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










Publication year range
1.
Nat Commun ; 12(1): 1616, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712620

ABSTRACT

The polyketide natural product reveromycin A (RM-A) exhibits antifungal, anticancer, anti-bone metastasis, anti-periodontitis and anti-osteoporosis activities by selectively inhibiting eukaryotic cytoplasmic isoleucyl-tRNA synthetase (IleRS). Herein, a co-crystal structure suggests that the RM-A molecule occupies the substrate tRNAIle binding site of Saccharomyces cerevisiae IleRS (ScIleRS), by partially mimicking the binding of tRNAIle. RM-A binding is facilitated by the copurified intermediate product isoleucyl-adenylate (Ile-AMP). The binding assays confirm that RM-A competes with tRNAIle while binding synergistically with L-isoleucine or intermediate analogue Ile-AMS to the aminoacylation pocket of ScIleRS. This study highlights that the vast tRNA binding site of the Rossmann-fold catalytic domain of class I aminoacyl-tRNA synthetases could be targeted by a small molecule. This finding will inform future rational drug design.


Subject(s)
Binding Sites/drug effects , Ligases/chemistry , Ligases/drug effects , Pyrans/antagonists & inhibitors , RNA, Transfer/drug effects , Spiro Compounds/antagonists & inhibitors , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/drug effects , Isoleucine , Isoleucine-tRNA Ligase/chemistry , Isoleucine-tRNA Ligase/drug effects , Ligands , Models, Molecular , Osteoporosis/drug therapy , RNA, Transfer/chemistry , Saccharomyces cerevisiae
2.
Exp Neurol ; 295: 23-35, 2017 09.
Article in English | MEDLINE | ID: mdl-28529112

ABSTRACT

Accumulating evidence supports that activation of inflammatory pathways is a crucial factor contributing to the pathogenesis of seizures. In particular, the activation of interleukin-1 beta (IL-1ß) system exerts proconvulsant effects in a large variety of seizure models. Myeloid differentiation factor 88 (MyD88) is a critical adaptor protein in the signaling cascade elicited by IL-1ß. The present study aimed to investigate the expression pattern of MyD88 in rat models of seizures and in patients with refractory temporal lobe epilepsy (TLE), and to study the role of MyD88 in epileptic seizures. Our results revealed that MyD88 was up-regulated in the hippocampus of rats in the lithium-pilocarpine model of acute seizures. Importantly, MyD88 overexpression was also significantly present in the brain from chronic epileptic rats and the temporal neocortex specimens from drug-resistant TLE patients. In the acute seizure model, both the behavioral and electrographic seizure activities were record and analyzed in rats for 90min, starting immediately after pilocarpine injection. ST2825, a synthetic MyD88 inhibitor, was administered intracerebroventricularly (2.5-5.0-10µg in 2µl) 20min before pilocarpine injection. We found that ST2825 at doses of 5µg and 10µg significantly inhibited the pilocarpine-induced behavioral and electrographic seizures. Moreover, 10µg ST2825 prevented the proconvulsant actions of IL-1ß. As previous evidence suggested that IL-1ß proconvulsant effects was mediated by enhancing the phosphorylation level of the NR2B subunit of N-methyl-d-aspartate (NMDA) receptor, we then probed whether this molecular was involved in the effect of the pharmacological inhibition. Our results revealed that 10µg ST2825 markedly reversed the increased Tyr1472-phosphorylation of the NR2B subunit of NMDA receptor observed in the proconvulsant conditions of IL-1ß and in seizures induced by pilocarpine alone. These findings indicate that altered expression of MyD88 might contribute to the pathogenesis of seizures and targeting of this adaptor protein might represent a novel therapeutic strategy to suppress seizure activities.


Subject(s)
Epilepsy/genetics , Epilepsy/physiopathology , Myeloid Differentiation Factor 88/biosynthesis , Myeloid Differentiation Factor 88/genetics , Seizures/genetics , Seizures/physiopathology , Adolescent , Adult , Animals , Anticonvulsants/pharmacology , Convulsants , Electroencephalography/drug effects , Epilepsy/chemically induced , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/physiopathology , Female , Heterocyclic Compounds, 2-Ring/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Injections, Intraventricular , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/pharmacology , Male , Middle Aged , Myeloid Differentiation Factor 88/antagonists & inhibitors , Pilocarpine , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/chemically induced , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacology , Young Adult
3.
Neuropharmacology ; 117: 1-13, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28093211

ABSTRACT

T-type voltage-gated Ca2+ channels (T-VGCCs) function in the pathophysiology of epilepsy, pain and sleep. However, their role in cognitive function remains unclear. We previously reported that the cognitive enhancer ST101, which stimulates T-VGCCs in rat cortical slices, was a potential Alzheimer's disease therapeutic. Here, we introduce a more potent T-VGCC enhancer, SAK3 (ethyl 8'-methyl-2',4-dioxo-2-(piperidin-1-yl)-2'H-spiro[cyclopentane-1,3'-imidazo [1,2-a]pyridin]-2-ene-3-carboxylate), and characterize its pharmacological properties in brain. Based on whole cell patch-clamp analysis, SAK3 (0.01-10 nM) significantly enhanced Cav3.1 currents in neuro2A cells ectopically expressing Cav3.1. SAK3 (0.1-10 nM nM) also enhanced Cav3.3 but not Cav3.2 currents in the transfected cells. Notably, Cav3.1 and Cav3.3 T-VGCCs were localized in cholinergic neurve systems in hippocampus and in the medial septum. Indeed, acute oral administration of SAK3 (0.5 mg/kg, p.o.), but not ST101 (0.5 mg/kg, p.o.) significantly enhanced acetylcholine (ACh) release in the hippocampal CA1 region of naïve mice. Moreover, acute SAK3 (0.5 mg/kg, p.o.) administration significantly enhanced hippocampal ACh levels in olfactory-bulbectomized (OBX) mice, rescuing impaired memory-related behaviors. Treatment of OBX mice with the T-VGCC-specific blocker NNC 55-0396 (12.5 mg/kg, i.p.) antagonized both enhanced ACh release and memory improvements elicited by SAK3 administration. We also observed that SAK3-induced ACh releases were significantly blocked in the hippocampus from Cav3.1 knockout (KO) mice. These findings suggest overall that T-VGCCs play a key role in cognition by enhancing hippocampal ACh release and that the cognitive enhancer SAK3 could be a candidate therapeutic in Alzheimer's disease.


Subject(s)
Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/physiology , Imidazoles/pharmacology , Spiro Compounds/pharmacology , Acetylcholine/metabolism , Animals , Behavior, Animal/drug effects , Benzimidazoles , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Calcium Channels, T-Type/genetics , Cells, Cultured , Cholinergic Neurons/physiology , Cyclopropanes , Dose-Response Relationship, Drug , Imidazoles/antagonists & inhibitors , Indans/pharmacology , Male , Memory/drug effects , Mice , Mice, Knockout , Naphthalenes , Nootropic Agents/pharmacology , Olfactory Bulb/surgery , Septal Nuclei/physiology , Spiro Compounds/antagonists & inhibitors
4.
Cell Death Dis ; 7: e2307, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27441659

ABSTRACT

Ferroptosis is an iron-dependent, oxidative cell death, and is distinct from apoptosis, necrosis and autophagy. In this study, we demonstrated that lysosome disrupting agent, siramesine and a tyrosine kinase inhibitor, lapatinib synergistically induced cell death and reactive oxygen species (ROS) in MDA MB 231, MCF-7, ZR-75 and SKBr3 breast cancer cells over a 24 h time course. Furthermore, the iron chelator deferoxamine (DFO) significantly reduced cytosolic ROS and cell death following treatment with siramesine and lapatinib. Furthermore, we determined that FeCl3 levels were elevated in cells treated with siramesine and lapatinib indicating an iron-dependent cell death, ferroptosis. To confirm this, we treated cells with a potent inhibitor of ferroptosis, ferrastatin-1 that effectively inhibited cell death following siramesine and lapatinib treatment. The increase levels of iron could be due to changes in iron transport. We found that the expression of transferrin, which is responsible for the transport of iron into cells, is increased following treatment with lapatinib alone or in combination with siramesine. Knocking down of transferrin resulted in decreased cell death and ROS after treatment. In addition, ferroportin-1 (FPN) is an iron transport protein, responsible for removal of iron from cells. We found its expression is decreased after treatment with siramesine alone or in combination with lapatinib. Overexpression FPN resulted in decreased ROS and cell death whereas knockdown of FPN increased cell death after siramesine and lapatinib treatment. This indicates a novel induction of ferroptosis through altered iron regulation by treating breast cancer cells with a lysosome disruptor and a tyrosine kinase inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Indoles/pharmacology , Iron/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Reactive Oxygen Species/agonists , Spiro Compounds/pharmacology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Cyclohexylamines/pharmacology , Deferoxamine/pharmacology , Drug Synergism , Female , Ferritins/genetics , Ferritins/metabolism , Gene Expression Regulation , Humans , Imidazoles/pharmacology , Indoles/antagonists & inhibitors , Iron Chelating Agents/pharmacology , Lapatinib , Lysosomes/drug effects , MCF-7 Cells , Phenylenediamines/pharmacology , Quinazolines/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Signal Transduction , Spiro Compounds/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transferrin/genetics , Transferrin/metabolism
5.
Eur J Pharmacol ; 754: 61-5, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25697471

ABSTRACT

Peripheral receptors may contribute to the effects of systemically administered centrally available analgesics. In the present study, we analysed the effect of local peripheral injection of the nociceptin/orphanin FQ peptide (NOP) receptor agonist Ro65-6570 and compared it to the µ-opioid peptide (MOP) receptor agonist morphine in streptozotocin-induced diabetic polyneuropathy in rats. Ro65-6570 and morphine were injected intraplantarly into the hind paw of diabetic rats, and mechanical withdrawal thresholds were determined in both paws (ipsi- and contralateral to the injection site). Ro65-6570 in the dose range of 7.1-71.4nmol/animal showed antihyperalgesic effects with maximal efficacy of 57.1±15.4% maximal possible effect (MPE) at the dose of 23.8nmol/animal. Intraplantar administration of morphine showed dose-dependent antihyperalgesic effects in the dose range of 25.8-257.8nmol/animal in a similar efficacy range with a maximal efficacy of 76.0±12.1% MPE at the dose of 257.8nmol/animal. Both compounds did not induce overt confounding side effects across the tested dose range. The NOP receptor antagonist J-113397 and the MOP receptor antagonist naloxone, intraplantarly co-administered with the respective agonists, selectively and completely prevented the antihyperalgesic action of the respective NOP and MOP receptor agonist. These results indicate that the activation of peripheral NOP and MOP receptors by Ro65-6570 and morphine, respectively, mediated antihyperalgesic effects in rats with diabetic polyneuropathy.


Subject(s)
Analgesics/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetic Neuropathies/drug therapy , Hyperalgesia/drug therapy , Imidazoles/therapeutic use , Morphine/therapeutic use , Receptors, Opioid, mu/agonists , Receptors, Opioid/agonists , Spiro Compounds/therapeutic use , Analgesics/pharmacology , Animals , Benzimidazoles/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/complications , Dose-Response Relationship, Drug , Hyperalgesia/complications , Imidazoles/antagonists & inhibitors , Imidazoles/pharmacology , Male , Morphine/antagonists & inhibitors , Morphine/pharmacology , Naloxone/pharmacology , Pain Threshold/drug effects , Piperidines/pharmacology , Rats , Receptors, Opioid/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacology , Nociceptin Receptor
6.
Drug Alcohol Depend ; 122(1-2): 119-26, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-21992874

ABSTRACT

BACKGROUND: Combining extinction training with cognitive-enhancing pharmacotherapy represents a novel strategy for improving the efficacy of exposure therapy for drug relapse prevention. We investigated if the selective glycine transporter-1 (GlyT-1) inhibitor RO4543338 could facilitate extinction of cocaine-conditioned responses and attenuate reacquisition of cocaine-seeking behavior. METHODS: Rats were trained to self-administer cocaine (0.3mg/kg), which was associated with a 2-s light cue under a second-order schedule of i.v. drug injection. Rats received vehicle, 30 or 45mg/kg of RO4543338 prior to three 1-h extinction-training sessions spaced at weekly intervals. Responses were extinguished by substituting saline for cocaine while maintaining response-contingent cue presentations. Reacquisition of cocaine-seeking behavior during self-administration sessions began 1 week after the last extinction session. Control experiments were conducted under conditions that precluded explicit extinction of cocaine-conditioned responses. RESULTS: Compared to vehicle, 30 and 45mg/kg RO4543338 significantly decreased responding early in extinction training and during subsequent reacquisition sessions. The latter effect persisted for at least five sessions. In control studies, reacquisition of cocaine-seeking behavior was not altered when RO4543338 was administered either prior to weekly self-administration control sessions or prior to weekly control sessions in which cocaine and cues were omitted and the levers retracted. CONCLUSIONS: As the GlyT-1 inhibitor facilitated cocaine-cue extinction learning and attenuated subsequent reacquisition of cocaine-seeking behavior, this class of compounds may have utility as a pharmacological adjunct to cocaine-cue exposure therapy in addicts.


Subject(s)
Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/drug effects , Extinction, Psychological/drug effects , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Imidazolidines/antagonists & inhibitors , Spiro Compounds/antagonists & inhibitors , Animals , Conditioning, Operant/drug effects , Cues , Male , Rats , Rats, Wistar , Self Administration
7.
Neuropsychopharmacology ; 37(2): 378-89, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21881568

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) peptide and its receptor (NOP receptor) have been implicated in a host of brain functions and diseases, but the contribution of this neuropeptide system to behavioral processes of relevance to psychosis has not been investigated. We examined the effect of the NOP receptor antagonists, Compound 24 and J-113397, and the synthetic agonist, Ro64-6198, on time function (2-2000 ms prepulse-pulse intervals) of acoustic (80 dB/10 ms prepulse) and visual (1000 Lux/20 ms prepulse) prepulse inhibition of startle reflex (PPI), a preattentive sensory filtering mechanism that is central to perceptual and mental integration. The effects of the dopamine D1-like receptor agonist, SKF-81297, the D2-like receptor agonist, quinelorane, and the mixed D1/D2 agonist, apomorphine, were studied for comparison. When acoustic stimulus was used as prepulse, BALB/cByJ mice displayed a monotonic time function of PPI, and consistent with previous studies, apomorphine and SKF-81279 induced PPI impairment, whereas quinelorane had no effect. None of the NOP receptor ligands was effective on acoustic PPI. When flash light was used as prepulse, BALB/cByJ mice displayed a bell-shaped time function of PPI and all dopamine agonists were active. Ro64-6198 was also effective in reducing visual PPI. NOP receptor antagonists showed no activity but blocked disruptive effect of Ro64-6198. Finally, coadministration of the typical antipsychotic, haloperidol, attenuated PPI impairment induced by Ro64-6198, revealing involvement of a dopaminergic component. These findings show that pharmacological stimulation of NOP or dopamine D2-like receptors is more potent in disrupting visual than acoustic PPI in mice, whereas D1-like receptor activation disrupts both. They further suggest that dysfunction of N/OFQ transmission may be implicated in the pathogenesis of psychotic manifestations.


Subject(s)
Dopamine Agonists/pharmacology , Receptors, Dopamine/physiology , Receptors, Opioid/physiology , Sensory Gating/physiology , Acoustic Stimulation/methods , Animals , Benzimidazoles/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Haloperidol/pharmacology , Imidazoles/antagonists & inhibitors , Imidazoles/pharmacology , Lysine/analogs & derivatives , Lysine/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Narcotic Antagonists , Photic Stimulation/methods , Piperidines/pharmacology , Receptors, Opioid/agonists , Reflex, Startle/drug effects , Reflex, Startle/physiology , Sensory Gating/drug effects , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacology , Nociceptin Receptor
8.
Nat Chem Biol ; 7(9): 639-47, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822274

ABSTRACT

Cephalostatin 1, OSW-1, ritterazine B and schweinfurthin A are natural products that potently, and in some cases selectively, inhibit the growth of cultured human cancer cell lines. The cellular targets of these small molecules have yet to be identified. We have discovered that these molecules target oxysterol binding protein (OSBP) and its closest paralog, OSBP-related protein 4L (ORP4L)--proteins not known to be involved in cancer cell survival. OSBP and the ORPs constitute an evolutionarily conserved protein superfamily, members of which have been implicated in signal transduction, lipid transport and lipid metabolism. The functions of OSBP and the ORPs, however, remain largely enigmatic. Based on our findings, we have named the aforementioned natural products ORPphilins. Here we used ORPphilins to reveal new cellular activities of OSBP. The ORPphilins are powerful probes of OSBP and ORP4L that will be useful in uncovering their cellular functions and their roles in human diseases.


Subject(s)
Biological Products/pharmacology , Cholestenones/pharmacology , Neoplasms/metabolism , Phenazines/pharmacology , Receptors, Steroid/metabolism , Saponins/pharmacology , Spiro Compounds/pharmacology , Steroids/pharmacology , Biological Products/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cholestenones/antagonists & inhibitors , Humans , Hydroxycholesterols/pharmacology , Lipid Metabolism/drug effects , Phenazines/antagonists & inhibitors , Receptors, Steroid/genetics , Saponins/antagonists & inhibitors , Signal Transduction/drug effects , Sphingomyelins/biosynthesis , Spiro Compounds/antagonists & inhibitors , Steroids/antagonists & inhibitors , Stilbenes/antagonists & inhibitors , Stilbenes/pharmacology
9.
Int J Neuropsychopharmacol ; 14(7): 977-89, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21029514

ABSTRACT

The nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor is a non-opioid branch of the opioid receptor family implicated in several neurological and psychological disorders, such as pain, anxiety, depression, involuntary movement, addiction, seizure and dementia. Heterogeneity of NOP receptors has been proposed based on the findings of splicing variants and from binding and functional studies. We have previously reported that Ro 64-6198, a NOP receptor agonist, activated a subset, but not all, of N/OFQ-sensitive NOP receptors in midbrain ventrolateral periaqueductal grey (vlPAG). In this study, we found that a new NOP receptor ligand, (+)-5a Compound ((3aS, 6aR)-1-(cis-4-isopropylcyclohexyl)-5'-methyl-2'-phenylhexahydrospiro[piperidine-4,1'-pyrrolo[3, 4-c]pyrrole]), also activated a subset of NOP receptors in vlPAG neurons. (+)-5a Compound (0.1-30 µm) concentration-dependently activated G-protein-coupled inwardly-rectifying potassium (GIRK) channels mediated through the NOP receptors in about 35% of the recorded vlPAG neurons. (+)-5a Compound (EC50: 605 nm) was less potent (1/12) and efficacious (47%) than N/OFQ. In (+)-5a Compound-insensitive neurons, Ro 64-6198 was also ineffective, and vice versa, but N/OFQ activated GIRK channels through NOP receptors. In (+)-5a Compound-sensitive neurons, (+)-5a Compound precluded the effect of Ro 64-6198. Immunofluorecent and morphometric studies showed that most of the (+)-5a Compound-sensitive neurons were multipolar with intensive dendritic arborization and immunoreactive to glutamic acid decarboxylase-67. It is suggested that (+)-5a Compound activates a subset of NOP receptors, similar to the Ro 64-6198-sensitive subset, in the vlPAG neurons which are mostly GABAergic. These results further support the presence of functional heterogeneity of NOP receptors in the midbrain PAG.


Subject(s)
Periaqueductal Gray/drug effects , Periaqueductal Gray/physiology , Receptors, Opioid/physiology , Animals , Dose-Response Relationship, Drug , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Glutamate Decarboxylase/metabolism , Imidazoles/antagonists & inhibitors , Imidazoles/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Opioid Peptides/pharmacology , Opioid Peptides/physiology , Patch-Clamp Techniques , Periaqueductal Gray/cytology , Periaqueductal Gray/metabolism , Pyridines/pharmacology , Rats , Rats, Wistar , Receptors, Opioid/agonists , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacology , Nociceptin Receptor , Nociceptin
10.
Br J Pharmacol ; 141(1): 132-40, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14662736

ABSTRACT

1. The ORL1 agonists nociceptin and Ro 64-6198 were compared in their ability to modify spontaneous locomotor activity in male NMRI mice not habituated to the test environment. 2. Higher doses of nociceptin (>5 nmol i.c.v.) reduced whereas lower doses (<1 nmol i.c.v.) stimulated locomotor activity. Both effects were blocked by the putative ORL1 antagonists [NPhe1]nociceptin(1-13)NH2 (10 nmol i.c.v.) and UFP101 (10 nmol, i.c.v.). The effects were also blocked by naloxone benzoylhydrazone (1 mg x kg(-1) s.c.), but not by the nonselective opioid antagonist naloxone (1 mg x kg(-1) s.c.). 3 In contrast to nociceptin, the synthetic ORL1 agonist Ro 64-6198 (0.01-1.0 mg x kg(-1) i.p.) produced monophasic inhibition of locomotor activity, which was insensitive to the treatment with [NPhe1]nociceptin(1-13)NH2 or naloxone benzoylhydrazone. Treatment with UFP101 abolished the locomotor inhibition induced by Ro 64-6198 (1.0 mg x kg(-1)), whereas naloxone (1.0 mg x kg(-1), s.c.) further increased the locomotor-inhibitory effects. 4. Naloxone benzoylhydrazone (0.3; 1.0 and 3.0 mg x kg(-1) s.c.) increased locomotor activity, although the effect was statistically significant only with the highest dose used. 5. Pretreatment with the tyrosine hydroxylase inhibitor H44-68 totally eliminated the motor-stimulatory effects of low doses of nociceptin, probably via dopamine depletion. 6. The results suggest that nociceptin stimulates locomotor activity at low doses if dopamine activity is intact. High doses of nociceptin and all the tested doses of Ro 64-6198 seem to interact with a functionally different subset of ORL1 receptors. In addition, the effects of Ro 64-6198 are modulated by tonic opioid receptor activity.


Subject(s)
Motor Activity/physiology , Naloxone/analogs & derivatives , Receptors, Opioid/physiology , Animals , Cerebrospinal Fluid/chemistry , Cerebrospinal Fluid/physiology , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Imidazoles/administration & dosage , Imidazoles/antagonists & inhibitors , Imidazoles/pharmacokinetics , Injections, Intraperitoneal , Injections, Intraventricular , Injections, Subcutaneous , Male , Methods , Methyltyrosines/administration & dosage , Methyltyrosines/pharmacokinetics , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Naloxone/administration & dosage , Naloxone/pharmacokinetics , Narcotic Antagonists , Opioid Peptides/administration & dosage , Opioid Peptides/antagonists & inhibitors , Opioid Peptides/chemistry , Opioid Peptides/pharmacokinetics , Receptors, Opioid/administration & dosage , Spiro Compounds/administration & dosage , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacokinetics , Tyrosine 3-Monooxygenase/administration & dosage , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/pharmacokinetics , Nociceptin Receptor , Nociceptin
11.
J Pharmacol Exp Ther ; 300(3): 1122-30, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861823

ABSTRACT

(2S,4R)-1-[5-Chloro-1-[(2,4-dimethoxyphenyl)sulfonyl]-3-(2-methoxy-phenyl)-2-oxo-2,3-dihydro-1H-indol-3-yl]-4-hydroxy-N,N-dimethyl-2-pyrrolidine carboxamide (SSR149415), the first selective, nonpeptide vasopressin V1b receptor antagonist yet described, has been characterized in vitro and in vivo. SSR149415 showed competitive nanomolar affinity for animal and human V1b receptors and exhibited much lower affinity for rat and human V1a, V2, and oxytocin receptors. Moreover, this compound did not interact with a large number of other receptors, enzymes, or ion channels. In vitro, SSR149415 behaved as a full antagonist and potently inhibited arginine vasopressin (AVP)-induced Ca2+ increase in Chinese hamster ovary cells expressing rat or human V1b receptors. The in vivo activity of SSR149415 has been studied in several models of elevated corticotropin secretion in conscious rats. SSR149415 inhibited exogenous AVP-induced increase in plasma corticotropin, from 3 mg/kg i.p. and 10 mg/kg p.o. upwards. Similarly, this compound antagonized AVP-potentiated corticotropin release provoked by exogenous corticoliberin at 3 mg/kg p.o. The effect lasted for more than 4 h at 10 mg/kg p.o. showing a long-lasting oral effect. SSR149415 (10 mg/kg p.o.) also blocked corticotropin secretion induced by endogenous AVP increase subsequent to body water loss. Moreover, 10 mg/kg i.p SSR149415 inhibited plasma corticotropin elevation after restraint-stress in rats by 50%. In the four-plate test, a mouse model of anxiety, SSR149415 (3 mg/kg p.o. upwards) displayed anxiolytic-like activity after acute and 7-day repeated administrations. Thus, SSR149415 is a potent, selective, and orally active V1b receptor antagonist. It represents a unique tool for exploring the functional role of V1b receptors and deserves to be clinically investigated in the field of stress and anxiety.


Subject(s)
Antidiuretic Hormone Receptor Antagonists , Adrenocorticotropic Hormone/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Arginine Vasopressin/pharmacology , CHO Cells , Cattle , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , Dehydration/metabolism , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism , In Vitro Techniques , Kinetics , Male , Morpholines/antagonists & inhibitors , Morpholines/pharmacology , Rats , Rats, Sprague-Dawley , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacology , Stress, Psychological/metabolism
12.
Neuroendocrinology ; 70(2): 101-6, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10461024

ABSTRACT

Growth hormone secretagogues (GHS) administered systemically selectively induce growth hormone (GH) release from the pituitary and the expression of Fos protein in arcuate nucleus neurons. Both the control of GH release and the expression of the GHS receptor in the arcuate nucleus are thought to be regulated, at least in part, by the negative feedback actions of GH. In this study, we utilized the immunocytochemical detection of Fos protein to examine the effects of morphine- and GH-releasing hormone (GHRH)-induced GH release on the activation of arcuate nucleus neurons following GHS administration. Given alone, two structurally different GHS induced significant amounts of Fos-LI in the arcuate nucleus of male rats, suggesting activation of cells in this region. Prior administration of morphine or GHRH significantly reduced the number of Fos-positive cells in the arcuate nucleus of rats injected with either GHS, although when given together, morphine and GHRH did not produce a greater reduction in Fos expression than when given alone. In no case was there a complete reduction in Fos expression, indicating that some arcuate nucleus neurons are not subject to the feedback effects of endogenous GH. These results provide evidence that, in the male rat, GH can feedback to the hypothalamus, altering the responsiveness of neurons involved in the central response to GHS.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Growth Hormone-Releasing Hormone/pharmacology , Hormone Antagonists , Indoles/antagonists & inhibitors , Morphine/pharmacology , Narcotics/pharmacology , Oligopeptides/antagonists & inhibitors , Spiro Compounds/antagonists & inhibitors , Animals , Electrophysiology , Feedback/drug effects , Gene Expression/drug effects , Genes, fos/drug effects , Hormones/pharmacology , Immunohistochemistry , Indoles/pharmacology , Male , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Spiro Compounds/pharmacology
13.
Neuroendocrinology ; 66(3): 188-94, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9380276

ABSTRACT

We set out to determine whether the central action of growth hormone (GH) secretagogues to induce Fos protein expression in the arcuate nucleus is influenced by central somatostatin action. Conscious male rats were injected i.v. with 100 micrograms sandostatin (octreotide, a long-acting somatostatin analogue) or saline, 10 min before an i.v. injection of either 50 micrograms GH-releasing peptide (GHRP-6), 50 micrograms MK-0677 (a non-peptide GH secretagogue) or saline. In a separate study, conscious male rats were injected i.c.v. with either 2 micrograms sandostatin or artificial cerebrospinal fluid (aCSF) vehicle 20 min before an i.v. injection of 50 micrograms GHRP-6. In all studies, rats were anaesthetized 90 min following GH secretagogue injection, perfused with fixative and the brains processed for the immunocytochemical detection of Fos protein. The number of Fos-positive nuclei detected in the arcuate nucleus of the i.v. sandostatin/i.v. GHRP-6 treated rats (28 +/- 5 nuclei/section) and the i.v. sandostatin/i.v. MK-0677-injected rats (8 +/- 2 nuclei/section) was significantly less than the i.v. saline/i.v. GHRP-6-treated group (56 +/- 5 nuclei/section) and the i.v. saline/ i.v. MK-0677-treated group (20 +/- 2 nuclei/section) respectively. Intracerebroventricular sandostatin injection attenuated the GHRP-6-induced Fos response, from 53 +/- 6 nuclei/section in the i.c.v. aCSF/i.v. GHRP-6 group, to 39 +/- 5 nuclei/section in the i.c.v. sandostatin/i.v. GHRP-6 group. Thus, the central action of GH secretagogues to induce Fos protein expression in the arcuate nucleus appears to be subject to central inhibitory control by somatostatin.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Growth Hormone/metabolism , Hormone Antagonists/pharmacology , Nerve Tissue Proteins/biosynthesis , Proto-Oncogene Proteins c-fos/biosynthesis , Somatostatin/pharmacology , Animals , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Indoles/antagonists & inhibitors , Injections, Intravenous , Injections, Intraventricular , Male , Octreotide/pharmacology , Oligopeptides/antagonists & inhibitors , Rats , Spiro Compounds/antagonists & inhibitors
14.
Article in English | MEDLINE | ID: mdl-9185324

ABSTRACT

Selective 5HT1a agonist binding to membranes from rabbit cerebral cortex was concentration-dependent and saturable; the Kd was 1.1 nM and Bmax of 480 fmols/mg protein. Scatchard as well as Hill plots were linear; the Hill coefficient was 0.96, suggesting a single, non-interacting binding site. Agonist binding was inhibited in a concentration-dependent fashion by gamma S GTP, a result consistent with the coupling of this binding site to the G protein signal transduction system. In competition experiments involving agonist and a series of agents with known affinities and specificities at 5HT1a receptors, a rank order relationship was found consistent with this binding site being a 5HT1a binding site. Direct comparisons of agonist and antagonist binding at rat cerebral cortex 5HT1a receptors and cloned human 5HT1a receptors also suggested that the rabbit binding site belongs to the 5HT1a class. The only rank order anomalies were with methiothepin in rabbit cerebral cortex, where a comparatively high Ki was observed and with buspirone in cloned human 5HT1a receptor, where a low Ki was determined; these anomalies bear further study in light of the comparative pharmacology of 5HT1a receptors. Finally, the natural product parthenolide was tested for affinity in the rabbit, rat, and human systems, where it uniformly was unable to displace agonist, suggesting that the 5HT1a receptor is not a target for this compound. Overall, these results suggest that a functional 5HT1a receptor exists in rabbit cerebral cortex.


Subject(s)
Cerebral Cortex/chemistry , Receptors, Serotonin/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/antagonists & inhibitors , Animals , Binding Sites , Binding, Competitive/physiology , CHO Cells , Cricetinae , Dioxanes/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/antagonists & inhibitors , Humans , Kinetics , Propranolol/pharmacology , Rabbits , Rats , Receptors, Serotonin, 5-HT1 , Serotonin Receptor Agonists/metabolism , Sesquiterpenes/pharmacology , Spiro Compounds/antagonists & inhibitors , Tritium
15.
Br J Pharmacol ; 122(4): 733-41, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9375971

ABSTRACT

1. We have examined the effects of the systemic administration of the selective 5-HT1A agonist alnespirone (S-20499) on in vivo 5-hydroxytryptamine (5-HT) release in the dorsal raphe nucleus, the median raphe nucleus and four forebrain areas innervated differentially by both (dorsal striatum, frontal cortex, ventral hippocampus and dorsal hippocampus). 2. Alnespirone (0.1-3 mg kg(-1), s.c.) dose-dependently reduced extracellular 5-HT in the six areas examined. In forebrain, the maximal reductions occurred in striatum and frontal cortex (maximal reduction to 23 and 29% of baseline, respectively). Those in dorsal and ventral hippocampus were more moderate (to ca 65% of baseline). In contrast, the decrease in 5-HT elicited in the median raphe nucleus was more marked than that in the dorsal raphe nucleus (to ca 30 and 60% of baseline, respectively). The selective 5-HT1A antagonist WAY-100635 (0.5 mg kg(-1), s.c.) prevented the decrease in 5-HT induced by alnespirone (0.3 mg kg(-1), s.c.) in frontal cortex. 3. 8-OH-DPAT (0.025, 0.1 and 0.3 mg kg(-1), s.c.) also reduced extracellular 5-HT in a regionally-selective manner (e.g., to 32% of baseline in striatum and to 69% in dorsal hippocampus at 0.1 mg kg(-1), s.c.). In midbrain, 8-OH-DPAT reduced the dialysate 5-HT slightly more in the median than in the dorsal raphe nucleus at all doses examined. 4. Doses of both compounds close to their respective ED50 values (0.3 mg kg(-1) alnespirone, 0.025 mg kg(-1) 8-OH-DPAT) reduced 5-HT to a comparable extent in all regions examined. However, the reductions attained at higher doses were more pronounced for 8-OH-DPAT. 5. These data show that the reduction of 5-HT release elicited by alnespirone and 8-OH-DPAT is more important in forebrain areas innervated by 5-hydroxytryptaminergic neurones of the dorsal raphe nucleus. This regional selectivity seems unlikely to be accounted for by differences in the sensitivity of 5-HT1A autoreceptors controlling 5-HT release, given the dissimilar effects of these two 5-HT1A agonists in regions rich in cell bodies and nerve terminals. This suggests the presence of complex mechanisms of control of 5-HT release by 5-HT1A receptors.


Subject(s)
8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Brain/drug effects , Extracellular Space/metabolism , Serotonin Receptor Agonists/pharmacology , Spiro Compounds/pharmacology , Animals , Area Under Curve , Brain/metabolism , Male , Microdialysis , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Spiro Compounds/antagonists & inhibitors , Spiro Compounds/pharmacokinetics
16.
Pharmacol Biochem Behav ; 55(1): 55-60, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8870038

ABSTRACT

The present experiments were performed to examine the effects of a new muscarinic M1-receptor agonist, (-)-YM796 ((-)-S-2,8-dimethyl-3-methylene-1-oxa-8-azaspiro[4.5]decane L-tartrate monohydrate), on yawning and oxytocin secretion from the posterior pituitary gland in rats YM796, at doses of 2.5-50 mg/kg (SC), elicited yawning. The yawning response was markedly increased by pretreatment with a beta-adrenoceptor antagonist, pindolol (20 mg/kg, IP), which per se did not elicit yawning. The yawning induced by YM796 (10 mg/kg, SC) in combination with pindolol (20 mg/kg, IP) was inhibited by scopolamine (0.5 mg/kg, SC), a muscarinic receptor antagonist, and pirenzcpine (300 micrograms/ rat, ICV) and EEDQ (N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline) (5 mg/kg, IP), muscarinic M1-receptor antagonists, but not by spiperone (0.5 mg/kg, SC), a dopamine D2-receptor antagonist, 4-DAMP (4-diphenylacetoxy-N-methylpiperidine methiodide) (100 micrograms/rat, ICV), a muscarinic M3-receptor antagonist, and [d(CH2)5, Tyr(Mc)2, Orn8]-vasotocin (100 ng/rat, ICV), an oxytocin receptor antagonist. YM796 at 2.5-50 mg/kg (SC) did not exert an action on prolactin levels but increased oxytocin secretion from the posterior pituitary gland in rats. This augmentation of oxytocin secretion by YM796 was inhibited by scopolamine (0.5 mg/kg, SC) and pirenzepine (3 mg/kg, SC), but not by mecamylamine (1 mg/kg, IP), a nicotinic receptor antagonist. The present findings obtained with YM796 suggest that the muscarinic M2-receptor stimulation participates in causing yawning behavior and oxytocin secretion in rats.


Subject(s)
Oxytocin/metabolism , Parasympathomimetics/pharmacology , Pituitary Gland, Posterior/metabolism , Spiro Compounds/pharmacology , Yawning/drug effects , Adrenergic beta-Antagonists/pharmacology , Animals , Dopamine Antagonists/pharmacology , In Vitro Techniques , Injections, Intraventricular , Male , Muscarinic Antagonists/pharmacology , Parasympathomimetics/administration & dosage , Pindolol/pharmacology , Pituitary Gland, Posterior/drug effects , Rats , Rats, Wistar , Receptors, Oxytocin/antagonists & inhibitors , Spiro Compounds/administration & dosage , Spiro Compounds/antagonists & inhibitors
17.
Biochem Pharmacol ; 43(4): 679-84, 1992 Feb 18.
Article in English | MEDLINE | ID: mdl-1540222

ABSTRACT

Paraherquamide was identified recently as a potent anthelmintic agent. In this paper we describe the identification and characterization of a specific, high-affinity paraherquamide binding site in a membrane preparation isolated from the free-living nematode, Caenorhabditis elegans. [3H] Paraherquamide bound specifically to C. elegans membranes with an apparent dissociation constant, Kd, of 263 nM. A series of paraherquamide analogs were examined, and their relative affinity for the paraherquamide binding site correlated with their nematocidal activity. Phenothiazines were the only other class of anthelmintics tested which inhibited specific [3H]paraherquamide binding. These results suggest that the anthelmintic activity of paraherquamide and phenothiazine is mediated via an interaction with a common binding site.


Subject(s)
Anthelmintics/pharmacology , Caenorhabditis/drug effects , Indolizines/pharmacology , Spiro Compounds/pharmacology , Animals , Binding Sites/drug effects , Binding, Competitive , Caenorhabditis/metabolism , Caenorhabditis/physiology , Indolizines/antagonists & inhibitors , Kinetics , Phenothiazines/pharmacology , Spiro Compounds/antagonists & inhibitors
18.
Psychopharmacology (Berl) ; 104(4): 444-50, 1991.
Article in English | MEDLINE | ID: mdl-1685794

ABSTRACT

In the present study, the effects of a new anxiolytic, DN-2327, were compared to those of diazepam and buspirone in rats in the elevated plus-maze test. Two indices of anxiety were obtained in this test: the number of entries into the open arms expressed as a percentage of the total number of arm entries and the percentage of time spent on the open arms. Both a typical anxiolytic, diazepam, at 2.5, 5 and 10 mg/kg, PO and a new anxiolytic, DN-2327, at 2.5 and 5 mg/kg, PO dose-dependently increased the two indices: the percentage of time spent on the open arms and the percentage of open-arm entries. On the other hand, pentylenetetrazol (PTZ) at 10 and 20 mg/kg, IP decreased the two indices dose dependently as did yohimbine at 1.5 and 3 mg/kg, IP. DN-2327 at 2.5 and 5 mg/kg, PO and diazepam at 5 and 10 mg/kg, PO dose dependently and significantly increased the two indices that were suppressed following administration of PTZ at 10 mg/kg, IP. The effects of both DN-2327, 5 mg/kg, PO, and diazepam, 10 mg/kg, PO, on the two indices were significantly antagonized by the benzodiazepine (BZD) receptor antagonist flumazenil, 20 mg/kg, IP. Buspirone (2.5-20 mg/kg, PO) did not affect either of the two responses but dose dependently decreased the number of rearings, although in the Vogel conflict test, the anti-conflict activity of buspirone was equipotent to that of diazepam and DN-2327 at the minimum effective dose (10 mg/kg, PO) of each drug.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/psychology , Buspirone/pharmacology , Diazepam/pharmacology , Exploratory Behavior/drug effects , Naphthyridines/pharmacology , Spiro Compounds/pharmacology , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Buspirone/antagonists & inhibitors , Conflict, Psychological , Diazepam/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Interactions , Electroshock , Flumazenil/pharmacology , Isoindoles , Male , Naphthyridines/antagonists & inhibitors , Pentylenetetrazole/pharmacology , Rats , Rats, Inbred Strains , Spiro Compounds/antagonists & inhibitors , Yohimbine/pharmacology
19.
Eur J Pharmacol ; 191(3): 391-400, 1990 Dec 04.
Article in English | MEDLINE | ID: mdl-1964908

ABSTRACT

The effects of MDL 73005EF (8-[2-(2,3-dihydro-1,4-benzodioxin-2-yl)methylamino]-8- azaspiro[4,5]decan-7,9-dione methyl sulphonate), a novel selective 5-HT1A receptor ligand with putative anxiolytic properties, were explored using models of central pre- and postsynaptic 5-HT1A receptor function in the male rat. MDL 73005EF dose dependently decreased the hippocampal 5-HT output measured by in vivo microdialysis in chloral hydrate-anaesthetised rats and this response was antagonised by the 5-HT1A/B receptor antagonist, pindolol. Local administration of MDL 73005EF had no effect on the hippocampal 5-HT output. MDL 73005EF failed to alter basal plasma adrenocorticotropin (ACTH) levels but, in common with pindolol, attenuated the ACTH response to the 5-HT1A receptor agonist, 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT). In contrast to 8-OH-DPAT, MDL 73005EF significantly increased plasma prolactin but apparently not through a 5-HT receptor-mediated mechanism. The results indicate that MDL 73005EF possesses mixed 5-HT1A receptor agonist/antagonist properties, acting as an agonist at presynaptic 5-HT1A receptors controlling 5-HT release and as an antagonist at postsynaptic 5-HT1A receptors mediating ACTH release.


Subject(s)
Central Nervous System/drug effects , Dioxins/pharmacology , Receptors, Serotonin/drug effects , Spiro Compounds/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/pharmacology , Animals , Dialysis , Dioxins/antagonists & inhibitors , Fenfluramine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neurosecretory Systems/drug effects , Pindolol/pharmacology , Prolactin/blood , Prolactin/pharmacology , Rats , Rats, Inbred Strains , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Spiro Compounds/antagonists & inhibitors , Synapses/drug effects , Tetrahydronaphthalenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...