious, we developed a simple method to determine the kinetic binding parameters of unlabeled competitors by a preincubation endpoint assay. The probe binding after preincubation of a competitor can be described by a single equation as a function of time. Simulations using the equation revealed the degree of IC50 change induced by preincubation of a competitor depended on the dissociation rate koff of the competitor but not on the association rate kon To validate the model, an in vitro binding assay was performed using a smoothened receptor (SMO) and [(3)H]TAK-441, a SMO antagonist. The equilibrium dissociation constants (KI) and koff of SMO antagonists determined by globally fitting the model to the concentration-response curves obtained with and without 24 h preincubation correlated well with those determined by other methods. This approach could be useful for early-stage optimization of drug candidates by enabling determination of binding kinetics in a high-throughput manner because it does not require kinetic measurements, an intermediate washout step during the reaction, or prior determination of competitors' KI values.
Collapse
Affiliation(s)
- Yuji Shimizu
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Kazumasa Ogawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Masaharu Nakayama
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
11
|
Sahlholm K, Zeberg H, Nilsson J, Ögren SO, Fuxe K, Århem P. The fast-off hypothesis revisited: A functional kinetic study of antipsychotic antagonism of the dopamine D2 receptor. Eur Neuropsychopharmacol 2016; 26:467-76. [PMID: 26811292 DOI: 10.1016/j.euroneuro.2016.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 12/02/2015] [Accepted: 01/11/2016] [Indexed: 11/28/2022]
Abstract
Newer, "atypical" antipsychotics carry a lower risk of motor side-effects than older, "typical" compounds. It has been proposed that a ~100-fold faster dissociation from the dopamine D2 receptor (D2R) distinguishes atypical from typical antipsychotics. Furthermore, differing antipsychotic D2R affinities have been suggested to reflect differences in dissociation rate constants (koff), while association rate constants (kon) were assumed to be similar. However, it was recently demonstrated that lipophilic accumulation of ligand in the cell interior and/or membrane can cause underestimation of koff, and as high-affinity D2R antagonists are frequently lipophilic, this may have been a confounding factor in previous studies. In the present work, a functional electrophysiology assay was used to measure the recovery of dopamine-mediated D2R responsivity from antipsychotic antagonism, using elevated concentrations of dopamine to prevent the potential bias of re-binding of lipophilic ligands. The variability of antipsychotic kon was also reexamined, capitalizing on the temporal resolution of the assay. kon was estimated from the experimental recordings using a simple mathematical model assumed to describe the binding process. The time course of recovery from haloperidol (typical antipsychotic) was only 6.4- to 2.5-fold slower than that of the atypical antipsychotics, amisulpride, clozapine, and quetiapine, while antipsychotic kons were found to vary more widely than previously suggested. Finally, affinities calculated using our kon and koff estimates correlated well with functional potency and with affinities reported from radioligand binding studies. In light of these findings, it appears unlikely that typical and atypical antipsychotics are primarily distinguished by their D2R binding kinetics.
Collapse
Affiliation(s)
- Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden.
| | - Hugo Zeberg
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Johanna Nilsson
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Peter Århem
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| |
Collapse
|
12
|
de Witte WEA, Wong YC, Nederpelt I, Heitman LH, Danhof M, van der Graaf PH, Gilissen RAHJ, de Lange ECM. Mechanistic models enable the rational use of in vitro drug-target binding kinetics for better drug effects in patients. Expert Opin Drug Discov 2015; 11:45-63. [PMID: 26484747 DOI: 10.1517/17460441.2016.1100163] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Drug-target binding kinetics are major determinants of the time course of drug action for several drugs, as clearly described for the irreversible binders omeprazole and aspirin. This supports the increasing interest to incorporate newly developed high-throughput assays for drug-target binding kinetics in drug discovery. A meaningful application of in vitro drug-target binding kinetics in drug discovery requires insight into the relation between in vivo drug effect and in vitro measured drug-target binding kinetics. AREAS COVERED In this review, the authors discuss both the relation between in vitro and in vivo measured binding kinetics and the relation between in vivo binding kinetics, target occupancy and effect profiles. EXPERT OPINION More scientific evidence is required for the rational selection and development of drug-candidates on the basis of in vitro estimates of drug-target binding kinetics. To elucidate the value of in vitro binding kinetics measurements, it is necessary to obtain information on system-specific properties which influence the kinetics of target occupancy and drug effect. Mathematical integration of this information enables the identification of drug-specific properties which lead to optimal target occupancy and drug effect in patients.
Collapse
Affiliation(s)
- Wilhelmus E A de Witte
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Yin Cheong Wong
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Indira Nederpelt
- b Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Laura H Heitman
- b Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Meindert Danhof
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Piet H van der Graaf
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Ron A H J Gilissen
- c A Division of Janssen Pharmaceutica N.V., Janssen Research and Development , Turnhoutseweg 30, Beerse 2340 , Belgium
| | - Elizabeth C M de Lange
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| |
Collapse
|
13
|
Vauquelin G, Van Liefde I, Swinney DC. Radioligand binding to intact cells as a tool for extended drug screening in a representative physiological context. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 17:28-34. [PMID: 26724334 DOI: 10.1016/j.ddtec.2015.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 06/05/2023]
Abstract
Radioligand binding assays on intact cells offer distinct advantages to those on membrane suspensions. Major pharmacological properties like drug affinity and binding kinetics are more physiologically relevant. Complex mechanisms can be studied with a wider choice of experimental approaches and so provide insights into induced-fit type binding, receptor internalisation and even into pharmacomicrokinetic phenomena like drug rebinding and partitioning into the membrane. Hence, intact cell binding constitutes a valuable addition to the pharmacologist's toolbox.
Collapse
Affiliation(s)
- Georges Vauquelin
- Dept. of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| | - Isabelle Van Liefde
- Dept. of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - David C Swinney
- Institute for Rare and Neglected Diseases Drug Discovery, 897 Independence Ave, Suite 2C, Mountain View, CA 94043, United States
| |
Collapse
|
14
|
Guo D, IJzerman AP, Heitman LH. Importance of Drug-Target Residence Time at G Protein-Coupled Receptors - a Case for the Adenosine Receptors. THERMODYNAMICS AND KINETICS OF DRUG BINDING 2015. [DOI: 10.1002/9783527673025.ch13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
15
|
Vauquelin G. On the 'micro'-pharmacodynamic and pharmacokinetic mechanisms that contribute to long-lasting drug action. Expert Opin Drug Discov 2015; 10:1085-98. [PMID: 26165720 DOI: 10.1517/17460441.2015.1067196] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Optimal drug therapy often requires continuing high levels of target occupancy. Besides the traditional pharmacokinetic (PK) contribution thereto, drug-target interactions that comprise successive 'microscopic' steps as well as the intervention of the cell membrane and other 'micro'-anatomical structures nearby may help attaining this objective. AREAS COVERED This article reviews the 'micro'-pharmacodynamic (PD) and PK mechanisms that may increase a drug's residence time. Special focus is on induced-fit- and bivalent ligand binding models as well as on the ability of the plasma membrane surrounding the target to act as a repository for the drug (e.g., microkinetic model), to actively participate in the binding process (e.g., exosite model) and, along with microanatomical elements like synapses and interstitial spaces, to act on the drug's diffusion properties (reduction in dimensionality and drug-rebinding models). EXPERT OPINION The PK profile, as well as the target dissociation kinetics of a drug, may fail to account for its long-lasting efficiency in intact tissues and in vivo. This lacuna could potentially be alleviated by incorporating some of the enumerated 'microscopic' mechanisms and, to unveil them, dedicated experiments on sufficiently physiologically relevant biological material like cell monolayers can already be implemented early on in the lead optimization process.
Collapse
Affiliation(s)
- Georges Vauquelin
- a Free University Brussels (VUB), Molecular and Biochemical Pharmacology Department , Pleinlaan 2, B-1050 Brussels, Belgium +32 2 6291955 ; +32 2 6291358 ;
| |
Collapse
|
16
|
Guo D, Hillger JM, IJzerman AP, Heitman LH. Drug-Target Residence Time-A Case for G Protein-Coupled Receptors. Med Res Rev 2014; 34:856-92. [DOI: 10.1002/med.21307] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden University; P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Julia M. Hillger
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden University; P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden University; P.O. Box 9502 2300 RA Leiden the Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry; Leiden Academic Centre for Drug Research; Leiden University; P.O. Box 9502 2300 RA Leiden the Netherlands
| |
Collapse
|
17
|
Veiksina S, Kopanchuk S, Rinken A. Budded baculoviruses as a tool for a homogeneous fluorescence anisotropy-based assay of ligand binding to G protein-coupled receptors: The case of melanocortin 4 receptors. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:372-81. [DOI: 10.1016/j.bbamem.2013.09.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/19/2013] [Accepted: 09/24/2013] [Indexed: 12/01/2022]
|
18
|
Typical and atypical antipsychotics do not differ markedly in their reversibility of antagonism of the dopamine D2 receptor. Int J Neuropsychopharmacol 2014; 17:149-55. [PMID: 24074141 DOI: 10.1017/s1461145713000801] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It has been suggested that the favorable side-effect profiles of atypical antipsychotics (e.g. clozapine and amisulpride) are related to their ∼100-fold faster dissociation from dopamine D2 receptors (D2R) compared with typical antipsychotics (e.g. haloperidol and chlorpromazine). Fast dissociation would entail rapidly reversible antagonism; however, this has not been thoroughly studied using functional assays. We compared the reversibilities of D2R antagonism by 17 compounds using an electrophysiological method to measure dopamine-evoked potassium channel activation via D2R. Varying rates and amplitudes of D2R response recovery were observed following antagonist removal. Whereas recovery rates differed 15-fold between atypical drugs, recovery from clozapine and amisulpride antagonism was, unexpectedly, less than twofold faster than from chlorpromazine. The recovery amplitude correlated with calculated water solubility and lipid/water distribution coefficients, suggesting variable drug partitioning into cell membranes. Our data do not support the notion that the rate of reversibility of D2R antagonism is what distinguishes atypical from typical antipsychotics.
Collapse
|
19
|
Targeted Materials. Drug Deliv 2014. [DOI: 10.1007/978-1-4939-1998-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
20
|
Vilums M, Zweemer AJM, Yu Z, de Vries H, Hillger JM, Wapenaar H, Bollen IAE, Barmare F, Gross R, Clemens J, Krenitsky P, Brussee J, Stamos D, Saunders J, Heitman LH, Ijzerman AP. Structure-kinetic relationships--an overlooked parameter in hit-to-lead optimization: a case of cyclopentylamines as chemokine receptor 2 antagonists. J Med Chem 2013; 56:7706-14. [PMID: 24028535 DOI: 10.1021/jm4011737] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Preclinical models of inflammatory diseases (e.g., neuropathic pain, rheumatoid arthritis, and multiple sclerosis) have pointed to a critical role of the chemokine receptor 2 (CCR2) and chemokine ligand 2 (CCL2). However, one of the biggest problems of high-affinity inhibitors of CCR2 is their lack of efficacy in clinical trials. We report a new approach for the design of high-affinity and long-residence-time CCR2 antagonists. We developed a new competition association assay for CCR2, which allows us to investigate the relation of the structure of the ligand and its receptor residence time [i.e., structure-kinetic relationship (SKR)] next to a traditional structure-affinity relationship (SAR). By applying combined knowledge of SAR and SKR, we were able to re-evaluate the hit-to-lead process of cyclopentylamines as CCR2 antagonists. Affinity-based optimization yielded compound 1 with good binding (Ki = 6.8 nM) but very short residence time (2.4 min). However, when the optimization was also based on residence time, the hit-to-lead process yielded compound 22a, a new high-affinity CCR2 antagonist (3.6 nM), with a residence time of 135 min.
Collapse
Affiliation(s)
- Maris Vilums
- Division of Medicinal Chemistry, Leiden Academic Center for Drug Research, Leiden University , Post Office Box 9502, 2300 RA Leiden, Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Guo D, van Dorp EJH, Mulder-Krieger T, van Veldhoven JPD, Brussee J, IJzerman AP, Heitman LH. Dual-Point Competition Association Assay. ACTA ACUST UNITED AC 2012; 18:309-20. [DOI: 10.1177/1087057112464776] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The concept of ligand-receptor binding kinetics is emerging as an important parameter in the early phase of drug discovery. Since the currently used kinetic assays are laborious and low throughput, we developed a method that enables fast and large format screening. It is a so-called dual-point competition association assay, which measures radioligand binding at two different time points in the absence or presence of unlabeled competitors. Specifically, this assay yields the kinetic rate index (KRI), which is a measure for the binding kinetics of the unlabeled ligands screened. As a prototypical drug target, the adenosine A1 receptor (A1R) was chosen for assay validation and optimization. A screen with 35 high-affinity A1R antagonists yielded seven compounds with a KRI value above 1.0, which indicated a relatively slow dissociation from the target. All other compounds had a KRI value below or equal to 1.0, predicting a relatively fast dissociation rate. Several compounds were selected for follow-up kinetic quantifications in classical kinetic assays and were shown to have kinetic rates that corresponded to their KRI values. The dual-point assay and KRI value may have general applicability at other G-protein-coupled receptors, as well as at drug targets from other protein families.
Collapse
Affiliation(s)
- Dong Guo
- Leiden University, Leiden, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
22
|
Vauquelin G, Van Liefde I. Radioligand dissociation measurements: potential interference of rebinding and allosteric mechanisms and physiological relevance of the biological model systems. Expert Opin Drug Discov 2012; 7:583-95. [DOI: 10.1517/17460441.2012.687720] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
23
|
Vauquelin G, Bostoen S, Vanderheyden P, Seeman P. Clozapine, atypical antipsychotics, and the benefits of fast-off D2 dopamine receptor antagonism. Naunyn Schmiedebergs Arch Pharmacol 2012; 385:337-72. [PMID: 22331262 DOI: 10.1007/s00210-012-0734-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 01/25/2012] [Indexed: 12/20/2022]
Abstract
Drug-receptor interactions are traditionally quantified in terms of affinity and efficacy, but there is increasing awareness that the drug-on-receptor residence time also affects clinical performance. While most interest has hitherto been focused on slow-dissociating drugs, D(2) dopamine receptor antagonists show less extrapyramidal side effects but still have excellent antipsychotic activity when they dissociate swiftly. Fast dissociation of clozapine, the prototype of the "atypical antipsychotics", has been evidenced by distinct radioligand binding approaches both on cell membranes and intact cells. The surmountable nature of clozapine in functional assays with fast-emerging responses like calcium transients is confirmatory. Potential advantages and pitfalls of the hitherto used techniques are discussed, and recommendations are given to obtain more precise dissociation rates for such drugs. Surmountable antagonism is necessary to allow sufficient D(2) receptor stimulation by endogenous dopamine in the striatum. Simulations are presented to find out whether this can be achieved during sub-second bursts in dopamine concentration or rather during much slower, activity-related increases thereof. While the antagonist's dissociation rate is important to distinguish between both mechanisms, this becomes much less so when contemplating time intervals between successive drug intakes, i.e., when pharmacokinetic considerations prevail. Attention is also drawn to the divergent residence times of hydrophobic antagonists like haloperidol when comparing radioligand binding data on cell membranes with those on intact cells and clinical data.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium.
| | | | | | | |
Collapse
|
24
|
Vauquelin G. Determination of drug–receptor residence times by radioligand binding and functional assays: experimental strategies and physiological relevance. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20015e] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
25
|
Demaegdt H, Gard P, De Backer JP, Lukaszuk A, Szemenyei E, Tóth G, Tourwé D, Vauquelin G. Binding of "AT4 receptor" ligands to insulin regulated aminopeptidase (IRAP) in intact Chinese hamster ovary cells. Mol Cell Endocrinol 2011; 339:34-44. [PMID: 21457753 DOI: 10.1016/j.mce.2011.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/17/2011] [Accepted: 03/22/2011] [Indexed: 01/03/2023]
Abstract
Insulin regulated aminopeptidase (IRAP) recognises "AT(4)-receptor" ligands like angiotensin IV (Ang IV) and peptidomimetics like AL-11. The metabolic stability and high affinity of [(3)H]AL-11 for catalytically active IRAP allowed its detection in Chinese hamster ovary (CHO-K1) cell membranes in the absence of chelators (Demaegdt et al., 2009). Here, we show that, contrary to [(3)H]Ang IV, [(3)H]AL-11 displays high affinity and specificity for IRAP in intact CHO-K1 cells as well. After binding to IRAP at the surface, [(3)H]AL-11 is effectively internalized by an endocytotic process. Unexpectedly, surface binding and internalization of [(3)H]AL-11 was not affected by pretreating the cells with Ang IV but declined with AL-11. In the latter case surface expression of IRAP even increased. After elimination of simpler explanations, it is proposed that metabolically stable "AT(4)-receptor" ligands undergo semi-continuous cycling between the cell surface and endosomal compartments. The in vivo efficacy of stable and unstable "AT(4)-receptor" ligands could therefore differ.
Collapse
Affiliation(s)
- Heidi Demaegdt
- Department of Molecular and Biochemical Pharmacology, Research Group of Experimental Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
26
|
De Mey JG, Compeer MG, Lemkens P, Meens MJ. ETA-receptor antagonists or allosteric modulators? Trends Pharmacol Sci 2011; 32:345-51. [DOI: 10.1016/j.tips.2011.02.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 02/27/2011] [Accepted: 02/28/2011] [Indexed: 01/14/2023]
|