Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Mol Pharmacol ; 91(2): 87-99, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27864425

ABSTRACT

The human histamine H3 receptor (hH3R) is subject to extensive gene splicing that gives rise to a large number of functional and nonfunctional isoforms. Despite the general acceptance that G protein-coupled receptors can adopt different ligand-induced conformations that give rise to biased signaling, this has not been studied for the H3R; further, it is unknown whether splice variants of the same receptor engender the same or differential biased signaling. Herein, we profiled the pharmacology of histamine receptor agonists at the two most abundant hH3R splice variants (hH3R445 and hH3R365) across seven signaling endpoints. Both isoforms engender biased signaling, notably for 4-[3-(benzyloxy)propyl]-1H-imidazole (proxyfan) [e.g., strong bias toward phosphorylation of glycogen synthase kinase 3ß (GSK3ß) via the full-length receptor] and its congener 3-(1H-imidazol-4-yl)propyl-(4-iodophenyl)-methyl ether (iodoproxyfan), which are strongly consistent with the former's designation as a "protean" agonist. The 80 amino acid IL3 deleted isoform hH3R365 is more permissive in its signaling than hH3R445: 2-(1H-imidazol-5-yl)ethyl imidothiocarbamate (imetit), proxyfan, and iodoproxyfan were all markedly biased away from calcium signaling, and principal component analysis of the full data set revealed divergent profiles for all five agonists. However, most interesting was the identification of differential biased signaling between the two isoforms. Strikingly, hH3R365 was completely unable to stimulate GSK3ß phosphorylation, an endpoint robustly activated by the full-length receptor. To the best of our knowledge, this is the first quantitative example of differential biased signaling via isoforms of the same G protein-coupled receptor that are simultaneously expressed in vivo and gives rise to the possibility of selective pharmacological targeting of individual receptor splice variants.


Subject(s)
Histamine Agonists/pharmacology , Receptors, Histamine H3/metabolism , Animals , Biological Assay , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Histamine Agonists/chemistry , Humans , Principal Component Analysis , Protein Isoforms/metabolism , Sequence Deletion
2.
Bioorg Med Chem ; 25(1): 38-52, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28029458

ABSTRACT

All clinically-used antipsychotics display similar affinity for both D2 (D2R) and D3 (D3R) receptors, and they likewise act as 5-HT2A receptor antagonists. They provide therapeutic benefit for positive symptoms, but no marked or consistent improvement in neurocognitive, social cognitive or negative symptoms. Since blockade of D3 and 5-HT6 (5-HT6R) receptors enhances neurocognition and social cognition, and potentially improves negative symptoms, a promising approach for improved treatment for schizophrenia would be to develop drugs that preferentially act at D3R versus D2R and likewise recognize 5-HT6R. Starting from the high affinity 5-HT6R ligands I and II, we identified compounds 11a and 14b that behave as 5-HT6R ligands with significant selectivity for D3R over D2R.


Subject(s)
Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacology , Drug Design , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Receptors, Serotonin/metabolism , Dopamine Antagonists/chemistry , Dopamine Antagonists/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Molecular Docking Simulation , Polycyclic Compounds/chemistry , Polycyclic Compounds/pharmacology , Schizophrenia/drug therapy , Serotonin Antagonists/chemistry , Serotonin Antagonists/pharmacology
3.
J Biol Chem ; 290(18): 11537-46, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25770211

ABSTRACT

Inasmuch as the neurohormone melatonin is synthetically derived from serotonin (5-HT), a close interrelationship between both has long been suspected. The present study reveals a hitherto unrecognized cross-talk mediated via physical association of melatonin MT2 and 5-HT2C receptors into functional heteromers. This is of particular interest in light of the "synergistic" melatonin agonist/5-HT2C antagonist profile of the novel antidepressant agomelatine. A suite of co-immunoprecipitation, bioluminescence resonance energy transfer, and pharmacological techniques was exploited to demonstrate formation of functional MT2 and 5-HT2C receptor heteromers both in transfected cells and in human cortex and hippocampus. MT2/5-HT2C heteromers amplified the 5-HT-mediated Gq/phospholipase C response and triggered melatonin-induced unidirectional transactivation of the 5-HT2C protomer of MT2/5-HT2C heteromers. Pharmacological studies revealed distinct functional properties for agomelatine, which shows "biased signaling." These observations demonstrate the existence of functionally unique MT2/5-HT2C heteromers and suggest that the antidepressant agomelatine has a distinctive profile at these sites potentially involved in its therapeutic effects on major depression and generalized anxiety disorder. Finally, MT2/5-HT2C heteromers provide a new strategy for the discovery of novel agents for the treatment of psychiatric disorders.


Subject(s)
Melatonin/metabolism , Protein Multimerization , Receptor, Melatonin, MT2/chemistry , Receptor, Serotonin, 5-HT2C/chemistry , Serotonin/metabolism , Signal Transduction , Acetamides/pharmacology , Arrestins/metabolism , Drug Synergism , Gene Expression Regulation/drug effects , HEK293 Cells , HeLa Cells , Humans , Melatonin/pharmacology , Protein Multimerization/drug effects , Protein Structure, Quaternary , Protein Transport/drug effects , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin/pharmacology , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Type C Phospholipases/metabolism , beta-Arrestins
4.
J Neurochem ; 136(5): 1037-51, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26685100

ABSTRACT

Dystrobrevin binding protein-1 (dysbindin-1), a candidate gene for schizophrenia, modulates cognition, synaptic plasticity and frontocortical circuitry and interacts with glutamatergic and dopaminergic transmission. Loss of dysbindin-1 modifies cellular trafficking of dopamine (DA) D2 receptors to increase cell surface expression, but its influence upon signaling has never been characterized. Further, the effects of dysbindin-1 upon closely related D3 receptors remain unexplored. Hence, we examined the impact of dysbindin-1 (isoform A) co-expression on the localization and coupling of human D2L and D3 receptors stably expressed in Chinese hamster ovary or SH-SY5Y cells lacking endogenous dysbindin-1. Dysbindin-1 co-transfection decreased cell surface expression of both D3 and D2L receptors. Further, while their affinity for DA was unchanged, dysbindin-1 reduced the magnitude and potency of DA-induced adenylate cylase recruitment/cAMP production. Dysbindin-1 also blunted the amplitude of DA-induced phosphorylation of ERK1/2 and Akt at both D2L and D3 receptors without, in contrast to cAMP, affecting the potency of DA. Interference with calveolin/clathrin-mediated processes of internalization prevented the modification by dysbindin-1 of ERK1/2 and adenylyl cyclase stimulation at D2L and D3 receptors. Finally, underpinning the specificity of the influence of dysbindin-1 on D2L and D3 receptors, dysbindin-1 did not modify recruitment of adenylyl cyclase by D1 receptors. These observations demonstrate that dysbindin-1 influences cell surface expression of D3 in addition to D2L receptors, and that it modulates activation of their signaling pathways. Accordingly, both a deficiency and an excess of dysbindin-1 may be disruptive for dopaminergic transmission, supporting its link to schizophrenia and other CNS disorders. Dysbindin-1, a candidate gene for schizophrenia, alters D2 receptors cell surface expression. We demonstrate that dysbindin-1 expression also influences cell surface levels of D3 receptors. Further, Dysbindin-1 reduces DA-induced adenylate cylase recruitment/cAMP production and modifies major signaling pathways (Akt and extracellular signal-regulated kinases1/2 (ERK1/2)) of both D2 and D3 receptors. Dysbindin-1 modulates thus D2 and D3 receptor signaling, supporting a link to schizophrenia.


Subject(s)
Adenylyl Cyclases/metabolism , Dystrophin-Associated Proteins/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Animals , CHO Cells , Carrier Proteins/metabolism , Cricetulus , Dopamine/metabolism , Dysbindin , Humans , Mice , Schizophrenia/metabolism , Signal Transduction/physiology
5.
Nat Chem Biol ; 10(7): 590-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880860

ABSTRACT

The serotonin6 receptor (5-HT6R) is a promising target for treating cognitive deficits of schizophrenia often linked to alterations of neuronal development. This receptor controls neurodevelopmental processes, but the signaling mechanisms involved remain poorly understood. Using a proteomic strategy, we show that 5-HT6Rs constitutively interact with cyclin-dependent kinase 5 (Cdk5). Expression of 5-HT6Rs in NG108-15 cells induced neurite growth and expression of voltage-gated Ca(2+) channels, two hallmarks of neuronal differentiation. 5-HT6R-elicited neurite growth was agonist independent and prevented by the 5-HT6R antagonist SB258585, which behaved as an inverse agonist. Moreover, it required receptor phosphorylation at Ser350 by Cdk5 and Cdc42 activity. Supporting a role of native 5-HT6Rs in neuronal differentiation, neurite growth of primary neurons was reduced by SB258585, by silencing 5-HT6R expression or by mutating Ser350 into alanine. These results reveal a functional interplay between Cdk5 and a G protein-coupled receptor to control neuronal differentiation.


Subject(s)
Cyclin-Dependent Kinase 5/genetics , Hippocampus/metabolism , Neurites/ultrastructure , Receptors, Serotonin/genetics , Animals , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 5/metabolism , Gene Expression Regulation, Developmental , Hippocampus/cytology , Hippocampus/growth & development , Humans , Ligands , Mice , Mutation , Neurites/metabolism , Phosphorylation , Piperazines/pharmacology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Serotonin/metabolism , Signal Transduction , Sulfonamides/pharmacology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
6.
Mol Cell Proteomics ; 13(5): 1273-85, 2014 May.
Article in English | MEDLINE | ID: mdl-24637012

ABSTRACT

The serotonin 5-HT(2A) receptor is a primary target of psychedelic hallucinogens such as lysergic acid diethylamine, mescaline, and psilocybin, which reproduce some of the core symptoms of schizophrenia. An incompletely resolved paradox is that only some 5-HT(2A) receptor agonists exhibit hallucinogenic activity, whereas structurally related agonists with comparable affinity and activity lack such a psychoactive activity. Using a strategy combining stable isotope labeling by amino acids in cell culture with enrichment in phosphorylated peptides by means of hydrophilic interaction liquid chromatography followed by immobilized metal affinity chromatography, we compared the phosphoproteome in HEK-293 cells transiently expressing the 5-HT(2A) receptor and exposed to either vehicle or the synthetic hallucinogen 1-[2,5-dimethoxy-4-iodophenyl]-2-aminopropane (DOI) or the nonhallucinogenic 5-HT(2A) agonist lisuride. Among the 5995 identified phosphorylated peptides, 16 sites were differentially phosphorylated upon exposure of cells to DOI versus lisuride. These include a serine (Ser(280)) located in the third intracellular loop of the 5-HT(2A) receptor, a region important for its desensitization. The specific phosphorylation of Ser(280) by hallucinogens was further validated by quantitative mass spectrometry analysis of immunopurified receptor digests and by Western blotting using a phosphosite specific antibody. The administration of DOI, but not of lisuride, to mice, enhanced the phosphorylation of 5-HT(2A) receptors at Ser(280) in the prefrontal cortex. Moreover, hallucinogens induced a less pronounced desensitization of receptor-operated signaling in HEK-293 cells and neurons than did nonhallucinogenic agonists. The mutation of Ser(280) to aspartic acid (to mimic phosphorylation) reduced receptor desensitization by nonhallucinogenic agonists, whereas its mutation to alanine increased the ability of hallucinogens to desensitize the receptor. This study reveals a biased phosphorylation of the 5-HT(2A) receptor in response to hallucinogenic versus nonhallucinogenic agonists, which underlies their distinct capacity to desensitize the receptor.


Subject(s)
Amphetamines/pharmacology , Hallucinogens/pharmacology , Lisuride/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Serine/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mice , Neurons/metabolism , Phosphorylation , Prefrontal Cortex/metabolism , Proteomics/methods , Signal Transduction/drug effects
7.
Bioorg Med Chem Lett ; 24(2): 510-4, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24374277

ABSTRACT

The tachykinin NK1 and NK3 receptors are a novel drug target for schizophrenia in order to treat not only the positive and cognitive symptoms, but also the associated co-morbid depression and sleep disturbances associated with the disease. A novel class of peptidomimetic derivatives based on a versatile phenylglycine central core was synthesized and tested in vitro as dual NK1/NK3 receptor antagonists. From this series emerged compounds with good NK1 receptor affinity, although only modest dual NK1/NK3 receptor affinity was observed with one of these analogs.


Subject(s)
Antipsychotic Agents/chemical synthesis , Drug Design , Neurokinin-1 Receptor Antagonists/chemical synthesis , Receptors, Neurokinin-1 , Receptors, Neurokinin-3/antagonists & inhibitors , Antipsychotic Agents/metabolism , Neurokinin-1 Receptor Antagonists/metabolism , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-3/metabolism , Structure-Activity Relationship
8.
J Biol Chem ; 287(12): 8864-78, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22291025

ABSTRACT

Human dopamine D(2long) and D(3) receptors were modified by N-terminal addition of SNAP or CLIP forms of O(6)-alkylguanine-DNA-alkyltransferase plus a peptide epitope tag. Cells able to express each of these four constructs only upon addition of an antibiotic were established and used to confirm regulated and inducible control of expression, the specificity of SNAP and CLIP tag covalent labeling reagents, and based on homogenous time-resolved fluorescence resonance energy transfer, the presence of cell surface D(2long) and D(3) receptor homomers. Following constitutive expression of reciprocal constructs, potentially capable of forming and reporting the presence of cell surface D(2long)-D(3) heteromers, individual clones were assessed for levels of expression of the constitutively expressed protomer. This was unaffected by induction of the partner protomer and the level of expression of the partner required to generate detectable cell surface D(2long)-D(3) heteromers was defined. Such homomers and heteromers were found to co-exist and using a reconstitution of function approach both homomers and heteromers of D(2long) and D(3) receptors were shown to be functional, potentially via trans-activation of associated G protein. These studies demonstrate the ability of dopamine D(2long) and D(3) receptors to form both homomers and heteromers, and show that in cells expressing each subtype a complex mixture of homomers and heteromers co-exists at steady state. These data are of potential importance both to disorders in which D(2long) and D(3) receptors are implicated, like schizophrenia and Parkinson disease, and also to drugs exerting their actions via these sites.


Subject(s)
Cell Membrane/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Cell Line , Cell Membrane/chemistry , Cell Membrane/genetics , Dimerization , Dopamine/metabolism , Humans , Protein Binding , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/genetics , Receptors, Dopamine D3/chemistry , Receptors, Dopamine D3/genetics
9.
J Neurochem ; 125(4): 532-44, 2013 May.
Article in English | MEDLINE | ID: mdl-23410496

ABSTRACT

D2/D3 dopamine receptors (D2R/D3R) agonists regulate Akt, but their effects display a complex time-course. In addition, the respective roles of D2R and D3R are not defined and downstream targets remain poorly characterized, especially in vivo. These issues were addressed here for D3R. Systemic administration of quinelorane, a D2R/D3R agonist, transiently increased phosphorylation of Akt and GSK-3ß in rat nucleus accumbens and dorsal striatum with maximal effects 10 min after injection. Akt activation was associated with phosphorylation of several effectors of the mammalian target of rapamycin complex 1 (mTORC1): p70S6 kinase, ribosomal protein-S6 (Ser240/244), and eukaryotic initiation factor-4E binding protein-1. The action of quinelorane was antagonized by a D2/D3R antagonist, raclopride, and the selective D3R antagonist S33084, inactive by themselves. Furthermore, no effect of quinerolane was seen in knock-out mice lacking D3R. In drd1a-EGFP transgenic mice, quinelorane activated Akt/GSK-3ß in both neurons expressing and lacking D1 receptor. Thus, the stimulation of D3R transiently activates the Akt/GSK-3ß pathway in the two populations of medium-size spiny neurons of the nucleus accumbens and dorsal striatum. This effect may contribute to the influence of D3R ligands on reward, cognition, and processes disrupted in schizophrenia, drug abuse, and Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , Glycogen Synthase Kinase 3/metabolism , Nucleus Accumbens/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Dopamine D3/metabolism , Animals , Corpus Striatum/drug effects , Dopamine Agonists/pharmacology , Glycogen Synthase Kinase 3 beta , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes , Nucleus Accumbens/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/genetics , Quinolines/pharmacology , Rats , Rats, Wistar , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases
10.
Biol Psychiatry Glob Open Sci ; 3(4): 1053-1061, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37881541

ABSTRACT

Background: Disrupted motivational control is a common-but poorly treated-feature of psychiatric disorders, arising via aberrant mesolimbic dopaminergic signaling. GPR88 is an orphan G protein-coupled receptor that is highly expressed in the striatum and therefore well placed to modulate disrupted signaling. While the phenotype of Gpr88 knockout mice suggests a role in motivational pathways, it is unclear whether GPR88 is involved in reward valuation and/or effort-based decision making in a sex-dependent manner and whether this involves altered dopamine function. Methods: In male and female Gpr88 knockout mice, we used touchscreen-based progressive ratio, with and without reward devaluation, and effort-related choice tasks to assess motivation and cost/benefit decision making, respectively. To explore whether these motivational behaviors were related to alterations in the striatal dopamine system, we quantified expression of dopamine-related genes and/or proteins and used [18F]DOPA positron emission tomography and GTPγ[35S] binding to assess presynaptic and postsynaptic dopamine function, respectively. Results: We showed that male and female Gpr88 knockout mice displayed greater motivational drive than wild-type mice, which was maintained following reward devaluation. Furthermore, we showed that cost/benefit decision making was impaired in male, but not female, Gpr88 knockout mice. Surprisingly, we found that Gpr88 deletion had no effect on striatal dopamine by any of the measures assessed. Conclusions: Our results highlight that GPR88 regulates motivational control but that disruption of such behaviors following Gpr88 deletion occurs independently of gross perturbations to striatal dopamine at a gene, protein, or functional level. This work provides further insights into GPR88 as a drug target for motivational disorders.

11.
J Pharmacol Exp Ther ; 340(3): 750-64, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22178752

ABSTRACT

Although most antidepressants suppress serotonin (5-HT) and/or noradrenaline reuptake, blockade of 5-HT(2C) receptors and α(2)-adrenoceptors likewise enhances monoaminergic transmission. These sites are targeted by the urea derivative N- [4-methoxy-3-(4-methylpiperazin-1-yl)phenyl]-1,2-dihydro-3-H-benzo[e]indole-3-carboxamide (S32212). S32212 was devoid of affinity for monoamine reuptake sites, yet displayed pronounced affinity (pK(i), 8.2) for constitutively active human 5-HT(2CINI) (h5-HT(2CINI)) receptors, behaving as an inverse agonist in reducing basal Gα(q) activation, [(3)H]inositol-phosphate production, and the spontaneous association of h5-HT(2CINI)-Renilla luciferase receptors with ß-arrestin2-yellow fluorescent protein. Furthermore, upon 18-h pretreatment, S32212 enhanced the plasma membrane expression of h5-HT(2CINI) receptors as visualized by confocal microscopy and quantified by enzyme-linked immunosorbent assay. Its actions were prevented by the neutral antagonist 6-chloro-5-methyl-N-[6-(2-methylpyridin-3-yloxy)pyridin-3-yl]indoline-1-carboxamide (SB242,084), which also impeded the induction by long-term exposure to S32212 of otherwise absent Ca(2+) mobilization in mouse cortical neurones. In vivo, S32212 blunted the inhibitory influence of the 5-HT(2C) agonist 2-(3-chlorobenzyloxy)-6-(1-piperazinyl)pyrazine (CP809,101) on ventrotegmental dopaminergic neurones. S32212 also blocked 5-HT-induced Gα(q) and phospholipase C activation at the h5-HT(2A) and, less potently, h5-HT(2B) receptors and suppressed the discriminative stimulus properties of the 5-HT(2A) agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane in rats. S32212 manifested marked affinity for human α(2A)- (pK(i) 7.2), α(2B)- (pK(i) 8.2), and α(2C)- (pK(i) 7.4) adrenoceptors, at which it abolished noradrenaline-induced recruitment of Gα(i3), Gα(o), adenylyl cyclase, and extracellular-regulated kinase1/2. Moreover, S32212 dose-dependently abolished the discriminative stimulus effects of the α(2)-adrenoceptor agonist (S)-spiro[(1-oxa-2-amino-3-azacyclopent-2-ene)-4,2'-(1',2',3',4'-tetrahydronaphthalene)] (S18616). Finally, S32212 displayed negligible affinity for α(1A)-adrenoceptors, histamine H(1) receptors, and muscarinic M(1) receptors. In conclusion, S32212 behaves as an inverse agonist at h5-HT(2C) receptors and as an antagonist at human α(2)-adrenoceptors (and h5-HT(2A) receptors). Its promising profile in preclinical models potentially relevant to the treatment of depression is described in J Pharmacol Exp Ther 340:765-780, 2012.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Antidepressive Agents/pharmacology , Indoles/pharmacology , Piperazines/pharmacology , Receptor, Serotonin, 5-HT2C/drug effects , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Drug Inverse Agonism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Penile Erection/drug effects , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2B/drug effects
12.
Eur J Pharmacol ; 925: 175016, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35545150

ABSTRACT

(+)-4-Propyl-9-hydroxynaphthoxazine ((+)PHNO) is a high affinity, preferential dopamine D3 versus D2 agonist employed in view of its high specificity and excellent signal-to-noise ratio as a radiotracer for positron emission tomography (PET) imaging. Surprisingly, its profile at other classes of monoamine receptor remains undocumented. In addition to hD3 and hD2L receptors, (+)PHNO revealed high affinity at hD4.4 but not hD1 or hD5 receptors. It also revealed significant affinity for several other G protein-coupled monoaminergic receptors, in particular h5-HT1A and h5-HT7. (+)PHNO behaved as a full agonist at hD4.4 and h5-HT1A receptors with potencies comparable to its actions at hD3 and hD2L receptors, and with less potency at 5-HT7 receptors. In binding assays with membranes derived from cells co-expressing hD3 and hD2L receptors and labeled with [3H]Nemonapride or [3H]Spiperone, the proportion of high affinity binding sites recognized by (+)PHNO was higher than an equivalent mixture of membranes from cells expressing hD3or hD2L receptors, suggesting that (+)PHNO promotes formation of hD3-hD2L heterodimers. Further, in cells co-expressing hD3 and hD2L receptors, (+)PHNO showed higher efficacy for inhibiting forskolin stimulated adenylyl cyclase and inducing adenylyl cyclase super-sensitization than in cells transfected with only hD2L receptors. In conclusion, (+)PHNO is a potent agonist at hD4.4, h5-HT1A and h5-HT7 as well as hD3 and hD2L receptors, and it potently activates dopamine hD3-hD2L heterodimers. These interactions should be considered when interpreting PET studies with [11C](+)PHNO and may be relevant to its functional and potential clinical properties in Parkinson's disease and other disorders.


Subject(s)
Dopamine , Receptors, Dopamine D2 , Adenylyl Cyclases , Dopamine/metabolism , Dopamine Agonists/pharmacology , Oxazines , Positron-Emission Tomography/methods , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism
13.
Mol Pharmacol ; 79(1): 91-105, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20952497

ABSTRACT

Although dopamine (DA) regulates the serine/threonine kinase Akt and its downstream substrate glycogen synthase kinase-3ß (GSK-3ß), the direct influence of dopaminergic receptors remains poorly characterized. Short-term incubation of Chinese hamster ovary (CHO)-expressed human (h)D(2L) and hD3) receptors with DA (maximal effect, 5-10 min) phosphorylated Akt (Thr308 and Ser473) and GSK-3ß (Ser9), actions blocked by the selective D2 and D3 antagonists, 3-[4-(4-chlorophenyl)-4-hydroxypiperidin-l-yl]methyl-1H-indole (L741,626) and (3aR,9bS)-N[4-(8-cyano-1,3a,4,9b-tetrahydro-3H-benzopyrano[3,4-c]pyrrole-2-yl)-butyl] (4-phenyl)benzamide (S33084), respectively. Similar findings were acquired with the specific D2/D3 receptor agonist quinelorane, which also enhanced (10 min after administration) levels of p-Akt and p-GSK-3ß in rat nucleus accumbens, an action blocked by the D2/D3 receptor antagonist raclopride. Akt and GSK-3ß phosphorylation mediated via CHO-expressed hD(2L) and hD3 receptors was prevented by pertussis toxin and by inhibitors of insulin-like growth factor-1 receptors as well as phosphatidylinositol 3-kinase and Src. Likewise, chelation of intracellular Ca²+ and interference with an "atypical" phorbol ester-insensitive protein kinase C (PKC) abolished recruitment of Akt and GSK-3ß. Inactivation of PKCµ blocked Akt and GSK-3ß phosphorylation at hD(2L) receptors. However, blockade of conventional PKC isoforms attenuated the actions of DA at hD3 receptors only. Furthermore, phospholipase C (PLC), calmodulin, and Akt inhibitors abolished DA-induced GSK-3ß phosphorylation by hD3 receptors, whereas phosphorylation by hD(2L) receptors partially involved calmodulin, Akt, and extracellular signal-regulated kinase (ERK) 1/2. In conclusion, at both hD(2L) and hD3 receptors, DA elicited a G(i/o)- and Ca²+/calmodulin-dependent phosphorylation of Akt and GSK-3ß via transactivation of insulin-like growth factor 1 receptor. However, significant differences were seen regarding the involvement of PLC, calmodulin, and ERK1/2.


Subject(s)
Cerebral Cortex/metabolism , Glycogen Synthase Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Signal Transduction/physiology , Animals , CHO Cells , Cloning, Molecular/methods , Cricetinae , Cricetulus , Glycogen Synthase Kinase 3 beta , Humans , Male , Phosphorylation/physiology , Rats , Rats, Wistar , Receptors, Dopamine D2/genetics , Receptors, Dopamine D3/genetics
14.
Int J Neuropsychopharmacol ; 14(6): 768-83, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20946699

ABSTRACT

The novel antidepressant, agomelatine, behaves as an agonist at melatonergic receptors, and as an antagonist at edited, human serotonin2C(VSV) receptors [h5-HT2C(VSV)Rs]. However, its actions at constitutively active 5-HT2CRs have yet to be characterized, an issue addressed herein. At unedited h5-HT2C(INI)Rs expressed in HEK-293 cells, 5-HT enhanced [35S]GTPγS binding to Gαq, whereas the inverse agonists SB206,553 and S32006 inhibited binding and, by analogy to the neutral antagonist, SB242,084, agomelatine exerted no effect alone. Mirroring these observations, 5-HT stimulated, whereas SB206,553 and S32006 inhibited, [3H]inositol phosphate formation. Both the agonist actions of 5-HT and the inverse agonist actions of SB206,553 and S32006 were abolished by agomelatine and SB242,084. As demonstrated by bioluminescence resonance energy transfer, 5-HT enhanced, whereas SB206,553 and S32006 decreased, association of 'h5-HT2C(INI)-Rluc-tagged' receptors with yellow-fluorescence-protein-coupled ß-arrestin2. These actions of 5-HT, SB206,553 and S32006 were prevented by agomelatine and SB242,084 were ineffective alone. As shown by ELISA and confocal microscopy, prolonged (18 h) exposure to SB206,553 or S32006 enhanced cell surface expression of N-terminal Flag-tagged h5-HT2C(INI)Rs: these effects were blocked by agomelatine and SB242,084, which were inactive alone. Finally, following pre-exposure to SB206,553 or S32006 for 18 h, 5-HT triggered 5-HT2CR-mediated elevations in cytosolic Ca2+ in primary cultures of mice cortical neurons. Agomelatine and SB242,084, inactive alone, prevented these actions of SB206,553 and S32006. In conclusion, agomelatine behaves as a neutral antagonist at constitutively active h5-HT2C(INI)Rs and native, cortical 5-HT2CRs. It will be of interest to determine whether the neutral antagonist properties of agomelatine are related to its favourable clinical profile of antidepressant properties with few side-effects and no discontinuation syndrome.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Receptors, Melatonin/agonists , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Arrestins/genetics , Arrestins/metabolism , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Humans , Kinetics , Ligands , Mice , Neurons/drug effects , Neurons/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Receptor, Serotonin, 5-HT2C/metabolism , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Serotonin/metabolism , beta-Arrestins
15.
Acta Neuropathol Commun ; 9(1): 177, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34727983

ABSTRACT

In Alzheimer's disease and related tauopathies, trans-synaptic transfer and accumulation of pathological tau from donor to recipient neurons is thought to contribute to disease progression, but the underlying mechanisms are poorly understood. Using complementary in vivo and in vitro models, we examined the relationship between these two processes and neuronal clearance. Accumulation of p62 (a marker of defective protein clearance) correlated with pathological tau accumulation in two mouse models of tauopathy spread; Entorhinal Cortex-tau (EC-Tau) mice where tau pathology progresses in time from EC to other brain regions, and PS19 mice injected with tau seeds. In both models and in several brain regions, p62 colocalized with human tau in a pathological conformation (MC1 antibody). In EC-Tau mice, p62 accumulated before overt tau pathology had developed and was associated with the presence of aggregation-competent tau seeds identified using a FRET-based assay. Furthermore, p62 accumulated in the cytoplasm of neurons in the dentate gyrus of EC-Tau mice prior to the appearance of MC1 positive tauopathy. However, MC1 positive tau was shown to be present at the synapse and to colocalize with p62 as shown by immuno electron microscopy. In vitro, p62 colocalized with tau inclusions in two primary cortical neuron models of tau pathology. In a three-chamber microfluidic device containing neurons overexpressing fluorescent tau, seeding of tau in the donor chamber led to tau pathology spread and p62 accumulation in both the donor and the recipient chamber. Overall, these data are in accordance with the hypothesis that the accumulation and trans-synaptic spread of pathological tau disrupts clearance mechanisms, preceding the appearance of obvious tau aggregation. A vicious cycle of tau accumulation and clearance deficit would be expected to feed-forward and exacerbate disease progression across neuronal circuits in human tauopathies.


Subject(s)
Brain/pathology , Neurons/pathology , Sequestosome-1 Protein/metabolism , Tauopathies/pathology , Animals , Brain/metabolism , Disease Progression , Humans , Mice , Neurons/metabolism , Tauopathies/metabolism
16.
Genes Brain Behav ; 20(2): e12710, 2021 02.
Article in English | MEDLINE | ID: mdl-33078498

ABSTRACT

The GPR88 orphan G protein-coupled receptor is expressed throughout the striatum, being preferentially localised in medium spiny neurons. It is also present in lower densities in frontal cortex and thalamus. Rare mutations in humans suggest a role in cognition and motor function, while common variants are associated with psychosis. Here we evaluate the influence of genetic deletion of GPR88 upon performance in translational tasks interrogating motivation, reward evaluation and cognitive function. In an automated radial arm maze 'N-back' working memory task, Gpr88 KO mice showed impaired correct responding, suggesting a role for GPR88 receptors in working memory circuitry. Associative learning performance was similar to wild-type controls in a touchscreen task but performance was impaired at the reversal learning stage, suggesting cognitive inflexibility. Gpr88 KO mice showed higher breakpoints, reduced latencies and lengthened session time in a progressive ratio task consistent with enhanced motivation. Simultaneously, locomotor hyperactivity was apparent in this task, supporting previous findings of actions of GPR88 in a cortico-striatal-thalamic motor loop. Evidence for a role of GPR88 in reward processing was demonstrated in a touchscreen-based equivalent of the Iowa gambling task. Although both Gpr88 KO and wild-type mice showed a preference for an optimum contingency choice, Gpr88 KO mice selected more risky choices at the expense of more advantageous lower risk options. Together these novel data suggest that striatal GPR88 receptors influence activity in a range of procedures integrated by prefrontal, orbitofrontal and anterior cingulate cortico-striatal-thalamic loops leading to altered cognitive, motivational and reward evaluation processes.


Subject(s)
Cognition , Memory, Short-Term , Receptors, G-Protein-Coupled/genetics , Reward , Animals , Corpus Striatum/metabolism , Corpus Striatum/physiology , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Motor Cortex/metabolism , Motor Cortex/physiology , Risk-Taking , Thalamus/metabolism , Thalamus/physiology
17.
Mol Pharmacol ; 78(5): 925-34, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20702763

ABSTRACT

In view of the therapeutic importance of dopamine D(3) and D(2) receptors, there remains considerable interest in novel ligands. Herein, we show that the tetrahydroisoquinoline 1H-indole-2-carboxylic acid {4-[2-(cyano-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl]-cyclohexyl}-amide (SB269,652) behaves as an atypical, allosteric antagonist at D(3) and D(2) receptors. Accordingly, SB269,652 potently (low nanomolar range) abolished specific binding of [(3)H]nemanopride and [(3)H]spiperone to Chinese hamster ovary-transfected D(3) receptors when radioligands were used at 0.2 and 0.5 nM, respectively. However, even at high concentrations (5 µM), SB269,652 only submaximally inhibited the specific binding of these radioligands when they were employed at 10-fold higher concentrations. By analogy, although SB269,652 potently blocked D(3) receptor-mediated activation of Gα(i3) and phosphorylation of extracellular-signal-regulated kinase (ERK)1/2, when concentrations of dopamine were increased by 10-fold, from 1 µM to 10 µM, SB269,652 only submaximally inhibited dopamine-induced stimulation of Gα(i3). SB269,652 (up to 10 µM) only weakly and partially (by approximately 20-30%) inhibited radioligand binding to D(2) receptors. Likewise, SB269,652 only submaximally suppressed D(2) receptor-mediated stimulation of Gα(i3) and Gα(qi5) (detected with the aequorin assay) and phosphorylation of ERK1/2 and Akt. Furthermore, SB269,652 only partially (35%) inhibited the dopamine-induced recruitment of ß-arrestin2 to D(2) receptors. Finally, Schild analysis using Gα(i3) assays, and studies of radioligand association and dissociation kinetics, supported allosteric actions of SB269,652 at D(3) and D(2) receptors.


Subject(s)
Dopamine D2 Receptor Antagonists , Indoles/pharmacology , Isoquinolines/pharmacology , Receptors, Dopamine D3/antagonists & inhibitors , Tetrahydroisoquinolines/pharmacology , Allosteric Regulation , Animals , Arrestins/metabolism , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Dopamine/pharmacology , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Transport , Quinpirole/pharmacology , Radioligand Assay , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/genetics , beta-Arrestins
18.
Neuropharmacology ; 162: 107829, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31666199

ABSTRACT

Parkinson's disease (PD) is characterized by progressive loss of midbrain dopaminergic neurons and treated with the dopamine precursor, 3,4-dihydroxy-l-phenylalanine (L-DOPA). Prolonged L-DOPA treatment is however associated with waning efficacy and the induction of L-DOPA induced dyskinesia (LID). GPR88 is an orphan G-protein Coupled Receptor (GPCR) expressed in dopaminoceptive striatal medium spiny neurons (MSNs) and their afferent corticostriatal glutamatergic neurons. Here, we studied the role of GPR88 in experimental parkinsonism and LID. Chronic L-DOPA administration to male GPR88 KO mice, subjected to unilateral 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle, resulted in more rotations than in their WT counterparts. Conversely, GPR88 KO mice had a lower abnormal involuntary movements (AIMs) score. These behavioral responses were accompanied by altered transcription of L-DOPA upregulated genes in lesioned GPR88 KO compared to WT striata. In accordance with a role for serotonin neurons in LID development, WT but not GPR88 KO striata exhibited 5-hydroxytryptamine displacement upon repeated L-DOPA treatment. Intact male GPR88 KO mice showed diminished tacrine-induced PD-like tremor and spontaneous hyperlocomotion. Dopamine and its metabolites were not increased in male GPR88 KO mice, but biosensor recordings revealed increased spontaneous/basal and evoked glutamate release in striata of male GPR88 KO mice. In conclusion, genetic deletion of GPR88 promotes l-DOPA-induced rotation and spontaneous locomotion yet suppresses the induction of LIDs and also reduces tremor. These data provide behavioral, neurochemical and molecular support that GPR88 antagonism may favour motor relief in PD patients without aggravating the induction of motor side effects.


Subject(s)
Antiparkinson Agents/pharmacology , Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/genetics , Levodopa/pharmacology , Locomotion/drug effects , Movement/drug effects , Parkinsonian Disorders/genetics , Receptors, G-Protein-Coupled/genetics , Adrenergic Agents/toxicity , Animals , Cholinesterase Inhibitors/toxicity , Corpus Striatum/drug effects , Dopamine Plasma Membrane Transport Proteins/genetics , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/physiopathology , GABAergic Neurons , Glutamic Acid/metabolism , Locomotion/genetics , Male , Medial Forebrain Bundle , Mice , Mice, Knockout , Neuronal Plasticity/genetics , Oxidopamine/toxicity , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/physiology , Serotonin/metabolism , Tacrine/toxicity , Tremor
19.
Neuropharmacology ; 177: 108099, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32525060

ABSTRACT

To date, there are no interventions that impede the inexorable progression of Alzheimer's disease (AD), and currently-available drugs cholinesterase (AChE) inhibitors and the N-Methyl-d-Aspartate receptor antagonist, memantine, offer only modest symptomatic benefit. Moreover, a range of mechanistically-diverse agents (glutamatergic, histaminergic, monoaminergic, cholinergic) have disappointed in clinical trials, alone and/or in association with AChE inhibitors. This includes serotonin (5-HT) receptor-6 antagonists, despite compelling preclinical observations in rodents and primates suggesting a positive influence on cognition. The emphasis has so far been on high selectivity. However, for a multi-factorial disorder like idiopathic AD, 5-HT6 antagonists possessing additional pharmacological actions might be more effective, by analogy to "multi-target" antipsychotics. Based on this notion, drug discovery programmes have coupled 5-HT6 blockade to 5-HT4 agonism and inhibition of AchE. Further, combined 5-HT6/dopamine D3 receptor (D3) antagonists are of especial interest since D3 blockade mirrors 5-HT6 antagonism in exerting broad-based pro-cognitive properties in animals. Moreover, 5-HT6 and dopamine D3 antagonists promote neurocognition and social cognition via both distinctive and convergent actions expressed mainly in frontal cortex, including suppression of mTOR over-activation and reinforcement of cholinergic and glutamatergic transmission. In addition, 5-HT6 blockade affords potential anti-anxiety, anti-depressive and anti-epileptic properties, and antagonising 5-HT6 receptors may be associated with neuroprotective ("disease-modifying") properties. Finally D3 antagonism may counter psychotic episodes and D3 receptors themselves offer a promising hub for multi-target agents. The present article reviews the status of "R and D" into multi-target 5-HT6 and D3 ligands for improved treatment of AD and other neurodegenerative disorders of aging. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Receptors, Dopamine D3/metabolism , Receptors, Serotonin/metabolism , Alzheimer Disease/psychology , Animals , Cognition/drug effects , Cognition/physiology , Cognitive Dysfunction/psychology , Dopamine Agents/administration & dosage , Dopamine Antagonists/administration & dosage , Humans , Receptors, Dopamine D3/antagonists & inhibitors , Recovery of Function/drug effects , Recovery of Function/physiology , Serotonin Antagonists/administration & dosage , Social Cognition
20.
Trends Pharmacol Sci ; 29(9): 454-64, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18676031

ABSTRACT

The broadly distributed monoaminergic neurotransmitter serotonin (5-hydroxytryptamine, 5-HT) exerts its actions via 14 classes of receptor. With the exception of 5-HT3 receptors, which gate a cation-permeable ion channel, all 5-HT receptors are coupled to G proteins. The core features of transduction via 5-HT receptors are well established, but much still remains to be learned, in particular, with regard to native populations in the brain. In this article, we survey the current knowledge of cellular signaling at G-protein-coupled 5-HT receptors and focus on several novel (and surprising) insights that have emerged over the past few years. We also highlight several promising directions for future research that should improve the understanding of serotonin signaling and ultimately permit its therapeutic exploitation in the control of central nervous system disorders. In view of the diversity of transduction mechanisms engaged by 5-HT, much of this discussion is relevant to other classes of G-protein-coupled receptors.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Receptors, Serotonin/physiology , Signal Transduction/physiology , Animals , Humans , Receptor Cross-Talk/drug effects , Receptors, G-Protein-Coupled/drug effects , Receptors, Serotonin/drug effects , Serotonin/pharmacology , Serotonin Agents/pharmacology , Serotonin Receptor Agonists/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL