Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
J Clin Invest ; 133(19)2023 10 02.
Article En | MEDLINE | ID: mdl-37561584

Biological aging can be described as accumulative, prolonged metabolic stress and is the major risk factor for cognitive decline and Alzheimer's disease (AD). Recently, we identified and described a quinone reductase 2 (QR2) pathway in the brain, in which QR2 acts as a removable memory constraint and metabolic buffer within neurons. QR2 becomes overexpressed with age, and it is possibly a novel contributing factor to age-related metabolic stress and cognitive deficit. We found that, in human cells, genetic removal of QR2 produced a shift in the proteome opposing that found in AD brains while simultaneously reducing oxidative stress. We therefore created highly specific QR2 inhibitors (QR2is) to enable evaluation of chronic QR2 inhibition as a means to reduce biological age-related metabolic stress and cognitive decline. QR2is replicated results obtained by genetic removal of QR2, while local QR2i microinjection improved hippocampal and cortical-dependent learning in rats and mice. Continuous consumption of QR2is in drinking water improved cognition and reduced pathology in the brains of AD-model mice (5xFAD), with a noticeable between-sex effect on treatment duration. These results demonstrate the importance of QR2 activity and pathway function in the healthy and neurodegenerative brain and what we believe to be the great therapeutic potential of QR2is as first-in-class drugs.


Alzheimer Disease , Quinone Reductases , Animals , Humans , Mice , Rats , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Hippocampus/metabolism , Oxidative Stress , Quinone Reductases/antagonists & inhibitors , Quinone Reductases/genetics , Quinone Reductases/metabolism , Stress, Physiological
2.
Nat Commun ; 11(1): 6038, 2020 11 27.
Article En | MEDLINE | ID: mdl-33247131

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) is threatening public health as it spreads worldwide across diverse environments. Its genetic hallmark, the mecA gene, confers resistance to many ß-lactam antibiotics. Here, we show that, in addition, mecA provides a broad selective advantage across diverse chemical environments. Competing fluorescently labelled wild-type and mecA-deleted CA-MRSA USA400 strains across ~57,000 compounds supplemented with subinhibitory levels of the ß-lactam drug cefoxitin, we find that mecA provides a widespread advantage across ß-lactam and non ß-lactam antibiotics, non-antibiotic drugs and even diverse natural and synthetic compounds. This advantage depends on the presence of cefoxitin and is strongly associated with the compounds' physicochemical properties, suggesting that it may be mediated by differential compounds permeability into the cell. Indeed, mecA protects the bacteria against increased cell-envelope permeability under subinhibitory cefoxitin treatment. Our findings suggest that CA-MRSA success might be driven by a cell-envelope mediated selective advantage across diverse chemical compounds.


Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Penicillin-Binding Proteins/metabolism , Cefoxitin/pharmacology , Cell Wall/drug effects , Cell Wall/metabolism , Logistic Models , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Multivariate Analysis , Permeability
3.
Assay Drug Dev Technol ; 16(7): 384-396, 2018 10.
Article En | MEDLINE | ID: mdl-30251873

G-protein-coupled receptors (GPCRs) have varying and diverse physiological roles, transmitting signals from a range of stimuli, including light, chemicals, peptides, and mechanical forces. More than 130 GPCRs are orphan receptors (i.e., their endogenous ligands are unknown), representing a large untapped reservoir of potential therapeutic targets for pharmaceutical intervention in a variety of diseases. Current deorphanization approaches are slow, laborious, and usually require some in-depth knowledge about the receptor pharmacology. In this study we describe a cell-based assay to identify small molecule probes of orphan receptors that requires no a priori knowledge of receptor pharmacology. Built upon the concept of pharmacochaperones, where cell-permeable small molecules facilitate the trafficking of mutant receptors to the plasma membrane, the simple and robust technology is readily accessible by most laboratories and is amenable to high-throughput screening. The assay consists of a target harboring a synthetic point mutation that causes retention of the target in the endoplasmic reticulum. Coupled with a beta-galactosidase enzyme-fragment complementation reporter system, the assay identifies compounds that act as pharmacochaperones causing forward trafficking of the mutant GPCR. The assay can identify compounds with varying mechanisms of action including agonists and antagonists. A universal positive control compound circumvents the need for a target-specific ligand. The veracity of the approach is demonstrated using the beta-2-adrenergic receptor. Together with other existing assay technologies to validate the signaling pathways and the specificity of ligands identified, this pharmacochaperone-based approach can accelerate the identification of ligands for these potentially therapeutically useful receptors.


High-Throughput Screening Assays/methods , Molecular Probes/analysis , Molecular Probes/chemistry , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Small Molecule Libraries/analysis , Small Molecule Libraries/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Ligands , Molecular Probes/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured
4.
SLAS Discov ; 22(7): 867-878, 2017 08.
Article En | MEDLINE | ID: mdl-28314120

Biased agonists are defined by their ability to selectively activate distinct signaling pathways of a receptor, and they hold enormous promise for the development of novel drugs that specifically elicit only the desired therapeutic response and avoid potential adverse effects. Unfortunately, most high-throughput screening (HTS) assays are designed to detect signaling of G protein-coupled receptors (GPCRs) downstream of either G protein or ß-arrestin-mediated signaling but not both. A comprehensive drug discovery program seeking biased agonists must employ assays that report on the activity of each compound at multiple discrete pathways, particularly for HTS campaigns. Here, we report a systematic approach to the identification of biased agonists of human apelin receptor (APJ). We synthesized 448 modified versions of apelin and screened them against a cascade of cell-based assays, including intracellular cAMP and ß-arrestin recruitment to APJ, simultaneously. The screen yielded potent and highly selective APJ agonists. Representative hits displaying preferential signaling via either G-protein or ß-arrestin were subjected to a battery of confirmation assays. These biased agonists will be useful as tools to probe the function and pharmacology of APJ and provide proof of concept of our systematic approach to the discovery of biased ligands. This approach is likely universally applicable to the search for biased agonists of GPCRs.


Apelin Receptors/agonists , Apelin Receptors/metabolism , Animals , CHO Cells , Cricetulus , GTP-Binding Proteins/metabolism , High-Throughput Screening Assays/methods , Humans , Ligands , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , beta-Arrestins/metabolism
5.
Biochemistry ; 56(3): 473-486, 2017 01 24.
Article En | MEDLINE | ID: mdl-28005346

GPR55 is a newly deorphanized class A G-protein-coupled receptor that has been implicated in inflammatory pain, neuropathic pain, metabolic disorder, bone development, and cancer. Few potent GPR55 ligands have been identified to date. This is largely due to an absence of information about salient features of GPR55, such as residues important for signaling and residues implicated in the GPR55 signaling cascade. The goal of this work was to identify residues that are key for the signaling of the GPR55 endogenous ligand, l-α-lysophosphatidylinositol (LPI), as well as the signaling of the GPR55 agonist, ML184 {CID 2440433, 3-[4-(2,3-dimethylphenyl)piperazine-1-carbonyl]-N,N-dimethyl-4-pyrrolidin-1-ylbenzenesulfonamide}. Serum response element (SRE) and serum response factor (SRF) luciferase assays were used as readouts for studying LPI and ML184 signaling at the GPR55 mutants. A GPR55 R* model based on the recent δ-opioid receptor (DOR) crystal structure was used to interpret the resultant mutation data. Two residues were found to be crucial for agonist signaling at GPR55, K2.60 and E3.29, suggesting that these residues form the primary interaction site for ML184 and LPI at GPR55. Y3.32F, H(170)F, and F6.55A/L mutation results suggested that these residues are part of the orthosteric binding site for ML184, while Y3.32F and H(170)F mutation results suggest that these two residues are part of the LPI binding pocket. Y3.32L, M3.36A, and F6.48A mutation results suggest the importance of a Y3.32/M3.36/F6.48 cluster in the GPR55 signaling cascade. C(10)A and C(260)A mutations suggest that these residues form a second disulfide bridge in the extracellular domain of GPR55, occluding ligand extracellular entry in the TMH1-TMH7 region of GPR55. Taken together, these results provide the first set of discrete information about GPR55 residues important for LPI and ML184 signaling and for GPR55 activation. This information should aid in the rational design of next-generation GPR55 ligands and the creation of the first high-affinity GPR55 radioligand, a tool that is sorely needed in the field.


Lysophospholipids/chemistry , Piperazines/chemistry , Pyrrolidines/chemistry , Receptors, G-Protein-Coupled/chemistry , Recombinant Fusion Proteins/chemistry , Serum Response Element , Amino Acid Motifs , Binding Sites , Crystallography, X-Ray , Gene Expression , HEK293 Cells , Humans , Kinetics , Ligands , Lysophospholipids/pharmacology , Molecular Docking Simulation , Mutation , Piperazines/pharmacology , Protein Binding , Pyrrolidines/pharmacology , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serum Response Factor/chemistry , Serum Response Factor/genetics , Serum Response Factor/metabolism , Signal Transduction , Glycine max , Structural Homology, Protein , Thermodynamics
6.
Bioorg Med Chem Lett ; 26(7): 1827-1830, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-26916440

A series of 1,3,4-oxadiazol-2-ones was synthesized and tested for activity as antagonists at GPR55 in cellular beta-arrestin redistribution assays. The synthesis was designed to be modular in nature so that a sufficient number of analogues could be rapidly accessed to explore initial structure-activity relationships. The design of analogues was guided by the docking of potential compounds into a model of the inactive form of GPR55. The results of the assays were used to learn more about the binding pocket of GPR55. With this oxadiazolone scaffold, it was determined that modification of the aryl group adjacent to the oxadiazolone ring was often detrimental and that the distal cyclopropane was beneficial for activity. These results will guide further exploration of this receptor.


Drug Design , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Arrestins/metabolism , CHO Cells , Cricetulus , Humans , Molecular Docking Simulation , Oxadiazoles/chemical synthesis , Piperidines/chemical synthesis , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship , beta-Arrestins
7.
J Med Chem ; 57(21): 8777-91, 2014 Nov 13.
Article En | MEDLINE | ID: mdl-25272206

We have recently identified 1,8-naphthyridin-2(1H)-one-3-carboxamide as a new scaffold very suitable for the development of new CB2 receptor potent and selective ligands. In this paper we describe a number of additional derivatives in which the same central scaffold has been variously functionalized in position 1 or 6. All new compounds showed high selectivity and affinity in the nanomolar range for the CB2 receptor. Furthermore, we found that their functional activity is controlled by the presence of the substituents at position C-6 of the naphthyridine scaffold. In fact, the introduction of substituents in this position determined a functionality switch from agonist to antagonists/inverse agonists. Finally, docking studies showed that the difference between the pharmacology of these ligands may be in the ability/inability to block the Toggle Switch W6.48(258) (χ1 g+ → trans) transition.


Naphthyridines/chemical synthesis , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Cell Line, Tumor , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Naphthyridines/chemistry , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/chemistry , Structure-Activity Relationship
8.
Br J Pharmacol ; 171(16): 3908-17, 2014 Aug.
Article En | MEDLINE | ID: mdl-24762058

BACKGROUND AND PURPOSE: GPR18 is a candidate cannabinoid receptor, but its classification as such is controversial. The rationale of the study presented herein was to consider the effects of N-arachidonoyl glycine (NAGly) and cannabinoids via differential G-protein coupled pathways, in addition to ß-arrestin signalling. Cellular localization of GPR18 receptors was also examined. EXPERIMENTAL APPROACH: Calcium mobilization and ERK1/2 phosphorylation were quantified in a cell line stably expressing GPR18 (HEK293/GPR18 cells). In addition, using the DiscoveRx PathHunter CHO-K1 GPR18 ß-arrestin cell line, recruitment of ß-arrestin was quantified. KEY RESULTS: Concentration-dependent increases in intracellular calcium and ERK1/2 phosphorylation were observed in the presence of NAGly, abnormal cannabidiol (AbnCBD), O-1602, O-1918 and Δ(9)-tetrahydrocannabinol (Δ(9)-THC) in HEK293/GPR18 cells. The initial rise in intracellular calcium in the presence of NAGly, O1918 and THC was blocked by either Gα(q) or Gα(i/o) inhibition. The ERK1/2 phosphorylation was inhibited by Pertussis toxin and N-arachidonoyl-L-serine (NARAS). Recruitment of ß-arrestin in the PathHunter CHO-K1 GPR18 cell line revealed a differential pattern of GPR18 activation; of all the ligands tested, only Δ(9)-THC produced a concentration-dependent response. The localization of GPR18 receptors within the HEK293/GPR18 cells is both intracellular, and on the plasma membrane. CONCLUSIONS AND IMPLICATIONS: These findings suggest that GPR18 activation involves several signal transduction pathways indicative of biased agonism, thereby providing a plausible explanation for the apparent discrepancies in GPR18 activation found in the literature. Additionally, the results presented herein provide further evidence for GPR18 as a candidate cannabinoid receptor.


Cannabinoid Receptor Agonists/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Arachidonic Acids/pharmacology , Arrestins/metabolism , CHO Cells , Calcium/metabolism , Cannabidiol/analogs & derivatives , Cricetulus , Cyclohexanes/pharmacology , Dronabinol/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, G-Protein-Coupled/metabolism , Resorcinols/pharmacology , Signal Transduction , beta-Arrestins
9.
Biochemistry ; 52(52): 9456-69, 2013 Dec 31.
Article En | MEDLINE | ID: mdl-24274581

GPR55 is a class A G protein-coupled receptor (GPCR) that has been implicated in inflammatory pain, neuropathic pain, metabolic disorder, bone development, and cancer. Initially deorphanized as a cannabinoid receptor, GPR55 has been shown to be activated by non-cannabinoid ligands such as l-α-lysophosphatidylinositol (LPI). While there is a growing body of evidence of physiological and pathophysiological roles for GPR55, the paucity of specific antagonists has limited its study. In collaboration with the Molecular Libraries Probe Production Centers Network initiative, we identified a series of GPR55 antagonists using a ß-arrestin, high-throughput, high-content screen of ~300000 compounds. This screen yielded novel, GPR55 antagonist chemotypes with IC50 values in the range of 0.16-2.72 µM [Heynen-Genel, S., et al. (2010) Screening for Selective Ligands for GPR55: Antagonists (ML191, ML192, ML193) (Bookshelf ID NBK66153; PMID entry 22091481)]. Importantly, many of the GPR55 antagonists were completely selective, with no agonism or antagonism against GPR35, CB1, or CB2 up to 20 µM. Using a model of the GPR55 inactive state, we studied the binding of an antagonist series that emerged from this screen. These studies suggest that GPR55 antagonists possess a head region that occupies a horizontal binding pocket extending into the extracellular loop region, a central ligand portion that fits vertically in the receptor binding pocket and terminates with a pendant aromatic or heterocyclic ring that juts out. Both the region that extends extracellularly and the pendant ring are features associated with antagonism. Taken together, our results provide a set of design rules for the development of second-generation GPR55 selective antagonists.


Drug Evaluation, Preclinical , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Binding Sites , Humans , Inhibitory Concentration 50 , Ligands , Models, Molecular , Protein Binding , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/metabolism
10.
J Neuroimmune Pharmacol ; 7(4): 856-65, 2012 Dec.
Article En | MEDLINE | ID: mdl-22454039

The role of cannabinoid receptors in inflammation has been the topic of many research endeavors. Despite this effort, to date the involvement of the endocannabinoid system (ECS) in inflammation remains obscure. The ambiguity of cannabinoid involvement may be explained by the existence of cannabinoid receptors, other than CB(1) and CB(2), or a consequence of interaction of endocannabinoids with other signaling systems. GPR55 has been proposed to be a cannabinoid receptor; however the interaction of the endocannabinoid system with GPR55 remains elusive. Consequently this study set about to examine the effects of the endocannabinoids, anandamide (AEA) and virodhamine, on GPR55 mediated signaling. Specifically, we assessed changes in ß-arrestin2 (ßarr2) distribution and GPR55 receptor internalization following activation by lysophosphatidylinositol (LPI), the synthetic cannabinoid ligand SR141716A, and new selective synthetic GPR55 agonists. Data obtained from the experiments presented herein demonstrate that AEA and virodhamine modulate agonist-mediated recruitment of ßarr2. AEA and virodhamine act as partial agonists; enhancing the agonist effect at low concentrations and inhibiting it at high concentrations. Furthermore, both virodhamine and AEA significantly attenuated agonist-induced internalization of GPR55. These effects are attributed to the expression of GPR55, and not CB(1) and CB(2) receptors, as we have established negligible expression of CB(1) and CB(2) in these GPR55-transfected U2OS cells. The identification of select endocannabinoids as GPR55 modulators will aide in elucidating the function of GPR55 in the ECS.


Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Cannabinoids/pharmacology , Endocannabinoids/pharmacology , Polyunsaturated Alkamides/pharmacology , Receptors, G-Protein-Coupled/drug effects , Animals , Arrestins/metabolism , CHO Cells , Cell Line , Cell Survival/drug effects , Cricetinae , Cricetulus , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Microscopy, Confocal , RNA/genetics , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/antagonists & inhibitors , beta-Arrestins
11.
Biochemistry ; 50(25): 5633-47, 2011 Jun 28.
Article En | MEDLINE | ID: mdl-21534610

Marijuana is the most widely abused illegal drug, and its spectrum of effects suggests that several receptors are responsible for the activity. Two cannabinoid receptor subtypes, CB1 and CB2, have been identified, but the complex pharmacological properties of exogenous cannabinoids and endocannabinoids are not fully explained by their signaling. The orphan receptor GPR55 binds a subset of CB1 and CB2 ligands and has been proposed as a cannabinoid receptor. This designation, however, is controversial as a result of recent studies in which lysophosphatidylinositol (LPI) was identified as a GPR55 agonist. Defining a biological role for GPR55 requires GPR55 selective ligands that have been unavailable. From a ß-arrestin, high-throughput, high-content screen of 300000 compounds run in collaboration with the Molecular Libraries Probe Production Centers Network initiative (PubChem AID1965), we identified potent GPR55 selective agonists. By modeling of the GPR55 activated state, we compared the GPR55 binding conformations of three of the novel agonists obtained from the screen, CID1792197, CID1172084, and CID2440433 (PubChem Compound IDs), with that of LPI. Our modeling indicates the molecular shapes and electrostatic potential distributions of these agonists mimic those of LPI; the GPR55 binding site accommodates ligands that have inverted-L or T shapes with long, thin profiles that can fit vertically deep in the receptor binding pocket while their broad head regions occupy a horizontal binding pocket near the GPR55 extracellular loops. Our results will allow the optimization and design of second-generation GPR55 ligands and provide a means for distinguishing GPR55 selective ligands from those interacting with cannabinoid receptors.


Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Amino Acid Sequence , Arrestins/chemistry , Arrestins/genetics , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Design , HEK293 Cells , Humans , Ligands , Models, Chemical , Molecular Sequence Data , Protein Structure, Secondary , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/metabolism , Static Electricity , beta-Arrestins
12.
Mol Pharmacol ; 78(4): 560-8, 2010 Oct.
Article En | MEDLINE | ID: mdl-20826425

Known agonists of the orphan receptor GPR35 are kynurenic acid, zaprinast, 5-nitro-2-(3-phenylproplyamino) benzoic acid, and lysophosphatidic acids. Their relatively low affinities for GPR35 and prominent off-target effects at other pathways, however, diminish their utility for understanding GPR35 signaling and for identifying potential therapeutic uses of GPR35. In a screen of the Prestwick Library of drugs and drug-like compounds, we have found that pamoic acid is a potent GPR35 agonist. Pamoic acid is considered by the Food and Drug Administration as an inactive compound that enables long-acting formulations of numerous drugs, such as the antihelminthics oxantel pamoate and pyrantel pamoate; the psychoactive compounds hydroxyzine pamoate (Vistaril) and imipramine pamoate (Tofranil-PM); and the peptide hormones triptorelin pamoate (Trelstar) and octreotide pamoate (OncoLar). We have found that pamoic acid induces a G(i/o)-linked, GPR35-mediated increase in the phosphorylation of extracellular signal-regulated kinase 1/2, recruitment of ß-arrestin2 to GPR35, and internalization of GPR35. In mice, it attenuates visceral pain perception, indicating an antinociceptive effect, possibly through GPR35 receptors. We have also identified in collaboration with the Sanford-Burnham Institute Molecular Libraries Probe Production Center new classes of GPR35 antagonist compounds, including the nanomolar potency antagonist methyl-5-[(tert-butylcarbamothioylhydrazinylidene)methyl]-1-(2,4-difluorophenyl)pyrazole-4-carboxylate (CID2745687). Pamoic acid and potent antagonists such as CID2745687 present novel opportunities for expanding the chemical space of GPR35, elucidating GPR35 pharmacology, and stimulating GPR35-associated drug development. Our results indicate that the unexpected biological functions of pamoic acid may yield potential new uses for a common drug constituent.


Analgesics/administration & dosage , Arrestins/metabolism , Drug Delivery Systems/methods , Extracellular Signal-Regulated MAP Kinases/metabolism , Naphthols/administration & dosage , Receptors, G-Protein-Coupled/metabolism , Animals , Arrestins/agonists , Cell Line , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , Male , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Receptors, G-Protein-Coupled/agonists , Renilla , beta-Arrestins
13.
J Biol Chem ; 285(34): 26097-106, 2010 Aug 20.
Article En | MEDLINE | ID: mdl-20522546

A role for Zn(2+) in accelerating wound healing is established, yet, the signaling pathways linking Zn(2+) to tissue repair are not well known. We show that in the human HaCaT keratinocytes extracellular Zn(2+) induces a metabotropic Ca(2+) response that is abolished by silencing the expression of the G-protein-coupled receptor GPR39, suggesting that this Zn(2+)-sensing receptor, ZnR, is mediating the response. Keratinocytic-ZnR signaling is highly selective for Zn(2+) and can be triggered by nanomolar concentrations of this ion. Interestingly, Zn(2+) was also released following cellular injury, as monitored by a specific non-permeable fluorescent Zn(2+) probe, ZnAF-2. Chelation of Zn(2+) and scavenging of ATP from conditioned medium, collected from injured epithelial cultures, was sufficient to eliminate the metabotropic Ca(2+) signaling. The signaling triggered by Zn(2+), via ZnR, or by ATP further activated MAP kinase and induced up-regulation of the sodium/proton exchanger NHE1 activity. Finally, activation of ZnR/GPR39 signaling or application of ATP enhanced keratinocytes scratch closure in an in vitro model. Thus our results indicate that extracellular Zn(2+), which is either applied or released following injury, activates ZnR/GPR39 to promote signaling leading to epithelial repair.


Keratinocytes/pathology , Receptors, G-Protein-Coupled/physiology , Wound Healing , Zinc/metabolism , Adenosine Triphosphate , Calcium Signaling , Cell Line , Epithelial Cells , Humans , Keratinocytes/physiology , Molecular Probes , Pyridines
14.
Pharmacol Ther ; 126(3): 301-13, 2010 Jun.
Article En | MEDLINE | ID: mdl-20298715

GPR55 has recently attracted much attention as another member of the cannabinoid family, potentially explaining physiological effects that are non-CB1/CB2 mediated. However, the data gathered so far are conflicting with respect to its pharmacology. We review the primary literature to date on GPR55, describing its discovery, structure, pharmacology and potential physiological functions. The CB1 receptor antagonist/inverse agonist AM251 has been shown to be a GPR55 agonist in all reports in which it was evaluated, as has the lysophospholipid, lysophosphatidylinositol (LPI). Whether GPR55 responds to the endocannabinoid ligands anandamide and 2-arachidonylglycerol and the phytocannabinoids, delta-9-tetrahydrocannabidiol and cannabidiol, is cell type and tissue-dependent. GPR55 has been shown to utilize G(q), G(12), or G(13) for signal transduction; RhoA and phospholipase C are activated. Experiments with mice in which GPR55 has been inactivated reveal a role for this receptor in neuropathic and inflammatory pain as well as in bone physiology. Thus delineating the pharmacology of this receptor and the discovery of selective agonists and antagonists merits further study and could lead to new therapeutics.


Cannabinoid Receptor Agonists , Cannabinoid Receptor Antagonists , Cannabinoids/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Humans , Receptors, Cannabinoid/physiology , Receptors, G-Protein-Coupled/physiology
15.
J Biol Chem ; 284(43): 29817-27, 2009 Oct 23.
Article En | MEDLINE | ID: mdl-19723626

The cannabinoid receptor 1 (CB(1)) and CB(2) cannabinoid receptors, associated with drugs of abuse, may provide a means to treat pain, mood, and addiction disorders affecting widespread segments of society. Whether the orphan G-protein coupled receptor GPR55 is also a cannabinoid receptor remains unclear as a result of conflicting pharmacological studies. GPR55 has been reported to be activated by exogenous and endogenous cannabinoid compounds but surprisingly also by the endogenous non-cannabinoid mediator lysophosphatidylinositol (LPI). We examined the effects of a representative panel of cannabinoid ligands and LPI on GPR55 using a beta-arrestin-green fluorescent protein biosensor as a direct readout of agonist-mediated receptor activation. Our data demonstrate that AM251 and SR141716A (rimonabant), which are cannabinoid antagonists, and the lipid LPI, which is not a cannabinoid receptor ligand, are GPR55 agonists. They possess comparable efficacy in inducing beta-arrestin trafficking and, moreover, activate the G-protein-dependent signaling of protein kinase CbetaII. Conversely, the potent synthetic cannabinoid agonist CP55,940 acts as a GPR55 antagonist/partial agonist. CP55,940 blocks GPR55 internalization, the formation of beta-arrestin GPR55 complexes, and the phosphorylation of ERK1/2; CP55,940 produces only a slight amount of protein kinase CbetaII membrane recruitment but does not stimulate membrane remodeling like LPI, AM251, or rimonabant. Our studies provide a paradigm for measuring the responsiveness of GPR55 to a variety of ligand scaffolds comprising cannabinoid and novel compounds and suggest that at best GPR55 is an atypical cannabinoid responder. The activation of GPR55 by rimonabant may be responsible for some of the off-target effects that led to its removal as a potential obesity therapy.


Cannabinoid Receptor Agonists , Cannabinoids/pharmacology , Receptors, Cannabinoid/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Analgesics/pharmacology , Arrestins/genetics , Arrestins/metabolism , Cannabinoids/metabolism , Cell Line , Cyclohexanols/pharmacology , Humans , Ligands , Lysophospholipids/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Obesity/drug therapy , Obesity/metabolism , Phosphorylation/drug effects , Piperidines/adverse effects , Piperidines/pharmacology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C beta , Pyrazoles/adverse effects , Pyrazoles/pharmacology , Receptors, Cannabinoid/genetics , Receptors, G-Protein-Coupled/genetics , Rimonabant , beta-Arrestins
16.
Biochem Biophys Res Commun ; 332(3): 845-52, 2005 Jul 08.
Article En | MEDLINE | ID: mdl-15913555

Taste and salivary secretion disorders have been linked to zinc deficiency, indeed zinc is found in secretory granules in the salivary gland. The signaling role for the zinc release in this tissue, however, is poorly understood. Here, we address the signaling pathways and physiological role of the zinc-sensing receptor, ZnR, in the ductal salivary gland cell line, HSY. Exposure of these cells to zinc triggered intracellular Ca2+ release from thapsigargin-sensitive stores. The G alpha q inhibitor, YM-254890 (1 microM), eliminated the Zn2+-dependent Ca2+ response, demonstrating that ZnR is a G alpha q-coupled receptor. Dose-response curves yielded an apparent K0.5 of 36 microM and a Hill coefficient of 7 in the absence of extracellular Ca2+, and K0.5 of 55 microM with a Hill coefficient of 3 in its presence. This indicates that although Zn2+ is essential for ZnR activation, Ca2+ may affect the receptor co-operativity. The homologous desensitization pattern of ZnR was characterized by pre-exposure of cells to Zn2+ at concentrations found to activate the receptor. Re-exposure of cells to Zn2+ elicited an attenuated Zn2+-dependent Ca2+ response for at least 3 h, indicating that the ZnR is strongly desensitized by Zn2+. Finally, we studied the paracrine affects of ZnR using a co-culture consisting of the HSY cells and vascular smooth muscle cells (VSMCs). While no Zn2+-dependent Ca2+ release was observed in VSMC alone, application of Zn2+ to the co-culture induced a Ca2+ rise in both HSY cells and VSMC. This Ca2+ rise was inhibited by the ATP scavenger, apyrase. Taken together, our results demonstrate that ZnR activity is monitored in salivary cells and is modulated by extracellular Ca2+. We further show that ZnR enhances secretion of ATP, thereby linking zinc to key signaling pathways involved in modification of salivary secretions by the ductal cells.


Adenosine Triphosphate/metabolism , Receptors, G-Protein-Coupled/metabolism , Zinc/metabolism , Calcium/metabolism , Calcium/pharmacology , Calcium Signaling , Cell Communication/physiology , Cell Line , Humans , Salivary Glands/drug effects , Salivary Glands/metabolism , Signal Transduction , Zinc/pharmacology
17.
J Biol Chem ; 279(50): 51804-16, 2004 Dec 10.
Article En | MEDLINE | ID: mdl-15355987

Extracellular zinc promotes cell proliferation and its deficiency leads to impairment of this process, which is particularly important in epithelial cells. We have recently characterized a zinc-sensing receptor (ZnR) linking extracellular zinc to intracellular release of calcium. In the present study, we addressed the role of extracellular zinc, acting via the ZnR, in regulating the MAP kinase pathway and Na+/H+ exchange in colonocytes. We demonstrate that Ca2+ release, mediated by the ZnR, induces phosphorylation of ERK1/2, which is highly metal-specific, mediated by physiological concentrations of extracellular Zn2+ but not by Cd2+, Fe2+, Ni2+, or Mn2+. Desensitization of the ZnR by Zn2+, is followed by approximately 90% inhibition of the Zn2+ -dependent ERK1/2 phosphorylation, indicating that the ZnR is a principal link between extracellular Zn2+ and ERK1/2 activation. Application of both the IP3 pathway and PI 3-kinase antagonists largely inhibited Zn2+ -dependent ERK1/2 phosphorylation. The physiological significance of the Zn2+ -dependent activation of ERK1/2 was addressed by monitoring Na+/H+ exchanger activity in HT29 cells and in native colon epithelium. Preincubation of the cells with zinc was followed by robust activation of Na+/H+ exchange, which was eliminated by cariporide (0.5 microm); indicating that zinc enhances the activity of NHE1. Activation of NHE1 by zinc was totally blocked by the ERK1/2 inhibitor, U0126. Prolonged acidification, in contrast, stimulates NHE1 by a distinct pathway that is not affected by extracellular Zn2+ or inhibitors of the MAP kinase pathway. Desensitization of ZnR activity eliminates the Zn2+ -dependent, but not the prolonged acidification-dependent activation of NHE1, indicating that Zn2+ -dependent activation of H+ extrusion is specifically mediated by the ZnR. Our results support a role for extracellular zinc, acting through the ZnR, in regulating multiple signaling pathways that affect pH homeostasis in colonocytes. Furthermore activation of both, ERK and NHE1, by extracellular zinc may provide the mechanism linking zinc to enhanced cell proliferation.


Extracellular Signal-Regulated MAP Kinases/metabolism , Sodium-Hydrogen Exchangers/metabolism , Zinc/metabolism , Zinc/pharmacology , Animals , Calcium/metabolism , Cation Transport Proteins/metabolism , Cell Line , Colon/cytology , Colon/drug effects , Colon/metabolism , Extracellular Fluid/metabolism , Humans , In Vitro Techniques , Inositol 1,4,5-Trisphosphate/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rats , Rats, Wistar , Signal Transduction , Sodium-Hydrogen Exchanger 1
...