Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters








Publication year range
1.
Naunyn Schmiedebergs Arch Pharmacol ; 394(9): 1829-1867, 2021 09.
Article in English | MEDLINE | ID: mdl-33991216

ABSTRACT

Manfred Göthert, who had served Naunyn-Schmiedeberg's Arch Pharmacol as Managing Editor from 1998 to 2005, deceased in June 2019. His scientific oeuvre encompasses more than 20 types of presynaptic receptors, mostly on serotoninergic and noradrenergic neurones. He was the first to identify presynaptic receptors for somatostatin and ACTH and described many presynaptic receptors, known from animal preparations, also in human tissue. In particular, he elucidated the pharmacology of presynaptic 5-HT receptors. A second field of interest included ligand-gated and voltage-dependent channels. The negative allosteric effect of anesthetics at peripheral nACh receptors is relevant for the peripheral clinical effects of these drugs and modified the Meyer-Overton hypothesis. The negative allosteric effect of ethanol at NMDA receptors in human brain tissue occurred at concentrations found in the range of clinical ethanol intoxication. Moreover, the inhibitory effect of gabapentinoids on P/Q Ca2+ channels and the subsequent decrease in AMPA-induced noradrenaline release may contribute to their clinical effect. Another ligand-gated ion channel, the 5-HT3 receptor, attracted the interest of Manfred Göthert from the whole animal via isolated preparations down to the cellular level. He contributed to that molecular study in which 5-HT3 receptor subtypes were disclosed. Finally, he found altered pharmacological properties of 5-HT receptor variants like the Arg219Leu 5-HT1A receptor (which was also shown to be associated with major depression) and the Phe124Cys 5-HT1B receptor (which may be related to sumatriptan-induced vasospasm). Manfred Göthert was a brilliant scientist and his papers have a major impact on today's pharmacology.


Subject(s)
Receptors, Serotonin/metabolism , Serotonin/metabolism , Allosteric Regulation , Animals , History, 20th Century , History, 21st Century , Humans , Ligand-Gated Ion Channels/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Nicotinic/metabolism , Receptors, Serotonin/history , Serotonin/history
2.
J Physiol Pharmacol ; 66(3): 343-53, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26084216

ABSTRACT

The activation of cannabinoid CB1 receptors decreases and increases blood pressure (BP) in anaesthetized and conscious rats, respectively. The aim of our study was to check the possible involvement of CB1 receptors in the paraventricular nucleus of the hypothalamus (PVN) in the cardiovascular effects of cannabinoids in rats. Methanandamide (metabolically stable analogue of the endocannabinoid anandamide) and the synthetic cannabinoid receptor agonist CP55940 were microinjected into the PVN of urethane-anaesthetized rats twice (S1 and S2, 20 min apart). Receptor antagonists were administered intravenously (i.v.) 5 min before S1. Methanandamide and CP55940 decreased blood pressure by 15 - 20%. The CB1 receptor antagonist AM251 reversed the depressor effect into a pressor response of 20 - 30%. The pressor effect of CP55940 observed in the presence of AM251 i.v. was reduced by AM251 given additionally into the PVN but not by the i.v. injection of the CB2 antagonist SR144528 or the vanilloid TRPV1 antagonist ruthenium red. In the presence of the peripherally restricted CB1 receptor antagonist AM6545, CP55940 given into the PVN increased BP by 40%. AM6545 reversed the decrease in BP induced by CP55940 i.v. into a marked increase. Bilateral chemical lesion of the PVN by kainic acid abolished all cardiovascular effects of CP55940 i.v. In conclusion, the cannabinoid CP55940 administered to the PVN of urethane-anaesthetized rats can induce depressor and pressor effects. The direction of the response probably depends on the sympathetic tone. The centrally induced hypertensive response of CP55940 can, in addition, be masked by peripheral CB1 receptors.


Subject(s)
Cannabinoids/pharmacology , Cyclohexanols/pharmacology , Paraventricular Hypothalamic Nucleus/physiology , Receptor, Cannabinoid, CB1/physiology , Amidohydrolases/metabolism , Anesthesia , Animals , Arachidonic Acids/metabolism , Arachidonic Acids/pharmacology , Blood Pressure/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Endocannabinoids/metabolism , Heart Rate/drug effects , Male , Microinjections , Morpholines/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Pyrazoles/pharmacology , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors
3.
Naunyn Schmiedebergs Arch Pharmacol ; 381(4): 349-60, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20198363

ABSTRACT

Intravenous (i.v.) injection of the endocannabinoid anandamide induces triphasic cardiovascular responses, including a pressor effect mediated via unknown central and peripheral mechanism(s). The aim of the present study was to determine the central mechanism(s) responsible for the pressor response to anandamide. For this purpose, the influence of antagonists at thromboxane A(2) TP (sulotroban, daltroban, SQ 29548), NMDA (MK-801) and beta(2)-adrenergic receptors (ICI 118551) on the pressor effect induced by i.v. and intracerebroventricularly (i.c.v.) administered anandamide was examined in urethane-anaesthetized rats. Anandamide (1.5-3 micromol/kg, i.v.) or its stable analogue methanandamide (0.75 micromol/kg, i.v.) increased blood pressure by 25%. Anandamide (0.03 mumol per animal i.c.v.) caused a pure pressor effect (by 20%) but only in the presence of antagonists of CB(1) and TRPV1 receptors. The effects of cannabinoids (i.v. or i.c.v.) were diminished by i.v. daltroban, sulotroban (10 mumol/kg each), and/or SQ 29548 (1 mumol/kg). The effect of anandamide i.v. was reduced by SQ 29548 (0.02 mumol per animal i.c.v.) and by the thromboxane A(2) synthesis inhibitor furegrelate i.c.v. (1.8 micromol per animal). ICI 118551, MK-801 (1 micromol/kg i.v. each), and bilateral adrenalectomy diminished the effect of anandamide i.c.v. Sulotroban (i.v.) failed to affect the response to anandamide (i.v.) in pithed rats, and anandamide and methanandamide did not bind to TP receptors in rat platelets. The present study suggests that central beta(2)-adrenergic, NMDA and thromboxane A(2) receptors are involved in the anandamide-induced adrenal secretion of catecholamines and their pressor effect in urethane-anaesthetized rats.


Subject(s)
Arachidonic Acids/pharmacology , Polyunsaturated Alkamides/pharmacology , Receptors, Adrenergic, beta-2/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Animals , Arachidonic Acids/administration & dosage , Arachidonic Acids/metabolism , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Catecholamines/metabolism , Dose-Response Relationship, Drug , Endocannabinoids , Injections, Intraventricular , Male , Polyunsaturated Alkamides/administration & dosage , Polyunsaturated Alkamides/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta-2/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/drug effects
4.
Naunyn Schmiedebergs Arch Pharmacol ; 381(1): 21-31, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20012265

ABSTRACT

Prostaglandin E(2) (PGE(2)) and its analogue sulprostone inhibit noradrenaline and serotonin release in rodent tissues. We examined whether the receptor involved is blocked by the EP(3) antagonist L-826,266, whether such receptors also occur on central cholinergic neurones and retinal dopaminergic cells, whether PGE(2) is produced by the degradation of the endocannabinoid virodhamine and whether EP(3) receptor activation stimulates (35)S-GTPgammaS binding. Transmitter release was studied as electrically evoked tritium overflow in superfused tissues preincubated with (3)H-noradrenaline (which in the guinea pig retina labels dopaminergic cells), (3)H-serotonin or (3)H-choline. (35)S-GTPgammaS binding, a measure of G protein activation, was studied in mouse and guinea pig hippocampal membranes. L-826,266 antagonised the effect of sulprostone on noradrenaline release in the rat cortex, yielding a Schild plot-based pA(2) value of 7.56. Apparent pA(2) values in mouse cortex and rat vas deferens (noradrenaline release) and rat cortex (serotonin release) were 7.55, 7.87 and 7.67, respectively. PGE(2) did not affect acetylcholine release in rat brain and dopamine release in guinea pig retina. In seven mice tissues, noradrenaline release was inhibited by sulprostone but not affected by virodhamine. (35)S-GTPgammaS binding was not altered by sulprostone but stimulated by the cannabinoid agonist WIN 55,212-2. Prostaglandins of the E series inhibit monoamine release via EP(3) receptors at which L-826,266 is a competitive antagonist. EP(3) receptors that inhibit transmitter release are not present on central cholinergic neurones and retinal dopaminergic cells. Virodhamine is not converted to PGE(2). An EP(3) receptor model based on (35)S-GTPgammaS binding could not be identified.


Subject(s)
Acrylamides/pharmacology , Biogenic Monoamines/antagonists & inhibitors , Biogenic Monoamines/physiology , Naphthalenes/pharmacology , Prostaglandins E/pharmacology , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/physiology , Animals , Female , Guinea Pigs , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Receptors, Prostaglandin E, EP3 Subtype , Tissue Distribution/drug effects , Tissue Distribution/physiology
5.
Br J Pharmacol ; 157(8): 1474-82, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19681871

ABSTRACT

BACKGROUND AND PURPOSE: We examined whether cannabinoid CB(1) and histamine H(3) receptors resemble alpha(2)-adrenoceptors in that their presynaptically mediated cardiovascular effects are less marked in urethane- than in pentobarbitone-anaesthetized pithed rats. EXPERIMENTAL APPROACH: Effects of the cannabinoid agonist CP-55,940 and the H(3) receptor agonist imetit on electrically induced tachycardic and vasopressor responses, respectively, was compared in pithed rats anaesthetized with urethane or pentobarbitone. The affinity of urethane for the three receptors was measured by radioligand binding studies in rat brain cortex membranes and its potency assessed in superfused mouse tissues preincubated with (3)H-noradrenaline. KEY RESULTS: The neurogenic tachycardic response was less markedly inhibited by CP-55,940 in urethane- than in pentobarbitone-anaesthetized pithed rats. Imetit inhibited the neurogenic vasopressor response after pentobarbitone but not after urethane. The catecholamine-induced tachycardic and vasopressor response did not differ between rats anaesthetized with either compound. Urethane 10 mM (plasma concentration reached under anaesthesia) did not affect binding to CB(1) or H(3) receptors and alpha(2) adrenoceptors, nor did it alter the inhibitory effect of agonists at the three receptors on electrically evoked (3)H-noradrenaline release. CONCLUSIONS AND IMPLICATIONS: Urethane, but not pentobarbitone, abolished the H(3) receptor-mediated vascular response in pithed rats and attenuated the CB(1) receptor-mediated cardiac response much more than pentobarbitone. The weaker effects of CB(1), H(3) and alpha(2) receptor agonists cannot be explained by antagonism by urethane at the three receptors in vitro. Pentobarbitone, but not urethane, is suitable as an anaesthetic for investigations of inhibitory presynaptic receptor function in pithed and anaesthetized rats.


Subject(s)
Anesthetics/pharmacology , Pentobarbital/pharmacology , Receptors, Presynaptic/physiology , Urethane/pharmacology , Adrenergic alpha-2 Receptor Agonists , Animals , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Cerebral Cortex/metabolism , Cyclohexanols/pharmacology , Decerebrate State , Electric Stimulation , Histamine Agonists/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Radioligand Assay , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/physiology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Vasoconstrictor Agents/pharmacology
6.
J Physiol Pharmacol ; 60(1): 51-60, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19439807

ABSTRACT

Bupranolol is an antagonist at the cardiostimulatory low-affinity state of b(1)-adrenoceptors and we were interested whether this effect is shared by its fluorine (GD-6), methyl (DZ-51) and isopropyl analogue (DZ-13) and by the analogue hydroxylated at the tertiary butyl moiety (DZ-52). (-)-Bupranolol and compounds (-)-GD-6, (+)-GD-6, (-)-DZ-13, (+)-DZ-13, DZ-51 and DZ-52 antagonized the CGP 12177-induced tachycardia in pithed rats with "apparent pA(2) values" of 6.1, 6.1, 4.6, 5.5, 4.6, 5.1 and 5.3, respectively. For comparison, their potencies and affinities at the high-affinity state of b(1)-adrenoceptors were determined, using the xamoterol-induced tachycardia in pithed rats and [(3)H]CGP 12177 binding to rat cerebrocortical membranes. The respective "apparent pA(2) values" in the functional experiments were 7.9, 8.1, 5.4, 8.4, 5.7, 7.3 and 6.8 and the pK(i) values in the binding experiments were 8.8, 8.4, 6.9, 8.5, 6.7, 8.4 and 8.2. In conclusion, (-)-bupranolol and its fluorine analogue (-)-GD-6 are equipotent at the low-affinity state of beta(1)-adrenoceptors. The stereoselectivity of GD-6 and DZ-13 suggests that the low-affinity state is indeed a receptor.


Subject(s)
Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-Antagonists/pharmacology , Bupranolol/pharmacology , Tachycardia/prevention & control , Animals , Binding Sites , Bupranolol/analogs & derivatives , Cerebral Cortex/metabolism , Male , Propanolamines/pharmacology , Rats , Rats, Wistar , Stereoisomerism , Xamoterol/pharmacology
7.
Br J Pharmacol ; 155(7): 1034-42, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18806815

ABSTRACT

BACKGROUND AND PURPOSE: The endocannabinoid virodhamine is a partial agonist at the cannabinoid CB(1) receptor and a full agonist at the CB(2) receptor, and relaxes rat mesenteric arteries through endothelial cannabinoid receptors. Its concentration in the periphery exceeds that of the endocannabinoid anandamide. Here, we examined the influence of virodhamine on the human pulmonary artery. EXPERIMENTAL APPROACH: Isolated human pulmonary arteries were obtained during resections for lung carcinoma. Vasorelaxant effects of virodhamine were examined on endothelium-intact vessels precontracted with 5-HT or KCl. KEY RESULTS: Virodhamine, unlike WIN 55,212-2, relaxed 5-HT-precontracted vessels concentration dependently. The effect of virodhamine was reduced by endothelium denudation, two antagonists of the endothelial cannabinoid receptor, cannabidiol and O-1918, and a high concentration of the CB(1) receptor antagonist rimonabant (5 muM), but only slightly attenuated by the NOS inhibitor L-NAME and not affected by a lower concentration of rimonabant (100 nM) or by the CB(2) and vanilloid receptor antagonists SR 144528 and capsazepine, respectively. The COX inhibitor indomethacin and the fatty acid amide hydrolase inhibitor URB597 and combined administration of selective blockers of small (apamin) and intermediate and large (charybdotoxin) conductance Ca(2+)-activated K(+) channels attenuated virodhamine-induced relaxation. The vasorelaxant potency of virodhamine was lower in KCl- than in 5-HT-precontracted preparations. CONCLUSIONS AND IMPLICATIONS: Virodhamine relaxes the human pulmonary artery through the putative endothelial cannabinoid receptor and indirectly through a COX-derived vasorelaxant prostanoid formed from the virodhamine metabolite, arachidonic acid. One or both of these mechanisms may stimulate vasorelaxant Ca(2+)-activated K(+) channels.


Subject(s)
Arachidonic Acids/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Pulmonary Artery/drug effects , Receptors, Cannabinoid/drug effects , Arachidonic Acid/metabolism , Arachidonic Acids/administration & dosage , Cannabinoids , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , In Vitro Techniques , Male , Middle Aged , Potassium Channels, Calcium-Activated/drug effects , Potassium Channels, Calcium-Activated/metabolism , Potassium Chloride/pharmacology , Pulmonary Artery/metabolism , Receptors, Cannabinoid/metabolism , Serotonin/pharmacology , Vasodilation/drug effects
8.
J Physiol Pharmacol ; 59(1): 3-15, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18441383

ABSTRACT

We studied whether cannabinoid CB(1) receptors occur on the sympathetic neurones innervating the guinea-pig atrium and renal cortex. Atrial and cortical kidney pieces preincubated with [(3)H]-noradrenaline were superfused and the electrically (3 Hz)-evoked tritium overflow was examined. The evoked overflow in atrium was inhibited by the cannabinoid agonist WIN 55,212-2 maximally by 35%; its concentration-response curve was shifted to the right by the CB(1) antagonist rimonabant (pA(2) 8.3), which, by itself, did not affect the evoked overflow. The evoked overflow in the renal cortex was not altered by WIN 55,212-2. The muscarinic agonist oxotremorine and prostaglandin E(2) inhibited the evoked overflow maximally by 55 and 65% in atrium and by 80 and 55% in kidney, respectively. Furthermore, the nucleotide sequence of the guinea-pig CB(1) receptor was determined (GenBank DQ355990). The deduced amino acid sequence has a high homology to the corresponding sequence of man (98.7%) and rat or mouse (99.2%). In conclusion, presynaptic CB(1) receptors leading to inhibition of noradrenaline release occur in guinea-pig atrium but not renal cortex. The deduced amino acid sequence of the guinea-pig CB(1) receptor shows a homology of 99% to the CB(1) receptor sequence of rodents and humans.


Subject(s)
Amino Acid Sequence , Norepinephrine/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Electric Stimulation , Guinea Pigs , Heart Atria/metabolism , Kidney Cortex/metabolism , Male , Molecular Sequence Data , Neurons/metabolism , Sequence Analysis, Protein , Species Specificity , Sympathetic Nervous System/metabolism
9.
Handb Exp Pharmacol ; (184): 409-34, 2008.
Article in English | MEDLINE | ID: mdl-18064421

ABSTRACT

Presynaptic receptors for four families of neuropeptides will be discussed: opioids, neuropeptide Y, adrenocorticotropic hormone (ACTH), and orexins. Presynaptic receptors for the opioids (micro, delta, kappa, and ORL(1)) and neuropeptide Y (Y(2)) inhibit transmitter release from a variety of neurones, both in the peripheral and central nervous systems. These receptors, which were also identified in human tissue, are coupled to G(i/o) proteins and block voltage-dependent Ca(2+) channels, activate voltage-dependent K(+) channels, and/or interfere with the vesicle release machinery. Presynaptic receptors for ACTH (MC(2) receptors) have so far been identified almost exclusively in cardiovascular tissues from rabbits, where they facilitate noradrenaline release; they are coupled to G(s) protein and act via stimulation of adenylyl cyclase. Presynaptic receptors for orexins (most probably OX(2) receptors) have so far almost exclusively been identified in the rat and mouse brain, where they facilitate the release of glutamate and gamma-aminobutyric acid (GABA); they are most probably linked to G(q) and directly activate the vesicle release machinery or act via a transduction mechanism upstream of the release process. Agonists and antagonists at opioid receptors owe at least part of their therapeutic effects to actions on presynaptic receptors. Therapeutic drugs targeting neuropeptide Y and orexin receptors and presynaptic ACTH receptors so far are not available.


Subject(s)
Neuropeptides/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Presynaptic/metabolism , Animals , Humans , Orexin Receptors , Receptors, Corticotropin/drug effects , Receptors, Corticotropin/metabolism , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Receptors, Presynaptic/drug effects
10.
J Physiol Pharmacol ; 59 Suppl 8: 91-107, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19258666

ABSTRACT

Endocannabinoids (e.g. anandamide, 2-arachidonoylglycerol or virodhamine) regulate the function of the cardiovascular system mainly in the following way: 1) by acting via CB(1) receptors, 2) by activation of CB(2) receptors, and 3) by modifying the function of vanilloid TRPV1, serotonin 5-HT(3) and alpha(7)-subunit-containing nicotinic acetylcholine receptors. Endocannabinoids are implicated in the pathogenesis of hypertension and of hypotension associated with haemorrhagic, endotoxic, and cardiogenic shock, and with advanced liver cirrhosis. There is also evidence for their involvement in the control of atherosclerosis.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Cardiovascular System/metabolism , Endocannabinoids , Shock, Cardiogenic/physiopathology , Animals , Humans , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptors, Nicotinic/metabolism , Receptors, Serotonin, 5-HT3/metabolism , TRPV Cation Channels/metabolism
11.
J Physiol Pharmacol ; 57(3): 317-28, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17033087

ABSTRACT

The aim of our study was to examine whether non beta(1)-/beta(2)-adrenoceptors participate in the relaxation of the human pulmonary artery. For this purpose the vasodilatory effect of the non-conventional partial beta-adrenoceptor agonist cyanopindolol was examined. Cyanopindolol (1-300 microM), studied in the presence of the beta(1)-/beta(2)-adrenoceptor antagonist propranolol, relaxed the human pulmonary artery preconstricted with serotonin 1 microM in a concentration-dependent manner (maximally by about 80%). This effect was diminished by bupranolol 10 microM (an antagonist of beta(1)-beta(3)-adrenoceptors and the low affinity state of the beta(1)-adrenoceptor) and CGP 20712 10 microM (known to antagonize the low-affinity state of the beta(1)-adrenoceptor at high concentrations). In further experiments, the effect of beta-adrenoceptor ligands on the serotonin-induced vasoconstriction was examined. The concentration-response curve for serotonin was not affected by cyanopindolol 30 microM, bupranolol 10 microM and CGP 20712 10 microM but shifted to the right by cyanopindolol 100 and 300 microM; the serotonin 5-HT(2A) receptor antagonist ketanserin 0.3 microM abolished the maximum contraction elicited by serotonin. In conclusion, the present study reveals that the vasodilatory effect of cyanopindolol in the human pulmonary artery consists of two components, i.e. activation of a propranolol-insensitive atypical beta-adrenoceptor and antagonism against 5-HT(2A) receptors.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Pindolol/analogs & derivatives , Pulmonary Artery/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Analysis of Variance , Bupranolol/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Imidazoles/pharmacology , In Vitro Techniques , Male , Middle Aged , Pindolol/pharmacology , Pulmonary Artery/physiology , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2A/physiology , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2/drug effects , Serotonin 5-HT2 Receptor Antagonists , Vasoconstriction/drug effects , Vasodilation/physiology
12.
Naunyn Schmiedebergs Arch Pharmacol ; 372(2): 139-46, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16195872

ABSTRACT

Cannabinoids exert complex effects on blood pressure related to their interference with cardiovascular centres in the central nervous system and to their direct influence on vascular muscle, vascular endothelium and heart. In view of the relative lack of information on the occurrence of CB1 receptors on the vascular postganglionic sympathetic nerve fibres, the aim of the present study was to examine whether cannabinoid receptor ligands affect the electrically evoked tritium overflow in superfused vessels (tissue pieces) from the guinea-pig, the rat and the mouse preincubated with 3H-noradrenaline. The cannabinoid receptor agonist WIN 55,212-2 (R(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl)methyl]-pyrrolo[1,2,3-de]1,4-benzoxazinyl](1-naphthalenyl) methanone) inhibited the evoked tritium overflow in the guinea-pig aorta, but not in that of the rat or mouse. The concentration-response curve of WIN 55,212-2 was shifted to the right by the CB1 receptor antagonist rimonabant, yielding an apparent pA2 value of 7.9. The most pronounced (near-maximum) inhibition obtained at the highest WIN 55,212-2 concentration applied (3.2 microM) amounted to 40%. WIN 55,212-2 also inhibited the evoked overflow in guinea-pig pulmonary artery, basilar artery and portal vein, again in a manner sensitive to antagonism by rimonabant. The latter did not affect the evoked overflow by itself in the four vessels, but did increase the electrically evoked tritium overflow from superfused guinea-pig hippocampal slices preincubated with 3H-choline and from superfused guinea-pig retina discs preincubated with 3H-noradrenaline (labelling dopaminergic cells in this tissue). The inhibitory effect of 3.2 microM WIN 55,212-2 on the evoked overflow from the guinea-pig aorta was comparable in size to that obtained with agonists at the histamine H3, kappa opioid (KOP) and ORL1 (NOP) receptor (1 or 10 microM, producing the respective near-maximum effects) whereas prostaglandin E2 1 microM caused a higher near-maximum inhibition of 70%. Prostaglandin E2 also induced an inhibition by 65 and 80% in the rat and mouse aorta respectively, indicating that the present conditions are basically suitable for detecting presynaptic receptor-mediated inhibition of noradrenaline release. The results show that the postganglionic sympathetic nerve fibres in the guinea-pig aorta, but not in the rat or mouse aorta, are endowed with presynaptic inhibitory cannabinoid CB1 receptors; such receptors also occur in guinea-pig pulmonary artery, basilar artery and portal vein. These CB1 receptors are not subject to an endogenous tone and the extent of inhibition obtainable via these receptors is within the same range as that of several other presynaptic heteroreceptors, but markedly lower than that obtainable via receptors for prostaglandin E2.


Subject(s)
Adrenergic Fibers/metabolism , Norepinephrine/metabolism , Presynaptic Terminals/metabolism , Receptor, Cannabinoid, CB1/metabolism , Adrenergic Fibers/drug effects , Animals , Aorta , Benzoxazines , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Guinea Pigs , In Vitro Techniques , Male , Mice , Morpholines/pharmacology , Naphthalenes/pharmacology , Piperidines/pharmacology , Presynaptic Terminals/drug effects , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/drug effects , Rimonabant , Species Specificity , Tritium
13.
Handb Exp Pharmacol ; (168): 327-65, 2005.
Article in English | MEDLINE | ID: mdl-16596780

ABSTRACT

The CB1 cannabinoid receptor is widely distributed in the central and peripheral nervous system. Within the neuron, the CB1 receptor is often localised in axon terminals, and its activation leads to inhibition of transmitter release. The consequence is inhibition of neurotransmission via a presynaptic mechanism. Inhibition of glutamatergic, GABAergic, glycinergic, cholinergic, noradrenergic and serotonergic neurotransmission has been observed in many regions of the central nervous system. In the peripheral nervous system, CB1 receptor-mediated inhibition of adrenergic, cholinergic and sensory neuroeffector transmission has been frequently observed. It is characteristic for the ubiquitous operation of CB1 receptor-mediated presynaptic inhibition that antagonistic components of functional systems (for example, the excitatory and inhibitory inputs of the same neuron) are simultaneously inhibited by cannabinoids. Inhibition of voltage-dependent calcium channels, activation of potassium channels and direct interference with the synaptic vesicle release mechanism are all implicated in the cannabinoid-evoked inhibition of transmitter release. Many presynaptic CB1 receptors are subject to an endogenous tone, i.e. they are constitutively active and/or are continuously activated by endocannabinoids. Compared with the abundant data on presynaptic inhibition by cannabinoids, there are only a few examples for cannabinoid action on the somadendritic parts of neurons in situ.


Subject(s)
Cannabinoids/pharmacology , Synaptic Transmission/drug effects , Acetylcholine/metabolism , Animals , Dopamine/metabolism , Humans , Ion Channels/drug effects , Neural Inhibition/drug effects , Norepinephrine/metabolism , Receptor, Cannabinoid, CB1/analysis , Receptor, Cannabinoid, CB1/physiology , Receptors, Presynaptic/physiology , gamma-Aminobutyric Acid/metabolism
14.
Br J Pharmacol ; 137(8): 1355-61, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12466246

ABSTRACT

1. We determined the effects of nociceptin/orphanin FQ and the NOP receptor ligands acetyl-Arg-Tyr-Tyr-Arg-Ile-Lys-NH(2) (Ac-RYYRIK-NH(2)) and naloxone benzoylhydrazone on transmitter release in vitro. 2. The electrically evoked tritium overflow from guinea-pig and mouse striatal slices and guinea-pig retinal discs preincubated with [(3)H]-dopamine was inhibited by nociceptin/orphanin FQ (pEC(50) 7.9, 7.6 and 8.6; E(max) 30, 50 and 55%). Ac-RYYRIK-NH(2) 0.032 microM and naloxone benzoylhydrazone 5 microM antagonized the effect of nociceptin/orphanin FQ in striatal slices of the guinea-pig (apparent pA(2) 9.1 and 6.8) and the mouse (apparent pA(2) 9.2 and 7.5) and strongly attenuated the effect of nociceptin/orphanin FQ 0.1 microM in guinea-pig retinal discs. Ac-RYYRIK-NH(2) 0.032 microM did not affect the evoked overflow by itself whereas naloxone benzoylhydrazone 5 microM inhibited it in each tissue. 3. The electrically evoked tritium overflow from mouse brain cortex slices preincubated with [(3)H]-noradrenaline was inhibited by nociceptin/orphanin FQ (pEC(50) 7.9, E(max) 85%), Ac-RYYRIK-NH(2) (pEC(50) 8.3, E(max) 47%) but not affected by naloxone benzoylhydrazone 5 microM. Ac-RYYRIK-NH(2) and naloxone benzoylhydrazone showed apparent pA(2) values of 8.6 and 6.9. 4. In conclusion, the inhibitory effect of nociceptin/orphanin FQ on dopamine release in the striatum and retina and on noradrenaline release in the cerebral cortex is mediated via NOP receptors. Ac-RYYRIK-NH(2) behaves as an extremely potent NOP receptor antagonist in the striatum and retina and as a partial agonist in the cortex.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Opioid Peptides/metabolism , Receptors, Opioid/physiology , Retina/metabolism , Animals , Corpus Striatum/drug effects , Dopamine Antagonists/metabolism , Dopamine Antagonists/pharmacology , Guinea Pigs , In Vitro Techniques , Male , Mice , Opioid Peptides/pharmacology , Receptors, Opioid/agonists , Retina/drug effects , Nociceptin Receptor , Nociceptin
15.
J Physiol Pharmacol ; 53(3): 301-24, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12369730

ABSTRACT

Nociceptin is the endogenous ligand of the opioid OP4 or ORL1 (opioid receptor-like1) receptor. It decreases blood pressure and heart rate in anesthetized and conscious rats and mice after its intravenous and intracerebroventricular injection in a manner sensitive to OP4 but not to OP1-3 (or delta, kappa and mu opioid) receptor antagonists. OP4 receptors involved in the cardiovascular effects of nociceptin were identified on sensory afferent fibres, in brain areas including the nucleus tractus solitarii and the rostral ventrolateral medulla, on preganglionic and/or postganglionic sympathetic and parasympathetic nerve fibres innervating blood vessels and heart or directly on these target organs. These receptors do not seem to be tonically activated but may play a role in the pathophysiology of inflammation, arterial hypertension and cardiac or brain circulatory ischemia.


Subject(s)
Cardiovascular Physiological Phenomena , Opioid Peptides/physiology , Receptors, Opioid/physiology , Animals , Cardiovascular Physiological Phenomena/drug effects , Cardiovascular System/innervation , Nervous System Physiological Phenomena , Opioid Peptides/pharmacology , Nociceptin Receptor , Nociceptin
16.
Trends Pharmacol Sci ; 22(11): 565-72, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11698100

ABSTRACT

Cannabis (marijuana) is not only a frequently abused drug but also has the potential for the development of useful agents for the treatment of emesis, anorexia and multiple sclerosis. In this article, the effects of modulation of transmitter release by cannabinoids in both the CNS and the PNS of various species, including humans, will be discussed. Cannabinoids inhibit neurotransmitter release via specific presynaptic cannabinoid CB1 receptors. Studies using either the CB1 receptor antagonist and inverse agonist SR141716 or CB1-receptor-deficient mice suggest that numerous presynaptic cannabinoid receptors are tonically activated by endogenous cannabinoids and/or are constitutively active. CB1-receptor-mediated inhibition of transmitter release might explain, for example, reinforcing properties and memory impairment caused by cannabinoids.


Subject(s)
Cannabinoids/pharmacology , Neurotransmitter Agents/metabolism , Receptors, Drug/metabolism , Receptors, Presynaptic/metabolism , Animals , Humans , Receptors, Cannabinoid , Receptors, Drug/drug effects , Receptors, Presynaptic/drug effects
17.
Naunyn Schmiedebergs Arch Pharmacol ; 364(1): 14-20, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11485033

ABSTRACT

We have shown previously that histaprodifen and its Nalpha-substituted analogues methylhistaprodifen and dimethylhistaprodifen are highly potent H1-receptor agonists in vivo. The aim of the present study was to examine the influence of four newly synthesized histaprodifen analogues, 3-fluoro-methylhistaprodifen (1), Nalpha-imidazolylethylhistaprodifen (2), bis-histaprodifen (3) and Nalpha-methyl-bis-histaprodifen (4), on the cardiovascular system in the pithed and in the anaesthetized rat. In pithed and vagotomized rats, diastolic blood pressure (which was increased to 80-85 mmHg by vasopressin infusion) was decreased dose dependently by methylhistaprodifen (the reference compound) and by compounds 1-4. The maximum decrease was about 47-50 mmHg for methylhistaprodifen and compounds 1, 2 and 3. Their potencies, expressed as pED50 (the negative logarithm of the dose in mole per kilogram body weight that decreased diastolic blood pressure by 25 mmHg), were 8.31, 8.23, 8.26 and 7.84, respectively. With compound 4 the maximal effect was not achieved at doses up to 1 micromol/kg (the latter dose decreased blood pressure by about 30 mmHg; pED50 approximately 6.5). The vasodepressor effect of the five compounds was attenuated by the H1-receptor antagonist dimetindene (1 micromol/kg) but was not changed by combined administration of the H2- and H3-receptor antagonists ranitidine and thioperamide (1 micromol/kg each), by combined administration of the alpha1- and alpha2-adrenoceptor antagonists prazosin and rauwolscine (1 micromol/kg each) or by the beta-adrenoceptor antagonist propranolol (3 micromol/kg). In anaesthetized rats methylhistaprodifen and compounds 1-4 induced almost the same fall in blood pressure as in pithed and vagotomized animals; the effects were sensitive to blockade by dimetindene (1 micromol/kg). Higher doses of compounds 1 and 2 (1 micromol/kg) increased heart rate in pithed and vagotomized rats in a manner sensitive to propranolol (3 micromol/kg) but insensitive to dimetindene (3 micromol/kg). The same dose of methylhistaprodifen and of compounds 3 and 4 failed to affect heart rate. We conclude that the agonistic potency of compounds 1 and 2 at H1-receptors in the cardiovascular system of the rat equals that of methylhistaprodifen, the most potent histamine H1-receptor agonist available until recently. Compounds 1 and 2 exhibit sympathomimetic activity at high doses.


Subject(s)
Blood Pressure/drug effects , Heart Rate/drug effects , Histamine/analogs & derivatives , Histamine/pharmacology , Methylhistamines/pharmacology , Anesthesia , Animals , Cardiovascular System/drug effects , Histamine/chemistry , Male , Methylhistamines/chemistry , Rats , Rats, Wistar , Sympathomimetics/pharmacology , Vagotomy
18.
Naunyn Schmiedebergs Arch Pharmacol ; 364(3): 233-41, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11521166

ABSTRACT

We studied the effects of nociceptin, the endogenous ligand of the opioid OP4 receptor, and of two cannabinoid receptor agonists WIN 55,212-2 and CP-55,940 (0.001-1 micromol/kg each) on the neurogenic tachycardia and bradycardia in pithed rats. Electrical stimulation (1 Hz, 1 ms, 50 V for 10 s) of the preganglionic sympathetic nerve fibres and injection of nicotine 2 micromol/kg or isoprenaline 0.5 nmol/kg increased heart rate by about 70 beats/min (bpm) in pithed rats pretreated with atropine 1.5-2 micromol/kg. The electrically induced tachycardia was reduced dose dependently by nociceptin, WIN 55,212-2 and CP-55,940 (by 60, 30 and 20% at the highest dose, respectively). The OP4 and cannabinoid receptor agonists diminished the nicotine- but not the isoprenaline-stimulated increase in heart rate. In pithed rats pretreated with propranolol 3 micromol/kg, vagal stimulation (5 Hz, 1 ms, 15 V for 10 s) or injection of methacholine (5-10 nmol/kg) decreased heart rate by about 30 bpm. Nociceptin, but not WIN 55,212-2 or CP-55,940 decreased the vagal bradycardia dose dependently (the inhibitory effect of 1 micromol/kg was about 40%). Nociceptin failed to modify the methacholine-induced decrease in heart rate. The OP4 receptor antagonists naloxone benzoylhydrazone 5 micromol/kg and/or [Phe1Psi(CH2-NH)Gly2]-nociceptin(1-13)NH2 0.7 micromol/kg, but not the OP(1-3) receptor antagonist naloxone 10 micromol/kg, diminished the inhibitory action of nociceptin on the neurogenic tachycardia and bradycardia. The inhibitory effect of both cannabinoid receptor agonists on the neurogenic tachycardia was abolished by the CB1 receptor antagonist SR 141716 0.1 micromol/kg. The present data suggest that the postganglionic sympathetic nerve fibres innervating the rat heart are endowed with presynaptic opioid OP4 and cannabinoid CB1 receptors, activation of which inhibits the neurogenic tachycardia. The parasympathetic nerve fibres innervating the heart and causing bradycardia are endowed with presynaptic opioid OP4 but not cannabinoid receptors.


Subject(s)
Autonomic Nervous System/drug effects , Bradycardia/drug therapy , Morpholines/pharmacology , Naphthalenes/pharmacology , Opioid Peptides/pharmacology , Receptors, Drug/drug effects , Receptors, Opioid/drug effects , Tachycardia/drug therapy , Vasodilator Agents/pharmacology , Analysis of Variance , Animals , Benzoxazines , Blood Pressure/drug effects , Cyclohexanols/pharmacology , Cyclohexanols/therapeutic use , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Male , Morpholines/therapeutic use , Naphthalenes/therapeutic use , Opioid Peptides/therapeutic use , Rats , Rats, Wistar , Receptors, Cannabinoid , Tachycardia/etiology , Nociceptin
19.
Br J Pharmacol ; 133(8): 1243-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11498509

ABSTRACT

1. Dopamine release in the retina is subject to modulation via autoreceptors, which belong to the D(2) receptor family (encompassing the D(2), D(3) and D(4) receptors). The aim of the present study was to determine the receptor subtype (D(2) vs D(3)) involved in the inhibition of dopamine release in guinea-pig retinal discs, using established (haloperidol, (S)-nafadotride) and novel dopamine receptor antagonists (ST-148, ST-198). 2. hD(2L) and hD(3) receptors were expressed in CHO cells and the pK(i) values determined in binding studies with [(125)I]-iodosulpride were: haloperidol 9.22 vs 8.54; ST-148 7.85 vs 6.60; (S)-nafadotride 8.52 vs 9.51; ST-198 6.14 vs 7.92. 3. The electrically evoked tritium overflow from retinal discs preincubated with [(3)H]-noradrenaline (which represents quasi-physiological dopamine release) was inhibited by the dopamine receptor agonists B-HT 920 (talipexole) and quinpirole (maximally by 82 and 71%; pEC(50) 5.80 and 5.83). The concentration-response curves of these agonists were shifted to the right by haloperidol (apparent pA(2) 8.69 and 8.23) and ST-148 (7.52 and 7.66). (S)-Nafadotride 0.01 microM and ST-198 0.32 microM did not affect the concentration-response curve of B-HT 920. 4. The dopamine autoreceptor in the guinea-pig retina can be classified as a D(2) receptor. ST-148 and ST-198 show an improved selectivity for D(2) and D(3) receptors when compared to haloperidol and (S)-nafadotride, respectively.


Subject(s)
Dopamine D2 Receptor Antagonists , Receptors, Dopamine D2/analysis , Retina/chemistry , Retina/drug effects , Animals , Azepines/metabolism , Azepines/pharmacology , Binding Sites , CHO Cells , Cricetinae , Dopamine/metabolism , Dopamine Agonists/metabolism , Dopamine Agonists/pharmacology , Dopamine Antagonists/chemistry , Dopamine Antagonists/metabolism , Dopamine Antagonists/pharmacology , Guinea Pigs , Haloperidol/metabolism , Haloperidol/pharmacology , Male , Naphthalenes/metabolism , Naphthalenes/pharmacology , Norepinephrine/metabolism , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Quinpirole/metabolism , Quinpirole/pharmacology , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , Retina/metabolism , Substrate Specificity
20.
Br J Pharmacol ; 132(8): 1665-72, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309237

ABSTRACT

We determined the affinities of eight novel histamine H(3)-receptor ligands (ethers and carbamates) for H(3)-receptor binding sites and their agonistic/antagonistic effects in two functional H(3)-receptor models. The compounds differ from histamine in that the ethylamine chain is replaced by a propyloxy chain; in the three ethers mentioned below (FUB 335, 373 and 407), R is n-pentyl, 3-methylbutyl and 3,3-dimethylbutyl, respectively. The compounds monophasically inhibited [(3)H]-N(alpha)-methylhistamine binding to mouse cerebral cortex membranes (pK(i) 7.51 - 9.53). The concentration-response curve of histamine for its inhibitory effect on the electrically evoked [(3)H]-noradrenaline overflow from mouse cortex slices was shifted to the right by these compounds (apparent pA(2) 6.61 - 8.00). Only FUB 373 and 407 inhibited the evoked overflow by themselves (intrinsic activities 0.3 and 0.4); these effects were counteracted by the H(3)-receptor antagonist clobenpropit. [(35)S]-GTPgammaS binding to mouse cortex membranes was stimulated by the H(3)-receptor agonist (R)-alpha-methylhistamine in a manner sensitive to clobenpropit. Among the novel compounds only FUB 373 and 407 stimulated [(35)S]-GTPgammaS binding (intrinsic activities 0.6 and 0.4). In conclusion, the novel compounds are partial H(3)-receptor agonists (FUB 373 and 407) or H(3)-receptor antagonists; comparison with FUB 335 shows that the transition from antagonist to agonist is caused by a slight structural change. A protonated N atom in the side chain is not necessary for agonism at H(3) receptors, proposing a receptor-ligand interaction different from that of classical agonists.


Subject(s)
Histamine Agonists/pharmacology , Histamine Antagonists/pharmacology , Receptors, Histamine H3/drug effects , Amines/chemistry , Animals , Brain Chemistry/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Electric Stimulation , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Histamine/pharmacology , Histamine Agonists/chemistry , In Vitro Techniques , Methylhistamines/pharmacology , Mice , Norepinephrine/metabolism , Perfusion , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL