Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Phys Rev Lett ; 132(20): 205102, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38829091

ABSTRACT

We present laboratory measurements showing the two-dimensional (2D) structure of energy conversion during magnetic reconnection with a guide field over the electron and ion diffusion regions, resolving the separate energy deposition on electrons and ions. We find that the electrons are energized by the parallel electric field at two locations, at the X line and around the separatrices. On the other hand, the ions are energized ballistically by the perpendicular electric field in the vicinity of the high-density separatrices. An energy balance calculation by evaluating the terms of the Poynting theorem shows that 40% of the magnetic energy is converted to particle energy, 2/3 of which is transferred to ions and 1/3 to electrons. Further analysis suggests that the energy deposited on particles manifests mostly in the form of thermal kinetic energy in the diffusion regions.

2.
Mol Pharmacol ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821630

ABSTRACT

Bipolar disorder impacts millions of patients in the United States but the mechanistic understanding of its pathophysiology and therapeutics is incomplete. Atypical antipsychotic serotonin2A (5-HT2A) receptor antagonists, such as quetiapine and olanzapine, and mood-stabilizing voltage-gated sodium channel (VGSC) blockers, such as lamotrigine, carbamazepine, and valproate, show therapeutic synergy and are often prescribed in combination for the treatment of bipolar disorder. Combination therapy is a complex task for clinicians and patients, often resulting in unexpected difficulties with dosing, drug tolerances, and decreased patient compliance. Thus, an unmet need for bipolar disorder treatment is to develop a therapeutic agent that targets both 5-HT2A receptors and VGSCs. Towards this goal, we developed a novel small molecule that simultaneously antagonizes 5-HT2A receptors and blocks sodium current. The new compound, N-(4-bromo-2,5-dimethoxyphenethyl)-6-(4-phenylbutoxy)hexan-1-amine (XOB) antagonizes 5-HT-stimulated, Gq-mediated, calcium flux at 5-HT2A receptors at low micromolar concentrations while displaying negligible affinity and activity at 5-HT1A, 5-HT2B, and 5-HT2C receptors. At similar concentrations, XOB administration inhibits sodium current in heterologous cells and results in reduced action potential (AP) firing and VGSC-related AP properties in mouse prefrontal cortex layer V pyramidal neurons. Thus, XOB represents a new, proof-of-principle tool that can be used for future preclinical investigations and therapeutic development. This polypharmacology approach of developing a single molecule to act upon two targets, which are currently independently targeted by combination therapies, may lead to safer alternatives for the treatment of psychiatric disorders that are increasingly being found to benefit from the simultaneous targeting of multiple receptors. Significance Statement We synthesized a novel small molecule (XOB) that simultaneously antagonizes two key therapeutic targets of bipolar disorder, 5-HT2A receptors and voltage-gated sodium channels (VGSCs), in heterologous cells, and inhibits the intrinsic excitability of mouse prefrontal cortex layer V pyramidal neurons in brain slices. XOB represents a valuable new proof-of-principle tool for future preclinical investigations and provides a novel molecular approach to the pharmacological treatment of complex neuropsychiatric disease, which often requires a combination of therapeutics for sufficient patient benefit.

3.
Phys Rev Lett ; 132(14): 145101, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38640378

ABSTRACT

The lower hybrid drift wave (LHDW) has been a candidate for anomalous resistivity and electron heating inside the electron diffusion region of magnetic reconnection. In a laboratory reconnection layer with a finite guide field, quasielectrostatic LHDW (ES-LHDW) propagating along the direction nearly perpendicular to the local magnetic field is excited in the electron diffusion region. ES-LHDW generates large density fluctuations (δn_{e}, about 25% of the mean density) that are correlated with fluctuations in the out-of-plane electric field (δE_{Y}, about twice larger than the mean reconnection electric field). With a small phase difference (∼30°) between two fluctuating quantities, the anomalous resistivity associated with the observed ES-LHDW is twice larger than the classical resistivity and accounts for 20% of the mean reconnection electric field. After we verify the linear relationship between δn_{e} and δE_{Y}, anomalous electron heating by LHDW is estimated by a quasilinear analysis. The estimated electron heating is about 2.6±0.3 MW/m^{3}, which exceeds the classical Ohmic heating of about 2.0±0.2 MW/m^{3}. This LHDW-driven heating is consistent with the observed trend of higher electron temperatures when the wave amplitude is larger. Presented results provide the first direct estimate of anomalous resistivity and electron heating power by LHDW, which demonstrates the importance of wave-particle interactions in magnetic reconnection.

4.
Front Mol Biosci ; 10: 1198079, 2023.
Article in English | MEDLINE | ID: mdl-37363403

ABSTRACT

Vascular endothelial cells are exposed to mechanical forces due to their presence at the interface between the vessel wall and flowing blood. The patterns of these mechanical forces (laminar vs. turbulent) regulate endothelial cell function and play an important role in determining endothelial phenotype and ultimately cardiovascular health. One of the key transcriptional mediators of the positive effects of laminar flow patterns on endothelial cell phenotype is the zinc-finger transcription factor, krüppel-like factor 2 (KLF2). Given its importance in maintaining a healthy endothelium, we sought to identify endothelial regulators of the KLF2 transcriptional program as potential new therapeutic approaches to treating cardiovascular disease. Using an approach that utilized both bioinformatics and targeted gene knockdown, we identified endothelial GPCRs capable of modulating KLF2 expression. Genetic screening using siRNAs directed to these GPCRs identified 12 potential GPCR targets that could modulate the KLF2 program, including a subset capable of regulating flow-induced KLF2 expression in primary endothelial cells. Among these targets, we describe the ability of several GPCRs (GPR116, SSTR3, GPR101, LGR4) to affect KLF2 transcriptional activation. We also identify these targets as potential validated targets for the development of novel treatments targeting the endothelium. Finally, we highlight the initiation of drug discovery efforts for LGR4 and report the identification of the first known synthetic ligands to this receptor as a proof-of-concept for pathway-directed phenotypic screening to identify novel drug targets.

5.
ACS Pharmacol Transl Sci ; 5(9): 694-709, 2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36110374

ABSTRACT

G protein-coupled receptors (GPCRs) are highly druggable targets that adopt numerous conformations. A ligand's ability to stabilize specific conformation(s) of its cognate receptor determines its efficacy or ability to produce a biological response. Identifying ligands that produce different receptor conformations and potentially discrete pharmacological effects (e.g., biased agonists, partial agonists, antagonists, allosteric modulators) is a major goal in drug discovery and necessary to develop drugs with better effectiveness and fewer side effects. Fortunately, direct measurements of ligand efficacy, via receptor conformational changes are possible with the recent development of conformational biosensors. In this review, we discuss classical efficacy models, including the two-state model, the ternary-complex model, and multistate models. We describe how nanobody-, transducer-, and receptor-based conformational biosensors detect and/or stabilize specific GPCR conformations to identify ligands with different levels of efficacy. In particular, conformational biosensors provide the potential to identify and/or characterize therapeutically desirable but often difficult to measure conformations of receptors faster and better than current methods. For drug discovery/development, several recent proof-of-principle studies have optimized conformational biosensors for high-throughput screening (HTS) platforms. However, their widespread use is limited by the fact that few sensors are reliably capable of detecting low-frequency conformations and technically demanding assay conditions. Nonetheless, conformational biosensors do help identify desirable ligands such as allosteric modulators, biased ligands, or partial agonists in a single assay, representing a distinct advantage over classical methods.

6.
J Clin Invest ; 132(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35439173

ABSTRACT

CD13, an ectoenzyme on myeloid and stromal cells, also circulates as a shed, soluble protein (sCD13) with powerful chemoattractant, angiogenic, and arthritogenic properties, which require engagement of a G protein-coupled receptor (GPCR). Here we identify the GPCR that mediates sCD13 arthritogenic actions as the bradykinin receptor B1 (B1R). Immunofluorescence and immunoblotting verified high expression of B1R in rheumatoid arthritis (RA) synovial tissue and fibroblast-like synoviocytes (FLSs), and demonstrated binding of sCD13 to B1R. Chemotaxis, and phosphorylation of Erk1/2, induced by sCD13, were inhibited by B1R antagonists. In ex vivo RA synovial tissue organ cultures, a B1R antagonist reduced secretion of inflammatory cytokines. Several mouse arthritis models, including serum transfer, antigen-induced, and local innate immune stimulation arthritis models, were attenuated in Cd13-/- and B1R-/- mice and were alleviated by B1R antagonism. These results establish a CD13/B1R axis in the pathogenesis of inflammatory arthritis and identify B1R as a compelling therapeutic target in RA and potentially other inflammatory diseases.


Subject(s)
Arthritis, Rheumatoid , CD13 Antigens/metabolism , Synoviocytes , Animals , Arthritis, Rheumatoid/pathology , Bradykinin/metabolism , Bradykinin/pharmacology , Disease Models, Animal , Fibroblasts/metabolism , Mice , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B1/metabolism , Receptors, G-Protein-Coupled/metabolism , Synovial Membrane/pathology , Synoviocytes/metabolism
7.
Front Chem ; 9: 671483, 2021.
Article in English | MEDLINE | ID: mdl-34692635

ABSTRACT

Allosteric modulators (AMs) of G-protein coupled receptors (GPCRs) are desirable drug targets because they can produce fewer on-target side effects, improved selectivity, and better biological specificity (e.g., biased signaling or probe dependence) than orthosteric drugs. An underappreciated source for identifying AM leads are peptides and proteins-many of which were evolutionarily selected as AMs-derived from endogenous protein-protein interactions (e.g., transducer/accessory proteins), intramolecular receptor contacts (e.g., pepducins or extracellular domains), endogenous peptides, and exogenous libraries (e.g., nanobodies or conotoxins). Peptides offer distinct advantages over small molecules, including high affinity, good tolerability, and good bioactivity, and specific disadvantages, including relatively poor metabolic stability and bioavailability. Peptidomimetics are molecules that combine the advantages of both peptides and small molecules by mimicking the peptide's chemical features responsible for bioactivity while improving its druggability. This review 1) discusses sources and strategies to identify peptide/peptidomimetic AMs, 2) overviews strategies to convert a peptide lead into more drug-like "peptidomimetic," and 3) critically analyzes the advantages, disadvantages, and future directions of peptidomimetic AMs. While small molecules will and should play a vital role in AM drug discovery, peptidomimetics can complement and even exceed the advantages of small molecules, depending on the target, site, lead, and associated factors.

8.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33846240

ABSTRACT

Positive allosteric modulators (PAMs) of the mu-opioid receptor (MOR) have been hypothesized as potentially safer analgesics than traditional opioid drugs. This is based on the idea that PAMs will promote the action of endogenous opioid peptides while preserving their temporal and spatial release patterns and so have an improved therapeutic index. However, this hypothesis has never been tested. Here, we show that a mu-PAM, BMS-986122, enhances the ability of the endogenous opioid Methionine-enkephalin (Met-Enk) to stimulate G protein activity in mouse brain homogenates without activity on its own and to enhance G protein activation to a greater extent than ß-arrestin recruitment in Chinese hamster ovary (CHO) cells expressing human mu-opioid receptors. Moreover, BMS-986122 increases the potency of Met-Enk to inhibit GABA release in the periaqueductal gray, an important site for antinociception. We describe in vivo experiments demonstrating that the mu-PAM produces antinociception in mouse models of acute noxious heat pain as well as inflammatory pain. These effects are blocked by MOR antagonists and are consistent with the hypothesis that in vivo mu-PAMs enhance the activity of endogenous opioid peptides. Because BMS-986122 does not bind to the orthosteric site and has no inherent agonist action at endogenously expressed levels of MOR, it produces a reduced level of morphine-like side effects of constipation, reward as measured by conditioned place preference, and respiratory depression. These data provide a rationale for the further exploration of the action and safety of mu-PAMs as an innovative approach to pain management.


Subject(s)
Allosteric Regulation/physiology , Pain/drug therapy , Receptors, Opioid, mu/metabolism , Allosteric Regulation/drug effects , Analgesia/methods , Analgesics , Analgesics, Opioid/pharmacology , Animals , CHO Cells , Cricetulus , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Morphine , Narcotic Antagonists , Pain Management/methods , Proof of Concept Study , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/drug effects
9.
Eur J Pharmacol ; 854: 1-8, 2019 Jul 05.
Article in English | MEDLINE | ID: mdl-30951717

ABSTRACT

Cannabinoid CB1 and CB2 receptors are activated by Δ9-tetrahydrocannabinol, a psychoactive component of marijuana. The cannabinoid CB1 receptor is primarily located in the brain and is responsible for the psychoactive side effects, whereas the cannabinoid CB2 receptor is located in immune cells and is an attractive target for immune-related maladies. We identify small molecules that selectively bind to the cannabinoid CB2 receptor and can be further developed into therapeutics. The affinity of three molecules, ABK5, ABK6, and ABK7, to the cannabinoid CB2 receptor was determined with radioligand competition binding. The potency of G-protein coupling was determined with GTPγS binding. The three compounds bound selectively to the cannabinoid CB2 receptor, and no binding to the cannabinoid CB1 receptor was detected up to 10 µM. Immunoblotting studies show that the amount of ERK1/2 and MEK phosphorylation increased in a Gi/o-dependent manner. Furthermore, an immune cell line (Jurkat cells) was treated with ABK5, and as a result, inhibited cell proliferation. These three compounds are novel cannabinoid CB2 receptor agonists and hold promise to be further developed to treat inflammation and the often-associated pain.


Subject(s)
Receptor, Cannabinoid, CB2/agonists , Binding, Competitive , Drug Evaluation, Preclinical , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Humans , Jurkat Cells , Ligands , Receptor, Cannabinoid, CB2/metabolism
10.
SLAS Discov ; 23(4): 375-383, 2018 04.
Article in English | MEDLINE | ID: mdl-29257918

ABSTRACT

The endocannabinoid system (ECS) plays a diverse role in human physiology ranging from the regulation of mood and appetite to immune modulation and the response to pain. Drug development that targets the cannabinoid receptors (CB1 and CB2) has been explored; however, success in the clinic has been limited by the psychoactive side effects associated with modulation of the neuronally expressed CB1 that are enriched in the CNS. CB2, however, are expressed in peripheral tissues, primarily in immune cells, and thus development of CB2-selective drugs holds the potential to modulate pain among other indications without eliciting anxiety and other undesirable side effects associated with CB1 activation. As part of a collaborative effort among industry and academic laboratories, we performed a high-throughput screen designed to discover selective agonists or positive allosteric modulators (PAMs) of CB2. Although no CB2 PAMs were identified, 167 CB2 agonists were discovered here, and further characterization of four select compounds revealed two with high selectivity for CB2 versus CB1. These results broaden drug discovery efforts aimed at the ECS and may lead to the development of novel therapies for immune modulation and pain management with improved side effect profiles.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Receptor, Cannabinoid, CB2/agonists , Animals , CHO Cells , Cricetulus , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Pain/drug therapy , Pain/metabolism , Receptor, Cannabinoid, CB1/agonists
11.
Mol Pharmacol ; 93(2): 157-167, 2018 02.
Article in English | MEDLINE | ID: mdl-29233847

ABSTRACT

Allosteric modulators of G protein-coupled receptors, including opioid receptors, have been proposed as possible therapeutic agents with enhanced selectivity. BMS-986122 is a positive allosteric modulator (PAM) of the µ-opioid receptor (µ-OR). BMS-986187 is a structurally distinct PAM for the δ-opioid receptor (δ-OR) that has been reported to exhibit 100-fold selectivity in promoting δ-OR over µ-OR agonism. We used ligand binding and second-messenger assays to show that BMS-986187 is an effective PAM at the µ-OR and at the κ-opioid receptor (κ-OR), but it is ineffective at the nociceptin receptor. The affinity of BMS-986187 for δ-ORs and κ-ORs is approximately 20- to 30-fold higher than for µ-ORs, determined using an allosteric ternary complex model. Moreover, we provide evidence, using a silent allosteric modulator as an allosteric antagonist, that BMS-986187 and BMS-986122 bind to a similar region on all three traditional opioid receptor types (µ-OR, δ-OR, and κ-OR). In contrast to the dogma surrounding allosteric modulators, the results indicate a possible conserved allosteric binding site across the opioid receptor family that can accommodate structurally diverse molecules. These findings have implications for the development of selective allosteric modulators.


Subject(s)
Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Allosteric Regulation/drug effects , Allosteric Site , Animals , CHO Cells , Cell Line, Tumor , Cricetulus , HEK293 Cells , Humans , Narcotic Antagonists/pharmacology , Radioligand Assay , Rats , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/drug effects , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/drug effects , Sodium/metabolism , Sulfones/pharmacology , Xanthones/pharmacology
12.
Assay Drug Dev Technol ; 15(7): 320-329, 2017.
Article in English | MEDLINE | ID: mdl-29120673

ABSTRACT

Homogeneous functional assays that utilize competition binding technology are widely used for determining pharmacological properties such as intrinsic activity and potency. One example is time-resolved fluorescence resonance energy transfer (TR-FRET) 3',5'-cyclic adenosine monophosphate (cAMP) assays, where labeled cAMP (tracer) and a labeled anti-cAMP antibody bind together to produce a TR-FRET signal when the two constituents are proximal to each other. This signal is disrupted when unlabeled and cellularly generated cAMP competes with the tracer cAMP for binding to the labeled antibody. It is important that the resulting assay signal, usually expressed as a TR-FRET ratio, be transformed to cAMP concentration using a cAMP standard curve. However, examples are still generated in the literature wherein investigators have used the ratiometric signal (not transformed using a standard curve) to determine values for intrinsic activity and potency of ligands. Untransformed raw data often produce reasonable looking sigmoidal concentration response curves, perhaps tempting investigators to use the raw data instead of the transformed data for applying pharmacological models. In this article, we describe the correct procedure for determining the potency and intrinsic activity of ligands that result in changes in cAMP levels using a lysate dilution assay of GLP-1 (7-36)-mediated TR-FRET cAMP accumulation and simulated data. We also highlight how the inappropriate use of raw signal data can dramatically affect interpretation of intrinsic activity and potency of ligands, and how this can adversely affect drug discovery programs. These findings apply not only to cAMP functional assays but also to other functional cellular signaling assays that utilize competition binding technologies.


Subject(s)
Binding, Competitive/physiology , Cyclic AMP/metabolism , Drug Partial Agonism , Fluorescence Resonance Energy Transfer/methods , Glucagon-Like Peptide 1/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Ligands
13.
J Med Chem ; 60(14): 6166-6190, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28635286

ABSTRACT

Agonism of the 5-HT2C receptor represents one of the most well-studied and clinically proven mechanisms for pharmacological weight reduction. Selectivity over the closely related 5-HT2A and 5-HT2B receptors is critical as their activation has been shown to lead to undesirable side effects and major safety concerns. In this communication, we report the development of a new screening paradigm that utilizes an active site mutant D134A (D3.32) 5-HT2C receptor to identify atypical agonist structures. We additionally report the discovery and optimization of a novel class of nonbasic heterocyclic amide agonists of 5-HT2C. SAR investigations around the screening hits provided a diverse set of potent agonists at 5-HT2C with high selectivity over the related 5-HT2A and 5-HT2B receptor subtypes. Further optimization through replacement of the amide with a variety of five- and six-membered heterocycles led to the identification of 6-(1-ethyl-3-(quinolin-8-yl)-1H-pyrazol-5-yl)pyridazin-3-amine (69). Oral administration of 69 to rats reduced food intake in an ad libitum feeding model, which could be completely reversed by a selective 5-HT2C antagonist.


Subject(s)
Arginine/analogs & derivatives , Flavones/chemistry , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Agonists/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Arginine/chemical synthesis , Arginine/chemistry , Arginine/pharmacology , Brain/metabolism , Caco-2 Cells , Cell Membrane Permeability , Feeding Behavior/drug effects , Flavones/chemical synthesis , Flavones/pharmacology , HEK293 Cells , Humans , Male , Membranes, Artificial , Mice, Knockout , Microsomes, Liver/metabolism , Mutation , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2B/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Serotonin 5-HT2 Receptor Agonists/chemical synthesis , Serotonin 5-HT2 Receptor Agonists/pharmacokinetics , Serotonin 5-HT2 Receptor Agonists/pharmacology , Structure-Activity Relationship
14.
Nat Commun ; 8: 14611, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28327612

ABSTRACT

Fear and emotional learning are modulated by endogenous opioids but the cellular basis for this is unknown. The intercalated cells (ITCs) gate amygdala output and thus regulate the fear response. Here we find endogenous opioids are released by synaptic stimulation to act via two distinct mechanisms within the main ITC cluster. Endogenously released opioids inhibit glutamate release through the δ-opioid receptor (DOR), an effect potentiated by a DOR-positive allosteric modulator. Postsynaptically, the opioids activate a potassium conductance through the µ-opioid receptor (MOR), suggesting for the first time that endogenously released opioids directly regulate neuronal excitability. Ultrastructural localization of endogenous ligands support these functional findings. This study demonstrates a new role for endogenously released opioids as neuromodulators engaged by synaptic activity to regulate moment-to-moment neuronal communication and excitability. These distinct actions through MOR and DOR may underlie the opposing effect of these receptor systems on anxiety and fear.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Fear/physiology , Interneurons/metabolism , Opioid Peptides/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Glutamic Acid/metabolism , In Vitro Techniques , Male , Patch-Clamp Techniques , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid/metabolism , Synapses/metabolism
15.
Bioorg Med Chem ; 25(2): 496-513, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27914948

ABSTRACT

Triazolopyridine ethers with mGlu2 positive allosteric modulator (PAM) activity are disclosed. The synthesis, in vitro activity, and metabolic stability data for a series of analogs is provided. The effort resulted in the discovery of a potent, selective, and brain penetrant lead molecule BMT-133218 ((+)-7m). After oral administration at 10mg/kg, BMT-133218 demonstrated full reversal of PCP-stimulated locomotor activity and prevented MK-801-induced working memory deficits in separate mouse models. Also, reversal of impairments in executive function were observed in rat set-shifting studies at 3 and 10mg/kg (p.o.). Extensive plasma protein binding as the result of high lipophilicity likely limited activity at lower doses. Optimized triazolopyridine ethers offer utility as mGlu2 PAMs for the treatment of schizophrenia and merit further preclinical investigation.


Subject(s)
Ethers/pharmacology , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Schizophrenia/drug therapy , Triazoles/pharmacology , Administration, Oral , Allosteric Regulation/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Ethers/administration & dosage , Ethers/chemistry , Haplorhini , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/metabolism , Structure-Activity Relationship , Triazoles/administration & dosage , Triazoles/chemistry
16.
Curr Protoc Pharmacol ; 74: 9.20.1-9.20.23, 2016 Sep 16.
Article in English | MEDLINE | ID: mdl-27636112

ABSTRACT

Allosteric ligands modulate the activity of receptor targets by binding to sites that are distinct from the orthosteric (native agonist) binding site. Allosteric modulators have potential therapeutic advantages over orthosteric agonists and antagonists, including improved selectivity, and maintenance of the spatial and temporal fidelity of native signaling patterns. The identification of allosteric ligands presents unique challenges because of the requirement for screening in the presence of an orthosteric agonist, the small signal window that is produced by many allosteric modulators, the proclivity of allosteric modulators to exhibit activity switching within a chemotype (e.g., one compound may be a positive allosteric modulator while a close analog is a negative allosteric modulator), and probe dependence (differential interactions with different orthosteric agonists). Described in this unit are emerging strategies for the identification of allosteric ligands by high-throughput screening (HTS), including the use of multiple-add/multiple-read HTS assays and tool molecule-based screening formats. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Allosteric Regulation , High-Throughput Screening Assays/methods , Allosteric Site , Animals , Humans , Ligands , Signal Transduction
17.
ACS Chem Biol ; 11(5): 1220-9, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26841170

ABSTRACT

Available crystal structures of opioid receptors provide a high-resolution picture of ligand binding at the primary ("orthosteric") site, that is, the site targeted by endogenous ligands. Recently, positive allosteric modulators of opioid receptors have also been discovered, but their modes of binding and action remain unknown. Here, we use a metadynamics-based strategy to efficiently sample the binding process of a recently discovered positive allosteric modulator of the δ-opioid receptor, BMS-986187, in the presence of the orthosteric agonist SNC-80, and with the receptor embedded in an explicit lipid-water environment. The dynamics of BMS-986187 were enhanced by biasing the potential acting on the ligand-receptor distance and ligand-receptor interaction contacts. Representative lowest-energy structures from the reconstructed free-energy landscape revealed two alternative ligand binding poses at an allosteric site delineated by transmembrane (TM) helices TM1, TM2, and TM7, with some participation of TM6. Mutations of amino acid residues at these proposed allosteric sites were found to either affect the binding of BMS-986187 or its ability to modulate the affinity and/or efficacy of SNC-80. Taken together, these combined experimental and computational studies provide the first atomic-level insight into the modulation of opioid receptor binding and signaling by allosteric modulators.


Subject(s)
Allosteric Regulation/drug effects , Benzamides/pharmacology , Piperazines/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Xanthones/pharmacology , Allosteric Site/drug effects , Humans , Molecular Docking Simulation , Protein Binding , Receptors, Opioid, delta/chemistry , Thermodynamics
18.
J Pharmacol Exp Ther ; 356(2): 293-304, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26582730

ABSTRACT

The muscarinic acetylcholine receptor subtype 1 (M1) receptors play an important role in cognition and memory, and are considered to be attractive targets for the development of novel medications to treat cognitive impairments seen in schizophrenia and Alzheimer's disease. Indeed, the M1 agonist xanomeline has been shown to produce beneficial cognitive effects in both Alzheimer's disease and schizophrenia patients. Unfortunately, the therapeutic utility of xanomeline was limited by cholinergic side effects (sweating, salivation, gastrointestinal distress), which are believed to result from nonselective activation of other muscarinic receptor subtypes such as M2 and M3. Therefore, drug discovery efforts targeting the M1 receptor have focused on the discovery of compounds with improved selectivity profiles. Recently, allosteric M1 receptor ligands have been described, which exhibit excellent selectivity for M1 over other muscarinic receptor subtypes. In the current study, the following three compounds with mixed agonist/positive allosteric modulator activities that are highly functionally selective for the M1 receptor were tested in rats, dogs, and cynomologous monkeys: (3-((1S,2S)-2-hydrocyclohexyl)-6-((6-(1-methyl-1H-pyrazol-4-yl)pyridin-3-yl)methyl)benzo[h]quinazolin-4(3H)-one; 1-((4-cyano-4-(pyridin-2-yl)piperidin-1-yl)methyl)-4-oxo-4H-quinolizine-3-carboxylic acid; and (R)-ethyl 3-(2-methylbenzamido)-[1,4'-bipiperidine]-1'-carboxylate). Despite their selectivity for the M1 receptor, all three compounds elicited cholinergic side effects such as salivation, diarrhea, and emesis. These effects could not be explained by activity at other muscarinic receptor subtypes, or by activity at other receptors tested. Together, these results suggest that activation of M1 receptors alone is sufficient to produce unwanted cholinergic side effects such as those seen with xanomeline. This has important implications for the development of M1 receptor-targeted therapeutics since it suggests that dose-limiting cholinergic side effects still reside in M1 receptor selective activators.


Subject(s)
Muscarinic Agonists/metabolism , Muscarinic Agonists/pharmacology , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Dogs , Dose-Response Relationship, Drug , Humans , Macaca fascicularis , Male , Mice , Rats , Rats, Sprague-Dawley
19.
J Chem Inf Model ; 55(9): 1836-43, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26347990

ABSTRACT

With the hope of discovering effective analgesics with fewer side effects, attention has recently shifted to allosteric modulators of the opioid receptors. In the past two years, the first chemotypes of positive or silent allosteric modulators (PAMs or SAMs, respectively) of µ- and δ-opioid receptor types have been reported in the literature. During a structure-guided lead optimization campaign with µ-PAMs BMS-986121 and BMS-986122 as starting compounds, we discovered a new chemotype that was confirmed to display µ-PAM or µ-SAM activity depending on the specific substitutions as assessed by endomorphin-1-stimulated ß-arrestin2 recruitment assays in Chinese Hamster Ovary (CHO)-µ PathHunter cells. The most active µ-PAM of this series was analyzed further in competition binding and G-protein activation assays to understand its effects on ligand binding and to investigate the nature of its probe dependence.


Subject(s)
Drug Discovery , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry , Allosteric Regulation , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Delivery Systems , Ligands , Models, Biological , Molecular Structure , Protein Binding/drug effects , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology
20.
Methods Mol Biol ; 1335: 223-40, 2015.
Article in English | MEDLINE | ID: mdl-26260604

ABSTRACT

The continued evolution of our understanding of G protein-coupled receptor (GPCR) signaling has revealed new opportunities for drug discovery. Specifically, biased agonism at GPCRs and allosteric modulation of GPCRs both represent emerging areas of GPCR biology that hold promise for the development of novel GPCR-targeted therapeutics that may provide greater therapeutic efficacy and/or improved side-effect profiles. To obtain initial chemical leads, high-throughput screening (HTS) of a large compound library for the desired activity is often deployed during the early stages of a discovery program. The identification of allosteric modulators, in particular, poses significant challenges for HTS. We describe several HTS protocols designed for the identification of GPCR ligands, with a particular focus on the identification of allosteric modulators.


Subject(s)
Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Allosteric Regulation/drug effects , Calcium/metabolism , Cell Line , Receptors, G-Protein-Coupled/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...