Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell Signal ; 63: 109383, 2019 11.
Article in English | MEDLINE | ID: mdl-31376526

ABSTRACT

A previous study observed bell-shaped concentration-response isotherms for activation of Gαi3 G-protein subunits by high efficacy 5-HT1A receptor agonists in a Chinese hamster ovary (CHO) cell line expressing high levels of these receptors. This suggested that a signaling switch took place in that cell line (from Gαi3 to activation of other G-proteins) but it was unclear if such effects are observed for 5-HT1A receptors in other cellular environments. Here, using an antibody capture-based [35S]GTPγS binding assay for Gαi3 activation, we investigated whether efficacious 5-HT1A receptor agonists (5-HT, F13714, befiradol, NLX-101), prototypical agonists ((+) and (-)8-OH-DPAT), and partial agonist, antagonists, inverse agonists (pindolol, WAY100635, spiperone) produced similar effects on 5 cell lines expressing different levels of human 5-HT1A receptors. In membranes from cell lines (HeLa, C6-glia and CHO-low) expressing moderate receptor levels (between 1 and 4 pmol/mg of protein), 5-HT, F13714, befiradol and NLX-101 elicited classical sigmoid concentration-response isotherms. In contrast, in cell lines (CHO-high, HEK-293F) expressing high receptor levels (>9 pmol/mg) these agonists elicited bell-shaped concentration-response isotherms that peaked at nanomolar-range concentrations and then returned to baseline or below. Spiperone elicited inverse agonist inhibitory sigmoid isotherms in all membrane preparations while WAY100635 was mostly 'silent' for Gαi3 activation. The other compounds elicited diverse responses in the different cell lines suggesting that other factors, in addition to receptor expression levels, could be influencing Gαi3 activation. These data indicate that Gαi3 G-protein activation by 5-HT1A receptor ligands is highly dependent on receptor expression levels and on cellular background. Moreover, the induction of bell-shape concentration-response isotherms by 5-HT and other high-efficacy agonists is consistent with a switch in signaling to other G-protein-mediated signaling cascades, possibly elicited by receptor conformational changes.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Animals , CHO Cells , Cricetulus , HEK293 Cells , HeLa Cells , Humans , Ligands , Rats , Signal Transduction
2.
Int J Cardiol ; 209: 258-65, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26901787

ABSTRACT

BACKGROUND: The MESAMI 1 trial was a bicentric pilot study designed to test the feasibility and safety of intramyocardially injected autologous bone marrow-derived mesenchymal stromal cells (MSCs) for the treatment of ischemic cardiomyopathy. METHODS AND RESULTS: The study included 10 patients with chronic myocardial ischemia, left ventricular (LV) ejection fractions (EFs) of ≤35%, and reversible perfusion defects who were on stable optimal medical therapy and were not candidates for revascularization. MSCs (mean: 61.5×10(6) cells per patient) were injected into 10-16 viable sites at the border of the LV scar via a NOGA-guided catheter. Both primary endpoints, feasibility (successful harvest, expansion, and injection of autologous MSCs) and safety (absence of severe adverse events [SAEs]) were met in all 10 patients at the 1-month follow-up time point, and none of the SAEs reported during the full 2-year follow-up period were attributable to the study intervention. The results of secondary efficacy endpoint analyses identified significant improvements from baseline to Month 12 in LVEF (29.4±2.0% versus 35.7±2.5%; p=0.003), LV end-systolic volume (167.8±18.8mL versus 156.1±28.6mL; p=0.04), 6-min walk test and NYHA functional class. CONCLUSIONS: Our results suggest that autologous MSCs can be safely administered to the hearts of patients with severe, chronic, reversible myocardial ischemia and impaired cardiac function and may be associated with improvements in cardiac performance, LV remodeling, and patient functional status. A randomized, double blind, multicenter, placebo-controlled clinical trial (MESAMI 2) will evaluate the efficacy of this treatment approach in a larger patient population. CLINICAL TRIAL REGISTRATION: Unique identifier: NCT01076920.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Myocardial Ischemia/diagnostic imaging , Myocardial Ischemia/therapy , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/therapy , Cells, Cultured , Feasibility Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Myocardium , Pilot Projects , Prospective Studies , Single Photon Emission Computed Tomography Computed Tomography , Transplantation, Autologous , Treatment Outcome
3.
Neuropharmacology ; 70: 338-47, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23499664

ABSTRACT

Levomilnacipran (LVM; F2695) is the more active enantiomer of the serotonin/norepinephrine (5-HT/NE) reuptake inhibitor (SNRI) milnacipran and is currently under development for the treatment of major depressive disorder. LVM was benchmarked against two other SNRIs, duloxetine and venlafaxine, in biochemical, neurochemical and pharmacological assays. LVM exhibited high affinity for human NE (Ki = 92.2 nM) and 5-HT (11.2 nM) transporters, and potently inhibited NE (IC50 = 10.5 nM) and 5-HT (19.0 nM) reuptake (human transporter) in vitro. LVM had 2-fold greater potency for norepinephrine relative to serotonin reuptake inhibition (i.e. NE/5-HT potency ratio: 0.6) and 17 and 27 times higher selectivity for NE reuptake inhibition compared with venlafaxine and duloxetine, respectively. LVM did not exhibit affinity for 23 off-target receptors. LVM (i.p.) increased cortical extracellular levels of 5-HT, and NE (minimal effective doses: MEDs = 20 and 10 mg/kg, respectively). In anti-depressive/anti-stress models, i.p. LVM diminished immobility time in the mouse forced swim (MED = 20 mg/kg) and tail suspension (MED = 2.5 mg/kg) tests, and reduced shock-induced ultrasonic vocalizations in rats (MED = 5 mg/kg). Duloxetine and venlafaxine were less potent (MEDs ≥ 10 mg/kg). At doses active in these three therapeutically-relevant models, LVM (i.p.) did not significantly affect spontaneous locomotor activity. In summary, LVM is a potent, selective inhibitor of NE and 5-HT transporters with preferential activity at the former. It is efficacious in models of anti-depressive/anti-stress activity, with minimal potential for locomotor side effects.


Subject(s)
Anxiety/drug therapy , Behavior, Animal/drug effects , Cyclopropanes/pharmacology , Depression/drug therapy , Neurotransmitter Transport Proteins/antagonists & inhibitors , Adrenergic Uptake Inhibitors , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cyclohexanols/pharmacology , Cyclopropanes/therapeutic use , Dopamine/metabolism , Duloxetine Hydrochloride , Humans , Male , Mice , Milnacipran , Motor Activity/drug effects , Norepinephrine/metabolism , Rats , Serotonin/metabolism , Synaptosomes/drug effects , Thiophenes/pharmacology , Venlafaxine Hydrochloride
4.
Cell Transplant ; 21(9): 2009-19, 2012.
Article in English | MEDLINE | ID: mdl-22525800

ABSTRACT

Ischemia-reperfusion and immunosuppressive therapy are a major cause of progressive renal failure after kidney transplantation. Recent studies have shown that administration of bone marrow mesenchymal stem cells (MSCs) improves kidney functional recovery in the acute phase of post ischemia-reperfusion injury. In the present study, we used an original model of renal ischemia-reperfusion in immunosuppressed rats (NIRC) to investigate the effects of bone marrow MSCs on progression of chronic renal failure and the mechanisms potentially involved. Left renal ischemia-reperfusion (IR) was induced in unilateral nephrectomized Lewis rats. After IR, rats were treated daily with cyclosporine (10 mg/kg SC) for 28 days. MSCs were injected into the kidney at day 7 after IR. At day 28 after IR, kidneys were removed for histomorphological, biochemical, and gene expression analysis. The effect of conditioned media from MSCs on epithelial-mesenchymal transition was studied in vitro on HK2 cells. Our results show that, as compared to untreated NIRC rats, rats treated by intrarenal injection of MSCs 7 days after IR displayed improvement in renal function, reduction of interstitial fibrosis, and decrease in chronic tubule injury. These effects were associated with a decrease in interstitial α-SMA accumulation and MMP2 activity, markers of fibroblast/fibroblast-like cell activation, and renal remodeling, respectively. Finally, experiments in vitro showed that MSC-conditioned medium prevented epithelial-mesenchymal transition induced by TGF-ß in HK2 cells. In conclusion, our results show that, in immunosuppressed animals, a single intrarenal administration of MSCs reduced renal fibrosis and promoted the recovery of renal function.


Subject(s)
Bone Marrow Cells/cytology , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Kidney Diseases/surgery , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Reperfusion Injury/surgery , Adult , Animals , Disease Models, Animal , Female , Fibrosis/pathology , Humans , Kidney/blood supply , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Middle Aged , Rats , Rats, Inbred Lew , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
5.
Cell Transplant ; 19(12): 1623-33, 2010.
Article in English | MEDLINE | ID: mdl-20719065

ABSTRACT

Mesenchymal stem cells (MSCs) may be used as a cell source for cell therapy of solid organs due to their differentiation potential and paracrine effect. Nevertheless, optimization of MSC-based therapy needs to develop alternative strategies to improve cell administration and efficiency. One option is the use of alginate microencapsulation, which presents an excellent biocompatibility and an in vivo stability. As MSCs are hypoimmunogenic, it was conceivable to produce microparticles with [alginate-poly-L-lysine-alginate (APA) microcapsules] or without (alginate microspheres) a surrounding protective membrane. Therefore, the aim of this study was to determine the most suitable microparticles to encapsulate MSCs for engraftment on solid organ. First, we compared the two types of microparticles with 4 × 10(6) MSCs/ml of alginate. Results showed that each microparticle has distinct morphology and mechanical resistance but both remained stable over time. However, as MSCs exhibited a better viability in microspheres than in microcapsules, the study was pursued with microspheres. We demonstrated that viable MSCs were still able to produce the paracrine factor bFGF and did not present any chondrogenic or osteogenic differentiation, processes sometimes reported with the use of polymers. We then proved that microspheres could be implanted under the renal capsule without degradation with time or inducing impairment of renal function. In conclusion, these microspheres behave as an implantable scaffold whose biological and functional properties could be adapted to fit with clinical applications.


Subject(s)
Alginates , Biocompatible Materials/metabolism , Drug Compounding , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Microspheres , Animals , Cell Culture Techniques , Cells, Cultured , Glucuronic Acid , Hexuronic Acids , Materials Testing , Mesenchymal Stem Cells/cytology , Particle Size , Rats , Rats, Inbred Lew
6.
Br J Pharmacol ; 158(1): 232-42, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19508400

ABSTRACT

BACKGROUND AND PURPOSE: Xaliproden (SR57746A) is a 5-HT(1A) receptor agonist and neurotrophic agent that reduces oxaliplatin-mediated neuropathy in clinical trials. The present study investigated its profile on in vitro transduction, neurochemical responses and acute nociceptive pain tests in rats. EXPERIMENTAL APPROACH: Xaliproden was tested on models associated with 5-HT(1A) receptor activation including G-protein activation, extracellular dopamine and 5-HT levels measured by microdialysis and formalin-induced pain. Activation of 5-HT(1A) receptors was confirmed by antagonism with WAY100635. KEY RESULTS: Xaliproden exhibited high affinity for rat (r) and human (h) 5-HT(1A) receptors (pK(i)= 8.84 and 9.00). In [(35)S]GTPgammaS (guanosine 5'-O-(3-[(35)S]thio)triphosphate) assays it activated both hippocampal r5-HT(1A)[pEC(50)/E(MAX) of 7.58/61% (%5-HT)] and recombinant h5-HT(1A) receptors (glioma C6-h5-HT(1A): 7.39/62%; HeLa-h5-HT(1A): 7.24/93%). In functional [(35)S]GTPgammaS autoradiography, xaliproden induced labelling in structures enriched with 5-HT(1A) receptors (hippocampus, lateral septum, prefrontal and entorhinal cortices). Xaliproden inhibited in vivo binding of [(3)H]WAY100635 to 5-HT(1A) receptors in mouse frontal cortex and hippocampus (ID(50): 3.5 and 3.3 mg x kg(-1), p.o. respectively). In rat, it increased extracellular dopamine levels in frontal cortex and reduced hippocampal 5-HT levels (ED(50): 1.2 and 0.7 mg x kg(-1), i.p. respectively). In a rat pain model, xaliproden inhibited paw licking and elevation (ED(50): 1 and 3 mg x kg(-1), i.p. respectively) following formalin injection in the paw. All effects were reversed by pretreatment with WAY100635. CONCLUSIONS AND IMPLICATIONS: These results indicate that activation of 5-HT(1A) receptors is the principal mechanism of action of xaliproden and provide further support for the utility of 5-HT(1A) receptor activation as an anti-nociceptive strategy.


Subject(s)
GTP-Binding Proteins/metabolism , Naphthalenes/pharmacology , Neurotransmitter Agents/metabolism , Pain Measurement/drug effects , Pyridines/pharmacology , Receptor, Serotonin, 5-HT1A/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , HeLa Cells , Humans , Male , Mice , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Serotonin 5-HT1 Receptor Agonists
7.
Br J Pharmacol ; 156(2): 338-53, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19154445

ABSTRACT

BACKGROUND AND PURPOSE: Activation of post-synaptic 5-HT(1A) receptors may provide enhanced therapy against depression. We describe the signal transduction profile of F15599, a novel 5-HT(1A) receptor agonist. EXPERIMENTAL APPROACH: F15599 was compared with a chemical congener, F13714, and with (+)8-OH-DPAT in models of signal transduction in vitro and ex vivo. KEY RESULTS: F15599 was highly selective for 5-HT(1A) receptors in binding experiments and in [(35)S]-GTPgammaS autoradiography of rat brain, where F15599 increased labelling in regions expressing 5-HT(1A) receptors. In cell lines expressing h5-HT(1A) receptors, F15599 more potently stimulated extracellular signal-regulated kinase (ERK1/2) phosphorylation, compared with G-protein activation, internalization of h5-HT(1A) receptors or inhibition of cAMP accumulation. F13714, (+)8-OH-DPAT and 5-HT displayed a different rank order of potency for these responses. F15599 stimulated [(35)S]-GTPgammaS binding more potently in frontal cortex than raphe. F15599, unlike 5-HT, more potently and efficaciously stimulated G(alphai) than G(alphao) activation. In rat prefrontal cortex (a region expressing post-synaptic 5-HT(1A) receptors), F15599 potently activated ERK1/2 phosphorylation and strongly induced c-fos mRNA expression. In contrast, in raphe regions (expressing pre-synaptic 5-HT(1A) receptors) F15599 only weakly or did not induce c-fos mRNA expression. Finally, despite its more modest affinity in vitro, F15599 bound to 5-HT(1A) receptors in vivo almost as potently as F13714. CONCLUSIONS AND IMPLICATIONS: F15599 showed a distinctive activation profiles for 5-HT(1A) receptor-mediated signalling pathways, unlike those of reference agonists and consistent with functional selectivity at 5-HT(1A) receptors. In rat, F15599 potently activated signalling in prefrontal cortex, a feature likely to underlie its beneficial effects in models of depression and cognition.


Subject(s)
Piperidines/pharmacology , Pyrimidines/pharmacology , Serotonin 5-HT1 Receptor Agonists , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Aminopyridines , Animals , Autoradiography , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Extracellular Signal-Regulated MAP Kinases/metabolism , In Vitro Techniques , Male , Phosphorylation , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/physiology , Signal Transduction
8.
Br J Pharmacol ; 151(2): 237-52, 2007 May.
Article in English | MEDLINE | ID: mdl-17375087

ABSTRACT

BACKGROUND AND PURPOSE: Combining 5-HT(1A) receptor activation with dopamine D(2)/D(3) receptor blockade should improve negative symptoms and cognitive deficits in schizophrenia. We describe the in vitro profile of F15063 (N-[(2,2-dimethyl-2,3-dihydro-benzofuran-7-yloxy)ethyl]-3-(cyclopent-1-enyl)-benzylamine). EXPERIMENTAL APPROACH: F15063 was characterised in tests of binding affinity and in cellular models of signal transduction at monoamine receptors. KEY RESULTS: Affinities (receptor and pK(i) values) of F15063 were: rD(2) 9.38; hD(2L) 9.44; hD(2S) 9.25; hD(3) 8.95; hD(4) 8.81; h5-HT(1A) 8.37. F15063 had little affinity (40-fold lower than D(2)) at other targets. F15063 antagonised dopamine-activated G-protein activation at hD(2), rD(2) and hD(3) receptors with potency (pK (b) values 9.19, 8.29 and 8.74 in [(35)S]GTP gamma S binding experiments) similar to haloperidol. F15063 did not exhibit any hD(2) receptor agonism, even in tests of ERK1/2 phosphorylation and G-protein activation in cells with high receptor expression. In contrast, like (+/-)8-OH-DPAT, F15063 efficaciously activated h5-HT(1A) (E(max) 70%, pEC(50) 7.57) and r5-HT(1A) receptors (52%, 7.95) in tests of [(35)S]GTP gamma S binding, cAMP accumulation (90%, 7.12) and ERK1/2 phosphorylation (93%, 7.13). F15063 acted as a partial agonist for [(35)S]GTP gamma S binding at hD(4) (29%, 8.15) and h5-HT(1D) receptors (35%, 7.68). In [(35)S]GTP gamma S autoradiography, F15063 activated G-proteins in hippocampus, cortex and septum (regions enriched in 5-HT(1A) receptors), but antagonised quinelorane-induced activation of D(2)/D(3) receptors in striatum. CONCLUSIONS AND IMPLICATIONS: F15063 antagonised dopamine D(2)/D(3) receptors, a property underlying its antipsychotic-like activity, whereas activation of 5-HT(1A) and D(4) receptors mediated its actions in models of negative symptoms and cognitive deficits of schizophrenia (see companion papers).


Subject(s)
Antipsychotic Agents/pharmacology , Benzofurans/pharmacology , Benzylamines/pharmacology , Cyclopentanes/pharmacology , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/metabolism , Benzofurans/chemistry , Benzofurans/metabolism , Benzylamines/chemistry , Benzylamines/metabolism , Binding, Competitive/drug effects , CHO Cells , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Cyclopentanes/chemistry , Cyclopentanes/metabolism , Dopamine Agonists/chemistry , Dopamine Agonists/metabolism , Dopamine Antagonists/chemistry , Dopamine Antagonists/metabolism , Dose-Response Relationship, Drug , HeLa Cells , Humans , Male , Molecular Structure , Phosphorylation/drug effects , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Dopamine/metabolism , Serotonin 5-HT1 Receptor Agonists , Serotonin Receptor Agonists/chemistry , Serotonin Receptor Agonists/metabolism , Spodoptera , Swine
9.
Neuropharmacology ; 52(3): 1003-14, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17178132

ABSTRACT

Cloned, human dopamine D(1) receptors recruit multiple effectors but the G-protein subtype(s) activated by cerebral populations remain poorly defined, a question addressed using a rapid immunocapture technique. In rat striatum, dopamine (DA) and four selective, benzazepine agonists at D(1) receptors concentration-dependently enhanced [(35)S]GTPgammaS binding to Galphas/olf. For all drugs, Galphaq was also recruited with similar potencies and efficacies. Comparable observations were made in the cortex wherein profiles of Galphas/olf vs Galphaq activation were also highly correlated. In contrast to Galphas/olf and Galphaq, Galphao and Galphai were activated neither in the striatum nor in the cortex, except for SKF82958. As compared to DA, both SKF81297 and SKF82958 were full agonists at Gs/olf and Gq in cortex and striatum, whereas SKF38393 behaved as a partial agonist. Likewise, the "atypical" agonist, SKF83959 only partially activated Galphaq and also Gs/olf in these two regions. In both striatum and cortex, the selective D(1) receptor antagonist, SCH23390, abolished the recruitment of Galphaq and Galphas by DA, and the action of DA was partially attenuated by SKF83959. These findings demonstrate that, in native CNS tissue, DA and other D(1) receptor agonists activate Galphas and Galphaq with similar potencies and efficacies, suggesting their recruitment via pharmacologically-indistinguishable populations of D(1) receptors, and show that SPA technology is well-adapted to study the coupling of native DA receptors.


Subject(s)
Antibodies/metabolism , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, Dopamine D1/metabolism , Animals , Antibodies/pharmacology , Antibody Specificity , Benzazepines/pharmacology , Binding Sites, Antibody , Binding, Competitive/drug effects , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , GTP-Binding Protein alpha Subunits, Gq-G11/immunology , GTP-Binding Protein alpha Subunits, Gs/immunology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Radioligand Assay/methods , Rats
10.
Naunyn Schmiedebergs Arch Pharmacol ; 368(3): 188-99, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12923612

ABSTRACT

This study employed [(35)S]guanosine 5'- O-(3-thiotriphosphate) ([(35)S]GTPgammaS) binding to compare the actions of antipsychotic agents known to stimulate cloned, human 5-HT(1A) receptors with those of reference agonists at postsynaptic 5-HT(1A) receptors. In rat hippocampal membranes, the following order of efficacy was observed (maximum efficacy, E(max), values relative to 5-HT=100): (+)8-OH-DPAT (85), flesinoxan (62), eltoprazine (60), S14506 (59), S16924 (48), buspirone (41), S15535 (22), clozapine (22), ziprasidone (21), pindolol (7), p-MPPI (0), WAY100,635 (0), spiperone (0). Despite differences in species and tissue source, the efficacy and potency (pEC(50)) of agonists (with the exception of clozapine) correlated well with those determined previously at human 5-HT(1A) receptors expressed in Chinese hamster ovary (CHO) cells. In contrast, clozapine was more potent at hippocampal membranes. The selective antagonists p-MPPI and WAY100,635 abolished stimulation of binding by (+)8-OH-DPAT, clozapine and S16924 (p-MPPI), indicating that these actions were mediated specifically by 5-HT(1A) receptors. Clozapine and S16924 also attenuated 5-HT- and (+)8-OH-DPAT-stimulated [(35)S]GTPgammaS binding, consistent with partial agonist properties. In [(35)S]GTPgammaS autoradiographic studies, 5-HT-induced stimulation, mediated through 5-HT(1A) receptors, was more potent in the septum (pEC(50) approximately 6.5) than in the dentate gyrus of the hippocampus (pEC(50) approximately 5) suggesting potential differences in coupling efficiency or G protein expression. Though clozapine (30 and 100 microM) did not enhance [(35)S]GTPgammaS labelling in any structure, S16924 (10 micro M) modestly increased [(35)S]GTPgammaS labelling in the dentate gyrus. On the other hand, both these antipsychotic agents attenuated 5-HT (10 microM)-stimulated [(35)S]GTPgammaS binding in the dentate gyrus and septum. In conclusion, clozapine, S16924 and ziprasidone act as partial agonists for G protein activation at postsynaptic 5-HT(1A) receptors in the hippocampus. These data support a role of postsynaptic 5-HT(1A) receptors in the functional profiles of certain antipsychotic agents.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/pharmacology , GTP-Binding Proteins/metabolism , Hippocampus/metabolism , Pyrrolidines/pharmacology , Serotonin 5-HT1 Receptor Agonists , Animals , Autoradiography , Binding, Competitive , CHO Cells , Cell Membrane/metabolism , Cricetinae , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Ligands , Male , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin/metabolism , Serotonin 5-HT1 Receptor Antagonists
11.
J Pharmacol Exp Ther ; 306(3): 954-64, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12750432

ABSTRACT

Agomelatine (S20098) displayed pKi values of 6.4 and 6.2 at native (porcine) and cloned, human (h)5-hydroxytryptamine (5-HT)2C receptors, respectively. It also interacted with h5-HT2B receptors (6.6), whereas it showed low affinity at native (rat)/cloned, human 5-HT2A (<5.0/5.3) and 5-HT1A (<5.0/5.2) receptors, and negligible (<5.0) affinity for other 5-HT receptors. In antibody capture/scintillation proximity assays, agomelatine concentration dependently and competitively abolished h5-HT2C receptor-mediated activation of Gq/11 and Gi3 (pA2 values of 6.0 and 6.1). As measured by [3H]phosphatidylinositol depletion, agomelatine abolished activation of phospholipase C by h5-HT2C (pKB value of 6.1) and h5-HT2B (pKB value of 6.6) receptors. In vivo, it dose dependently blocked induction of penile erections by the 5-HT2C agonists (S)-2-(6-chloro-5-fluoroindol-1-yl)-1-methylethylamine (Ro60,0175) and 1-methyl-2-(5,8,8-trimethyl-8H-3-aza-cyclopenta[a]inden-3-yl) ethylamine (Ro60,0332). Furthermore, agomelatine dose dependently enhanced dialysis levels of dopamine in frontal cortex of freely moving rats, whereas they were unaffected in nucleus accumbens and striatum. Although the electrical activity of ventrotegmental dopaminergic neurons was unaffected agomelatine, it abolished their inhibition by Ro60,0175. Extracellular levels of noradrenaline in frontal cortex were also dose dependently enhanced by agomelatine in parallel with an acceleration in the firing rate of adrenergic cell bodies in the locus coeruleus. These increases in noradrenaline and dopamine levels were unaffected by the selective melatonin antagonist N-[2-(5-ethyl-benzo[b]thien-3-yl)ethyl] acetamide (S22153) and likely flect blockade of 5-HT2C receptors inhibitory to frontocortical dopaminergic and adrenergic pathways. Correspondingly, distinction to agomelatine, melatonin showed negligible activity 5-HT2C receptors and failed to modify the activity of adrenergic and dopaminergic pathways. In conclusion, in contrast to melatonin, agomelatine behaves as an antagonist at 5-HT2B and 5-HT2C receptors: blockade of the latter reinforces frontocortical adrenergic and dopaminergic transmission.


Subject(s)
Acetamides/pharmacology , Frontal Lobe/drug effects , Melatonin/pharmacology , Neurons/drug effects , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacology , Animals , Binding Sites , Corpus Striatum/metabolism , Dopamine/metabolism , Electrophysiology , Frontal Lobe/metabolism , Male , Neurons/physiology , Norepinephrine/metabolism , Nucleus Accumbens/metabolism , Penile Erection/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2A , Receptor, Serotonin, 5-HT2C , Receptors, Adrenergic/metabolism , Receptors, Dopamine/metabolism , Synaptic Transmission/drug effects , Tritium
12.
Brain Res ; 920(1-2): 41-54, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11716810

ABSTRACT

Unilateral 6-hydroxydopamine (6-OHDA) lesions of substantia nigra pars compacta (SNPC) neurons in rats induce behavioural hypersensitivity to dopaminergic agonists. However, the role of specific dopamine receptors is unclear, and potential alterations in their transduction mechanisms remain to be evaluated. The present study addressed these issues employing the dopaminergic agonist, quinelorane, which efficaciously stimulated G-protein activation (as assessed by [35S]GTPgammaS binding) at cloned hD2 (and hD3) receptors. At rat striatal membranes, dopamine stimulated [35S]GTPgammaS binding by 1.9-fold over basal, but its actions were only partially reversed by the selective D2/D3 receptor antagonist, raclopride, indicating the involvement of other receptor subtypes. In contrast, quinelorane-induced stimulation (48% of the effect of dopamine) was abolished by raclopride, and by the D2 receptor antagonist, L741,626. Further, novel antagonists selective for D3 and D4 receptors, S33084 and S18126, respectively, blocked the actions of quinelorane at concentrations corresponding to their affinities for D2 receptors. Quinelorane potently induced contralateral rotation in unilaterally 6-OHDA-lesioned rats, an effect abolished by raclopride and L741,626, but not by D3 and D4 receptor-selective doses of S33084 and S18126, respectively. In functional ([35S]GTPgammaS) autoradiography experiments, quinelorane stimulated G-protein activation in caudate putamen and, to a lesser extent, in nucleus accumbens and cingulate cortex of naive rats. In unilaterally SNPC-lesioned rats, quinelorane-induced G-protein activation in the caudate putamen on the non-lesioned side was similar to that seen in naive animals (approximately 50% stimulation), but significantly greater on the lesioned side (approximately 80%). This increase was both pharmacologically and regionally specific since it was reversed by raclopride, and was not observed in nucleus accumbens or cingulate cortex. In conclusion, the present data indicate that, in rat striatum, the actions of quinelorane are mediated primarily by D2 receptors, and suggest that behavioural hypersensitivity to this agonist, induced by unilateral SNPC lesions, is associated with an increase in D2, but not D3 or D4, receptor-mediated G-protein activation.


Subject(s)
GTP-Binding Proteins/metabolism , Neostriatum/metabolism , Receptors, Dopamine D2/physiology , Substantia Nigra/physiology , Sympathectomy, Chemical , Animals , Autoradiography , Binding, Competitive/drug effects , Biotransformation , Brain Chemistry/drug effects , Dopamine/metabolism , Dopamine Agonists/metabolism , Dopamine Antagonists/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Neostriatum/drug effects , Oxidopamine , Quinolines/metabolism , Rats , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3 , Recombinant Proteins/metabolism , Stereotyped Behavior/drug effects , Substantia Nigra/drug effects , Sympatholytics
13.
Br J Pharmacol ; 133(8): 1387-95, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11498526

ABSTRACT

1. Rat foetal liver contains large amounts of alpha2-adrenoceptors. The present work aimed to identify the receptor subtype and the cell type accounting for high expression and to clarify the mechanisms responsible for the sharp decrease in hepatic receptivity occurring during the late stage of foetal development. 2. Binding experiments indicated that the density of alpha2-adrenoceptors in the foetal liver (embryonic day 18; 615+/-155 fmol mg(-1) of protein) is 18 fold higher than in newborn or adult (35.2+/-4.3 fmol mg(-1)). A high amount of receptor is also found in the placenta (443+/-53 fmol mg(-1)). In both tissues, the rank order of antagonists to inhibit radioligand binding matched the pharmacological profile of the alpha2B-adrenoceptor and exclusively RNG transcripts were detected by RNase protection assays. 3. Isolation of cell fractions from foetal liver showed that alpha2B-adrenoceptor is primarily expressed by haematopoietic cells. Consistent with this view, the receptor is found to be abundant in foetal blood, carried by reticulocytes. The expression in blood gradually declines to zero at 3 weeks of age and it is not recovered following induction of reticulocytosis in adults. 4. In foetal reticulocytes, a low proportion of the receptor population is coupled to G-protein. The alpha2-agonist UK14304 has a marginal effect on cyclic AMP level but significantly increases arachidonic acid release. The function of the receptor remains to be elucidated. However, together with observations on alpha2B-knockout mice, the current finding strongly suggests a role for alpha2B-adrenoceptor during foetal haematopoiesis in rodents.


Subject(s)
Cell Lineage , Erythrocytes/metabolism , Liver/embryology , Liver/metabolism , Placenta/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-Agonists/metabolism , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Animals, Newborn , Arachidonic Acid/metabolism , Binding, Competitive , Cyclic AMP/metabolism , Erythrocytes/cytology , Erythropoietin/pharmacology , Hematopoiesis/drug effects , Heterotrimeric GTP-Binding Proteins/metabolism , Lung/metabolism , Mice , RNA/genetics , RNA/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/genetics , Reticulocyte Count , Spleen/metabolism , Substrate Specificity
14.
Neuropsychopharmacology ; 25(3): 410-22, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11522469

ABSTRACT

The actions of diverse antipsychotics at cloned h5-HT(1B) and h5-HT(1D) receptors were examined employing [3H]-GR125,743 and [35S]-GTPgammaS for determination of affinities and efficacies, respectively. Compared with hD(2) receptors, haloperidol, chlorpromazine and olanzapine showed markedly (>100-fold) lower affinity for h5-HT(1D) and h5-HT(1B) receptors at which they expressed inverse agonist properties. Clozapine, risperidone and ocaperidone likewise behaved as inverse agonists at h5-HT(1B) and h5-HT(1D) receptors but their affinities were only approximately 10-fold lower than at hD(2) receptors. Moreover, ziprasidone, S16924 and ORG5222 interacted at h5-HT(1B) and h5-HT(1D) receptors with affinities similar to hD(2) sites. While S16924 and ORG5222 were inverse agonists at h5-HT(1B) and h5-HT(1D) sites, ziprasidone was an inverse agonist at h5-HT(1D) receptors yet a partial agonist at h5-HT(1B) receptors. These actions of antipsychotics were abolished by the selective, neutral antagonist, S18127. In conclusion, with the exception of ziprasidone, all antipsychotics were inverse agonists at h5-HT(1B) and h5-HT(1D) receptors, although they differed markedly in their potency at these sites as compared to hD(2) receptors.


Subject(s)
Antipsychotic Agents/pharmacology , Receptors, Serotonin/drug effects , Serotonin Receptor Agonists/pharmacology , Animals , CHO Cells , Cloning, Molecular , Cricetinae , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guinea Pigs , Rats , Receptor, Serotonin, 5-HT1B , Receptor, Serotonin, 5-HT1D , Receptors, Dopamine D2/drug effects , Recombinant Proteins/pharmacology
15.
Psychopharmacology (Berl) ; 156(1): 58-62, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11465634

ABSTRACT

RATIONALE: The benzazepine and "selective" dopamine D1 receptor antagonist, SCH23390 [(R)-(+)-8-chloro-2,3,4,5-tetrahydro-3-methyl-5-phenyl-1H-benzazepine-7-ol], shows significant affinity at native serotonin (5-HT)2C receptors. OBJECTIVES: We examined its functional actions at cloned human (h)5-HT2C receptors (VSV isoform) stably expressed in CHO cells. METHODS: Since 5-HT2C receptors are positively coupled to phospholipase C (PLC), their activation was determined by depletion of membrane-bound pools of pre-labelled [3H]phosphotidylinositol ([3H]PI). RESULTS: SCH23390 showed high affinity (Ki, 9.3 nM) at h5-HT2C sites and depleted [3H]PI with an EC50 of 2.6 nM. Its efficacy was equivalent to that of 5-HT. [3H]PI depletion elicited by SCH23390 was concentration-dependently abolished by the selective 5-HT2C antagonist, SB242,084, with a K(B) of 0.55 nM. Further, in the presence of a fixed concentration of SB242,084 (10 nM), the concentration-response curve for SCH23390 was shifted to the right without loss of maximal effect, yielding a K(B) of 0.57 nM. CONCLUSIONS: SCH23390 is a potent and high efficacy agonist at h5-HT2C receptors. Activation of 5-HT2C receptors by SCH23390 may contribute to its functional properties both in animals and in humans.


Subject(s)
Benzazepines/pharmacology , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Serotonin/metabolism , Animals , Benzazepines/metabolism , Binding, Competitive/drug effects , Binding, Competitive/physiology , CHO Cells , Cricetinae , Dopamine Antagonists/metabolism , Dose-Response Relationship, Drug , Humans , Rats , Receptor, Serotonin, 5-HT2C , Receptors, Dopamine D1/metabolism , Receptors, Serotonin/physiology
16.
Eur J Pharmacol ; 424(1): 13-7, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11470255

ABSTRACT

At h5-HT1A receptors, stably transfected into Chinese Hamster Ovary Cells (CHO-h5-HT1A), the selective 5-HT1A receptor agonist, (+)8-hydroxy-dipropyl-amino-tetralin, ((+)8-OH-DPAT), transiently activated mitogen-activated protein kinase (MAPK) with a pEC50 of 8.5. The arylalkylamine, (-)-pindolol, also behaved as an agonist with a maximal effect of 57% relative to (+)8-OH-DPAT (100%), and with a pEC50 of 7.2. The selective 5-HT(1A) receptor antagonist, N-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-N-(2-pyridinyl)cyclo-hexane carboxamide (WAY100,635), blocked (+)8-OH-DPAT- and (-)-pindolol-induced MAPK activation with pK(B)s of 9.7 and 9.9, respectively, whereas the selective 5-HT(1B) receptor antagonist, 1'-Methyl-5-[2'-methyl-4'-(5-methyl-1,2,4-oxadiazol-3-yl)biphenyl-4-ylcarbonyl]-2,3,6,7-tetrahydro-5H-spiro[furo[2,3-f]indole-3,4'-piperidine] (SB224,289) was inactive. Pertussis toxin blocked the actions of (+)8-OH-DPAT and (-)-pindolol demonstrating implication of G(i)/G(o) proteins. Thus, stimulation of MAPK provides an intracellular marker and signal for expression of the agonist actions of (-)-pindolol at h5-HT1A receptors.


Subject(s)
Mitogen-Activated Protein Kinases/drug effects , Pindolol/pharmacology , Receptors, Serotonin/drug effects , Serotonin Receptor Agonists/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Mitogen-Activated Protein Kinases/metabolism , Piperazines/pharmacology , Pyridines/pharmacology , Receptors, Serotonin/metabolism , Receptors, Serotonin, 5-HT1 , Serotonin Antagonists/pharmacology
17.
J Pharmacol Exp Ther ; 297(3): 876-87, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356907

ABSTRACT

Compared with cloned, human (h)D(2) receptors (pK(i) = 6.9), the antiparkinsonian agent piribedil showed comparable affinity for halpha(2A)- (7.1) and halpha(2C)- (7.2) adrenoceptors (ARs), whereas its affinity for halpha(2B)-ARs was less marked (6.5). At halpha(2A)- and halpha(2C)-ARs, piribedil antagonized induction of [(35)S]guanosine-5'-O-(3-thio)triphosphate (GTPgammaS) binding by norepinephrine (NE) with pK(b) values of 6.5 and 6.9, respectively. Furthermore, Schild analysis of the actions of piribedil at halpha(2A)-ARs indicated competitive antagonism, yielding a pA(2) of 6.5. At a porcine alpha(2A)-AR-Gi1alpha-Cys351C (wild-type) fusion protein, piribedil competitively abolished (pA(2) = 6.5) GTPase activity induced by epinephrine. However, at a alpha(2A)-AR-Gi1alpha-Cys351I (mutant) fusion protein of amplified sensitivity, although still acting as a competitive antagonist (pA(2) = 6.2) of epinephrine, piribedil itself manifested weak partial agonist properties. Similarly, piribedil weakly induced mitogen-activated protein kinase phosphorylation via wild-type halpha(2A)-ARs, although attenuating its phosphorylation by NE. As demonstrated by functional [(35)S]GTPgammaS autoradiography in rats, piribedil antagonized activation by NE of alpha(2)-ARs in cortex, amygdala, and septum. Antagonist properties were also expressed in a dose-dependent enhancement of the firing rate of adrenergic neurons in locus ceruleus (0.125-4.0 mg/kg i.v.). Furthermore, piribedil (2.5-4.0 mg/kg s.c.) accelerated hippocampal NE synthesis, elevated dialysis levels of NE in hippocampus and frontal cortex, and blocked hypnotic-sedative properties of the alpha(2)-AR agonist xylazine. Finally, piribedil showed only modest affinity for rat alpha(1)-ARs (5.9) and weakly antagonized NE-induced activation of phospholipase C via halpha(1A)-ARs (pK(b) = 5.6). In conclusion, piribedil displays essentially antagonist properties at cloned, human and cerebral, rat alpha(2)-ARs. Blockade of alpha(2)-ARs may, thus, contribute to its clinical antiparkinsonian profile.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists , Antiparkinson Agents/pharmacology , Piribedil/pharmacology , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Agonists/pharmacology , Animals , Binding, Competitive/drug effects , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Frontal Lobe/drug effects , Frontal Lobe/metabolism , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Locus Coeruleus/cytology , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacology , Phosphatidylinositols/metabolism , Phosphorylation/drug effects , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/genetics , Receptors, Adrenergic, alpha-2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Serotonin/metabolism
18.
J Pharmacol Exp Ther ; 295(3): 1192-205, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11082457

ABSTRACT

S18616 ((S)-spiro[(1-oxa-2-amino-3-azacyclopent-2-ene)-4, 2'-(8'-chloro-1',2',3',4'-tetrahydronaphthalene)]) displayed high affinity at native rat alpha(2)-adrenoceptors (AR)s (pK(i), 9.8), native human (h)alpha(2A)-ARs (9.6), and cloned halpha(2A)- (9.5), halpha(2B)- (9.2), and halpha(2C)- (9.0) ARs. It showed 40-fold lower affinity for halpha(1A)-ARs (8.4) and >/=100-fold lower affinity for rat alpha(1)-ARs (7.1), halpha(1B)-ARs (7.7), halpha(1D)-ARs (7.6), imidazoline(1) (7.4), and imidazoline(2) (7.4) sites and >100-fold lower affinity for all other (>50) sites. At halpha(2A)-ARs, in guanosine-5'-O-(3-[(35)S]thio)triphosphate binding studies, S18616 was a potent (partial) agonist: log effective concentration (pEC(50)), 9.3/maximal effect, 51%. This observation was corroborated employing a halpha(2A)-Gi1alpha fusion protein/GTPase assay (9.0/40%) in which the actions of S18616 were blocked by pertussis toxin. Employing guanosine-5'-O-(3-[(35)S]thio)triphosphate binding assays, S18616 was also a partial agonist at halpha(2C)-ARs (8.2/63%) but a full agonist (8.4/124%) at halpha(2B)-ARs. At halpha(2A)-, halpha(2B)-, and halpha(2C)-ARs, the selective alpha(2)-AR antagonist, atipamezole, abolished the actions of S18616: pK(b) values of 9.1, 9. 1, and 9.4, respectively. As determined by depletion of membrane-bound [(3)H]phosphatidyl inositols, S18616 behaved as a (less potent) agonist (7.8/79%) at halpha(1A)-ARs, an action abolished by prazosin (pK(b), 8.9). Reflecting alpha(2)-AR agonist properties, S18616 potently (>/=1 microg/kg, s.c.) and dose dependently elicited hypothermia and antinociception (nine diverse models) in rodents. These actions were dose dependently inhibited by chemically diverse alpha(2)- versus alpha(1)-AR antagonists, atipamezole, idazoxan, RX821,002, and BRL44418 (a preferential alpha(2A)-AR ligand). In contrast, the actions of S18616 were unaffected by the alpha(1)-AR antagonists, ARC239 and prazosin (which preferentially block alpha(2B/2C)- versus alpha(2A)-ARs). Although the affinity of dexmedetomidine at alpha(2)-ARs was lower than S18616; it displayed a similar receptor and functional profile. Clonidine displayed lower efficacy than S18616, was substantially less potent, and had marked affinity for imidazoline(1) sites and alpha(1)-ARs. In conclusion, S18616 is a novel, selective, and highly potent agonist at alpha(2)-ARs.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Analgesics, Non-Narcotic/pharmacology , Body Temperature/drug effects , Clonidine/pharmacology , Dexmedetomidine/pharmacology , Oxazoles/pharmacology , Animals , CHO Cells , Cricetinae , Humans , Imidazoles/pharmacology , Imidazoline Receptors , Male , Mice , Rats , Rats, Wistar , Receptors, Drug/metabolism
19.
Mol Pharmacol ; 58(4): 821-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10999953

ABSTRACT

The heterologous regulation of the alpha2C-adrenergic receptor (alpha2C-AR) was investigated in the HepG2 cell line. Binding of [(3)H]MK912 (alpha2-antagonist) to membranes from cells submitted to various treatments showed that exposure to insulin, phorbol 12-myristate 13-acetate, or dexamethasone did not affect receptor density. On the other hand, treatment with forskolin resulted in a large reduction of alpha2C-AR number. The effect of forskolin was mimicked by 8-br-cAMP and was abolished by the protein kinase A inhibitor, H89. The action of cAMP was slow (t(1/2) = 23 h), dose-dependent, and additive to the receptor down-regulation elicited by the alpha2-agonist, UK14304. Furthermore, the diminution of receptor was not caused by an increased rate of its degradation but resulted from a decrease in the steady state amounts of alpha2C4-mRNA. As assessed by experiments in the presence of actinomycin D, the stability of alpha2C4-mRNA was not affected by 8-br-cAMP or forskolin. By contrast, the activity of a luciferase construct containing the entire promoter region of the alpha2C4 gene (1.9 kilobase pairs) was inhibited, indicating that the primary mechanism of action of the two compounds is at the transcriptional level. Deletions in the 5'-end of this construct showed that the elements responsible for cAMP responsiveness lie within a 242-base-pair fragment of the gene promoter (nucleotides -236/+6 relative to transcription start). Band-shift experiments indicated that nuclear factors bind to this region in a cAMP-dependent manner. The determination of the actual cis- and trans-acting elements involved will be the object of future investigation, but the present study provides evidence for transcriptional regulation of human alpha2C-AR by cAMP.


Subject(s)
Cyclic AMP/pharmacology , Down-Regulation/drug effects , Receptors, Adrenergic, alpha-2/genetics , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Colforsin/pharmacology , Humans , Kinetics , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Receptors, Adrenergic, alpha-2/biosynthesis , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured
20.
Gut ; 47(2): 242-50, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10896916

ABSTRACT

BACKGROUND AND AIMS: Previous studies on rodents have suggested that catecholamines stimulate proliferation of the intestinal epithelium through activation of alpha(2) adrenoceptors located on crypt cells. The occurrence of this effect awaits demonstration in humans and the molecular mechanisms involved have not yet been elucidated. Here, we examined the effect of alpha(2) agonists on a clone of Caco2 cells expressing the human alpha(2A) adrenoceptor. METHODS: Cells were transfected with a bicistronic plasmid containing the alpha2C10 and neomycin phosphotransferase genes. G418 resistant clones were assayed for receptor expression using radioligand binding. Receptor functionality was assessed by testing its ability to couple Gi proteins and to inhibit cAMP production. Mitogen activated protein kinase (MAPK) phosphorylation was followed by western blot, and cell proliferation was estimated by measuring protein and DNA content. RESULTS: Permanent transfection of Caco2 cells allowed us to obtain a clone (Caco2-3B) expressing alpha(2A) adrenoceptors at a density similar to that found in normal human intestinal epithelium. Caco2-3B retained morphological features and brush border enzyme expression characteristic of enterocytic differentiation. The receptor was coupled to Gi2/Gi3 proteins and its stimulation caused marked diminution of forskolin induced cAMP production. Treatment of Caco2-3B with UK14304 (alpha(2) agonist) induced a rapid increase in the phosphorylation state of MAPK, extracellular regulated protein kinase 1 (Erk1), and 2 (Erk2). This event was totally abolished in pertussis toxin treated cells and in the presence of kinase inhibitors (genistein or PD98059). It was unaffected by protein kinase C downregulation but correlated with a transient increase in Shc tyrosine phosphorylation. Finally, sustained exposure of Caco2-3B to UK14304 resulted in modest but significant acceleration of cell proliferation. None of these effects was observed in the parental cell line Caco2. CONCLUSION: The results obtained in the present study support a regulatory role for alpha(2) adrenoceptors in intestinal cell proliferation.


Subject(s)
Enterocytes/cytology , Intestinal Mucosa/cytology , Receptors, Adrenergic, alpha-2/physiology , Adrenergic alpha-2 Receptor Agonists , Blotting, Western , Caco-2 Cells/cytology , Cell Division/physiology , Colforsin/metabolism , Cyclic AMP/biosynthesis , Humans , Mitogen-Activated Protein Kinases/physiology , Phosphorylation , Radioligand Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...