Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
ACS Med Chem Lett ; 14(1): 18-25, 2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36655130

ABSTRACT

GPR55 is an orphan G-protein coupled receptor involved in various pathophysiological conditions. However, there are only a few noncannabinoid GPR55 ligands reported so far. The lack of potent and selective GPR55 ligands precludes a deep exploration of this receptor. The studies presented here focused on a thienopyrimidine scaffold based on the GPR55 antagonist ML192, previously discovered by high-throughput screening. The GPR55 activities of the new synthesized compounds were assessed using ß-arrestin recruitment assays in Chinese hamster ovary cells overexpressing human GPR55. Some derivatives were identified as GPR55 antagonists with functional efficacy and selectivity versus CB1 and CB2 cannabinoid receptors.

2.
Cannabis Cannabinoid Res ; 8(5): 731-748, 2023 10.
Article in English | MEDLINE | ID: mdl-35792570

ABSTRACT

Background: Activation of signaling effectors by G-protein coupled receptors (GPCRs) depends on different molecular mechanisms triggered by conserved amino acid residues. Although studies have focused on the G-protein signaling state, the mechanism for ß-arrestin signaling by CB1 is not yet well defined. Studies have indicated that transmembrane helix 7 (TMH7) and the highly conserved NPXXY motif can be subject to different conformational changes in response to biased ligands and could therefore participate in a molecular mechanism to trigger ß-arrestin recruitment. Objective: To investigate the effect of mutations in the NPXXY motif on different signaling pathways activated by the CB1 receptor. Materials and Methods: Point mutations of the NPXXY motif and associated residues were generated in the CB1 receptor using site-directed mutagenesis and transfection into HEK-293 cells. Signaling by wild-type and mutant receptors was analyzed by quantifying inhibition of cAMP, and by ß-arrestin recruitment assays. Results: We found that N7.49 and Y7.53 are essential for ß-arrestin recruitment by CB1. N7.49A and Y7.53F impair ß-arrestin signaling, with no effect on G-protein signaling. We found a regulatory role for residue I2.43; I2.43 interacts with Y7.53, affecting its positioning. Reducing steric bulk at I2.43 (I2.43A) enhances ß-arrestin1 recruitment, while introducing a polar residue (I2.43T) reduces ß-arrestin recruitment. Conclusions: These findings point to a novel mechanism for ß-arrestin recruitment, implicating amino acids in the NPXXY motif as critical for the putative ß-arrestin biased conformational state of Class A GPCRs.


Subject(s)
Receptor, Cannabinoid, CB1 , beta-Arrestin 1 , Humans , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism , Cannabinoids , GTP-Binding Proteins/metabolism , HEK293 Cells , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
3.
J Mol Neurosci ; 72(9): 1859-1874, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35779192

ABSTRACT

KLS-13019, a novel devised cannabinoid-like compound, was explored for anti-inflammatory actions in dorsal root ganglion cultures relevant to chemotherapy-induced peripheral neuropathy (CIPN). Time course studies with 3 µM paclitaxel indicated > 1.9-fold increases in immunoreactive (IR) area for cell body GPR55 after 30 min as determined by high content imaging. To test for reversibility of paclitaxel-induced increases in GPR55, cultures were treated for 8 h with paclitaxel alone and then a dose response to KLS-13019 added for another 16 h. This "reversal" paradigm indicated established increases in cell body GPR55 IR areas were decreased back to control levels. Because GPR55 had previously reported inflammatory actions, IL-1ß and NLRP3 (inflammasome-3 marker) were also measured in the "reversal" paradigm. Significant increases in all inflammatory markers were produced after 8 h of paclitaxel treatment alone that were reversed to control levels with KLS-13019 treatment. Accompanying studies using alamar blue indicated that decreased cellular viability produced by paclitaxel treatment was reverted back to control levels by KLS-13019. Similar studies conducted with lysophosphatidylinositol (GPR55 agonist) in DRG or hippocampal cultures demonstrated significant increases in neuritic GPR55, NLRP3 and IL-1ß areas that were reversed to control levels with KLS-13019 treatment. Studies with a human GPR55-ß-arrestin assay in Discover X cells indicated that KLS-13019 was an antagonist without agonist activity. These studies indicated that KLS-13019 has anti-inflammatory properties mediated through GPR55 antagonist actions. Together with previous studies, KLS-13019 is a potent neuroprotective, anti-inflammatory cannabinoid with therapeutic potential for high efficacy treatment of neuropathic pain.


Subject(s)
Cannabinoids , Neuralgia , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cannabinoids/therapeutic use , Ganglia, Spinal/metabolism , Hippocampus/metabolism , Humans , NLR Family, Pyrin Domain-Containing 3 Protein , Neuralgia/drug therapy , Paclitaxel/pharmacology , Receptors, Cannabinoid/metabolism
4.
J Natl Cancer Inst Monogr ; 2021(58): 99-106, 2021 11 28.
Article in English | MEDLINE | ID: mdl-34850900

ABSTRACT

Plant-based, synthetic, and endogenous cannabinoids have been shown to control a diverse array of biological processes, including regulation of cell fate across cancers. Their promise as broad-based antitumor agents in preclinical models has led to the initiation of pilot clinical trials. Session 5 of the National Cancer Institute's Cannabis, Cannabinoids and Cancer Research Symposium provides an overview of this research topic. Overall, the presentations highlight cannabinoid signal transduction and specific molecular mechanisms underlying cannabinoid antitumor activity. They also demonstrate the broad-based antitumor activity of the plant-based, synthetic, and endogenous cannabinoid compounds. Importantly, evidence is presented demonstrating when cannabinoids may be contraindicated as a treatment for cancer, as in the case of human papilloma virus-meditated oropharynx cancer or potentially other p38 MAPK pathway-driven cancers. Finally, it is discussed that a key to advancing cannabinoids into the clinic is to conduct well-designed, large-scale clinical trials to determine whether cannabinoids are effective antitumor agents in cancer patients.


Subject(s)
Cannabinoids , Medical Marijuana , Neoplasms , Biology , Cannabinoids/pharmacology , Clinical Trials as Topic , Humans , Neoplasms/drug therapy , Neoplasms/prevention & control
5.
Molecules ; 26(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34500853

ABSTRACT

The CB1 cannabinoid receptor is a G-protein coupled receptor highly expressed throughout the central nervous system that is a promising target for the treatment of various disorders, including anxiety, pain, and neurodegeneration. Despite the wide therapeutic potential of CB1, the development of drug candidates is hindered by adverse effects, rapid tolerance development, and abuse potential. Ligands that produce biased signaling-the preferential activation of a signaling transducer in detriment of another-have been proposed as a strategy to dissociate therapeutic and adverse effects for a variety of G-protein coupled receptors. However, biased signaling at the CB1 receptor is poorly understood due to a lack of strongly biased agonists. Here, we review studies that have investigated the biased signaling profile of classical cannabinoid agonists and allosteric ligands, searching for a potential therapeutic advantage of CB1 biased signaling in different pathological states. Agonist and antagonist bound structures of CB1 and proposed mechanisms of action of biased allosteric modulators are used to discuss a putative molecular mechanism for CB1 receptor activation and biased signaling. Current studies suggest that allosteric binding sites on CB1 can be explored to yield biased ligands that favor or hinder conformational changes important for biased signaling.


Subject(s)
Cannabinoid Receptor Agonists/chemistry , Receptor, Cannabinoid, CB1/chemistry , Allosteric Site , Central Nervous System/metabolism , Humans , Indoles/chemistry , Ligands , Models, Molecular , Piperidines/chemistry , Pregnenolone/chemistry , Protein Binding , Protein Conformation , Signal Transduction
6.
J Med Chem ; 64(12): 8104-8126, 2021 06 24.
Article in English | MEDLINE | ID: mdl-33826336

ABSTRACT

We apply the magic methyl effect to improve the potency/efficacy of GAT211, the prototypic 2-phenylindole-based cannabinoid type-1 receptor (CB1R) agonist-positive allosteric modulator (ago-PAM). Introducing a methyl group at the α-position of nitro group generated two diastereomers, the greater potency and efficacy of erythro, (±)-9 vs threo, (±)-10 constitutes the first demonstration of diastereoselective CB1R-allosteric modulator interaction. Of the (±)-9 enantiomers, (-)-(S,R)-13 evidenced improved potency over GAT211 as a CB1R ago-PAM, whereas (+)-(R,S)-14 was a CB1R allosteric agonist biased toward G protein- vs ß-arrestin1/2-dependent signaling. (-)-(S,R)-13 and (+)-(R,S)-14 were devoid of undesirable side effects (triad test), and (+)-(R,S)-14 reduced intraocular pressure with an unprecedentedly long duration of action in a murine glaucoma model. (-)-(S,R)-13 docked into both a CB1R extracellular PAM and intracellular allosteric-agonist site(s), whereas (+)-(R,S)-14 preferentially engaged only the latter. Exploiting G-protein biased CB1R-allosteric modulation can offer safer therapeutic candidates for glaucoma and, potentially, other diseases.


Subject(s)
Cannabinoid Receptor Agonists/therapeutic use , Glaucoma/drug therapy , Indoles/therapeutic use , Receptor, Cannabinoid, CB1/agonists , Allosteric Site , Animals , CHO Cells , Cannabinoid Receptor Agonists/chemical synthesis , Cannabinoid Receptor Agonists/metabolism , Cricetulus , HEK293 Cells , Hippocampus/cytology , Humans , Indoles/chemical synthesis , Indoles/metabolism , Intraocular Pressure/drug effects , Ligands , Male , Mice, Inbred C57BL , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Neurons/drug effects , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB1/metabolism , Stereoisomerism , Structure-Activity Relationship
7.
J Med Chem ; 63(23): 14216-14227, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32914978

ABSTRACT

GPR18 is a G-protein-coupled receptor that belongs to the orphan class A family. Even though it shares low sequence homology with the cannabinoid receptors CB1R and CB2R, a growing body of research suggests its relationship with the endocannabinoid system, not only because it is able to recognize cannabinoid ligands but also because of its expression and ability to heteromerize with CBRs. In this review, we aim to analyze the biological relevance, reported modulators, and structural features of GPR18. In order to guide future drug design in this field, highlights from molecular modeling of GPR18 will be provided.


Subject(s)
Cannabinoids/metabolism , Receptors, G-Protein-Coupled/metabolism , Drug Design , Humans , Protein Conformation , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/therapeutic use
8.
J Med Chem ; 63(2): 542-568, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31756109

ABSTRACT

Cannabinoid 1 receptor (CB1R) allosteric ligands hold a far-reaching therapeutic promise. We report the application of fluoro- and nitrogen-walk approaches to enhance the drug-like properties of GAT211, a prototype CB1R allosteric agonist-positive allosteric modulator (ago-PAM). Several analogs exhibited improved functional potency (cAMP, ß-arrestin 2), metabolic stability, and aqueous solubility. Two key analogs, GAT591 (6r) and GAT593 (6s), exhibited augmented allosteric-agonist and PAM activities in neuronal cultures, improved metabolic stability, and enhanced orthosteric agonist binding (CP55,940). Both analogs also exhibited good analgesic potency in the CFA inflammatory-pain model with longer duration of action over GAT211 while being devoid of adverse cannabimimetic effects. Another analog, GAT592 (9j), exhibited moderate ago-PAM potency and improved aqueous solubility with therapeutic reduction of intraocular pressure in murine glaucoma models. The SAR findings and the enhanced allosteric activity in this class of allosteric modulators were accounted for in our recently developed computational model for CB1R allosteric activation and positive allosteric modulation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Fluorine/chemistry , Indoles/chemistry , Nitrogen/chemistry , Receptor, Cannabinoid, CB1/drug effects , Allosteric Regulation/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biotransformation , Freund's Adjuvant , HEK293 Cells , Humans , Indoles/pharmacokinetics , Indoles/pharmacology , Inflammation/chemically induced , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Receptor, Cannabinoid, CB1/agonists , Stereoisomerism , Structure-Activity Relationship
9.
Drug Alcohol Depend ; 206: 107637, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31734036

ABSTRACT

BACKGROUND: Cocaine has a variety of negative effects on the central nervous system, including reports of decreased barrier function of brain microvascular endothelial cells. However, few studies have directly shown the effects of cocaine on blood-brain barrier (BBB) function in vivo. The miniature integrated fluorescence microscope (i.e., miniscope) technology was used to visualize cocaine-induced changes in BBB permeability in awake, freely-moving rats. METHODS: The miniscope was implanted in the prefrontal cortex of adult male rats. After recovery and acclimation, rats received an injection of cocaine (5-20 mg/kg ip) 15 minutes following iv infusion of sodium fluorescein, a low molecular weight tracer. Fluorescence intensity was recordedin vivo via the miniscope for 30 minutes or 24 hours post cocaine administration and served as an indicator of BBB permeability. RESULTS: Results demonstrate that cocaine increased the sodium fluorescein extravasation in brain microcirculation in a dose-dependent manner 30 minutes, but not 24 hours after administration. CONCLUSION: We report for the first time using direct visualization of brain microcirculation with the miniscope technology in awake, freely-moving rats, that acute cocaine administration produced a transient increase in the BBB permeability.


Subject(s)
Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Cocaine/pharmacokinetics , Microscopy, Fluorescence , Animals , Blood-Brain Barrier/physiopathology , Brain/blood supply , Brain/diagnostic imaging , Endothelial Cells/drug effects , Fluorescein , Male , Rats
10.
Neuroscience ; 414: 88-98, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31279825

ABSTRACT

GPR55, an atypical cannabinoid receptor activated by lysophosphatidylinositol (LPI) has been involved in various physiological and pathological processes. We examined the effect of GPR55 activation on rat brain microvascular endothelial cells (RBMVEC), an essential component of the blood-brain barrier (BBB). GPR55 was detected in RBMVEC by western blot and immunocytochemistry. Treatment of RBMVEC with LPI increased cytosolic Ca2+ concentration, [Ca2+]i, in a concentration-dependent manner; the effect was abolished by the GPR55 antagonist, ML-193. Repetitive application of LPI induced tachyphylaxis. LPI-induced increase in [Ca2+]i was not sensitive to U-73122, a phospholipase C inhibitor, but was abolished by the blockade of voltage-gated Ca2+ channels or in Ca2+-free saline, indicating that Ca2+ influx was involved in this response. LPI induced a biphasic change in RBMVEC membrane potential: a fast depolarization followed by a long-lasting hyperpolarization. The hyperpolarization phase was prevented by apamin and charibdotoxin, inhibitors of small- and intermediate-conductance Ca2+-activated K+ channels (KCa). Immunofluorescence studies indicate that LPI produced transient changes in tight and adherens junctions proteins and F-actin stress fibers. LPI decreased the electrical resistance of RBMVEC monolayer assessed with Electric Cell-Substrate Impedance Sensing (ECIS) in a dose-dependent manner. In vivo studies indicate that systemic administration of LPI increased the permeability of the BBB, assessed with Evans Blue method. Taken together, our results indicate that GPR55 activation modulates the function of endothelial cells of brain microvessels, produces a transient reduction in endothelial barrier function and increases BBB permeability.


Subject(s)
Blood-Brain Barrier/drug effects , Calcium Signaling/drug effects , Endothelial Cells/drug effects , Lysophospholipids/pharmacology , Receptors, Cannabinoid/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood-Brain Barrier/metabolism , Calcium/metabolism , Cell Line , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Male , Membrane Potentials/drug effects , Microvessels/drug effects , Microvessels/metabolism , Rats , Rats, Sprague-Dawley
11.
Cell Rep ; 26(2): 330-337.e4, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30625315

ABSTRACT

Sigma-1 receptors (Sig-1Rs) are integral ER membrane proteins. They bind diverse ligands, including psychoactive drugs, and regulate many signaling proteins, including the inositol 1,4,5-trisphosphate receptors (IP3Rs) that release Ca2+ from the ER. The endogenous ligands of Sig-1Rs are unknown. Phospholipase D (PLD) cleaves phosphatidylcholine to choline and phosphatidic acid (PA), with PA assumed to mediate all downstream signaling. We show that choline is also an intracellular messenger. Choline binds to Sig-1Rs, it mimics other Sig-1R agonists by potentiating Ca2+ signals evoked by IP3Rs, and it is deactivated by metabolism. Receptors, by stimulating PLC and PLD, deliver two signals to IP3Rs: IP3 activates IP3Rs, and choline potentiates their activity through Sig-1Rs. Choline is also produced at synapses by degradation of acetylcholine. Choline uptake by transporters activates Sig-1Rs and potentiates Ca2+ signals. We conclude that choline is an endogenous agonist of Sig-1Rs linking extracellular stimuli, and perhaps synaptic activity, to Ca2+ signals.


Subject(s)
Calcium Signaling , Choline/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Receptors, sigma/metabolism , Animals , Cell Line , Humans , MCF-7 Cells , Mice , Phospholipase D/metabolism , Sigma-1 Receptor
12.
Neuroscience ; 377: 105-113, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29522856

ABSTRACT

Platelet-activating factor (PAF) is a potent phospholipid mediator that exerts various pathophysiological effects by interacting with a G protein-coupled receptor. PAF has been reported to increase the permeability of the blood-brain barrier (BBB) via incompletely characterized mechanisms. We investigated the effect of PAF on rat brain microvascular endothelial cells (RBMVEC), a critical component of the BBB. PAF produced a dose-dependent increase in cytosolic Ca2+ concentration; the effect was prevented by the PAF receptor antagonist, WEB2086. The effect of PAF on cytosolic Ca2+ was abolished in Ca2+-free saline or in the presence of L-type voltage-gated Ca2+ channel inhibitor, nifedipine, indicating that Ca2+ influx is critical for PAF-induced increase in cytosolic Ca2+. PAF produced RBMVEC depolarization; the effect was inhibited by WEB2086. In cells loaded with [(4-amino-5-methylamino-2',7'-difluoro-fluorescein)diacetate] (DAF-FM), a nitric oxide (NO)-sensitive fluorescent dye, PAF increased the NO level; the effect was prevented by WEB2086, nifedipine or by l-NAME, an inhibitor of NO synthase. Immunocytochemistry studies indicate that PAF reduced the immunostaining of ZO-1, a tight junction-associated protein, increased F-actin fibers, and produced intercellular gaps. PAF produced a decrease in RBMVEC monolayer electrical resistance assessed with Electric Cell-Substrate Impedance Sensing (ECIS), indicative of a disruption of endothelial barrier function. In vivo studies indicate that PAF increased the BBB permeability, assessed with sodium fluorescein and Evans Blue methods, via PAF receptor-dependent mechanisms, consequent to Ca2+ influx and increased NO levels. Our studies reveal that PAF alters the BBB permeability by multiple mechanisms, which may be relevant for central nervous system (CNS) inflammatory disorders.


Subject(s)
Brain/blood supply , Brain/metabolism , Endothelial Cells/metabolism , Microvessels/metabolism , Platelet Activating Factor/metabolism , Animals , Brain/drug effects , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cations, Divalent/metabolism , Cell Survival/physiology , Cells, Cultured , Cytosol/metabolism , Endothelial Cells/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microvessels/drug effects , Rats
13.
Bioorg Med Chem ; 25(24): 6427-6434, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29079014

ABSTRACT

In this work, we explored the molecular framework of the known CB1R allosteric modulator PSNCBAM-1 with the aim to generate new bioactive analogs and to deepen the structure-activity relationships of this type of compounds. In particular, the introduction of a NH group between the pyridine ring and the phenyl nucleus generated the amino-phenyl-urea derivative SN15b that behaved as a positive allosteric modulator (PAM), increasing the CB1R binding affinity of the orthosteric ligand CP55,940. The functional activity was evaluated using serum response element (SRE) assay, which assesses the CB1R-dependent activation of the MAPK/ERK signaling pathway. SN15b and the biphenyl-urea analog SC4a significantly inhibited the response produced by CP55,940 in the low µM range, thus behaving as negative allosteric modulators (NAMs). The new derivatives presented here provide further insights about the modulation of CB1R binding and functional activity by allosteric ligands.


Subject(s)
Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Allosteric Regulation/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship
14.
Neuroscience ; 365: 23-32, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-28951324

ABSTRACT

Bradykinin (BK), a component of the kallikrein-kininogen-kinin system exerts multiple effects via B1 and B2 receptor activation. In the cardiovascular system, bradykinin has cardioprotective and vasodilator properties. We investigated the effect of BK on cardiac-projecting neurons of nucleus ambiguus, a key site for the parasympathetic cardiac regulation. BK produced a dose-dependent increase in cytosolic Ca2+ concentration. Pretreatment with HOE140, a B2 receptor antagonist, but not with R715, a B1 receptor antagonist, abolished the response to BK. A selective B2 receptor agonist, but not a B1 receptor agonist, elicited an increase in cytosolic Ca2+ similarly to BK. Inhibition of N-type voltage-gated Ca2+ channels with ω-conotoxin GVIA had no effect on the Ca2+ signal produced by BK, while pretreatment with ω-conotoxin MVIIC, a blocker of P/Q-type of Ca2+ channels, significantly diminished the effect of BK. Pretreatment with xestospongin C and 2-aminoethoxydiphenyl borate, antagonists of inositol 1,4,5-trisphosphate receptors, abolished the response to BK. Inhibition of ryanodine receptors reduced the BK-induced Ca2+ increase, while disruption of lysosomal Ca2+ stores with bafilomycin A1 did not affect the response. BK produced a dose-dependent depolarization of nucleus ambiguus neurons, which was prevented by the B2 receptor antagonist. In vivo studies indicate that microinjection of BK into nucleus ambiguus elicited bradycardia in conscious rats via B2 receptors. In summary, in cardiac vagal neurons of nucleus ambiguus, BK activates B2 receptors promoting Ca2+ influx and Ca2+ release from endoplasmic reticulum, and membrane depolarization; these effects are translated in vivo by bradycardia.


Subject(s)
Bradykinin/pharmacology , Heart Rate/drug effects , Medulla Oblongata/cytology , Neurons/drug effects , Vagus Nerve/physiology , Vasodilator Agents/pharmacology , Animals , Animals, Newborn , Barbiturates/metabolism , Bradykinin/analogs & derivatives , Bradykinin Receptor Antagonists/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Isoxazoles/metabolism , Male , Medulla Oblongata/drug effects , Membrane Potentials/drug effects , Rats , Rats, Sprague-Dawley , Vagus Nerve/drug effects
15.
Adv Pharmacol ; 80: 169-206, 2017.
Article in English | MEDLINE | ID: mdl-28826534

ABSTRACT

The CB1 and CB2 cannabinoid receptors (CB1R, CB2R) are members of the G protein-coupled receptor (GPCR) family that were identified over 20 years ago. CB1Rs and CB2Rs mediate the effects of Δ9-tetrahydrocannabinol (Δ9-THC), the principal psychoactive constituent of marijuana, and subsequently identified endogenous cannabinoids (endocannabinoids) anandamide and 2-arachidonoyl glycerol. CB1Rs and CB2Rs have both similarities and differences in their pharmacology. Both receptors recognize multiple classes of agonist and antagonist compounds and produce an array of distinct downstream effects. Natural polymorphisms and alternative splice variants may also contribute to their pharmacological diversity. As our knowledge of the distinct differences grows, we may be able to target select receptor conformations and their corresponding pharmacological responses. This chapter will discuss their pharmacological characterization, distribution, phylogeny, and signaling pathways. In addition, the effects of extended agonist exposure and how that affects signaling and expression patterns of the receptors are considered.


Subject(s)
Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Alternative Splicing/genetics , Animals , Humans , Phylogeny , Polymorphism, Genetic , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Signal Transduction/drug effects
16.
Methods Enzymol ; 593: 23-42, 2017.
Article in English | MEDLINE | ID: mdl-28750805

ABSTRACT

With the approach of the 30th year since the pioneering discovery of a cannabinoid receptor in rat brain (Devane et al., 1988), the field of cannabinoid pharmacology and physiology has impacted human physiology at multiple levels. The development of highly specific and potent orthosteric ligands, as well as the blossoming field of allosteric ligand development, has placed the endocannabinoid system in the forefront as a modulator of a multitude of physiologic processes. Reproducibility among laboratories is especially important due to the development of novel tools to investigate the role(s) of the endocannabinoid system in human physiology, and to clarify the roles for medicinal marijuana. Any definitive role in normal, or diseased states, must be satisfied through the demonstration of a specific receptor-mediated event. This chapter provides working protocols for the study of cannabinoid receptor-ligand binding, as well as immediate and downstream G protein-dependent signaling assays to assess receptor function.


Subject(s)
Receptors, Cannabinoid/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , HEK293 Cells , Humans , Protein Binding , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/isolation & purification , Reproducibility of Results , Scintillation Counting , Signal Transduction , Sulfur Radioisotopes/chemistry
17.
Biochem Biophys Res Commun ; 490(4): 1389-1393, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28698140

ABSTRACT

Interest in lipoamino acids as endogenous modulators of G-protein coupled receptors has escalated due to their involvement in a variety of physiologic processes. In particular, a role for these amino acid conjugates has emerged in the endocannabinoid system. The study presented herein investigated the effects of N-arachidonoyl glycine (NAGly) on a candidate endocannabinoid receptor, GPR55. Our novel findings reveal that NAGly induces concentration dependent increases in calcium mobilization and mitogen-activated protein kinase activities in HAGPR55/CHO cells. These increases were attenuated by the selective GPR55 antagonist ML193 (N-[4-[[(3,4-Dimethyl-5-isoxazolyl)amino]sulfonyl]phenyl]-6,8-dimethyl-2-(2-pyridinyl)-4-quinolinecarboxamide), supporting receptor mediated signaling. To our knowledge this is the first report identifying GPR55 as a target of the endogenous lipoamino acid, NAGly.


Subject(s)
Arachidonic Acids/pharmacology , Calcium/metabolism , Glycine/analogs & derivatives , Receptors, G-Protein-Coupled/genetics , Animals , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Gene Expression Regulation , Glycine/pharmacology , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Kinetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Quinolines/pharmacology , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism
18.
Bioorg Med Chem ; 25(16): 4355-4367, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28673732

ABSTRACT

GPR55, a G protein-coupled receptor, is an attractive target to alleviate inflammatory and neuropathic pain and treat osteoporosis and cancer. Identifying a potent and selective ligand will aid to further establish the specific physiological roles and pharmacology of the receptor. Towards this goal, a targeted library of 22 compounds was synthesized in a modular fashion to obtain structure-activity relationship information. The general route consisted of coupling a variety of p-aminophenyl sulfonamides to isothiocyanates to form acylthioureas. For the synthesis of a known naphthyl ethyl alcohol motif, route modification led to a shorter and more efficient process. The 22 analogues were analyzed for their ability to serve as agonists at GPR55 and valuable information for both ends of the molecule was ascertained.


Subject(s)
Drug Design , Receptors, G-Protein-Coupled/agonists , Thiourea/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Structure , Receptors, Cannabinoid , Structure-Activity Relationship , Thiourea/analogs & derivatives , Thiourea/chemical synthesis
19.
Drug Alcohol Depend ; 178: 7-14, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28623807

ABSTRACT

BACKGROUND: HIV-1 infection and drug abuse are frequently co-morbid and their association greatly increases the severity of HIV-1-induced neuropathology. While nucleus accumbens (NAcc) function is severely perturbed by drugs of abuse, little is known about how HIV-1 infection affects NAcc. METHODS: We used calcium and voltage imaging to investigate the effect of HIV-1 trans-activator of transcription (Tat) on rat NAcc. Based on previous neuronal studies, we hypothesized that Tat modulates intracellular Ca2+ homeostasis of NAcc neurons. RESULTS: We provide evidence that Tat triggers a Ca2+ signaling cascade in NAcc medium spiny neurons (MSN) expressing D1-like dopamine receptors leading to neuronal depolarization. Firstly, Tat induced inositol 1,4,5-trisphsophate (IP3) receptor-mediated Ca2+ release from endoplasmic reticulum, followed by Ca2+ and Na+ influx via transient receptor potential canonical channels. The influx of cations depolarizes the membrane promoting additional Ca2+ entry through voltage-gated P/Q-type Ca2+ channels and opening of tetrodotoxin-sensitive Na+ channels. By activating this mechanism, Tat elicits a feed-forward depolarization increasing the excitability of D1-phosphatidylinositol-linked NAcc MSN. We previously found that cocaine targets NAcc neurons directly (independent of the inhibition of dopamine transporter) only when IP3-generating mechanisms are concomitantly initiated. When tested here, cocaine produced a dose-dependent potentiation of the effect of Tat on cytosolic Ca2+. CONCLUSION: We describe for the first time a HIV-1 Tat-triggered Ca2+ signaling in MSN of NAcc involving TRPC and depolarization and a potentiation of the effect of Tat by cocaine, which may be relevant for the reward axis in cocaine-abusing HIV-1-positive patients.


Subject(s)
Neurons/physiology , Nucleus Accumbens/physiology , Receptors, Dopamine D1/metabolism , tat Gene Products, Human Immunodeficiency Virus/physiology , Animals , Calcium/metabolism , Cells, Cultured , Cocaine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Female , Male , Neurons/metabolism , Nucleus Accumbens/drug effects , Rats , Signal Transduction/physiology , Sodium/metabolism , tat Gene Products, Human Immunodeficiency Virus/pharmacology
20.
Brain Res ; 1657: 297-303, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28043808

ABSTRACT

The pituitary adenylyl cyclase-activating polypeptide (PACAP) and its G protein-coupled receptors, PAC1, VPAC1 and VPAC2 form a system involved in a variety of biological processes. Although some sympathetic stimulatory effects of this system have been reported, its central cardiovascular regulatory properties are poorly characterized. VPAC1 receptors are expressed in the nucleus ambiguus (nAmb), a key center controlling cardiac parasympathetic tone. In this study, we report that selective VPAC1 activation in rhodamine-labeled cardiac vagal preganglionic neurons of the rat nAmb produces inositol 1,4,5-trisphosphate receptor-mediated Ca2+ mobilization, membrane depolarization and activation of P/Q-type Ca2+ channels. In vivo, this pathway converges onto transient reduction in heart rate of conscious rats. Therefore we demonstrate a VPAC1-dependent mechanism in the central parasympathetic regulation of the heart rate, adding to the complexity of PACAP-mediated cardiovascular modulation.


Subject(s)
Medulla Oblongata/metabolism , Neurons/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Bradycardia/chemically induced , Bradycardia/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Female , Heart Rate/drug effects , Heart Rate/physiology , Male , Medulla Oblongata/cytology , Medulla Oblongata/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuroanatomical Tract-Tracing Techniques , Neurons/cytology , Neurons/drug effects , Parasympatholytics/pharmacology , Rats, Sprague-Dawley , Receptors, Vasoactive Intestinal Polypeptide, Type I/agonists , Vagus Nerve/cytology , Vagus Nerve/drug effects , Vagus Nerve/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...