Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
SLAS Discov ; 26(7): 835-850, 2021 08.
Article in English | MEDLINE | ID: mdl-34112012

ABSTRACT

The analysis framework used to quantify drug potency in vitro (e.g., Kd or Ki) was initially developed for classical pharmacology bioassays, for example, organ bath experiments testing moderate-affinity natural products. Modern drug discovery can infringe the assumptions of the classical pharmacology analysis equations, owing to the reduction of assay volume in miniaturization, target overexpression, and the increase of compound-target affinity in medicinal chemistry. These assumptions are that (1) the compound concentration greatly exceeds the target concentration (i.e., minimal ligand depletion), and (2) the compound is at equilibrium with the receptor (i.e., rapid ligand binding kinetics). Unappreciated infringement of these assumptions can lead to substantial underestimation of compound affinity, which negatively impacts the drug discovery process, from early-stage lead optimization to prediction of human dosing. This study evaluates the real-world impact of these factors on the target interaction assays used in drug discovery using literature examples, database searches, and simulations. The ranges of compound affinity and the assay types that are prone to depletion and equilibration artifacts are identified. Importantly, the highest-affinity compounds, usually the highest value chemical matter in drug discovery, are the most affected. Methods and simulation tools are provided to enable investigators to evaluate, manage, and minimize depletion or equilibration artifacts. This study enables the correct application of pharmacological data analysis to accurately quantify affinity using modern drug discovery assay technology.


Subject(s)
Drug Discovery/methods , In Vitro Techniques , Pharmacology/methods , Computational Biology/methods , Humans , Kinetics , Ligands
2.
Front Cell Neurosci ; 15: 814547, 2021.
Article in English | MEDLINE | ID: mdl-35110998

ABSTRACT

Neurons integrate inputs over different time and space scales. Fast excitatory synapses at boutons (ms and µm), and slow modulation over entire dendritic arbors (seconds and mm) are all ultimately combined to produce behavior. Understanding the timing of signaling events mediated by G-protein-coupled receptors is necessary to elucidate the mechanism of action of therapeutics targeting the nervous system. Measuring signaling kinetics in live cells has been transformed by the adoption of fluorescent biosensors and dyes that convert biological signals into optical signals that are conveniently recorded by microscopic imaging or by fluorescence plate readers. Quantifying the timing of signaling has now become routine with the application of equations in familiar curve fitting software to estimate the rates of signaling from the waveform. Here we describe examples of the application of these methods, including (1) Kinetic analysis of opioid signaling dynamics and partial agonism measured using cAMP and arrestin biosensors; (2) Quantifying the signaling activity of illicit synthetic cannabinoid receptor agonists measured using a fluorescent membrane potential dye; (3) Demonstration of multiplicity of arrestin functions from analysis of biosensor waveforms and quantification of the rates of these processes. These examples show how temporal analysis provides additional dimensions to enhance the understanding of GPCR signaling and therapeutic mechanisms in the nervous system.

3.
Sci Rep ; 10(1): 12263, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704081

ABSTRACT

In classical pharmacology, bioassay data are fit to general equations (e.g. the dose response equation) to determine empirical drug parameters (e.g. EC50 and Emax), which are then used to calculate chemical parameters such as affinity and efficacy. Here we used a similar approach for kinetic, time course signaling data, to allow empirical and chemical definition of signaling by G-protein-coupled receptors in kinetic terms. Experimental data are analyzed using general time course equations (model-free approach) and mechanistic model equations (mechanistic approach) in the commonly-used curve-fitting program, GraphPad Prism. A literature survey indicated signaling time course data usually conform to one of four curve shapes: the straight line, association exponential curve, rise-and-fall to zero curve, and rise-and-fall to steady-state curve. In the model-free approach, the initial rate of signaling is quantified and this is done by curve-fitting to the whole time course, avoiding the need to select the linear part of the curve. It is shown that the four shapes are consistent with a mechanistic model of signaling, based on enzyme kinetics, with the shape defined by the regulation of signaling mechanisms (e.g. receptor desensitization, signal degradation). Signaling efficacy is the initial rate of signaling by agonist-occupied receptor (kτ), simply the rate of signal generation before it becomes affected by regulation mechanisms, measurable using the model-free analysis. Regulation of signaling parameters such as the receptor desensitization rate constant can be estimated if the mechanism is known. This study extends the empirical and mechanistic approach used in classical pharmacology to kinetic signaling data, facilitating optimization of new therapeutics in kinetic terms.


Subject(s)
Models, Biological , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Dose-Response Relationship, Drug , Drug Discovery , Pharmacokinetics , Signal Transduction/drug effects
4.
Sci Rep ; 10(1): 1766, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019973

ABSTRACT

The kinetics/dynamics of signaling are of increasing value for G-protein-coupled receptor therapeutic development, including spatiotemporal signaling and the kinetic context of biased agonism. Effective application of signaling kinetics to developing new therapeutics requires reliable kinetic assays and an analysis framework to extract kinetic pharmacological parameters. Here we describe a platform for measuring arrestin recruitment kinetics to GPCRs using a high quantum yield, genetically encoded fluorescent biosensor, and a data analysis framework to quantify the recruitment kinetics. The sensor enabled high temporal resolution measurement of arrestin recruitment to the angiotensin AT1 and vasopressin V2 receptors. The analysis quantified the initial rate of arrestin recruitment (kτ), a biologically-meaningful kinetic drug efficacy parameter, by fitting time course data using routine curve-fitting methods. Biased agonism was assessed by comparing kτ values for arrestin recruitment with those for Gq signaling via the AT1 receptor. The kτ ratio values were in good agreement with bias estimates from existing methods. This platform potentially improves and simplifies assessment of biased agonism because the same assay modality is used to compare pathways (potentially in the same cells), the analysis method is parsimonious and intuitive, and kinetic context is factored into the bias measurement.


Subject(s)
Biosensing Techniques/methods , Protein Binding/physiology , Signal Transduction/physiology , Angiotensin I/metabolism , Arrestins/metabolism , Cell Line , HEK293 Cells , Humans , Kinetics , Ligands , Receptors, G-Protein-Coupled/metabolism , Receptors, Vasopressin/metabolism
5.
Drug Discov Today ; 25(1): 7-14, 2020 01.
Article in English | MEDLINE | ID: mdl-31557449

ABSTRACT

In drug discovery, it is essential to accurately measure drug-target binding affinity. Here, we revisit the fact that target binding kinetics impact the measurement of affinity, using a case study: development of corticotropin-releasing factor antagonists. Slow dissociation of the drug-target complex results in affinity assays being far from equilibrium, which results in erroneous estimates of affinity. This scenario can impair prediction of human dosing, assessment of target selectivity, identification of high-affinity ligands and determination of SAR. We describe strategies to detect lack of equilibration in affinity assays and methods to correctly measure affinity of slowly dissociating compounds. These considerations will facilitate drug discovery by ensuring reliable measurement of drug-target binding affinity.


Subject(s)
Drug Discovery , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , Humans , Kinetics , Protein Binding , Receptors, Corticotropin-Releasing Hormone/metabolism
6.
Article in English | MEDLINE | ID: mdl-28818556

ABSTRACT

INTRODUCTION: Measuring unlabeled ligand receptor binding kinetics is valuable in optimizing and understanding drug action. Unfortunately, deriving equations for estimating kinetic parameters is challenging because it involves calculus; integration can be a frustrating barrier to the pharmacologist seeking to measure simple rate parameters. Here, a well-known tool for simplifying the derivation, the Laplace transform, is applied to models of receptor-ligand interaction. The method transforms differential equations to a form in which simple algebra can be applied to solve for the variable of interest, for example the concentration of ligand-bound receptor. METHODS: The goal is to provide instruction using familiar examples, to enable investigators familiar with handling equilibrium binding equations to derive kinetic equations for receptor-ligand interaction. First, the Laplace transform is used to derive the equations for association and dissociation of labeled ligand binding. Next, its use for unlabeled ligand kinetic equations is exemplified by a full derivation of the kinetics of competitive binding equation. Finally, new unlabeled ligand equations are derived using the Laplace transform. These equations incorporate a pre-incubation step with unlabeled or labeled ligand. RESULTS: Four equations for measuring unlabeled ligand kinetics were compared and the two new equations verified by comparison with numerical solution. Importantly, the equations have not been verified with experimental data because no such experiments are evident in the literature. Equations were formatted for use in the curve-fitting program GraphPad Prism 6.0 and fitted to simulated data. DISCUSSION: This description of the Laplace transform method will enable pharmacologists to derive kinetic equations for their model or experimental paradigm under study. Application of the transform will expand the set of equations available for the pharmacologist to measure unlabeled ligand binding kinetics, and for other time-dependent pharmacological activities.


Subject(s)
Binding, Competitive , Models, Biological , Pharmacokinetics , Ligands , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Software
7.
J Pharmacol Exp Ther ; 361(3): 454-461, 2017 06.
Article in English | MEDLINE | ID: mdl-28404690

ABSTRACT

The vesicular monoamine transporter 2 (VMAT2) is an integral presynaptic protein that regulates the packaging and subsequent release of dopamine and other monoamines from neuronal vesicles into the synapse. Valbenazine (NBI-98854), a novel compound that selectively inhibits VMAT2, is approved for the treatment of tardive dyskinesia. Valbenazine is converted to two significant circulating metabolites in vivo, namely, (+)-α-dihydrotetrabenazine (R,R,R-HTBZ) and a mono-oxy metabolite, NBI-136110. Radioligand-binding studies were conducted to assess and compare valbenazine, tetrabenazine, and their respective metabolites in their abilities to selectively and potently inhibit [3H]-HTBZ binding to VMAT2 in rat striatal, rat forebrain, and human platelet homogenates. A broad panel screen was conducted to evaluate possible off-target interactions of valbenazine, R,R,R-HTBZ, and NBI-136110 at >80 receptor, transporter, and ion channel sites. Radioligand binding showed R,R,R-HTBZ to be a potent VMAT2 inhibitor in homogenates of rat striatum (Ki = 1.0-2.8 nM), rat forebrain (Ki = 4.2 nM), and human platelets (Ki = 2.6-3.3 nM). Valbenazine (Ki = 110-190 nM) and NBI-136110 (Ki = 160-220 nM) also exhibited inhibitory effects on VMAT2, but with lower potency than R,R,R-HTBZ. Neither valbenazine, R,R,R-HTBZ, nor NBI-136110 had significant off-target interactions at serotonin (5-HT1A, 5-HT2A, 5-HT2B) or dopamine (D1 or D2) receptor sites. In vivo studies measuring ptosis and prolactin secretion in the rat confirmed the specific and dose-dependent interactions of tetrabenazine and R,R,R-HTBZ with VMAT2. Evaluations of potency and selectivity of tetrabenazine and its pharmacologically active metabolites were also performed. Overall, the pharmacologic characteristics of valbenazine appear consistent with the favorable efficacy and tolerability findings of recent clinical studies [KINECT 2 (NCT01733121), KINECT 3 (NCT02274558)].


Subject(s)
Tetrabenazine/analogs & derivatives , Valine/analogs & derivatives , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , Vesicular Monoamine Transport Proteins/metabolism , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , CHO Cells , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cricetinae , Cricetulus , HEK293 Cells , Humans , Protein Binding/drug effects , Protein Binding/physiology , Rats , Tetrabenazine/metabolism , Tetrabenazine/pharmacology , Valine/metabolism , Valine/pharmacology
8.
J Pharmacol Exp Ther ; 341(2): 518-31, 2012 May.
Article in English | MEDLINE | ID: mdl-22357972

ABSTRACT

Corticotropin-releasing factor (CRF) receptor antagonists are under preclinical and clinical investigation for stress-related disorders. In this study the impact of receptor-ligand binding kinetics on CRF1 receptor antagonist pharmacology was investigated by measuring the association rate constant (k1), dissociation rate constant (k₋1), and kinetically derived affinity at 37°C. Three aspects of antagonist pharmacology were reevaluated: comparative binding activity of advanced compounds, in vivo efficacy, and structure-activity relationships. Twelve lead compounds, with little previously noted difference of affinity, varied substantially in their kinetic binding activity with a 510-fold range of kinetically derived affinity (k₋1/k1), 170-fold range of k₋1, and 13-fold range of k1. The k₋1 values indicated previous affinity measurements were not close to equilibrium, resulting in compression of the measured affinity range. Dissociation was exceptionally slow for three ligands (k₋1 t(1/2) of 1.6-7.2 h at 37°C). Differences of binding behavior were consistent with in vivo pharmacodynamics (suppression of adrenocorticotropin in adrenalectomized rats). Ligand concentration-effect relationships correlated with their kinetically derived affinity. Two ligands that dissociated slowly (53 and 130 min) produced prolonged suppression, whereas only transient suppression was observed with a more rapidly dissociating ligand (16 min). Investigating the structure-activity relationship indicated exceptionally low values of k1, approaching 100,000-fold less than the diffusion-limited rate. Retrospective interpretation of medicinal chemistry indicates optimizing specific elements of chemical structure overcame kinetic barriers in the association pathway, for example, constraint of the pendant aromatic orthogonal to the ligand core. Collectively, these findings demonstrate receptor binding kinetics provide new dimensions for understanding and potentially advancing the pharmacology of CRF1 receptor antagonists.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Binding, Competitive , HEK293 Cells , Humans , Kinetics , Ligands , Protein Binding , Radioligand Assay/methods , Rats , Receptors, Corticotropin-Releasing Hormone/chemistry , Retrospective Studies , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 20(24): 7259-64, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21074436

ABSTRACT

Antagonists of the corticotropin-releasing factor (CRF) neuropeptide may prove effective in treating stress and anxiety related disorders. In an effort to identify antagonists with improved physico-chemical properties a new series of CRF(1) antagonists were designed to substitute the propyl groups at the C7 position of the pyrazolo[1,5-a]pyrimidine core of 1 with heterocycles. Compound (S)-8d was identified as a high affinity ligand with a pK(i) value of 8.2 and a functional CRF(1) antagonist with pIC(50) value of 7.0 in the in vitro CRF ACTH production assay.


Subject(s)
Azabicyclo Compounds/chemistry , Oxadiazoles/chemistry , Pyrazoles/chemistry , Pyridines/chemistry , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/pharmacokinetics , Humans , Microsomes, Liver/metabolism , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacokinetics , Protein Binding , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
10.
J Recept Signal Transduct Res ; 29(2): 84-93, 2009.
Article in English | MEDLINE | ID: mdl-19308787

ABSTRACT

We introduce a novel experimental method to determine both the extent of ex vivo receptor occupancy of administered compound and its dissociation rate constant (k4). [Here, we reference k4 as the rate of offset of unlabeled ligand in convention with Motulsky and Mahan (1)]. We derived a kinetic rate equation based on the dissociation rate constant for an unlabeled compound competing for the same site as a labeled compound and describe a model to simulate fractional occupancy. To validate our model, we performed in vitro kinetics and ex vivo occupancy experiments in rat cortex with varying concentrations of (R)-dimethindene, a sedating antihistamine. Brain tissue was removed at various times post oral administration, and histamine H1 receptor ligand [3H]-doxepin binding to homogenates from drug-treated or vehicle-treated rats was measured at multiple time points at room temperature. Fractional occupancy and k4 for (R)-dimethindene binding to H1 receptors were calculated by using our proposed model. Rats dosed with 30 and 60 mg/kg (R)-dimethindene showed 42% and 67% occupancy of central H1 receptors, respectively. These results were comparable to occupancy data determined by equilibrium radioligand binding. In addition, drug k4 rate determined by using our ex vivo method was equivalent to k4 determined by in vitro competition kinetics (dissociation half-life t(1/2) approximately 30 min). The outlined method can be used to assess, by simulation and experiment, occupancy for compounds based on dissociation rate constants and contributes to current efforts in drug optimization to profile antagonist efficacy in terms of its kinetic drug-target binding parameters. Data described by the method may be analyzed with commercially available software. Suggested fitting procedures are given in the appendix.


Subject(s)
Dimethindene/metabolism , Histamine H1 Antagonists/metabolism , Radioligand Assay , Receptors, Drug/metabolism , Receptors, Histamine H1/metabolism , Animals , Binding, Competitive , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dimethindene/chemistry , Dimethindene/pharmacology , Doxepin/metabolism , Histamine H1 Antagonists/chemistry , Histamine H1 Antagonists/pharmacology , Kinetics , Male , Rats , Receptors, Drug/chemistry , Receptors, Drug/drug effects , Receptors, Histamine H1/chemistry
11.
Mol Pharmacol ; 73(5): 1371-80, 2008 May.
Article in English | MEDLINE | ID: mdl-18239030

ABSTRACT

Allosteric modulators of G-protein-coupled receptors can regulate conformational states involved in receptor activation ( Mol Pharmacol 58: 1412-1423, 2000 ). This hypothesis was investigated for the corticotropin-releasing factor type 1 (CRF(1)) receptor using a novel series of ligands with varying allosteric effect on CRF binding (inhibition to enhancement). For the G-protein-uncoupled receptor, allosteric modulation of CRF binding was correlated with nonpeptide ligand signaling activity; inverse agonists inhibited and agonists enhanced CRF binding. These data were quantitatively consistent with a two-state equilibrium underlying the modulation of CRF binding to the G-protein-uncoupled receptor. We next investigated the allosteric effect on CRF-stimulated G-protein coupling. Ligands inhibited CRF-stimulated cAMP accumulation regardless of their effect on the G-protein-uncoupled state. The modulators reduced CRF E(max) values, suggesting that they reduced the efficacy of a CRF-bound active state to couple to G-protein. Consistent with this hypothesis, the modulators inhibited binding to a guanine nucleotide-sensitive state. Together, the results are quantitatively consistent with a model in which 1) the receptor exists in three predominant states: an inactive state, a weakly active state, and a CRF-bound fully active state; 2) allosteric inverse agonists stabilize the inactive state, and allosteric agonists stabilize the weakly active state; and 3) antagonism of CRF signaling results from destabilization of the fully active state. These findings imply that nonpeptide ligands differentially modulate conformational states involved in CRF(1) receptor activation and suggest that different conformational states can be targeted in designing nonpeptide ligands to inhibit CRF signaling.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, Corticotropin-Releasing Hormone/metabolism , Allosteric Regulation/drug effects , Amphibian Proteins , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Iodine Radioisotopes , Ligands , Models, Chemical , Peptide Hormones , Peptides/metabolism , Peptides/pharmacology , Protein Binding/drug effects , Protein Conformation/drug effects , Rats , Signal Transduction/drug effects
12.
Curr Neuropharmacol ; 5(3): 168-79, 2007 Sep.
Article in English | MEDLINE | ID: mdl-19305799

ABSTRACT

Class B GPCR's are activated by peptide ligands, typically 30-40 amino acid residues, that are involved in major physiological functions such as glucose homeostasis (glucagon and glucagon-like peptide 1), calcium homeostasis and bone turnover (parathyroid hormone and calcitonin), and control of the stress axis (corticotropin-releasing factor). Peptide therapeutics have been developed targeting these receptors but development of nonpeptide ligands, enabling oral administration, has proved challenging. Allosteric modulation of these receptors provides a potential route to developing nonpeptide ligands that inhibit, activate, or potentiate activation of these receptors. Here the known mechanisms of allosteric modulators targeting Class B GPCR's are reviewed, particularly nonpeptide antagonists of the corticotropin-releasing factor 1 receptor and allosteric enhancers of the glucagon-like peptide-1 receptor. Also discussed is the potential for antagonist ligands to operate by competitive inhibition of one of the peptide binding sites, analogous to the Charniere mechanism. These mechanisms are then used to discuss potential strategies and management of pharmacological complexity in the future development of allosteric modulators for Class B GPCR's.

13.
Biochem Pharmacol ; 72(7): 838-49, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-16930559

ABSTRACT

Numerous nonpeptide ligands have been developed for the human gonadotropin-releasing hormone (GnRH) receptor as potential agents for treatment of disorders of the reproductive-endocrine axis. While the equilibrium binding of these ligands has been studied in detail, little is known of the kinetics of their receptor interaction. In this study we evaluated the kinetic structure-activity relationships (SAR) of uracil-series antagonists by measuring their association and dissociation rate constants. These constants were measured directly using a novel radioligand, [3H] NBI 42902, and indirectly for unlabeled ligands. Receptor association and dissociation of [3H] NBI 42902 was monophasic, with an association rate constant of 93+/-10 microM(-1) min(-1) and a dissociation rate constant of 0.16+/-0.02 h(-1) (t(1/2) of 4.3 h). Four unlabeled compounds were tested with varying substituents at the 2-position of the benzyl group at position 1 of the uracil (-F, -SO(CH3), -SO2(CH3) and -CF3). The nature of the substituent did not appreciably affect the association rate constant but varied the dissociation rate constant >50-fold (t(1/2) ranging from 52 min for -SO(CH3) to >43 h for -CF3). This SAR was poorly resolved in standard competition assays due to lack of equilibration. The functional consequences of the varying dissociation rate were investigated by measuring antagonism of GnRH-stimulated [3H] inositol phosphates accumulation. Slowly dissociating ligands displayed insurmountable antagonism (decrease of the GnRH E(max)) while antagonism by more rapidly dissociating ligands was surmountable (without effect on the GnRH E(max)). Therefore, evaluating the receptor binding kinetics of nonpeptide antagonists revealed SAR, not evident in standard competition assays, that defined at least in part the mode of functional antagonism by the ligands. These findings are of importance for the future definition of nonpeptide ligand SAR and for the identification of potentially useful slowly dissociating antagonists for the GnRH receptor.


Subject(s)
Quantitative Structure-Activity Relationship , Receptors, LHRH/antagonists & inhibitors , Uracil/pharmacology , Binding, Competitive/drug effects , Humans , Kinetics , Ligands , Molecular Structure , Radioligand Assay/methods , Receptors, LHRH/metabolism , Thymine/analogs & derivatives , Thymine/metabolism , Tritium , Uracil/chemistry , Uracil/metabolism
14.
Biochem Pharmacol ; 72(2): 244-55, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16750175

ABSTRACT

The molecular interactions between non-peptide antagonists and the corticotropin-releasing factor type 1 (CRF1) receptor are poorly understood. A CRF1 receptor mutation has been identified that reduces binding affinity of the non-peptide antagonist NBI 27914 (M276I in transmembrane domain 5). We have investigated the mechanism of the mutation's effect using a combination of peptide and non-peptide ligands and receptor mutations. The M276I mutation reduced binding affinity of standard non-peptide antagonists 5-75-fold while having no effect on peptide ligand binding. We hypothesized that the side chain of isoleucine, beta-branched and so rotationally constrained when within an alpha-helix, introduces a barrier to non-peptide antagonist binding. In agreement with this hypothesis, mutation of M276 to the rotationally constrained valine produced similar reductions of affinity as M276I mutation, whereas mutation to leucine (with an unbranched beta-carbon) minimally affected non-peptide antagonist affinity. Mutation to alanine did not appreciably affect non-peptide antagonist affinity, implying the methionine side chain does not contribute directly to binding. Three observations suggested M276I/V mutations interfere with binding of the heterocyclic core of the compounds: (1) all compounds affected by M276I/V mutations possess a planar heterocyclic core. (2) None of the M276 mutations affected binding of an acylic compound. (3) The mutations differentially affected affinity of two compounds that differ only by core methylation. These findings imply that non-peptide antagonists, and specifically the heterocyclic core of such molecules, bind in the vicinity of M276 of the CRF1 receptor. M276 mutations did not affect peptide ligand binding and this residue is distant from determinants of peptide binding (predominantly in the extracellular regions), providing molecular evidence for non-overlapping (allosteric) binding sites for peptide and non-peptide ligands within the CRF1 receptor.


Subject(s)
Amino Acids/chemistry , Receptors, Corticotropin-Releasing Hormone/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Molecular Sequence Data , Mutation , Radioligand Assay , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/chemistry
15.
Biochemistry ; 44(44): 14494-508, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16262250

ABSTRACT

The melanocortin-4 (MC4) receptor is a potential therapeutic target for obesity and cachexia, for which nonpeptide agonists and antagonists are being developed, respectively. The aim of this study was to identify molecular interactions between the MC4 receptor and nonpeptide ligands, and to compare the mechanism of binding between agonist and antagonist ligands. Nonpeptide ligand interaction was affected by mutations that reduce peptide ligand binding (D122A, D126A, S190A, M200A, F261A, and F284A), confirming overlapping binding determinants for peptide and nonpeptide ligands. The common halogenated phenyl group of nonpeptide ligands was a determinant of F261A and F284A mutations' affinity-reducing effect, implying this group interacts with the aromatic side chains of these residues. All affected compounds contain this group, the mutations reduced binding of 2,4-dichloro-substituted compounds more than 4-chloro-substituted-compounds, and F284A mutation eliminated the affinity-enhancing effect of 2-chloro-substitution. F261A and F284A mutations reduced the affinity of antagonists more than agonists, suggesting that the stronger ligand interaction with these residues, the lower the ligand efficacy. Supporting this hypothesis, F261A mutation increased the efficacy of nonpeptide antagonist and partial agonist ligands. D122A and D126A mutations reduced nonpeptide ligand interaction. Removing the ligands' derivatized amide group eliminated the effect of the mutations. Interaction of agonists, which bear a common amine within this group, was strongly reduced by D126A mutation (550-3300-fold), suggesting an electrostatic interaction between the amine and the acidic group of D126. These postulated interactions with aromatic and acidic regions of the MC4 receptor are consistent with a molecular model of the receptor. Furthermore, the strength of interaction with the aromatic pocket, and potentially the acidic pocket, controls the signaling efficacy of the ligand.


Subject(s)
Receptor, Melanocortin, Type 4 , Amino Acid Sequence , Binding Sites , Cell Line , Cyclic AMP/metabolism , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/metabolism
16.
J Med Chem ; 48(18): 5780-93, 2005 Sep 08.
Article in English | MEDLINE | ID: mdl-16134945

ABSTRACT

Antagonists of the corticotropin-releasing factor (CRF) neuropeptide should prove to be effective in treating stress and anxiety-related disorders. In an effort to identify antagonists with improved physicochemical properties, new tricyclic CRF(1) antagonists were designed, synthesized, and tested for biological activity. As a result of studies aimed at establishing a relationship between structure and CRF(1) binding affinity, NBI 35965 (12a) was identified as a high-affinity antagonist with a pK(i) value of 8.5. Compound 12a proved to be a functional CRF(1) antagonist with pIC(50) values of 7.1 and 6.9 in the in vitro CRF-stimulated cAMP accumulation and ACTH production assays, respectively, and 12a also reduced CRF or stress induced ACTH production in vivo.


Subject(s)
Heterocyclic Compounds, 3-Ring/chemical synthesis , Pyrazoles/chemical synthesis , Pyridines/chemical synthesis , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Acenaphthenes , Adrenocorticotropic Hormone/biosynthesis , Animals , Cells, Cultured , Corticotropin-Releasing Hormone/metabolism , Cyclic AMP/biosynthesis , Drug Design , Female , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Molecular Conformation , Pituitary Gland, Anterior/cytology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 15(10): 2541-6, 2005 May 16.
Article in English | MEDLINE | ID: mdl-15863313

ABSTRACT

Optimization on a series of piperazinebenzylamines resulted in analogues with low nanomolar binding at the human MC4 receptor but weak affinity (Ki > 500 nM) at the MC3 receptor. Compound 14c was identified to be a potent MC4R antagonist (Ki = 3.2 nM) with a selectivity of 240-fold over MC3R. It proved to be an insurmountable antagonist in a cAMP assay. Compound 14c potently stimulated food intake in satiated mice when given by intracerebroventricular administration.


Subject(s)
Benzylamines/pharmacology , Feeding Behavior/drug effects , Piperazines/pharmacology , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Satiety Response , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Mice
18.
Drug Discov Today ; 10(6): 417-27, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15808821

ABSTRACT

Class B G-protein-coupled receptors are a small family of 15 peptide-binding receptors. This family includes at least six biologically attractive therapeutic targets for both peptide ligands (osteoporosis and Type II diabetes) and nonpeptide ligands (anxiety, depression and migraine). A general mechanism of peptide binding has emerged for this receptor family, termed the two-domain model. In this mechanism, the C-terminal ligand region binds the extracellular N-terminal domain of the receptor. This interaction acts as an affinity trap, promoting interaction of the N-terminal ligand region with the juxtamembrane domain of the receptor. Peptide binding to the juxtamembrane domain activates the receptor and stimulates intracellular signaling. Nonpeptide ligands bind the juxtamembrane or N-terminal domain and, in most cases, allosterically modulate peptide-ligand binding. Here, these mechanisms of peptide and nonpeptide ligand binding are reviewed, then applied in a discussion of the future strategies of drug development for Class B G-protein-coupled receptors.


Subject(s)
Peptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Allosteric Regulation , Animals , Drug Design , Humans , Ligands , Models, Molecular , Peptides/chemistry , Peptides/pharmacology , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/drug effects
19.
Endocrinology ; 146(6): 2766-73, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15774557

ABSTRACT

Cachexia is metabolic disorder characterized by anorexia, an increased metabolic rate, and loss of lean body mass. It is a relatively common disorder, and is a pathological feature of diseases such as cancer, HIV infection, and renal failure. Recent studies have demonstrated that cachexia brought about by a variety of illnesses can be attenuated or reversed by blocking activation of the melanocortin 4 subtype receptor (MC4-R) within the central nervous system. Although the potential use of central MC4-R antagonists for the treatment of cachexia was supported by these studies, utility was limited by the need to deliver these agents intracerebroventricularly. In the current study, we present a series of experiments demonstrating that peripheral administration of a small molecule MC4-R antagonist can effectively stimulate daytime (satiated) food intake as well as decrease basal metabolic rate in normal animals. Furthermore, this compound attenuated cachexia and preserved lean body mass in a murine cancer model. These data clearly demonstrate the potential of small molecule MC4-R antagonists in the treatment of cachexia and underscore the importance of melanocortin signaling in the development of this metabolic disorder.


Subject(s)
Cachexia/drug therapy , Carcinoma, Lewis Lung/complications , Lung Neoplasms/complications , Receptor, Melanocortin, Type 4/antagonists & inhibitors , alpha-MSH/analogs & derivatives , Animals , Cachexia/etiology , Cachexia/prevention & control , Cell Line , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Humans , Iodine Radioisotopes , Kidney/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Radioligand Assay , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 4/genetics , alpha-MSH/metabolism , alpha-MSH/pharmacology
20.
J Pharmacol Exp Ther ; 313(3): 1281-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15743921

ABSTRACT

Agonists of the melanocortin 4 (MC4) receptor have potential pharmaceutical benefit in the treatment of obesity and sexual dysfunction. In this study, we have compared the ability of a number of peptide and nonpeptide agonists to activate a FLAG-tagged human MC4 (FMC4) receptor, as measured by both cAMP accumulation and calcium mobilization using a fluorometric imaging plate reader (FLIPR). In addition, we have analyzed the ability of these agonists to cause receptor internalization, as measured by fluorescence-activated cell sorting analysis. The endogenous agonist alpha-melanocortin-stimulating hormone (alpha-MSH) increased cAMP accumulation, calcium mobilization, and receptor internalization in a dose-dependent manner in human embryonic kidney 293 cells expressing the FMC4 receptor. The activity of the other agonists varied considerably in these assays, and overall, the potency and intrinsic activity of the agonists in the cAMP accumulation assays did not correlate with their potency or intrinsic activity in either the FLIPR or receptor internalization assays. Agonists could be clearly separated into two functional classes based on their structure. Peptide agonists beta-MSH, des-acetyl-alpha-MSH, and [Nle(4), D-Phe(7)]-alpha-melanocortin-stimulating hormone exhibited 80 to 112% of the maximal alpha-MSH response in cAMP accumulation and 62 to 96% in FLIPR assays and were able to cause 75 to 118% of receptor internalization induced by alpha-MSH. Conversely, although the nonpeptide agonists exhibited 73 to 149% of the alpha-MSH response in the cAMP accumulation assays, they were significantly impaired in the FLIPR (7-40%) and receptor internalization (-5-38%) assays. These findings demonstrate an important difference in activation and internalization of the MC4 receptor by nonpeptide versus peptide agonists and provides evidence of agonist-specific conformational states.


Subject(s)
Receptor, Melanocortin, Type 4/agonists , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Fluorometry , Humans , Ligands , Melanocyte-Stimulating Hormones/metabolism , Protein Conformation , Receptor, Melanocortin, Type 4/chemistry , Receptor, Melanocortin, Type 4/physiology , alpha-MSH/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...