Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 204: 276-286, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37217089

ABSTRACT

We developed S1QEL1.719, a novel bioavailable S1QEL (suppressor of site IQ electron leak). S1QEL1.719 prevented superoxide/hydrogen peroxide production at site IQ of mitochondrial complex I in vitro. The free concentration giving half-maximal suppression (IC50) was 52 nM. Even at 50-fold higher concentrations S1QEL1.719 did not inhibit superoxide/hydrogen peroxide production from other sites. The IC50 for inhibition of complex I electron flow was 500-fold higher than the IC50 for suppression of superoxide/hydrogen peroxide production from site IQ. S1QEL1.719 was used to test the metabolic effects of suppressing superoxide/hydrogen peroxide production from site IQin vivo. C57BL/6J male mice fed a high-fat chow for one, two or eight weeks had increased body fat, decreased glucose tolerance, and increased fasting insulin concentrations, classic symptoms of metabolic syndrome. Daily prophylactic or therapeutic oral treatment of high-fat-fed animals with S1QEL1.719 decreased fat accumulation, strongly protected against decreased glucose tolerance and prevented or reversed the increase in fasting insulin level. Free exposures in plasma and liver at Cmax were 1-4 fold the IC50 for suppression of superoxide/hydrogen peroxide production at site IQ and substantially below levels that inhibit electron flow through complex I. These results show that the production of superoxide/hydrogen peroxide from mitochondrial site IQin vivo is necessary for the induction and maintenance of glucose intolerance caused by a high-fat diet in mice. They raise the possibility that oral administration of S1QELs may be beneficial in metabolic syndrome.


Subject(s)
Metabolic Syndrome , Superoxides , Mice , Male , Animals , Superoxides/metabolism , Hydrogen Peroxide/metabolism , Peroxides , Insulin , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Fasting , Adipose Tissue/metabolism , Glucose
2.
J Pharmacol Exp Ther ; 372(1): 107-118, 2020 01.
Article in English | MEDLINE | ID: mdl-31732698

ABSTRACT

Cystic fibrosis (CF) is the most common monogenic autosomal recessive disease in Caucasians caused by pathogenic mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene (CFTR). Significant small molecule therapeutic advances over the past two decades have been made to target the defective CFTR protein and enhance its function. To address the most prevalent defect of the defective CFTR protein (i.e., F508del mutation) in CF, two biomolecular activities are required, namely, correctors to increase the amount of properly folded F508delCFTR levels at the cell surface and potentiators to allow the effective opening, i.e., function of the F508delCFTR channel. Combined, these activities enhance chloride ion transport yielding improved hydration of the lung surface and subsequent restoration of mucociliary clearance. To enhance clinical benefits to CF patients, a complementary triple combination therapy consisting of two corrector molecules, type 1 (C1) and type 2, with additive mechanisms along with a potentiator are being investigated in the clinic for maximum restoration of mutated CFTR function. We report the identification and in vitro biologic characterization of ABBV-2222/GLPG2222 (4-[(2R,4R)-4-({[1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]carbonyl}amino)-7-(difluoromethoxy)-3,4-dihydro-2H-chromen-2-yl]benzoic acid),-a novel, potent, and orally bioavailable C1 corrector developed by AbbVie-Galapagos and currently in clinical trials-which exhibits substantial improvements over the existing C1 correctors. This includes improvements in potency and drug-drug interaction (DDI) compared with 3-(6-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanecarboxamido)-3-methylpyridin-2-yl)benzoic acid (VX-809, Lumacaftor) and improvements in potency and efficacy compared with 1-(2,2-difluoro-1,3-benzodioxol-5-yl)-N-[1-[(2R)-2,3-dihydroxypropyl]-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)indol-5-yl]cyclopropane-1-carboxamide (VX-661, Tezacaftor). ABBV-2222/GLPG2222 exhibits potent in vitro functional activity in primary patient cells harboring F508del/F508del CFTR with an EC50 value <10 nM. SIGNIFICANCE STATEMENT: To address the most prevalent defect of the defective CFTR protein (i.e., F508del mutation) in cystic fibrosis, AbbVie-Galapagos has developed ABBV-2222/GLPG2222, a novel, potent, and orally bioavailable C1 corrector of this protein. ABBV-2222/GLPG2222, which is currently in clinical trials, exhibits potent in vitro functional activity in primary patient cells harboring F508del/F508del CFTR and substantial improvements over the existing C1 correctors.


Subject(s)
Benzoates/pharmacology , Benzopyrans/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Protein Folding/drug effects , Animals , Binding Sites , Cell Membrane/metabolism , Cells, Cultured , Chlorides/metabolism , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , HEK293 Cells , Humans , Membrane Transport Modulators/pharmacology , Protein Binding , Protein Transport/drug effects , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
3.
J Pain ; 15(4): 387.e1-14, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24374196

ABSTRACT

UNLABELLED: Voltage-gated Ca(2+) channels play an important role in nociceptive transmission. There is significant evidence supporting a role for N-, T- and P/Q-type Ca(2+) channels in chronic pain. Here, we report that A-1264087, a structurally novel state-dependent blocker, inhibits each of these human Ca(2+) channels with similar potency (IC50 = 1-2 µM). A-1264087 was also shown to inhibit the release of the pronociceptive calcitonin gene-related peptide from rat dorsal root ganglion neurons. Oral administration of A-1264087 produces robust antinociceptive efficacy in monoiodoacetate-induced osteoarthritic, complete Freund adjuvant-induced inflammatory, and chronic constrictive injury of sciatic nerve-induced, neuropathic pain models with ED50 values of 3.0, 5.7, and 7.8 mg/kg (95% confidence interval = 2.2-3.5, 3.7-10, and 5.5-12.8 mg/kg), respectively. Further analysis revealed that A-1264087 also suppressed nociceptive-induced p38 and extracellular signal-regulated kinase 1/2 phosphorylation, which are biochemical markers of engagement of pain circuitry in chronic pain states. Additionally, A-1264087 inhibited both spontaneous and evoked neuronal activity in the spinal cord dorsal horn in complete Freund adjuvant-inflamed rats, providing a neurophysiological basis for the observed antihyperalgesia. A-1264087 produced no alteration of body temperature or motor coordination and no learning impairment at therapeutic plasma concentrations. PERSPECTIVE: The present results demonstrate that the neuronal Ca(2+) channel blocker A-1264087 exhibits broad-spectrum efficacy through engagement of nociceptive signaling pathways in preclinical pain models in the absence of effects on psychomotor and cognitive function.


Subject(s)
Analgesics/pharmacology , Azabicyclo Compounds/pharmacology , Calcium Channel Blockers/pharmacology , Leucine/analogs & derivatives , Neurons/metabolism , Nociception/drug effects , Spinal Cord/drug effects , Animals , Disease Models, Animal , Immunohistochemistry , Leucine/pharmacology , Male , Neurons/drug effects , Pain/metabolism , Patch-Clamp Techniques , Rats, Sprague-Dawley , Spinal Cord/metabolism
5.
Assay Drug Dev Technol ; 10(6): 542-50, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22428804

ABSTRACT

Ca(V)2.2 (N-type) calcium channels are key regulators of neurotransmission. Evidence from knockout animals and localization studies suggest that Ca(V)2.2 channels play a critical role in nociceptive transmission. Additionally, ziconotide, a selective peptide inhibitor of Ca(V)2.2 channels, is clinically used to treat refractory pain. However, the use of ziconotide is limited by its low therapeutic index, which is believed, at least in part, to be a consequence of ziconotide inhibiting Ca(V)2.2 channels regardless of the channel state. Subsequent efforts have focused on the discovery of state-dependent inhibitors that preferentially bind to the inactivated state of Ca(V)2.2 channels in order to achieve an improved safety profile relative to ziconotide. Much less attention has been paid to understanding the binding kinetics of these state-dependent inhibitors. Here, we describe a novel electrophysiology-based assay on an automated patch platform designed to differentiate Ca(V)2.2 inhibitors based on their combined state dependence and kinetics. More specifically, this assay assesses inactivated state block, closed state block, and monitors the kinetics of recovery from block when channels move between states. Additionally, a use-dependent assay is described that uses a train of depolarizing pulses to drive channels to a similar level of inactivation for comparison. This use-dependent protocol also provides information on the kinetics of block development. Data are provided to show how these assays can be utilized to screen for kinetic diversity within and across chemical classes.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Electrophysiology/methods , Animals , Automation , Biological Assay , Cell Line , Data Interpretation, Statistical , Drug Evaluation, Preclinical , Indoles/pharmacology , Kinetics , Patch-Clamp Techniques , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship , Triazines/pharmacology , Triazoles/pharmacology , omega-Conotoxins/pharmacology
6.
Bioorg Med Chem Lett ; 22(4): 1716-8, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22277280

ABSTRACT

A novel series of diphenyl lactam containing calcium channel blockers is described. Extensive SAR studies resulted in compounds with low molar activity and good plasma exposure after oral dosing. Compounds 2, 6 and 7 demonstrated significant efficacy in the capsaicin model of secondary hyperalgesia following oral administration.


Subject(s)
Biphenyl Compounds/chemical synthesis , Calcium Channel Blockers/chemical synthesis , Calcium Channels, N-Type/metabolism , Drug Discovery , Lactams/chemical synthesis , Administration, Oral , Animals , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacokinetics , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacokinetics , Inhibitory Concentration 50 , Lactams/chemistry , Lactams/pharmacokinetics , Molecular Structure , Piperazines/chemistry , Rats , Solubility
7.
Biochem Pharmacol ; 83(3): 406-18, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22153861

ABSTRACT

Blockade of voltage-gated Ca²âº channels on sensory nerves attenuates neurotransmitter release and membrane hyperexcitability associated with chronic pain states. Identification of small molecule Ca²âº channel blockers that produce significant antinociception in the absence of deleterious hemodynamic effects has been challenging. In this report, two novel structurally related compounds, A-686085 and A-1048400, were identified that potently block N-type (IC50=0.8 µM and 1.4 µM, respectively) and T-type (IC50=4.6 µM and 1.2 µM, respectively) Ca²âº channels in FLIPR based Ca²âº flux assays. A-686085 also potently blocked L-type Ca²âº channels (EC50=0.6 µM), however, A-1048400 was much less active in blocking this channel (EC50=28 µM). Both compounds dose-dependently reversed tactile allodynia in a model of capsaicin-induced secondary hypersensitivity with similar potencies (EC50=300-365 ng/ml). However, A-686085 produced dose-related decreases in mean arterial pressure at antinociceptive plasma concentrations in the rat, while A-1048400 did not significantly alter hemodynamic function at supra-efficacious plasma concentrations. Electrophysiological studies demonstrated that A-1048400 blocks native N- and T-type Ca²âº currents in rat dorsal root ganglion neurons (IC50=3.0 µM and 1.6 µM, respectively) in a voltage-dependent fashion. In other experimental pain models, A-1048400 dose-dependently attenuated nociceptive, neuropathic and inflammatory pain at doses that did not alter psychomotor or hemodynamic function. The identification of A-1048400 provides further evidence that voltage-dependent inhibition of neuronal Ca²âº channels coupled with pharmacological selectivity vs. L-type Ca²âº channels can provide robust antinociception in the absence of deleterious effects on hemodynamic or psychomotor function.


Subject(s)
Analgesics/administration & dosage , Calcium Channel Blockers/administration & dosage , Hemodynamics/physiology , Neurons/physiology , Pain Measurement , Piperidones/administration & dosage , Piperidones/chemistry , Administration, Oral , Animals , Dose-Response Relationship, Drug , HEK293 Cells , Hemodynamics/drug effects , Humans , Male , Neurons/drug effects , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
8.
Inflamm Res ; 60(7): 683-93, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21394563

ABSTRACT

OBJECTIVE: The aim of this study was to compare a diverse set of peptide and small-molecule calcium channel blockers for inactivated-state block of native and recombinant N-type calcium channels using fluorescence-based and automated patch-clamp electrophysiology assays. METHODS: The pharmacology of calcium channel blockers was determined at N-type channels in IMR-32 cells and in HEK cells overexpressing the inward rectifying K(+) channel Kir2.1. N-type channels were opened by increasing extracellular KCl. In the Kir2.1/N-type cell line the membrane potential could be modulated by adjusting the extracellular KCl, allowing determination of resting and inactivated-state block of N-type calcium channels. The potency and degree of state-dependent inhibition of these blockers were also determined by automated patch-clamp electrophysiology. RESULTS: N-type-mediated calcium influx in IMR-32 cells was determined for a panel of blockers with IC(50) values of 0.001-7 µM and this positively correlated with inactivated-state block of recombinant channels measured using electrophysiology. The potency of several compounds was markedly weaker in the state-dependent fluorescence-based assay compared to the electrophysiology assay, although the degree of state-dependent blockade was comparable. CONCLUSIONS: The present data demonstrate that fluorescence-based assays are suitable for assessing the ability of blockers to selectively interact with the inactivated state of the N-type channel.


Subject(s)
Calcium Channel Blockers/metabolism , Calcium Channels, N-Type/metabolism , Animals , Calcium/metabolism , Calcium Channels, N-Type/pharmacology , Cell Line , Humans , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques
9.
J Med Chem ; 51(22): 7094-8, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18983139

ABSTRACT

cis-4-(Piperazin-1-yl)-5,6,7a,8,9,10,11,11a-octahydrobenzofuro[2,3-h]quinazolin-2-amine, 4 (A-987306) is a new histamine H(4) antagonist. The compound is potent in H(4) receptor binding assays (rat H(4), K(i) = 3.4 nM, human H(4) K(i) = 5.8 nM) and demonstrated potent functional antagonism in vitro at human, rat, and mouse H(4) receptors in cell-based FLIPR assays. Compound 4 also demonstrated H(4) antagonism in vivo in mice, blocking H(4)-agonist induced scratch responses, and showed anti-inflammatory activity in mice in a peritonitis model. Most interesting was the high potency and efficacy of this compound in blocking pain responses, where it showed an ED(50) of 42 mumol/kg (ip) in a rat post-carrageenan thermal hyperalgesia model of inflammatory pain.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzofurans/pharmacology , Hyperalgesia/drug therapy , Pain/prevention & control , Quinazolines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzofurans/chemical synthesis , Benzofurans/chemistry , Carrageenan , Disease Models, Animal , Drug Design , Drug Evaluation, Preclinical , Humans , Hyperalgesia/chemically induced , Ligands , Mice , Molecular Structure , Pain/physiopathology , Peritonitis/drug therapy , Quinazolines/chemical synthesis , Quinazolines/chemistry , Rats , Receptors, Histamine , Receptors, Histamine H4 , Stereoisomerism , Structure-Activity Relationship
10.
Eur J Pharmacol ; 601(1-3): 8-15, 2008 Dec 28.
Article in English | MEDLINE | ID: mdl-18977214

ABSTRACT

We have recently identified three splice isoforms of the histamine H(3) receptor in multiple brain regions of cynomolgus monkey (Macaca fascicularis). Two of the novel isoforms displayed a deletion in the third intracellular loop (H(3)(413) and H(3)(410)), the third isoform H(3)(335) displayed a deletion in the i3 intracellular loop and a complete deletion of the putative fifth transmembrane domain TM5. We have confirmed by RT-PCR the expression of full-length H(3)(445) mRNA as well as H(3)(413), H(3)(410), and H(3)(335) splice isoform mRNA in multiple monkey brain regions including the frontal, parietal and occipital cortex, parahippocampal gyrus, hippocampus, amygdala, caudate nucleus, putamen, thalamus, hypothalamus, and cerebellum. The full-length isoform H(3)(445) was predominant in all of the regions tested, followed by H(3)(335), with the H(3)(413) and H(3)(410) being of low abundance. When expressed in C6 cells, H(3)(445), H(3)(413), and H(3)(410) exhibit high affinity binding to the agonist ligand [(3)H]-(N)-alpha-methylhistamine with respective pK(D) values of 9.7, 9.7, and 9.6. As expected, the H(3)(335) isoform did not display any saturable binding with [(3)H]-(N)-alpha-methylhistamine. The histamine H(3) receptor agonists histamine, (R)-alpha-methylhistamine, imetit and proxyfan were able to activate calcium mobilization responses through H(3)(445), H(3)(413) and H(3)(410) receptors when they were co-expressed with the chimeric G alpha(qi5)-protein in HEK293 cells, while no response was elicited in cells expressing the H(3)(335) isoform. The existence of multiple H(3) receptor splice isoforms across species raises the possibility that isoform specific properties including ligand affinity, signal transduction coupling, and brain localization may differentially contribute to observed in vivo effects of histamine H(3) receptor antagonists.


Subject(s)
Gene Expression , RNA, Messenger/metabolism , Receptors, Histamine H3/metabolism , Animals , Brain/metabolism , Cell Line , Cloning, Molecular , Histamine Agonists/pharmacology , Humans , Ligands , Macaca fascicularis , Male , Protein Binding , Protein Isoforms , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Species Specificity
11.
J Med Chem ; 51(20): 6571-80, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18811133

ABSTRACT

A series of 2-aminopyrimidines was synthesized as ligands of the histamine H4 receptor (H4R). Working in part from a pyrimidine hit that was identified in an HTS campaign, SAR studies were carried out to optimize the potency, which led to compound 3, 4- tert-butyl-6-(4-methylpiperazin-1-yl)pyrimidin-2-ylamine. We further studied this compound by systematically modifying the core pyrimidine moiety, the methylpiperazine at position 4, the NH2 at position 2, and positions 5 and 6 of the pyrimidine ring. The pyrimidine 6 position benefited the most from this optimization, especially in analogs in which the 6- tert-butyl was replaced with aromatic and secondary amine moieties. The highlight of the optimization campaign was compound 4, 4-[2-amino-6-(4-methylpiperazin-1-yl)pyrimidin-4-yl]benzonitrile, which was potent in vitro and was active as an anti-inflammatory agent in an animal model and had antinociceptive activity in a pain model, which supports the potential of H 4R antagonists in pain.


Subject(s)
Histamine Antagonists/chemical synthesis , Histamine Antagonists/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Receptors, Histamine/metabolism , Animals , Biomarkers , Histamine Antagonists/chemistry , Humans , Hyperplasia/chemically induced , Hyperplasia/prevention & control , Ligands , Locomotion/drug effects , Mice , Molecular Structure , Pyrimidines/chemistry , Rats , Structure-Activity Relationship , Substrate Specificity
12.
Bioorg Med Chem Lett ; 18(1): 355-9, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18077160

ABSTRACT

Structure-activity relationships (SAR) were analyzed within a library of diverse yet simple compounds prepared as histamine H3 antagonists. The libraries were constructed with a variety of low molecular weight pyrrolidines, selected from (R)-2-methylpyrrolidine, (S)-2-methylpyrrolidine, and pyrrolidine.


Subject(s)
Histamine H3 Antagonists/chemistry , Histamine H3 Antagonists/pharmacology , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Animals , Humans , Kinetics , Rats , Structure-Activity Relationship
13.
J Biol Chem ; 282(5): 3146-56, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17121830

ABSTRACT

Salvinorin A, the active component of the hallucinogenic sage Salvia divinorum, is an apparently selective and highly potent kappa-opioid receptor (KOR) agonist. Salvinorin A is unique among ligands for peptidergic G protein-coupled receptors in being nonnitrogenous and lipid-like in character. To examine the molecular basis for the subtype-selective binding of salvinorin A, we utilized an integrated approach using chimeric opioid receptors, site-directed mutagenesis, the substituted cysteine accessibility method, and molecular modeling and dynamics studies. We discovered that helix 2 is required for salvinorin A binding to KOR and that two residues (Val-108(2.53) and Val-118(2.63)) confer subtype selectivity. Intriguingly, molecular modeling studies predicted that these loci exhibit an indirect effect on salvinorin A binding, presumably through rotation of helix 2. Significantly, and in agreement with our in silico predictions, substituted cysteine accessibility method analysis of helix 2 comparing KOR and the delta-opioid receptor, which has negligible affinity for salvinorin A, revealed that residues known to be important for salvinorin A binding exhibit a differential pattern of water accessibility. These findings imply that differences in the helical orientation of helix 2 are critical for the selectivity of salvinorin A binding to KOR and provide a structurally novel basis for ligand selectivity.


Subject(s)
Diterpenes/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Diterpenes, Clerodane , Genetic Vectors , Hallucinogens/metabolism , Humans , Kidney , Kinetics , Mice , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Radioligand Assay , Receptors, Opioid, kappa/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
14.
Mol Interv ; 6(5): 257-65, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17035666

ABSTRACT

The hallucinogenic plant Salvia divinorum (i.e., "magic mint") is a member of the Sage family that has been used for divination and shamanism by the Mazatecs. Over the past decade or so, S. divinorum has been increasingly used recreationally. The neoclerodane diterpene salvinorin A is the active component of S. divinorum, and recently, the kappa opioid receptor (KOR) has been identified, in vitro and in vivo, as its molecular target. The discovery of KOR as the molecular target of salvinorin A has opened up many opportunities for drug discovery and drug development for a number of psychiatric and non-psychiatric disorders.


Subject(s)
Diterpenes , Nervous System Diseases/drug therapy , Plant Extracts , Salvia/chemistry , Animals , Diterpenes/chemistry , Diterpenes/metabolism , Diterpenes/therapeutic use , Diterpenes, Clerodane , Drug Design , Hallucinogens/chemistry , Hallucinogens/therapeutic use , Humans , Models, Molecular , Molecular Structure , Plant Extracts/chemistry , Plant Extracts/metabolism , Plant Extracts/therapeutic use , Protein Binding , Protein Conformation , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Signal Transduction/physiology , Structure-Activity Relationship
15.
Neuropharmacology ; 50(5): 576-84, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16376953

ABSTRACT

Acute activation of D2 dopamine receptors inhibits adenylyl cyclase (EC 4.6.1.1), whereas persistent activation of these inhibitory receptors results in a compensatory increase in cyclic AMP accumulation. This sensitization of adenylyl cyclase is thought to involve enhanced Galpha(s)-adenylyl cyclase interactions; however, the absolute requirement of Galpha(s) has not been determined. The present study used a Galpha(s)-deficient cell line to examine directly the role of Galpha(s) in D2 dopamine receptor-induced sensitization of recombinant adenylyl cyclase type 1 (AC1) and 5 (AC5). In acute experiments, quinpirole activation of the D2 dopamine receptor inhibited AC1 and AC5 activity, indicating that the acute regulatory properties of AC1 and AC5 were retained in the absence of Galpha(s). Subsequent experiments revealed that short-term (2 h) activation of the D2 dopamine receptor resulted in significantly enhanced forskolin-stimulated AC1 activity in the absence of Galpha(s), whereas sensitization of forskolin-stimulated AC5 activity appeared to require Galpha(s). The Galpha(s)-independent sensitization of AC1 was explored further using AC1-selective activation protocols (A23187 and CCE) following short- and long-term agonist treatment. These studies revealed that persistent activation of D2 dopamine receptors sensitized AC1 activity to Ca2+ stimulation in cells devoid of endogenous Galpha(s) and demonstrate directly that sensitization of AC1 is Galpha(s)-independent.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Receptors, Dopamine D2/physiology , Animals , Calcimycin/pharmacology , Calcium/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP/pharmacokinetics , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Activation/drug effects , Hybrid Cells , Ionophores/pharmacology , Isoproterenol/pharmacology , Mice , Quinpirole/pharmacology , RNA, Messenger/biosynthesis , Rabbits , Reverse Transcriptase Polymerase Chain Reaction/methods , Transfection/methods , Tritium/pharmacokinetics
16.
J Org Chem ; 70(24): 10057-61, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16292839

ABSTRACT

[reaction: see text] Treatment of salvinorin A (1a) with KOH in MeOH gave the enedione 3, for which the dienone structure 7 was recently proposed. Also isolated, after methylation, were the secotriesters 4a-c. A mechanism for this unusual series of autoxidations is proposed. Surprisingly, 4a showed weak affinity at the kappa-opioid receptor. Divinatorins A-C (2a-c) showed no affinity at opioid receptors. Attempted reduction of 3 to a novel salvinorin diol (9d) was unsuccessful, but careful deacetylation of salvinorin C (9a) provided a viable route to this compound. A general method for identifying salvinorin 8-epimers by TLC is also presented.


Subject(s)
Diterpenes/chemistry , Diterpenes/chemical synthesis , Diterpenes, Clerodane , Hydroxides/chemistry , Methanol/chemistry , Molecular Conformation , Oxidation-Reduction , Potassium Compounds/chemistry , Stereoisomerism
17.
Biochemistry ; 44(24): 8643-51, 2005 Jun 21.
Article in English | MEDLINE | ID: mdl-15952771

ABSTRACT

Salvinorin A is a naturally occurring hallucinogenic diterpenoid from the plant Salvia divinorumthat selectively and potently activates kappa-opioid receptors (KORs). Salvinorin A is unique in that it is the only known lipid-like molecule that selectively and potently activates a G-protein coupled receptor (GPCR), which has as its endogenous agonist a peptide; salvinorin A is also the only known non-nitrogenous opioid receptor agonist. In this paper, we identify key residues in KORs responsible for the high binding affinity and agonist efficacy of salvinorin A. Surprisingly, we discovered that salvinorin A was stabilized in the binding pocket by interactions with tyrosine residues in helix 7 (Tyr313 and Tyr320) and helix 2 (Tyr119). Intriguingly, activation of KORs by salvinorin A required interactions with the helix 7 tyrosines Tyr312, Tyr313, and Tyr320 and with Tyr139 in helix 3. In contrast, the prototypical nitrogenous KOR agonist U69593 and the endogenous peptidergic agonist dynorphin A (1-13) showed differential requirements for these three residues for binding and activation. We also employed a novel approach, whereby we examined the effects of cysteine-substitution mutagenesis on the binding of salvinorin A and an analogue with a free sulfhydryl group, 2-thiosalvinorin B. We discovered that residues predicted to be in close proximity, especially Tyr313, to the free thiol of 2-thiosalvinorin B when mutated to Cys showed enhanced affinity for 2-thiosalvinorin B. When these findings are taken together, they imply that the diterpenoid salvinorin A utilizes unique residues within a commonly shared binding pocket to selectively activate KORs.


Subject(s)
Diterpenes/chemistry , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Amino Acid Sequence , Binding Sites , DNA, Complementary , Diterpenes, Clerodane , Humans , Kinetics , Models, Molecular , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Salvia
18.
IDrugs ; 8(6): 491-6, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15906196

ABSTRACT

An estimated 50% of currently marketed drugs target G protein-coupled receptors (GPCRs) for a wide variety of indications, including central nervous system (CNS) disorders. Although drug discovery efforts have focused on GPCRs, less than 10% of GPCRs are currently used as drug targets. Thus, GPCRs continue to represent a significant opportunity for future CNS drug development. Identifying the molecular targets of psychoactive compounds may result in the elucidation of novel targets for CNS drug discovery. This commentary will describe discovery-based approaches and provide several recent examples of novel ligand-receptor interactions discovered through systematic screening of the 'receptorome'.


Subject(s)
Central Nervous System Agents/pharmacology , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Animals , Drug Evaluation, Preclinical , Genome, Human , Humans
19.
Neuron ; 45(4): 489-96, 2005 Feb 17.
Article in English | MEDLINE | ID: mdl-15721235

ABSTRACT

The manifestations of Parkinson's disease are caused by reduced dopaminergic innervation of the striatum. Loss-of-function mutations in the DJ-1 gene cause early-onset familial parkinsonism. To investigate a possible role for DJ-1 in the dopaminergic system, we generated a mouse model bearing a germline disruption of DJ-1. Although DJ-1(-/-) mice had normal numbers of dopaminergic neurons in the substantia nigra, evoked dopamine overflow in the striatum was markedly reduced, primarily as a result of increased reuptake. Nigral neurons lacking DJ-1 were less sensitive to the inhibitory effects of D2 autoreceptor stimulation. Corticostriatal long-term potentiation was normal in medium spiny neurons of DJ-1(-/-) mice, but long-term depression (LTD) was absent. The LTD deficit was reversed by treatment with D2 but not D1 receptor agonists. Furthermore, DJ-1(-/-) mice displayed hypoactivity in the open field. Collectively, our findings suggest an essential role for DJ-1 in dopaminergic physiology and D2 receptor-mediated functions.


Subject(s)
Dopamine/deficiency , Hypokinesia/physiopathology , Oncogene Proteins/physiology , Parkinsonian Disorders/metabolism , Substantia Nigra/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Age Factors , Animals , Behavior, Animal , Blotting, Southern/methods , Blotting, Western/methods , Cell Count , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Disease Models, Animal , Dopamine Agonists/pharmacology , Dopamine Plasma Membrane Transport Proteins , Electric Stimulation/methods , Electrochemistry/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Germ-Line Mutation , Humans , Immunohistochemistry/methods , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/physiology , Parkinsonian Disorders/genetics , Protein Deglycase DJ-1 , Quinpirole/pharmacology , RNA, Messenger/biosynthesis , Radioligand Assay/methods , Receptors, Dopamine D2/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Substantia Nigra/cytology , Substantia Nigra/drug effects , Tyrosine 3-Monooxygenase/metabolism
20.
Mol Pharmacol ; 66(6): 1617-24, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15361543

ABSTRACT

Short-term activation of Galpha(i/o)-coupled receptors inhibits adenylyl cyclase, whereas persistent activation of Galpha(i/o)-coupled receptors results in a compensatory sensitization of adenylyl cyclase activity after subsequent activation by Galpha(s) or forskolin. Several indirect observations have suggested the involvement of increased Galpha(s)-adenylyl cyclase interactions in the expression of sensitization; however, evidence supporting a direct role for Galpha(s) has not been well established. In the present report, we used two genetic approaches to further examine the role of Galpha(s) in heterologous sensitization of Ca(2+)-sensitive type 1 adenylyl cyclase (AC1). In the first approach, we constructed Galpha(s)-insensitive mutants of AC1 (F293L and Y973S) that retained sensitivity to Ca2+ and forskolin activation. Persistent (2 h) activation of the D2 dopamine receptor resulted in a significant augmentation of basal or Ca(2+)- and forskolin-stimulated AC1 activity; however, sensitization of Galpha(s)-insensitive mutants of AC1 was markedly reduced compared with wild-type AC1. In the second strategy, we examined the requirement of an intact receptor-Galpha(s) signaling pathway for the expression of sensitization using dominant-negative Galpha(s) mutants (alpha3beta5 G226A/A366S or alpha3beta5 G226A/E268A/A366S) to disrupt D1 dopamine receptor activation of recombinant AC1. D1 dopamine receptor-Galpha(s) signaling was attenuated in the presence of alpha3beta5 G226A/A366S or alpha3beta5 G226A/E268A/A366S, but D2 agonist-induced sensitization of Ca(2+)-stimulated AC1 activity was not altered. Together, the present findings directly support the hypothesis that the expression of sensitization of AC1 involves Galpha(s)-adenylyl cyclase interactions.


Subject(s)
Adenylyl Cyclases/metabolism , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gs/physiology , Receptors, Dopamine D2/physiology , Amino Acid Substitution , Cell Line , Humans , Kidney , Quinpirole/pharmacology , Receptors, Dopamine D2/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...