Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Publication year range
1.
Mol Pharmacol ; 91(5): 518-532, 2017 05.
Article in English | MEDLINE | ID: mdl-28254957

ABSTRACT

Pairing orphan G protein­coupled receptors (GPCRs) with their cognate endogenous ligands is expected to have a major impact on our understanding of GPCR biology. It follows that the reproducibility of orphan receptor ligand pairs should be of fundamental importance to guide meaningful investigations into the pharmacology and function of individual receptors. GPR17 is an orphan receptor characterized by some as a dualistic uracil nucleotide/cysteinyl leukotriene receptor and by others as inactive toward these stimuli altogether. Whereas regulation of central nervous system myelination by GPR17 is well established, verification of activity of its putative endogenous ligands has proven elusive so far. Herein we report that uracil nucleotides and cysteinyl leukotrienes do not activate human, mouse, or rat GPR17 in various cellular backgrounds, including primary cells, using eight distinct functional assay platforms based on labelfree pathway-unbiased biosensor technologies, as well as canonical second-messenger or biochemical assays. Appraisal of GPR17 activity can neither be accomplished with co-application of both ligand classes, nor with exogenous transfection of partner receptors (nucleotide P2Y12, cysteinyl-leukotriene CysLT1) to reconstitute the elusive pharmacology. Moreover, our study does not support the inhibition of GPR17 by the marketed antiplatelet drugs cangrelor and ticagrelor, previously suggested to antagonize GPR17. Whereas our data do not disagree with a role of GPR17 per se as an orchestrator of central nervous system functions, they challenge the utility of the proposed (ant)agonists as tools to imply direct contribution of GPR17 in complex biologic settings.


Subject(s)
Cysteine/pharmacology , Leukotrienes/pharmacology , Receptors, G-Protein-Coupled/metabolism , Uracil Nucleotides/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Animals , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cricetulus , HEK293 Cells , Humans , Ligands , Mice , Nerve Tissue Proteins/metabolism , Rats , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Ticagrelor
2.
Mol Pharmacol ; 91(4): 348-356, 2017 04.
Article in English | MEDLINE | ID: mdl-28167741

ABSTRACT

Protean agonists are of great pharmacological interest as their behavior may change in magnitude and direction depending on the constitutive activity of a receptor. Yet, this intriguing phenomenon has been poorly described and understood, due to the lack of stable experimental systems and design strategies. In this study, we overcome both limitations: First, we demonstrate that modulation of the ionic strength in a defined experimental set-up allows for analysis of G protein-coupled receptor activation in the absence and presence of a specific amount of spontaneous receptor activity using the muscarinic M2 acetylcholine receptor as a model. Second, we employ this assay system to show that a dualsteric design principle, that is, molecular probes, carrying two pharmacophores to simultaneously adopt orthosteric and allosteric topography within a G protein-coupled receptor, may represent a novel approach to achieve protean agonism. We pinpoint three molecular requirements within dualsteric compounds that elicit protean agonism at the muscarinic M2 acetylcholine receptor. Using radioligand-binding and functional assays, we posit that dynamic ligand binding may be the mechanism underlying protean agonism of dualsteric ligands. Our findings provide both new mechanistic insights into the still enigmatic phenomenon of protean agonism and a rationale for the design of such compounds for a G protein-coupled receptor.


Subject(s)
Protein Engineering , Receptors, G-Protein-Coupled/agonists , Allosteric Regulation , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Ligands , Protein Binding , Receptor, Muscarinic M2/metabolism , Receptors, G-Protein-Coupled/metabolism , Tromethamine
3.
J Biol Chem ; 291(31): 16375-89, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27298318

ABSTRACT

G protein-coupled receptors constitute the largest family of membrane receptors and modulate almost every physiological process in humans. Binding of agonists to G protein-coupled receptors induces a shift from inactive to active receptor conformations. Biophysical studies of the dynamic equilibrium of receptors suggest that a portion of receptors can remain in inactive states even in the presence of saturating concentrations of agonist and G protein mimetic. However, the molecular details of agonist-bound inactive receptors are poorly understood. Here we use the model of bitopic orthosteric/allosteric (i.e. dualsteric) agonists for muscarinic M2 receptors to demonstrate the existence and function of such inactive agonist·receptor complexes on a molecular level. Using all-atom molecular dynamics simulations, dynophores (i.e. a combination of static three-dimensional pharmacophores and molecular dynamics-based conformational sampling), ligand design, and receptor mutagenesis, we show that inactive agonist·receptor complexes can result from agonist binding to the allosteric vestibule alone, whereas the dualsteric binding mode produces active receptors. Each agonist forms a distinct ligand binding ensemble, and different agonist efficacies depend on the fraction of purely allosteric (i.e. inactive) versus dualsteric (i.e. active) binding modes. We propose that this concept may explain why agonist·receptor complexes can be inactive and that adopting multiple binding modes may be generalized also to small agonists where binding modes will be only subtly different and confined to only one binding site.


Subject(s)
Molecular Dynamics Simulation , Receptor, Muscarinic M2/agonists , Receptor, Muscarinic M2/chemistry , Allosteric Regulation , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Ligands , Receptor, Muscarinic M2/metabolism
4.
J Pharmacol Exp Ther ; 360(2): 289-299, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28082514

ABSTRACT

Drug discovery strives for selective ligands to achieve targeted modulation of tissue function. Here we introduce engineered context-sensitive agonism as a postreceptor mechanism for tissue-selective drug action through a G protein-coupled receptor. Acetylcholine M2-receptor activation is known to mediate, among other actions, potentially dangerous slowing of the heart rate. This unwanted side effect is one of the main reasons that limit clinical application of muscarinic agonists. Herein we show that dualsteric (orthosteric/allosteric) agonists induce less cardiac depression ex vivo and in vivo than conventional full agonists. Exploration of the underlying mechanism in living cells employing cellular dynamic mass redistribution identified context-sensitive agonism of these dualsteric agonists. They translate elevation of intracellular cAMP into a switch from full to partial agonism. Designed context-sensitive agonism opens an avenue toward postreceptor pharmacologic selectivity, which even works in target tissues operated by the same subtype of pharmacologic receptor.


Subject(s)
Drug Discovery , Muscarinic Agonists/pharmacology , Receptor, Muscarinic M2/agonists , Receptor, Muscarinic M2/metabolism , Allosteric Regulation/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Female , Heart/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Mice , Muscarinic Agonists/adverse effects , Signal Transduction/drug effects
5.
Nat Chem Biol ; 10(1): 18-20, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24212135

ABSTRACT

We present a new concept of partial agonism at G protein-coupled receptors. We demonstrate the coexistence of two functionally distinct populations of the muscarinic M2 receptor stabilized by one dynamic ligand, which binds in two opposite orientations. The ratio of orientations determines the cellular response. Our concept allows predicting and virtually titrating ligand efficacy, which opens unprecedented opportunities for the design of drugs with graded activation of the biological system.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Binding Sites , Ligands , Receptors, G-Protein-Coupled/metabolism
6.
Nat Chem Biol ; 8(7): 631-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22634634

ABSTRACT

Differential targeting of heterotrimeric G protein versus ß-arrestin signaling are emerging concepts in G protein-coupled receptor (GPCR) research and drug discovery, and biased engagement by GPCR ligands of either ß-arrestin or G protein pathways has been disclosed. Herein we report on a new mechanism of ligand bias to titrate the signaling specificity of a cell-surface GPCR. Using a combination of biomolecular and virtual screening, we identified the small-molecule modulator Gue1654, which inhibits Gßγ but not Gα signaling triggered upon activation of Gα(i)-ßγ by the chemoattractant receptor OXE-R in both recombinant and human primary cells. Gue1654 does not interfere nonspecifically with signaling directly at or downstream of Gßγ. This hitherto unappreciated mechanism of ligand bias at a GPCR highlights both a new paradigm for functional selectivity and a potentially new strategy to develop pathway-specific therapeutics.


Subject(s)
Benzeneacetamides/metabolism , Benzothiazoles/metabolism , Biopolymers/metabolism , GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Humans , Ligands
7.
Drug Discov Today Technol ; 10(2): e245-52, 2013.
Article in English | MEDLINE | ID: mdl-24050275

ABSTRACT

Muscarinic acetylcholine receptors belong to Class Aseven transmembrane helical receptors and serve as important drug targets in the treatment of various diseases such as chronic obstructive pulmonary disease, overactive bladder, bronchial asthma and glaucoma. Despite intensive research the discovery of experimental ligands which activate or block specific muscarinic receptor subtypes has only been successful for the M1 and M4 subtypes but remains a challenging task at the other subtypes. In recent years, ligands have been introduced which bind simultaneously to the acetylcholine binding site, that is, the orthosteric site, and to an allosteric binding site. These so-called dualsteric ligands display M2 subtype preference due to the addressing of the allosteric binding site. As proven recently, dualsteric receptor activation goes along with a pronounced signaling bias which follows clear structure­bias-relationships. Dualsteric receptor targeting might represent a common strategy to generate functional selectivity.


Subject(s)
Receptor, Muscarinic M2/metabolism , Animals , Binding Sites , Ligands , Protein Binding , Receptor, Muscarinic M2/chemistry
8.
Angew Chem Int Ed Engl ; 52(2): 508-16, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23225228

ABSTRACT

Cell-membrane-spanning G protein coupled receptors (GPCRs) belong to the most important therapeutic target structures. Endogenous transmitters bind from the outer side of the membrane to the "orthosteric" binding site either deep in the binding pocket or at the extracellular N-terminal end of the receptor protein. Exogenous modulators that utilize a different, "allosteric", binding site unveil a pathway to receptor subtype-selectivity. However, receptor activation through the orthosteric area is often more powerful. Recently there has been evidence that orthosteric/allosteric, in other words "dualsteric", hybrid compounds unite subtype selectivity and receptor activation. These "bitopic" modulators channelreceptor activation and subsequent intracellular signaling into a subset of possible routes. This concept offers access to GPCR modulators with an unprecedented receptor-subtype and signaling selectivity profile and, as a consequence, to drugs with fewer side effects.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Allosteric Regulation , Allosteric Site , Drug Design , Humans , Ligands , Models, Molecular , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
10.
Med Res Rev ; 30(3): 463-549, 2010 May.
Article in English | MEDLINE | ID: mdl-19557759

ABSTRACT

Allosteric receptor ligands bind to a recognition site that is distinct from the binding site of the endogenous messenger molecule. As a consequence, allosteric agents may attach to receptors that are already transmitter-bound. Ternary complex formation opens an avenue to qualitatively new drug actions at G protein-coupled receptors (GPCRs), in particular receptor subtype selective potentiation of endogenous transmitter action. Consequently, suitable exploitation of allosteric recognition sites as alternative molecular targets could pave the way to a drug discovery paradigm different from those aimed at mimicking or blocking the effects of endogenous (orthosteric) receptor activators. The number of allosteric ligands reported to modulate GPCR function is steadily increasing and some have already reached routine clinical use. This review aims at introducing into this fascinating field of drug discovery and at providing an overview about the achievements that have already been made. Various case examples will be discussed in the framework of GPCR classification (family A, B, and C receptors). In addition, the behavior at muscarinic receptors of hybrid derivatives incorporating both an allosteric and an orthosteric fragment in a common molecular skeleton will be illustrated.


Subject(s)
Ligands , Receptors, G-Protein-Coupled/chemistry , Allosteric Site , Animals , Binding Sites , Chemistry, Pharmaceutical/methods , Drug Design , Humans , Models, Biological , Models, Chemical , Protein Binding , Protein Conformation , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
11.
FASEB J ; 23(2): 442-50, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18842964

ABSTRACT

Selective modulation of cell function by G protein-coupled receptor (GPCR) activation is highly desirable for basic research and therapy but difficult to achieve. We present a novel strategy toward this goal using muscarinic acetylcholine receptors as a model. The five subtypes bind their physiological transmitter in the highly conserved orthosteric site within the transmembrane domains of the receptors. Orthosteric muscarinic activators have no binding selectivity and poor signaling specificity. There is a less well conserved allosteric site at the extracellular entrance of the binding pocket. To gain subtype-selective receptor activation, we synthesized two hybrids fusing a highly potent oxotremorine-like orthosteric activator with M(2)-selective bis(ammonio)alkane-type allosteric fragments. Radioligand binding in wild-type and mutant receptors supplemented by receptor docking simulations proved M(2) selective and true allosteric/orthosteric binding. G protein activation measurements using orthosteric and allosteric blockers identified the orthosteric part of the hybrid to engender receptor activation. Hybrid-induced dynamic mass redistribution in CHO-hM(2) cells disclosed pathway-specific signaling. Selective receptor activation (M(2)>M(1)>M(3)) was verified in living tissue preparations. As allosteric sites are increasingly recognized on GPCRs, the dualsteric concept of GPCR targeting represents a new avenue toward potent agonists for selective receptor and signaling pathway activation.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Cell Line , GTP-Binding Proteins/agonists , GTP-Binding Proteins/metabolism , Ligands , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Radioligand Assay , Receptors, G-Protein-Coupled/chemistry , Signal Transduction/drug effects
12.
Eur J Pharmacol ; 883: 173183, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32534072

ABSTRACT

Although agonists and antagonists of muscarinic receptors have been known for long time, there is renewed interest in compounds (such as allosteric or bitopic ligands, or biased agonists) able to differently and selectively modulate these receptors. As a continuation of our previous research, we designed a new series of dimers of the well-known cholinergic agonist carbachol. The new compounds were tested on the five cloned human muscarinic receptors (hM1-5) expressed in CHO cells by means of equilibrium binding experiments, showing a dependence of the binding affinity on the length and position of the linker connecting the two monomers. Kinetic binding studies revealed that some of the tested compounds were able to slow the rate of NMS dissociation, suggesting allosteric behavior, also supported by docking simulations. Assessment of ERK1/2 phosphorylation on hM1, hM2 and hM3 activation showed that the new compounds are endowed with muscarinic antagonist properties. At hM2 receptors, some compounds were able to stimulate GTPγS binding but not cAMP accumulation, suggesting a biased behavior. Classification, Molecular and cellular pharmacology.


Subject(s)
Carbachol/pharmacology , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Receptors, Muscarinic/drug effects , Animals , CHO Cells , Carbachol/chemistry , Carbachol/metabolism , Cricetulus , Cyclic AMP/metabolism , Dimerization , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Kinetics , Molecular Docking Simulation , Molecular Structure , Muscarinic Agonists/chemistry , Muscarinic Agonists/metabolism , Muscarinic Antagonists/chemistry , Muscarinic Antagonists/metabolism , Phosphorylation , Protein Binding , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Signal Transduction , Structure-Activity Relationship
13.
ACS Pharmacol Transl Sci ; 3(5): 859-867, 2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33073186

ABSTRACT

Allosteric coupling describes a reciprocal process whereby G-protein-coupled receptors (GPCRs) relay ligand-induced conformational changes from the extracellular binding pocket to the intracellular signaling surface. Therefore, GPCR activation is sensitive to both the type of extracellular ligand and intracellular signaling protein. We hypothesized that ligand-specific allosteric coupling may result in preferential (i.e., biased) engagement of downstream effectors. However, the structural basis underlying ligand-dependent control of this essential allosteric mechanism is poorly understood. Here, we show that two sets of extended muscarinic acetylcholine receptor M1 agonists, which only differ in linker length, progressively constrain receptor signaling. We demonstrate that stepwise shortening of their chemical linker gradually hampers binding pocket closure, resulting in divergent coupling to distinct G-protein families. Our data provide an experimental strategy for the design of ligands with selective G-protein recognition and reveal a potentially general mechanism of ligand-specific allosteric coupling.

14.
J Recept Signal Transduct Res ; 29(3-4): 140-5, 2009.
Article in English | MEDLINE | ID: mdl-19586284

ABSTRACT

Increasing attention is paid in basic science and in drug discovery to pathway selective intracellular signaling as a novel approach to achieve precise control of cell function via G protein-coupled receptors (GPCRs). With respect to signaling, GPCRs are often promiscuous in that more than one intracellular biochemical pathway is activated upon receptor stimulation by the endogenous transmitter or by exogenous drugs. We studied signaling by a novel class of GPCR activators that were designed to bind simultaneously to the orthosteric transmitter-binding site and the allosteric site of muscarinic acetylcholine receptors. An optical biosensor technique was applied to measure activation-induced dynamic mass redistribution (DMR) in CHO cells stably expressing the muscarinic receptor subtype of interest. The use of tools to modulate signaling and measuring G protein activation directly proved that DMR is a valid and comfortable approach to gain real-time insight into intracellular signaling pathway activation and to identify signaling pathway-selective drugs.


Subject(s)
Biosensing Techniques , Drug Evaluation, Preclinical/methods , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Acetylcholine/pharmacology , Animals , Atropine/pharmacology , CHO Cells , Carbachol/analogs & derivatives , Carbachol/pharmacology , Cell Line , Cholinergic Agonists/pharmacology , Cricetinae , Cricetulus , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Oxotremorine/analogs & derivatives , Oxotremorine/pharmacology , Signal Transduction/physiology
16.
Neuropharmacology ; 136(Pt C): 427-437, 2018 07 01.
Article in English | MEDLINE | ID: mdl-28935216

ABSTRACT

Muscarinic acetylcholine receptors are G protein-coupled receptors (GPCRs) which are broadly expressed in the central nervous system (CNS) and other tissues in the periphery. They emerge as important drug targets for a number of diseases including Alzheimer's disease, Parkinson's disease, and schizophrenia. Muscarinic receptors are divided into five subtypes (M1-M5) of which M1-M4 have been crystalized. All subtypes possess at least one allosteric binding site which is located in the extracellular region of the receptor on top of the ACh (i.e. orthosteric) binding site. The former can be specifically targeted by chemical compounds (mostly small molecules) and binding of such allosteric modulators affects the affinity and/or efficacy of orthosteric ligands. This allows highly specific modulation of GPCR function and, from a drug discovery point of view, may be advantageous in terms of subtype selectivity and biased signaling. There is a plethora of allosteric modulators for all five muscarinic receptor subtypes. This review presents the basic principles of allosteric modulation of GPCRs on both the molecular and structural level focusing on allosteric modulators of the muscarinic receptor family. Further we discuss dualsteric (i.e. bitopic orthosteric/allosteric) ligands emphasizing their potential in modulating muscarinic receptor dynamics and signaling. The common mechanisms of muscarinic receptor allosteric modulation have been proven to be generalizable and are at play at many, if not all GPCRs. Given this paradigmatic role of muscarinic receptors we suggest that also new developments in muscarinic allosteric modulation may also be extended to other members of the GPCR superfamily. This article is part of the Special Issue entitled 'Neuropharmacology on Muscarinic Receptors'.


Subject(s)
Central Nervous System/metabolism , Receptors, Muscarinic/metabolism , Allosteric Regulation , Animals , Humans
17.
Naunyn Schmiedebergs Arch Pharmacol ; 391(11): 1295-1299, 2018 11.
Article in English | MEDLINE | ID: mdl-30032313

ABSTRACT

Muscarinic M2 and M4 receptors resemble each other in brain distribution, function, and Gi/o protein signaling. However, there is evidence from human recombinant receptors that the M4 receptor also couples to Gs protein whereas such an alternative signaling is of minor importance for its M2 counterpart. The question arises whether this property is shared by native receptors, e.g., the murine hippocampal M2- and the striatal M4-autoreceptor. To this end, the electrically evoked tritium overflow was studied in mouse hippocampal and striatal slices pre-incubated with 3H-choline. 3H-Acetylcholine release in either region was inhibited by the potent muscarinic receptor agonist iperoxo (pIC50 8.6-8.8) in an atropine-sensitive manner (apparent pA2 8.6-8.8); iperoxo was much more potent than oxotremorine (pIC50 6.5-6.6). In hippocampal slices, N-ethylmaleimide (NEM) 32 µM, which inactivates Gi/o proteins, tended to shift the concentration-response curve of iperoxo (pIC50 8.8) to the right (pIC50 8.5) and depressed its maximum from 85 to 69%. In striatal slices, the inhibitory effect of iperoxo declined at concentrations higher than 0.1 µM, yielding a biphasic curve with a pIC50 of 8.6 for the falling part and a pEC50 of 6.4 for the rising part of the curve. The inhibitory effect of iperoxo 10 µM (47%) after NEM pre-treatment was lower by about 35% compared to the maximum (74%) obtained without NEM. In conclusion, our data, which need to be confirmed by pertussis toxin, might suggest that in the striatum, unlike the hippocampus, stimulatory Gs protein comes into play at high concentrations of a muscarinic receptor agonist.


Subject(s)
Acetylcholine/metabolism , Autoreceptors/metabolism , Corpus Striatum/drug effects , Ethylmaleimide/pharmacology , Hippocampus/drug effects , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M4/metabolism , Animals , Corpus Striatum/metabolism , Hippocampus/metabolism , Isoxazoles/pharmacology , Male , Mice , Muscarinic Agonists/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptor, Muscarinic M2/agonists , Tritium
18.
Cell Chem Biol ; 25(6): 775-786.e5, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29706593

ABSTRACT

Identification of additional uses for existing drugs is a hot topic in drug discovery and a viable alternative to de novo drug development. HAMI3379 is known as an antagonist of the cysteinyl-leukotriene CysLT2 receptor, and was initially developed to treat cardiovascular and inflammatory disorders. In our study we identified HAMI3379 as an antagonist of the orphan G protein-coupled receptor GPR17. HAMI3379 inhibits signaling of recombinant human, rat, and mouse GPR17 across various cellular backgrounds, and of endogenous GPR17 in primary rodent oligodendrocytes. GPR17 blockade by HAMI3379 enhanced maturation of primary rat and mouse oligodendrocytes, but was without effect in oligodendrocytes from GPR17 knockout mice. In human oligodendrocytes prepared from inducible pluripotent stem cells, GPR17 is expressed and its activation impaired oligodendrocyte differentiation. HAMI3379, conversely, efficiently favored human oligodendrocyte differentiation. We propose that HAMI3379 holds promise for pharmacological exploitation of orphan GPR17 to enhance regenerative strategies for the promotion of remyelination in patients.


Subject(s)
Cell Differentiation/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Drug Repositioning , Oligodendroglia/cytology , Oligodendroglia/drug effects , Phthalic Acids/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Cyclohexanecarboxylic Acids/chemistry , Dose-Response Relationship, Drug , Humans , Indoles/chemistry , Indoles/pharmacology , Mice , Mice, Knockout , Molecular Structure , Phthalic Acids/chemistry , Propionates/chemistry , Propionates/pharmacology , Rats , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
19.
J Med Chem ; 50(23): 5685-95, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17944454

ABSTRACT

Gallamine and tacrine are allosteric antagonists at muscarinic M2 acetylcholine receptors and inhibitors of acetylcholinesterase. At both acetylcholine-binding proteins, gallamine and tacrine are known to occupy two different binding sites: in M2 receptors within the allosteric binding area and in acetylcholinesterase at its catalytic and its peripheral site. To find new ligands of both targets, we designed a gallamine-tacrine dimer and several derived hybrid compounds to address the two binding sites. Their M2 receptor allosteric and acetylcholinesterase inhibitory potential was determined. The hybrid compounds revealed an allosteric potency in the low nanomolar range exceeding the allosteric potency of gallamine and tacrine by factors of 100 and 4800, respectively. Cholinesterase inhibition was augmented by hybrid formation, and all compounds exhibited IC50 values in the lower nanomolar range. Thus, gallamine-tacrine hybrid formation is a valuable approach toward high affinity ligands concurrently targeting these acetylcholine-binding proteins.


Subject(s)
Cholinesterase Inhibitors/chemical synthesis , Gallamine Triethiodide/analogs & derivatives , Gallamine Triethiodide/chemical synthesis , Receptor, Muscarinic M2/antagonists & inhibitors , Tacrine/analogs & derivatives , Tacrine/chemical synthesis , Acetylcholinesterase/chemistry , Allosteric Regulation , Animals , Binding Sites , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/pharmacology , Drug Design , Gallamine Triethiodide/pharmacology , Humans , In Vitro Techniques , Ligands , Models, Molecular , Myocardium/metabolism , Radioligand Assay , Structure-Activity Relationship , Swine , Tacrine/pharmacology
20.
ACS Chem Biol ; 12(7): 1743-1748, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28585805

ABSTRACT

G protein-coupled receptors transmit extracellular signals across cell membranes via different G protein classes and ß-arrestins. Some pathways may be therapeutically beneficial, whereas others may be detrimental under certain pathophysiological conditions. For many GPCRs, biased agonists are available, which preferentially signal through one pathway or a subset of pathways, and harnessing biased agonism could be a potential novel therapeutic strategy. However, the incomplete mechanistic understanding of biased agonism hampers rational design of biased ligands. Using the muscarinic M2 receptor as a model system, we have analyzed the relationship between ligand-dependent conformational changes as revealed in all-atom MD simulations and the activation of specific G proteins. We find that the extent of closure of the extracellular, allosteric binding site interferes with the activation of certain G proteins. Our data allow the rational design of Gi-biased agonists at the M2 receptor and delineate a simple principle which may be translated to other GPRCs.


Subject(s)
Molecular Dynamics Simulation , Receptor, Muscarinic M2/chemistry , Receptor, Muscarinic M2/metabolism , Signal Transduction , Allosteric Regulation , Binding Sites , Crystallography, X-Ray , Drug Design , Ligands , Protein Conformation , Receptor, Muscarinic M2/agonists
SELECTION OF CITATIONS
SEARCH DETAIL