Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Curr Top Behav Neurosci ; 60: 1-28, 2023.
Article in English | MEDLINE | ID: mdl-35467293

ABSTRACT

Before 1990, the multiplicity of dopamine receptors beyond D1 and D2 had remained a controversial concept, despite its substantial clinical implications, at a time when it was widely accepted that dopamine interacted with only two receptor subtypes, termed D1 and D2, differing one from the other by their pharmacological specificity and opposite effects on adenylyl cyclase. It was also generally admitted that the therapeutic efficacy of antipsychotics resulted from blockade of D2 receptors. Thanks to molecular biology techniques, the D3 receptor could be characterized as a distinct molecular entity having a restricted anatomical gene expression and different signaling, which could imply peculiar functions in controlling cognitive and emotional behaviors. Due to the structural similarities of D2 and D3 receptors, the search for D3-selective compounds proved to be difficult, but nevertheless led to the identification of fairly potent and in vitro and in vivo selective compounds. The latter permitted to confirm a role of D3 receptors in motor functions, addiction, cognition, and schizophrenia, which paved the way for the development of new drugs for the treatment of psychiatric disorders.


Subject(s)
Antipsychotic Agents , Receptors, Dopamine D3 , Humans , Receptors, Dopamine D3/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction/physiology , Antipsychotic Agents/pharmacology , Dopamine
2.
ACS Chem Biol ; 17(3): 709-722, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35227060

ABSTRACT

Inhibiting receptor tyrosine kinases is commonly achieved by two main strategies targeting either the intracellular kinase domain by low molecular weight compounds or the extracellular ligand-binding domain by monoclonal antibodies. Identifying small molecules able to inhibit RTKs at the extracellular level would be highly desirable to gain exquisite selectivity but is believed to be challenging owing to the size of RTK endogenous ligands (cytokines, growth factors) and the topology of RTK extracellular domains. We here report the high-throughput screening of the French Chemical Library (48K compounds) for extracellular inhibitors of the Fms-like tyrosine kinase 3 (FLT3) receptor tyrosine kinase, by a homogeneous time-resolved fluorescence competition assay. A total of 679 small molecular weight ligands (1.4%) were confirmed to strongly inhibit (>75%) the binding of the fluorescent labeled FLT3 ligand (FL cytokine) to FLT3 overexpressed in HEK-293 cells, at two different concentrations (5 and 20 µM). Concentration-response curves, obtained for 111 lead-like molecules, confirmed the unexpected tolerance of the FLT3 extracellular domain for low molecular weight druggable inhibitors exhibiting submicromolar potencies, chemical diversity, and promising pharmacokinetic properties. Further investigation of one hit confirmed inhibitory properties in dorsal root ganglia neurons and in a mouse model of neuropathic pain.


Subject(s)
High-Throughput Screening Assays , fms-Like Tyrosine Kinase 3 , Animals , HEK293 Cells , Humans , Ligands , Mice
3.
Eur J Pharmacol ; 890: 173635, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33065094

ABSTRACT

F17464 (N-(3-{4-[4-(8-Oxo-8H-[1,3]-dioxolo-[4,5-g]-chromen-7-yl)-butyl]-piperazin-1-yl}-phenyl)-methanesulfonamide, hydrochloride) is a new potential antipsychotic with a unique profile. The compound exhibits high affinity for the human dopamine receptor subtype 3 (hD3) (Ki = 0.17 nM) and the serotonin receptor subtype 1a (5-HT1a) (Ki = 0.16 nM) and a >50 fold lower affinity for the human dopamine receptor subtype 2 short and long form (hD2s/l) (Ki = 8.9 and 12.1 nM, respectively). [14C]F17464 dynamic studies show a slower dissociation rate from hD3 receptor (t1/2 = 110 min) than from hD2s receptor (t1/2 = 1.4 min) and functional studies demonstrate that F17464 is a D3 receptor antagonist, 5-HT1a receptor partial agonist. In human dopaminergic neurons F17464 blocks ketamine induced morphological changes, an effect D3 receptor mediated. In vivo F17464 target engagement of both D2 and 5-HT1a receptors is demonstrated in displacement studies in the mouse brain. F17464 increases dopamine release in the rat prefrontal cortex and mouse lateral forebrain - dorsal striatum and seems to reduce the effect of MK801 on % c-fos mRNA medium expressing neurons in cortical and subcortical regions. F17464 also rescues valproate induced impairment in a rat social interaction model of autism. All the neurochemistry and behavioural effects of F17464 are observed in the dose range 0.32-2.5 mg/kg i.p. in both rats and mice. The in vitro - in vivo pharmacology profile of F17464 in preclinical models is discussed in support of a therapeutic use of the compound in schizophrenia and autism.


Subject(s)
Antipsychotic Agents/pharmacology , Benzopyrans/pharmacology , Dopamine Antagonists/pharmacology , Piperazines/pharmacology , Receptors, Dopamine D3/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Antipsychotic Agents/therapeutic use , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Behavior, Animal/drug effects , Benzopyrans/therapeutic use , Biogenic Monoamines/metabolism , Brain/drug effects , Brain/metabolism , Catalepsy/drug therapy , Cells, Cultured , Dopamine/metabolism , Dopamine Antagonists/therapeutic use , Dopaminergic Neurons/drug effects , Female , Genes, fos/drug effects , Male , Mice , Neuronal Plasticity/drug effects , Piperazines/therapeutic use , Prolactin/blood , Rats, Sprague-Dawley , Receptors, Dopamine D3/metabolism , Sulfonamides/therapeutic use , Valproic Acid/toxicity
4.
Psychopharmacology (Berl) ; 237(2): 519-527, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31773210

ABSTRACT

RATIONALE: F17464, a dopamine D3 receptor antagonist with relatively high D3 selectivity (70 fold vs D2 in vitro), exhibits an antipsychotic profile in preclinical studies, and therapeutic efficacy was demonstrated in a randomized placebo-controlled clinical trial in patients with schizophrenia (Bitter et al. Neuropsychopharmacology 44(11):1917-1924, 2019). OBJECTIVE: This open-label study in healthy male subjects aimed at characterizing F17464 binding to D3/D2 receptors and the time course of receptor occupancy using positron emission tomography (PET) imaging with a D3-preferring tracer, [11C]-(+)-PHNO. METHODS: PET scans were performed at baseline and following a single 30 mg or 15 mg dose of F17464 (3 subjects/dose), and blood samples were collected for pharmacokinetic analysis. Receptor occupancy was calculated based upon reduction in binding potential of the tracer following F17464 administration. The relationship between plasma F17464 concentration and D3/D2 receptor occupancy was modeled and the plasma concentration corresponding to 50% receptor occupancy (EC50) calculated. RESULTS: Both doses of F17464 robustly blocked [11C]-(+)-PHNO D3 receptor binding, with substantial occupancy from 1 h post-administration, which increased at 6-9 h (89-98% and 79-87% for the 30 mg and 15 mg groups, respectively) and remained detectable at 22 h. In contrast, D2 binding was only modestly blocked at all time points (< 18%). F17464 exhibited a combination of rapid peripheral kinetics and hysteresis (persistence of binding 22 h post-dose despite low plasma concentration). The best estimate of the EC50 was 19 ng ml-1 (~ 40 nM). CONCLUSION: Overall, F17464 was strongly D3-selective in healthy volunteers, a unique profile for an antipsychotic candidate drug.


Subject(s)
Antipsychotic Agents/metabolism , Brain/metabolism , Carbon Radioisotopes/metabolism , Positron-Emission Tomography/methods , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Adult , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Brain/drug effects , Dopamine Agonists/metabolism , Dopamine Agonists/pharmacology , Dopamine Antagonists/metabolism , Dopamine Antagonists/pharmacology , Healthy Volunteers , Humans , Male , Middle Aged , Protein Binding/drug effects , Protein Binding/physiology , Receptors, Dopamine D3/antagonists & inhibitors , Schizophrenia/drug therapy , Schizophrenia/metabolism
5.
Neuropsychopharmacology ; 44(11): 1917-1924, 2019 10.
Article in English | MEDLINE | ID: mdl-30822774

ABSTRACT

F17464, a highly potent preferential D3 antagonist, is a novel compound in development for schizophrenia treatment. This phase II, double-blind, randomized, placebo-controlled, parallel-group study in five European countries evaluated the efficacy and safety of F17464, 20 mg twice daily, versus placebo over 6 weeks in patients with acute exacerbation of schizophrenia. Change from baseline to Day 43 of the Positive and Negative Syndrome Scale (PANSS) total score was the primary outcome. The data from 134 randomized patients (67 per group) were analyzed (efficacy/safety). Using analysis of covariance (ANCOVA) after last observation carried forward (LOCF) imputation (primary analysis), the PANSS total score reduction was statistically significantly greater for F17464 than placebo treated subjects at endpoint (p = 0.014); using ANCOVA with Multiple Imputation (MI) method, the between-group difference was in favor of F17464 but did not reach statistical significance. Differences in PANSS positive and general psychopathology subscale score, Marder positive factor score, PANSS response, and PANSS resolution criteria were also statistically significant in favor of F17464 (p values < 0.05) using the LOCF method, with similar results as for the primary analysis using the MI method. Treatment-related adverse events (AEs) were reported in 49.3% and 46.3% of patients on F17464 and placebo, respectively. The most common AEs in F17464 group: insomnia, agitation, and increased triglycerides; worsening of schizophrenia/drug ineffective was less frequent in F17464. Interestingly, no weight gain, no extrapyramidal disorder except rare akathisia were observed under F17464. This 6-week trial demonstrated therapeutic efficacy of 40 mg/day F17464 in improving symptoms of acute exacerbation of schizophrenia with a favorable safety profile.


Subject(s)
Antipsychotic Agents/therapeutic use , Dopamine Antagonists/therapeutic use , Receptors, Dopamine D3/antagonists & inhibitors , Schizophrenia/drug therapy , Adult , Akathisia, Drug-Induced , Antipsychotic Agents/adverse effects , Dopamine Antagonists/adverse effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Sleep Initiation and Maintenance Disorders/chemically induced , Treatment Outcome
6.
Nat Commun ; 9(1): 1042, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29531216

ABSTRACT

Peripheral neuropathic pain (PNP) is a debilitating and intractable chronic disease, for which sensitization of somatosensory neurons present in dorsal root ganglia that project to the dorsal spinal cord is a key physiopathological process. Here, we show that hematopoietic cells present at the nerve injury site express the cytokine FL, the ligand of fms-like tyrosine kinase 3 receptor (FLT3). FLT3 activation by intra-sciatic nerve injection of FL is sufficient to produce pain hypersensitivity, activate PNP-associated gene expression and generate short-term and long-term sensitization of sensory neurons. Nerve injury-induced PNP symptoms and associated-molecular changes were strongly altered in Flt3-deficient mice or reversed after neuronal FLT3 downregulation in wild-type mice. A first-in-class FLT3 negative allosteric modulator, discovered by structure-based in silico screening, strongly reduced nerve injury-induced sensory hypersensitivity, but had no effect on nociception in non-injured animals. Collectively, our data suggest a new and specific therapeutic approach for PNP.


Subject(s)
Peripheral Nervous System Diseases/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Blotting, Western , Cells, Cultured , Ganglia, Spinal/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Neuralgia/genetics , Neuralgia/metabolism , Peripheral Nervous System Diseases/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Sensory Receptor Cells/metabolism , fms-Like Tyrosine Kinase 3/genetics
7.
Eur J Neurosci ; 45(1): 2-19, 2017 01.
Article in English | MEDLINE | ID: mdl-27600596

ABSTRACT

This review updates the existing knowledge suggesting a role for the D3 receptor in schizophrenia and drug addiction. The D3 receptor is expressed in brain regions controlling reward, emotions, and motivation. Antipsychotics bind in vitro to the D3 receptor with similar affinity as to the D2 receptor, and occupancy of D3 receptors in vivo by these compounds given acutely at clinical dosage have been demonstrated in Positron Emission Tomography (PET) studies. The D3 receptor modulates glutamatergic pathways from the prefrontal cortex to subcortical areas, either directly by interacting with N-methyl-D-aspartate (NMDA) receptors in the nucleus accumbens, or indirectly by controlling dopamine release from ventral tegmental area neurons. In animals, D3 receptor antagonists reverse behavioral manifestations of NMDA receptor blockade and improve cognitive performances in various paradigms. Two D3 receptor-selective compounds have reached clinical trials in schizophrenia, with negative results seemingly due to insufficient target engagement; the results with a third compound, F17464, have not been disclosed yet. There is converging evidence that D3 receptors do not control the reinforcing effects of drugs of abuse (with the exception of alcohol under low requirement), but rather affects the motivation to take the drugs under high requirement, reactivity to drug-associated cues, and drug-seeking behaviors triggered by stimuli associated with relapse in humans. D3 receptor expression measured by PET is upregulated in humans with various drug addictions. A single administration of the D3 receptor-selective antagonist, GSK598809, in humans transiently alleviated craving in smokers after overnight abstinence. The clinical development of D3-selective compounds will benefit from initial assessment of target engagement through the use of PET.


Subject(s)
Brain/drug effects , Dopamine Antagonists/pharmacology , Drug-Seeking Behavior/drug effects , Receptors, Dopamine D3/metabolism , Substance-Related Disorders/metabolism , Animals , Brain/metabolism , Humans , Receptors, Dopamine D2/metabolism
8.
J Comp Neurol ; 524(14): 2776-802, 2016 10 01.
Article in English | MEDLINE | ID: mdl-26918661

ABSTRACT

GPR88 is a neuronal cerebral orphan G-protein-coupled receptor (GPCR) that has been linked to various psychiatric disorders. However, no extensive description of its localization has been provided so far. Here, we investigate the spatiotemporal expression of the GPR88 in prenatal and postnatal rat tissues by using in situ hybridization and immunohistochemistry. GPR88 protein was initially detected at embryonic day 16 (E16) in the striatal primordium. From E16-E20 to adulthood, the highest expression levels of both protein and mRNA were observed in striatum, olfactory tubercle, nucleus accumbens, amygdala, and neocortex, whereas in spinal cord, pons, and medulla GPR88 expression remains discrete. We observed an intracellular redistribution of GPR88 during cortical lamination. In the cortical plate of the developing cortex, GPR88 presents a classical GPCR plasma membrane/cytoplasmic localization that shifts, on the day of birth, to nuclei of neurons progressively settling in layers V to II. This intranuclear localization remains throughout adulthood and was also detected in monkey and human cortex as well as in the amygdala and hypothalamus of rats. Apart from the central nervous system, GPR88 was transiently expressed at high levels in peripheral tissues, including adrenal cortex (E16-E21) and cochlear ganglia (E19-P3), and also at moderate levels in retina (E18-E19) and spleen (E21-P7). The description of the GPR88 anatomical expression pattern may provide precious functional insights into this novel receptor. Furthermore, the GRP88 nuclear localization suggests nonclassical GPCR modes of action of the protein that could be relevant for cortical development and psychiatric disorders. J. Comp. Neurol. 524:2776-2802, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Nucleus/metabolism , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cytoplasm/metabolism , Gene Expression Regulation, Developmental , Receptors, G-Protein-Coupled/biosynthesis , Age Factors , Animals , Animals, Newborn , Cerebral Cortex/chemistry , Cytoplasm/chemistry , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/analysis , Young Adult
9.
Mol Cell Biol ; 36(6): 1019-31, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26787837

ABSTRACT

Palmitoylation is involved in several neuropsychiatric and movement disorders for which a dysfunctional signaling of the dopamine D3 receptor (Drd3) is hypothesized. Computational modeling of Drd3's homologue, Drd2, has shed some light on the putative role of palmitoylation as a reversible switch for dopaminergic receptor signaling. Drd3 is presumed to be palmitoylated, based on sequence homology with Drd2, but the functional attributes afforded by Drd3 palmitoylation have not been studied. Since these receptors are major targets of antipsychotic and anti-Parkinsonian drugs, a better characterization of Drd3 signaling and posttranslational modifications, like palmitoylation, may improve the prospects for drug development. Using molecular dynamics simulations, we evaluated in silico how Drd3 palmitoylation could elicit significant remodeling of the C-terminal cytoplasmic domain to expose docking sites for signaling proteins. We tested this model in cellulo by using the interaction of Drd3 with the G-alpha interacting protein (GAIP) C terminus 1 (GIPC1) as a template. From a series of biochemical studies, live imaging, and analyses of mutant proteins, we propose that Drd3 palmitoylation acts as a molecular switch for Drd3-biased signaling via a GIPC1-dependent route, which is likely to affect the mode of action of antipsychotic drugs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Palmitates/metabolism , Receptors, Dopamine D3/metabolism , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Cell Membrane/metabolism , HEK293 Cells , Humans , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Interaction Maps , Protein Transport , Receptors, Dopamine D3/analysis , Receptors, Dopamine D3/genetics , Signal Transduction
10.
Prog Brain Res ; 211: 255-75, 2014.
Article in English | MEDLINE | ID: mdl-24968784

ABSTRACT

The dopamine D3 receptor is located in the limbic area and apparently mediates selective effects on motivation to take drugs and drug-seeking behaviors, so that there has been considerable interest on the possible use of D3 receptor ligands to treat drug addiction. However, only recently selective tools allowing studying this receptor have been developed. This chapter presents an overview of findings that were presented at a symposium on the conference Dopamine 2013 in Sardinia in May 2013. Novel neurobiological findings indicate that drugs of abuse can lead to significant structural plasticity in rodent brain and that this is dependent on the availability of functional dopamine D3 autoreceptor, whose activation increased phosphorylation in the ERK pathway and in the Akt/mTORC1 pathway indicating the parallel engagement of a series of intracellular signaling pathways all involved in cell growth and survival. Preclinical findings using animal models of drug-seeking behaviors confirm that D3 antagonists have a promising profile to treat drug addiction across drugs of abuse type. Imaging the D3 is now feasible in human subjects. Notably, the development of (+)-4-propyl-9-hydroxynaphthoxazine ligand used in positron emission tomography (PET) studies in humans allows to measure D3 and D2 receptors based on the area of the brain under study. This PET ligand has been used to confirm up-regulation of D3 sites in psychostimulant users and to reveal that tobacco smoking produces elevation of dopamine at the level of D3 sites. There are now novel antagonists being developed, but also old drugs such as buspirone, that are available to test the D3 hypothesis in humans. The first results of clinical investigations are now being provided. Overall, those recent findings support further exploration of D3 ligands to treat drug addiction.


Subject(s)
Brain/metabolism , Brain/physiopathology , Receptors, Dopamine D3/antagonists & inhibitors , Substance-Related Disorders/metabolism , Substance-Related Disorders/physiopathology , Animals , Humans , Ligands
11.
PLoS One ; 9(6): e100612, 2014.
Article in English | MEDLINE | ID: mdl-24967814

ABSTRACT

BACKGROUND: GABAA receptor (GABAAR) function is maintained by an endogenous phosphorylation mechanism for which the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is the kinase. This phosphorylation is specific to the long intracellular loop I2 of the α1 subunit at two identified serine and threonine residues. The phosphorylation state is opposed by an unknown membrane-bound phosphatase, which inhibition favors the phosphorylated state of the receptor and contributes to the maintenance of its function. In cortical nervous tissue from epileptogenic areas in patients with drug-resistant epilepsies, both the endogenous phosphorylation and the functional state of the GABAAR are deficient. METHODOLOGY/PRINCIPAL FINDINGS: The aim of this study is to characterize the membrane-bound phosphatases counteracting the endogenous phosphorylation of GABAAR. We have developed a new analytical tool for in vitro detection of the phosphatase activities in cortical washed membranes by liquid chromatography coupled to mass spectrometry. The substrates are two synthetic phosphopeptides, each including one of the identified endogenous phosphorylation sites of the I2 loop of GABAAR α1 subunit. We have shown the presence of multiple and atypical phosphatases sensitive to zinc ions. Patch-clamp studies of the rundown of the GABAAR currents on acutely isolated rat pyramidal cells using the phosphatase inhibitor okadaic acid revealed a clear heterogeneity of the phosphatases counteracting the function of the GABAAR. CONCLUSION/SIGNIFICANCE: Our results provide new insights on the regulation of GABAAR endogenous phosphorylation and function by several and atypical membrane-bound phosphatases specific to the α1 subunit of the receptor. By identifying specific inhibitors of these enzymes, novel development of antiepileptic drugs in patients with drug-resistant epilepsies may be proposed.


Subject(s)
Cell Membrane/enzymology , Enzyme Assays/methods , Mass Spectrometry , Phosphoric Monoester Hydrolases/metabolism , Receptors, GABA-A/metabolism , Zinc/metabolism , Amino Acid Sequence , Animals , Cattle , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Electrophysiological Phenomena , Humans , Mice , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Okadaic Acid/metabolism , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Phosphoric Monoester Hydrolases/chemistry , Phosphorylation , Rats , Receptors, GABA-A/chemistry , Substrate Specificity
12.
Naunyn Schmiedebergs Arch Pharmacol ; 386(2): 107-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23001156

ABSTRACT

This article, based on original data as well as on previously reported preclinical and clinical data that are reviewed, describes direct and indirect interactions of the D(3) receptor with N-methyl-D-aspartate receptor (NMDA) signaling and their functional consequences and therapeutic implications for schizophrenia. D(3) receptor immunoreactivity at ultrastructural level with electron microscopy was identified at presumably glutamatergic, asymmetric synapses of the medium-sized spiny neurons of the nucleus accumbens. This finding supports the existence of a direct interaction of the D(3) receptor with glutamate, in line with previously described interactions with NMDA signaling involving Ca(2+)/calmodulin-dependent protein kinase II at post-synaptic densities (Liu et al. 2009). Indirect interactions of the D(3) receptor with glutamate could involve a negative control exerted by the D(3) receptor on mesocortical dopamine neurons and the complex regulation of the glutamatergic pyramidal cells by dopamine in the prefrontal cortex. This could be exemplified here by the regulation of pyramidal cell activity in conditions of chronic NMDA receptor blockade with dizocilpine (MK-801). BP897, a D(3) receptor-selective partial agonist, reversed the dysregulation of cortical c-fos mRNA expression and pyramidal cell hyperexcitability, as measured by paired-pulse electrophysiology. At the behavioral level, blockade of the D(3) receptor, by known D(3) receptor antagonists or the novel D(3) receptor-selective antagonist F17141, produces antipsychotic-like effects in reversing hyperactivity and social interaction deficits induced by NMDA receptor blockade by MK-801 in mice. The glutamate-D(3) receptor interactions described here offer a conceptual framework for developing new D(3) receptor-selective drugs, which may appear as an original, efficacious, and safe way to potentially indirectly target glutamate in schizophrenia.


Subject(s)
Glutamic Acid/physiology , Receptors, Dopamine D3/physiology , Schizophrenia , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Dizocilpine Maleate/pharmacology , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Male , Mice , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/ultrastructure , Piperazines/pharmacology , Rats , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology
13.
Epilepsia ; 51 Suppl 3: 43-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20618399

ABSTRACT

We have shown that the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is the kinase involved in the endogenous phosphorylation of the alpha1 subunit of the gamma-aminobutyric acid (GABA)(A) receptor (GABA(A)R), maintaining GABA(A)-R function. GABA(A)R endogenous phosphorylation is opposed by one or several atypical phosphatases. We have shown in addition, using cerebral tissue obtained during epilepsy surgery and control tissue from patients undergoing brain tumor surgery, that both endogenous phosphorylation and GABA(A)R function are significantly reduced in the "epileptogenic" cerebral cortex when compared to control. This dysfunction likely contributes to seizure generation and/or transition from the interictal to the ictal state. The therapeutic challenge is to alleviate the endogenous phosphorylation deficiency of GABA(A)R in the epileptogenic cortical tissue, either through activating the endogenous kinase activity, or inhibiting dephosphorylation of the alpha1 subunit. Following the first trail, we have shown that spermine (the most effective polyamine) increases the GAPDH kinase activity on GABA(A)R and that subsequently such modulation potentiates its function as assessed by rundown studies on isolated neurons. Following the second trail, we have developed methods to identify these atypical membrane-bound phosphatases. Their activities were detected using two synthetic phosphopeptides corresponding to the alpha1 regions of phosphorylation by GAPDH. After purification, the active fractions are submitted to proteomic analysis by nanoLC-Maldi-TOF/TOF for protein identification. Two candidate proteins have been identified, which will be used as targets for high-throughput screening in order to develop original antiepileptic molecules.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy/drug therapy , Animals , Anticonvulsants/therapeutic use , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/physiopathology , Epilepsy/etiology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/physiology , Humans , Phosphorylation/drug effects , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Spermine/physiology
14.
Sleep ; 33(7): 930-42, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20614853

ABSTRACT

STUDY OBJECTIVE: Caffeine, an adenosine A1 and A2a receptor antagonist, is a widely consumed stimulant and also used for the treatment of hypersomnia; however, the wake-promoting potency of caffeine is often not strong enough, and high doses may induce side effects. Caffeine is metabolized to paraxanthine, theobromine, and theophylline. Paraxanthine is a central nervous stimulant and exhibits higher potency at A1 and A2 receptors, but has lower toxicity and lesser anxiogenic effects than caffeine. DESIGN: We evaluated the wake-promoting efficacy of paraxanthine, caffeine, and a reference wake-promoting compound, modafinil, in a mice model of narcolepsy, a prototypical disease model of hypersomnia. Orexin/ataxin-3 transgenic (TG) and wild-type (WT) mice were subjected to oral administration (at ZT 2 and ZT14) of 3 doses of paraxanthine, caffeine, modafinil, or vehicle. RESULTS: Paraxanthine, caffeine, and modafinil significantly promoted wakefulness in both WT and narcoleptic TG mice and proportionally reduced NREM and REM sleep in both genotypes. The wake-promoting potency of 100 mg/kg p.o. of paraxanthine during the light period administration roughly corresponds to that of 200 mg/kg p.o. of modafinil. The wake-promoting potency of paraxanthine is greater and longer lasting than that of the equimolar concentration of caffeine, when the drugs were administered during the light period. The wake-promotion by paraxanthine, caffeine, and modafinil are associated with an increase in locomotor activity and body temperature. However, the higher doses of caffeine and modafinil, but not paraxanthine, induced hypothermia and reduced locomotor activity, thereby confirming the lower toxicity of paraxanthine. Behavioral evaluations of anxiety levels in WT mice revealed that paraxanthine induced less anxiety than caffeine did. CONCLUSIONS: Because it is also reported to provide neuroprotection, paraxanthine may be a better wake-promoting agent for hypersomnia associated with neurodegenerative diseases.


Subject(s)
Body Temperature/drug effects , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Motor Activity/drug effects , Narcolepsy/drug therapy , Sleep/drug effects , Theophylline/pharmacology , Animals , Ataxin-3 , Benzhydryl Compounds/pharmacology , Disease Models, Animal , Female , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Modafinil , Narcolepsy/genetics , Neuropeptides , Nuclear Proteins , Orexins , Transcription Factors
15.
Cell Signal ; 22(1): 158-65, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19781635

ABSTRACT

We developed a cellular Bioluminescent Resonance Energy Transfer (BRET) assay based on the interaction of TrkB fused to Renilla luciferase with the intracellular adaptor protein Shc fused to Enhanced Yellow Fluorescent Protein (EYFP). The TrkB agonist Brain Derived Neurotrophic Factor (BDNF) induced a maximum BRET signal as of 10 min with an EC(50) value of 1.4 nM, similar to the other endogenous agonists NT-3 and NT-4/5, 1.5 nM and 0.34 nM, respectively. Interestingly, measure of the BRET signal with increasing expression of Shc-EYFP, in the presence or absence of BDNF, suggested a conformational change of preformed TrkB/Shc complexes rather than Shc recruitment. Furthermore, the Y516F TrkB mutant deficient to bind Shc as well as the kinase-dead K572R TrkB mutant was unable to respond to BDNF and exhibited a lower basal BRET signal than that of the wild-type TrkB receptor, again suggesting a preformed complex with constitutive activity. The double YY706/707FF TrkB mutant in the kinase activation loop also showed reduced basal activity but surprisingly kept its capacity to enhance BDNF-induced interaction with Shc, though with less efficacy. The Trk selective kinase inhibitors K252a and BMS-9 blocked BDNF-induced BRET signal with similar potency (100-150 nM), the preferential c-Met inhibitor PF-2341006 being one order of magnitude less potent. Remarkably, in the absence of BDNF, K252a and BMS-9 also reduced basal activity to the level of the Y516F TrkB mutant, suggesting that these compounds were able to reduce the TrkB constitutive activity. BRET responses of mutants and to kinase inhibitors thus reveal a complex level of interaction between TrkB and Shc and suggest that this BRET assay could be of great utility to test blockers of TrkB signalling in a physiologically relevant context.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Fluorescence Resonance Energy Transfer/methods , Receptor, trkB/analysis , Receptor, trkB/metabolism , Shc Signaling Adaptor Proteins/analysis , Shc Signaling Adaptor Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Models, Molecular , Mutation , Protein Binding , Protein Structure, Tertiary , Receptor, trkB/chemistry , Receptor, trkB/genetics , Shc Signaling Adaptor Proteins/chemistry
16.
Psychopharmacology (Berl) ; 207(2): 201-12, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19756528

ABSTRACT

RATIONALE: Dimethylaminoethanol pyroglutamate (DMAE p-Glu) is a compound resulting from the reaction between dimethylaminoethanol (an indirect precursor of acetylcholine) and pyroglutamic acid (a cyclic derivative of glutamic acid having procholinergic properties and promnesic effects in both animals and man). OBJECTIVES: The present study undertook preclinical and clinical evaluations to test a potential therapeutic utility for DMAE p-Glu in cognitive impairments related to central cholinergic deficit. MATERIALS AND METHODS: In preclinical study, DMAE p-Glu was studied in rats by intracerebral microdialysis in conscious freely moving animals, on performance of rats in the Morris water maze test of spatial memory, and on the deficit in passive avoidance behavior induced by scopolamine. The clinical study examined the effect of DMAE p-Glu on cognitive deficits induced by an intravenous injection of scopolamine in healthy young male subjects. RESULTS: In rat experiments, DMAE p-Glu increased the extracellular levels of choline and acetylcholine in the medial prefrontal cortex, as assessed by intracerebral microdialysis, improved performance in a test of spatial memory, and reduced scopolamine-induced memory deficit in passive avoidance behavior. Clinical study results show that scopolamine induced a memory deficit and that DMAE p-Glu produced a significant positive effect on scores in the Buschke test, as well as a slight but significant difference on choice reaction time. CONCLUSION: These results indicate that DMAE p-Glu reduces the deleterious effect of scopolamine on long-term memory in healthy volunteers and suggest that DMAE p-Glu might be effective in reducing memory deficits in patients with cognitive impairment.


Subject(s)
Cognition Disorders/drug therapy , Deanol/analogs & derivatives , Glutamates/pharmacology , Memory Disorders/drug therapy , Adult , Animals , Avoidance Learning/drug effects , Cognition Disorders/chemically induced , Cross-Over Studies , Deanol/pharmacology , Double-Blind Method , Humans , Male , Memory Disorders/chemically induced , Microdialysis/methods , Muscarinic Antagonists/toxicity , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Scopolamine/toxicity , Young Adult
17.
Eur J Neurosci ; 30(3): 397-414, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19656174

ABSTRACT

GPR88, an orphan G protein-coupled receptor, was designated Strg/GPR88 for striatum-specific G protein-coupled receptor (K. Mizushima et al. (2000)Genomics, 69, 314-321). In this study, we focused on striatal GPR88 protein localization using a polyclonal antibody. We established that the distribution of immunoreactivity in rat brain matched that of GPR88 transcripts and provided evidence for its exclusive neuronal expression. GPR88 protein is abundant throughout the striatum of rat and primate, with expression limited to the two subsets of striatal projection medium spiny neurons (MSNs) expressing preprotachykinin-substance P or preproenkephalin mRNAs. Ultrastructural immunolabelling revealed the GPR88 concentration at post-synaptic sites along the somatodendritic compartments of MSNs, with pronounced preference for dendrites and dendritic spines. The GPR88-rich expression, in both striatal output pathways, designates this receptor as a potential therapeutic target for diseases involving dysfunction of the basal ganglia, such as Parkinson's disease. Hence, we investigated changes of GPR88 expression in a model of Parkinson's disease (unilateral 6-hydroxydopamine-lesioned rats) following repeated L-DOPA treatment. In dopamine-depleted striatum, GPR88 expression was differentially regulated, i.e. decreased in striatopallidal and increased in striatonigral MSNs. L-DOPA treatment led to a normalization of GPR88 levels through dopamine D1 and D2 receptor-mediated mechanisms in striatopallidal and striatonigral MSNs, respectively. Moreover, the removal of corticostriatal inputs, by ibotenate infusion, downregulated GPR88 in striatopallidal MSNs. These findings provide the first evidence that GPR88 is confined to striatal MSNs and indicate that L-DOPA-mediated behavioural effects in hemiparkinsonian rats may involve normalization of striatal GPR88 levels probably through dopamine receptor-mediated mechanisms and modulations of corticostriatal pathway activity.


Subject(s)
Corpus Striatum/metabolism , Neurons, Afferent/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Animals , Dendrites/drug effects , Dendrites/metabolism , Dopamine/metabolism , Dopamine Agents/pharmacology , Fluorescent Antibody Technique , Glutamine/metabolism , Haplorhini , Immunohistochemistry , In Situ Hybridization , Levodopa/pharmacology , Male , Microscopy, Electron, Transmission , Neurons, Afferent/drug effects , Parkinsonian Disorders/metabolism , Rats , Rats, Wistar
18.
J Neurochem ; 107(3): 701-11, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18761710

ABSTRACT

Mouse models of MPTP intoxication have been used extensively to explore the molecular mechanisms of Parkinson's disease. However, these models present some limitations since; (i) Dopaminergic (DA) cell death occurs rapidly in contrast to the presumably slow evolution of the disease process. (ii) Some of the key histological features of the disease such as Lewy body like inclusions and long-term inflammatory changes are lacking. Fornai et al. [Proc. Natl Acad. Sci. USA 102 (2005), 3413] suggested that continuous delivery of MPTP with Alzet osmotic minipumps may possibly circumvent these problems. Our results show, however, that MPTP infusion via Alzet osmotic minipumps (40 mg/kg/day) produces only a transient depletion in striatal dopamine (DA) without causing dopaminergic cell loss in the substantia nigra. Neuronal cell loss occurred, however, if MPTP was infused concomitantly with probenecid, an uricosuric agent which potentiates the effects of the toxin injected via the i.p. route. Even under these conditions, dopaminergic cell loss was moderate (-25%) and other neurodegenerative changes characteristic of Parkinson's disease remained undetectable.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Disease Models, Animal , Infusion Pumps , Neurotoxins/administration & dosage , Parkinsonian Disorders/chemically induced , Probenecid/administration & dosage , Adjuvants, Pharmaceutic/administration & dosage , Animals , Brain/drug effects , Brain/pathology , Chromatography, High Pressure Liquid , Dopamine/analysis , Dopamine/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced
19.
Mol Pharmacol ; 74(4): 980-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18621927

ABSTRACT

Epidemiological evidence suggests that caffeine or its metabolites reduce the risk of developing Parkinson's disease, possibly by protecting dopaminergic neurons, but the underlying mechanism is not clearly understood. Here, we show that the primary metabolite of caffeine, paraxanthine (PX; 1, 7-dimethylxanthine), was strongly protective against neurodegeneration and loss of synaptic function in a culture system of selective dopaminergic cell death. In contrast, caffeine itself afforded only marginal protection. The survival effect of PX was highly specific to dopaminergic neurons and independent of glial cell line-derived neurotrophic factor (GDNF). Nevertheless, PX had the potential to rescue dopaminergic neurons that had matured initially with and were then deprived of GDNF. The protective effect of PX was not mediated by blockade of adenosine receptors or by elevation of intracellular cAMP levels, two pharmacological effects typical of methylxanthine derivatives. Instead, it was attributable to a moderate increase in free cytosolic calcium via the activation of reticulum endoplasmic ryanodine receptor (RyR) channels. Consistent with these observations, PX and also ryanodine, the preferential agonist of RyRs, were protective in an unrelated paradigm of mitochondrial toxin-induced dopaminergic cell death. In conclusion, our data suggest that PX has a neuroprotective potential for diseased dopaminergic neurons.


Subject(s)
Caffeine/metabolism , Neuroprotective Agents/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Theophylline/pharmacology , Animals , Apoptosis/physiology , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Dopamine/physiology , Embryo, Mammalian/cytology , Fluorescent Antibody Technique, Indirect , Hydrogen-Ion Concentration , Mesencephalon/cytology , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/agonists , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification , Rats , Rats, Wistar , Ryanodine/pharmacology , Solubility , Theophylline/agonists , Theophylline/chemical synthesis , Theophylline/chemistry , Theophylline/isolation & purification
20.
Int J Neuropsychopharmacol ; 11(1): 79-92, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17559709

ABSTRACT

Growing evidence supports the involvement of brain-derived neurotrophic factor (BDNF) in mood disorders and the mechanism of action of antidepressant drugs. However, the relationship between BDNF and serotonergic signalling is poorly understood. Heterozygous mutants BDNF +/- mice were utilized to investigate the influence of BDNF on the serotonin (5-HT) system and the activity of the serotonin transporter (SERT) in the hippocampus. The zero net flux method of quantitative microdialysis revealed that BDNF +/- heterozygous mice have increased basal extracellular 5-HT levels in the hippocampus and decreased 5-HT reuptake capacity. In keeping with these results, the selective serotonin reuptake inhibitor paroxetine failed to increase hippocampal extracellular 5-HT levels in BDNF +/- mice while it produced robust effects in wild-type littermates. Using in-vitro autoradiography and synaptosome techniques, we investigated the causes of attenuated 5-HT reuptake in BDNF +/- mice. A significant decrease in [3H]citalopram-binding-site density in the CA3 subregion of the ventral hippocampus and a significant reduction in [3H]5-HT uptake in hippocampal synaptosomes, revealed mainly a decrease in SERT function. However, 5-HT1A autoreceptors were not desensitized in BDNF +/- mice. These results provide evidence that constitutive reductions in BDNF modulate SERT function reuptake in the hippocampus.


Subject(s)
Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/genetics , Hippocampus/metabolism , Serotonin/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Autoradiography , Brain Chemistry/drug effects , Citalopram , Dose-Response Relationship, Drug , Electrophysiology , Hippocampus/drug effects , Male , Mice , Mice, Knockout , Microdialysis , Paroxetine , Phenotype , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Receptor, Serotonin, 5-HT1A/drug effects , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Receptor Agonists/pharmacology , Selective Serotonin Reuptake Inhibitors , Synaptic Transmission/genetics , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...