Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 436
Filter
1.
Neuroscience ; 248: 392-402, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23811399

ABSTRACT

Bradykinin (BK) and its receptors (B1 and B2) may exert a role in the pathophysiology of certain CNS diseases, including epilepsy. In healthy tissues, B2 receptors are constitutively and widely expressed and B1 receptors are absent or expressed at very low levels, but both receptors, particularly B1, are up-regulated under many pathological conditions. Available data support the notion that up-regulation of B1 receptors in brain areas like the amygdala, hippocampus and entorhinal cortex favors the development and maintenance of an epileptic condition. The role of B2 receptors, instead, is still unclear. In this study, we used two different models to investigate the susceptibility to seizures of B1 knockout (KO) and B2 KO mice. We found that B1 KO are more susceptible to seizures compared with wild-type (WT) mice, and that this may depend on B2 receptors, in that (i) B2 receptors are overexpressed in limbic areas of B1 KO mice, including the hippocampus and the piriform cortex; (ii) hippocampal slices prepared from B1 KO mice are more excitable than those prepared from WT controls, and this phenomenon is B2 receptor-dependent, being abolished by B2 antagonists; (iii) kainate seizure severity is attenuated by pretreatment with a non-peptide B2 antagonist in WT and (more effectively) in B1 KO mice. These data highlight the possibility that B2 receptors may have a role in the responsiveness to epileptogenic insults and/or in the early period of epileptogenesis, that is, in the onset of the molecular and cellular events that lead to the transformation of a normal brain into an epileptic one.


Subject(s)
Disease Susceptibility , Hippocampus/metabolism , Piriform Cortex/metabolism , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/metabolism , Seizures/metabolism , Animals , Bradykinin/metabolism , Bradykinin B1 Receptor Antagonists/pharmacology , Bradykinin B2 Receptor Antagonists/pharmacology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/physiopathology , Kainic Acid/toxicity , Mice , Mice, Knockout , Piriform Cortex/drug effects , Piriform Cortex/physiopathology , Receptor, Bradykinin B1/deficiency , Receptor, Bradykinin B2/deficiency , Seizures/chemically induced , Seizures/genetics
2.
Br J Pharmacol ; 154(6): 1163-5, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18536746

ABSTRACT

A surprising proportion of patients with inflammatory bowel disease (IBD) remain refractory to all classes of drugs presently in clinical use. Kinins are inflammatory mediators of potential relevance in IBD, because at least the kinin B1 receptor subtype is upregulated in human or animal intestinal inflammation and also both B1 and B2 receptors for kinins support inflammation and epithelial electrogenic ion transport that leads to secretory diarrhoea. In this issue of the BJP, Hara et al. report the therapeutic effect of a modern and selective nonpeptide kinin B1 receptor antagonist, SSR240612 ((2R)-2-(((3R)-3-(1,3-benzodioxol-5-yl)-3-(((6-methoxy-2-naphthyl)sulphonyl)amino)propanoyl)amino)-3-(4-((2R,6S)-2,6-dimethylpiperidinyl)methyl)phenyl)-N-isopropyl-N-methylpropanamide hydrochloride), with benefits such as decreased neutrophil influx and improved macroscopic tissue scoring. The results were corroborated using kinin B1 receptor gene-knockout mice. Further, kinin B1 receptor upregulation in this inflammatory model is partially dependent on TNF-alpha, a recognized target for IBD pharmacotherapy. More work is warranted to evaluate the value of the kinin B1 receptor antagonists as a novel anti-inflammatory therapeutic option for IBD.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Bradykinin B1 Receptor Antagonists , Inflammatory Bowel Diseases/drug therapy , Anti-Inflammatory Agents/pharmacology , Dioxoles/pharmacology , Dioxoles/therapeutic use , Humans , Kallikrein-Kinin System/physiology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
3.
Br J Pharmacol ; 154(2): 471-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18376418

ABSTRACT

BACKGROUND AND PURPOSE: Neuropeptide S (NPS) was recently identified as the endogenous ligand of an orphan receptor, now referred to as the NPS receptor. In vivo, NPS produces a unique behavioural profile by increasing wakefulness and exerting anxiolytic-like effects. In the present study, we further evaluated the effects of in vivo supraspinal NPS in mice. EXPERIMENTAL APPROACH: Effects of NPS, injected intracerebroventricularly (i.c.v.), on locomotor activity (LA), righting reflex (RR) recovery and on anxiety states (measured with the elevated plus maze (EPM) and stress-induced hyperthermia (SIH) tests) were assessed in Swiss mice. KEY RESULTS: NPS (0.01-1 nmol per mouse) caused a significant increase in LA in naive mice, in mice habituated to the test cages and in animals sedated with diazepam (5 mg kg(-1)). In the RR assay, NPS dose dependently reduced the proportion of animals losing the RR in response to diazepam (15 mg kg(-1)) and their sleeping time. In the EPM and SIH test, NPS dose dependently evoked anxiolytic-like effects by increasing the time spent by animals in the open arms and reducing the SIH response, respectively. CONCLUSIONS AND IMPLICATIONS: We provide further evidence that NPS acts as a novel modulator of arousal and anxiety-related behaviours by promoting a unique pattern of effects: stimulation associated with anxiolysis. Therefore, NPS receptor ligands may represent innovative drugs for the treatment of sleep and anxiety disorders.


Subject(s)
Anti-Anxiety Agents/metabolism , Anxiety/metabolism , Behavior, Animal , Central Nervous System Stimulants/metabolism , Neuropeptides/metabolism , Animals , Anti-Anxiety Agents/administration & dosage , Anxiety/prevention & control , Anxiety/psychology , Behavior, Animal/drug effects , Body Temperature Regulation , Caffeine/administration & dosage , Central Nervous System Stimulants/administration & dosage , Diazepam/administration & dosage , Dose-Response Relationship, Drug , Exploratory Behavior , Injections, Intraventricular , Male , Mice , Motor Activity , Neuropeptides/administration & dosage , Reflex , Sleep/drug effects , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Time Factors
4.
Neuropeptides ; 41(6): 375-87, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17988733

ABSTRACT

Diabetes Mellitus leads to pain neuropathy and cardiovascular complications which remain resistant to current therapies involving the control of glycaemia. This study aims at defining the contribution of kinin B(1) receptor (B(1)R) and the oxidative stress on sensory abnormalities and arterial hypertension in a rat model of insulin resistance. Rats were fed with 10% d-glucose for a chronic period of 12-14 weeks and the impact of a diet supplemented with alpha-lipoic acid, a potent antioxidant, was determined on tactile and cold allodynia, arterial hypertension and the expression of kinin B(1)R (real-time PCR and autoradiography) in several tissues. Acute effects of brain penetrant (LF22-0542) and peripherally acting (R-715) B(1)R antagonists were also assessed. Glucose-fed rats exhibited tactile and cold allodynia along with increases in systolic blood pressure between 4 and 12 weeks; these alterations were alleviated by alpha-lipoic acid. The latter regimen also decreased significantly increased plasma levels of insulin and glucose and insulin resistance (HOMA index) at 14 weeks. B(1)R mRNA was virtually absent in liver, aorta, lung, kidney and spinal cord isolated from control rats, yet B(1)R mRNA was markedly increased in all tissues in glucose-fed rats. Up-regulated B(1)R mRNA and B(1)R binding sites (spinal cord) were significantly reduced by alpha-lipoic acid in glucose-fed rats. LF22-0542 reduced tactile and cold allodynia (3h) and reversed arterial hypertension (3-48h) in glucose-fed rats. R-715 abolished tactile and cold allodynia but had not effect on blood pressure. Data suggest that the oxidative stress contributes to the induction and up-regulation of B(1)R in the model of insulin resistance induced by glucose feeding. The over expressed B(1)R contributes centrally to arterial hypertension and in the periphery to sensory abnormalities.


Subject(s)
Hypertension/etiology , Insulin Resistance , Oxidative Stress/physiology , Receptor, Bradykinin B1/physiology , Sensation Disorders/etiology , Animals , Disease Models, Animal , Glucose/administration & dosage , Glucose/pharmacology , Hypertension/metabolism , Kinins , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B1/genetics , Sensation Disorders/metabolism , Up-Regulation/genetics
5.
J Pept Res ; 66(1): 39-47, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15946194

ABSTRACT

In the present study we describe the in vitro pharmacological characterization of the nociceptin/orphanin FQ (N/OFQ) receptor (NOP) ligand Ac-RYYRWK-NH2 and the synthesis and biological evaluation of 13 Trp5 substituted Ac-RYYRWK-NH2 analogs. Results indicate that Ac-RYYRWK-NH2 behaves as a highly potent and selective partial agonist at the NOP receptors and that the whole indole moiety of the Trp5 side chain is not required, being a phenyl-ethyl side chain already sufficient for maintaining high potency.


Subject(s)
Amino Acids, Aromatic/chemistry , Oligopeptides/chemistry , Opioid Peptides/pharmacology , Tryptophan/chemistry , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Electric Stimulation , Male , Mice , Mice, Knockout , Oligopeptides/pharmacology , Opioid Peptides/agonists , Structure-Activity Relationship , Vas Deferens/drug effects , Vas Deferens/physiology , Nociceptin
6.
J Pept Res ; 63(6): 477-84, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15175020

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand for the G-protein coupled receptor referred to as N/OFQ peptide (NOP) receptor. NOP receptor activation by N/OFQ modulates several biological functions both at central and peripheral level. Structure activity relationship (SAR) studies demonstrated that the N/OFQ sequence can be divided into a N-terminal tetrapeptide 'message' crucial for receptor activation and a C-terminal 'address' important for receptor binding. On the basis of this message/address concept we synthesized some chimeric compounds in which we substituted the natural message domain with the nonselective nonpeptide NOP ligand (8-Naphthalen-1-yl-methyl-4-oxo-1-phenyl-1,3,8-triaza-spiro[4,5]dec-3-yl)-aceticacid methyl ester (NNC 63-0532) and used as address domain the peptide sequences Thr-NH2, N/OFQ(5-9)-NH2, N/OFQ(5-13)-NH2 and N/OFQ(5-17)-NH2. All the compounds were pharmacologically evaluated in the electrically stimulated guinea-pig ileum. NNC 63-0532 produced a concentration-dependent inhibition of the electrically induced twitches showing, in comparison with N/OFQ, lower potency and higher maximal effects. In addition, contrary to N/OFQ, the effects of NNC 63-0532 were insensitive to the NOP selective antagonist [Nphe1, Arg14, Lys15]N/OFQ-NH2 (UFP-101) while prevented by naloxone. Similar results were obtained with NNC 63-0532/Thr-NH2 and NNC 63-0532/N/OFQ(1-9)-NH2. On the contrary, the inhibitory effects of NNC 63-0532/N/OFQ(5-13)-NH2 and NNC 63-0532/N/OFQ(5-17)-NH2 were slightly antagonized by UFP-101 while naloxone prevented the effects of the high but not of the low concentrations of the two ligands. These data indicate that it is possible to functionalize with the N/OFQ address sequence a nonpeptide NOP ligand for increasing its binding to the NOP receptor. Moreover, these results corroborate the idea that the 5-13 sequence represents the crucial core of the N/OFQ address domain.


Subject(s)
Opioid Peptides/chemistry , Receptors, Opioid/agonists , Acetates/chemistry , Acetates/pharmacology , Amino Acid Sequence , Animals , Drug Design , Guinea Pigs , Ileum/drug effects , Ligands , Male , Molecular Sequence Data , Naloxone/chemistry , Naloxone/pharmacology , Opioid Peptides/chemical synthesis , Opioid Peptides/metabolism , Opioid Peptides/pharmacology , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship , Nociceptin Receptor , Nociceptin
7.
Naunyn Schmiedebergs Arch Pharmacol ; 369(6): 547-53, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15197534

ABSTRACT

Receptor antagonist and knockout studies have demonstrated that blockade of signalling via nociceptin/orphanin FQ (N/OFQ) and its receptor (NOP) has antidepressant-like effects in mice submitted to the forced swimming test (FST). The aim of the present study was to explore further the antidepressant-like properties of the NOP antagonist UFP-101 in different species (mouse and rat) and using different assays [FST and tail suspension test (TST)], and to investigate the mechanism(s) involved in its actions.UFP-101 (10 nmol i.c.v.) reduced immobility time of Swiss mice in the TST (mean+/-SEM) from 179+/-11 to 111+/-10 s. N/OFQ (1 nmol i.c.v.) was without effect per se, but fully prevented the effect of UFP-101. The spontaneous immobility time of NOP(-/-) CD1-C57BL/6J-129 mice in the TST was much lower than that of wild-type (NOP(+/+)) littermates (75+/-11 vs. 144+/-17 s) or of Swiss mice. UFP-101 (10 nmol i.c.v.) decreased immobility time (-65%) and increased climbing time (71%) in rats submitted to the FST. In rat brain slices, N/OFQ (100 nM) triggered robust K(+)-dependent hyperpolarizing currents in locus coeruleus and dorsal raphe neurons. UFP-101 (3 microM) fully prevented N/OFQ-induced currents, but was inactive per se. Fluoxetine, desipramine (both 30 mg/kg i.p.) and UFP-101 (10 nmol i.c.v.) reduced immobility time of mice in the FST. The serotonin synthesis inhibitor p-chlorophenylalanine methylester (PCPA, 4 x 100 mg/kg per day i.p.) prevented the antidepressant-like effects of fluoxetine and UFP-101 (but not desipramine), whereas N-(2-chloroethyl)- N-ethyl-2-bromobenzylamine (DSP-4, neurotoxic for noradrenergic neurons; 50 mg/kg i.p., 7 days beforehand), suppressed only the effect of desipramine. Neither pretreatment affected spontaneous immobility time per se.Thus, UFP-101 exhibits pronounced antidepressant-like effects in different species and animal models, possibly by preventing the inhibitory effects of endogenous N/OFQ on brain monoaminergic (in particular serotonergic) neurotransmission. Participation of the N/OFQ-NOP receptor system in mood modulation sets new potential targets for antidepressant drug development.


Subject(s)
Antidepressive Agents/pharmacology , Narcotic Antagonists , Opioid Peptides/pharmacology , Animals , Brain/drug effects , Brain/physiology , Electrophysiology , Hindlimb Suspension/physiology , Male , Mice , Mice, Knockout , Rats , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Signal Transduction/drug effects , Swimming/physiology , Nociceptin Receptor , Nociceptin
8.
Eur J Neurosci ; 17(9): 1987-90, 2003 May.
Article in English | MEDLINE | ID: mdl-12752799

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ), the endogenous ligand of the NOP receptor, regulates several central functions such as pain transmission, learning and memory, fear and anxiety and feeding and locomotor activity. It has been recently reported that NOP receptor antagonists induce antidepressant-like effects in the mouse forced swimming test (FST), i.e. reduce immobility time. This assay was used in the present study for further investigating the involvement of the NOP receptor in depression states. In male Swiss mice, intracerebroventricular injection (i.c.v) of the novel NOP receptor antagonist, UFP-101 (1-10 nmol) dose-dependently reduced the immobility time (control 192 +/- 14 s, UFP-101 91 +/- 15 s). The effect of 3 or 10 nmol UFP-101 was fully or partially reversed, respectively, by the coadministration of 1 nmol N/OFQ, which was inactive per se. NOP receptor knockout mice showed a reduced immobility time compared with their wild-type littermates (wild-type 215 +/- 10 s, knockout 143 +/- 12 s). Moreover, i.c.v. injected UFP-101 (10 nmol) significantly reduced immobility time in wild-type mice but not in NOP receptor knockout animals. In conclusion, these results, obtained using a combined pharmacological and genetic approach, indicate that blockade of the N/OFQ-NOP receptor signalling in the brain produces antidepressant-like effects in the mouse FST. These findings support the NOP receptor as a candidate target for the development of innovative antidepressant drugs.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Depression/metabolism , Narcotic Antagonists , Receptors, Opioid/genetics , Signal Transduction/genetics , Animals , Antidepressive Agents/pharmacology , Dose-Response Relationship, Drug , Immobilization/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Opioid/deficiency , Receptors, Opioid/physiology , Signal Transduction/drug effects , Swimming/physiology , Nociceptin Receptor
9.
Can J Physiol Pharmacol ; 80(5): 407-12, 2002 May.
Article in English | MEDLINE | ID: mdl-12056546

ABSTRACT

We have studied the effects of naloxone benzoylhydrazone (NalBzoH) at recombinant human OP4 receptors expressed in Chinese hamster ovary (CHO) cells (CHOhOP4) and native OP4 sites in isolated tissues from various species. In CHOhOP4 membranes, nociceptin (NC) and NalBzoH displaced [125I]Tyr14-NC with pKi values of 10.1 and 7.3. In the presence of 100 microM GDP, NC stimulated GTPgamma35S binding (pEC50 = 8.5). NalBzoH was ineffective but antagonized the effects of NC (pA2 = 6.9). At 5 microM GDP, there was an increase in potency (pEC50 = 9.3) and efficacy (4.3-fold) of NC. NalBzOH was a partial agonist (pEC50 = 7.0, Emax = 13% relative to NC). In CHOhOP4 cells, NC and NalBzoH inhibited cAMP formation with pEC50 and Emax values of 9.8 and 100% and 6.0 and 44%, respectively. In the rat vas deferens, NalBzoH (10 microM) did not modify electrically induced twitches but competitively antagonized the inhibitory action of NC (pA2 = 6.2). In the mouse vas deferens (mVD) and guinea pig ileum (gpI), NalBzoH inhibited twitches with pEC50 and Emax values of 7.6 and 78% and 8.5 and 77%, respectively. The effect of 3 microM NalBzoH was fully inhibited by 3 microM naloxone in mVD and 30 microM in gpI. Under these conditions, NalBzoH antagonized the actions of NC in both preparations with pA2 values of 6.3 and 6.8, respectively. Collectively, these data demonstrate that NalBzoH is a nonselective OP4 ligand with system-dependent behaviour.


Subject(s)
Naloxone/analogs & derivatives , Naloxone/pharmacology , Narcotic Antagonists , Recombinant Proteins/antagonists & inhibitors , Animals , CHO Cells/drug effects , CHO Cells/metabolism , Cricetinae , Dose-Response Relationship, Drug , Electric Stimulation , Guinea Pigs , Humans , In Vitro Techniques , Male , Mice , Naloxone/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/metabolism , Nociceptin Receptor
10.
Life Sci ; 71(4): 363-70, 2002 Jun 14.
Article in English | MEDLINE | ID: mdl-12044836

ABSTRACT

Recently, the cloning of a novel preprotachykinin gene (PPT-C) has been reported. This gene codes for a novel peptide named hemokinin 1 (HK-1). In contrast with the known tachykinins, which are exclusively expressed in neuronal tissues, PPT-C mRNA was detected primarily in hematopoietic cells. In this study, we pharmacologically characterised the effects of HK-1 using three tachykinin monoreceptor systems, namely the rabbit jugular vein (rbJV) for NK(1), the rabbit pulmonary artery (rbPA) for NK(2), and rat portal vein (rPV) for NK(3) receptors. In all these preparations substance P (SP), neurokinin A (NKA) and neurokinin B (NKB) elicited concentration dependent contractions showing similar maximal effects and the following rank order of potency: SP > NKA = NKB in the rbJV, NKA > NKB >> SP in the rbPA, and NKB > NKA > SP in the rPV. In those vessels HK-1 behaved as a full agonist displaying potencies similar (rbPA and rPV) or slightly higher (rbJV) than those of SP. In the rbJV, SR 140333, a selective NK(1) receptor antagonist, antagonised the effects of HK-1 and SP with similar high potencies (pK(B) 9.3 and 9.5, respectively). Similar results were obtained with the pseudopeptide NK(1) antagonist, MEN 11467 (pK(B) 8.8 and 8.6, respectively). Taken together, these data indicate that HK-1 behaves as a NK(1) preferring receptor agonist.


Subject(s)
Jugular Veins/drug effects , Protein Precursors/pharmacology , Receptors, Neurokinin-1/metabolism , Tachykinins/pharmacology , Animals , Jugular Veins/metabolism , Male , Portal Vein/drug effects , Portal Vein/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Rabbits , Rats , Receptors, Neurokinin-2/metabolism , Receptors, Neurokinin-3/metabolism
11.
Can J Physiol Pharmacol ; 80(4): 281-6, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12025962

ABSTRACT

Peptide and nonpeptide compounds have been shown to interact specifically with B2 receptors of three different species, namely human, rabbit, and pig. Peptide agonists and nonpeptide antagonists show marked differences in potencies and suggest the existence of B2 receptor subtypes. This conclusion is based on data obtained with the modified agonist peptide LF 150943 whose potency (pEC50 9.4) is at least 100-fold higher in rabbit than in humans (7.4) and pig (6.7). The same conclusion can be drawn from data obtained with antagonists that are more potent in humans (LF 160687, pA2 9.2) than in rabbit (8.7) and pig (8.2) or with antagonists (S 1567) that show the opposite potency order, being much weaker in humans (pA2 6.9) than in rabbit (7.6) and pig (9.4). Two other compounds (FR 173657 and FR 172357) show similar pharmacological spectra as S 1567 and differ from LF 160687.


Subject(s)
Bradykinin Receptor Antagonists , Receptors, Bradykinin/agonists , Adult , Animals , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Ligands , Male , Rabbits , Receptor, Bradykinin B2 , Receptors, Bradykinin/physiology , Swine , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
12.
Can J Physiol Pharmacol ; 80(4): 293-302, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12025964

ABSTRACT

In this study, we describe the in vitro and in vivo activities of a series of cyclic peptide analogues of the selective kinin B2 receptor antagonist MEN11270 on Chinese hamster ovary cells expressing the human B2 receptor (hB2R), the human isolated umbilical vein (hUV), the isolated guinea pig ileum (gpI), and bradykinin (BK) induced bronchoconstriction (BC) and hypotension in anaesthetized guinea pigs. Substitutions in the backbone of MEN1 1270 (H-DArg-Arg-Pro-Hyp-Gly-Thi-c(Dab-DTic-Oic-Arg)c(7gamma-10alpha)) aimed to increase the potency in inhibiting bronchospasm versus hypotension following the topical (intratracheal (i.t.)) or systemic (intravenous (i.v.)) application of these antagonists. A series of analogues were left unprotected from N-terminal cleavage by aminopeptidases (MEN12739, MEN13052, MEN13346, and MEN13371): these compounds maintained sizeable affinities for the hB2R (pKi = 9.4, 9.6, 9.7, and 8.6, respectively) and antagonist activities toward BK in the hUV (pA2 = 7.9, 8.3, 8.2, and 7.5) and gpI assays (pK(B) = 7.4, 7.8, 7.9, and 7.9), but the inhibition of BK-induced BC and hypotension in vivo was negligible following either i.v. or i.t. administration. Two analogues (MEN12388 and MEN13405) could be potential substrates of angiotensin-converting enzyme: these have good activity in the hB2R (pKi = 9.5 and 8.9, respectively), hUV (pA2 = 8.2 for MEN12388), and gpI assays (pK(B) = 8.4 and 8.0) but an in vivo activity 10- to 30-fold lower than the parent compound MEN1 1270 (pKi = 9.4, pA2 = 8.1, pKB = 8.3) when given by either the i.v. or the i.t. route. Other analogues were functionalized with a quaternary ammonium Lys derivative (MEN13031, MEN12374, and the previously mentioned MEN13052) or with an ethyl group on Arg (MEN13655 and the previously mentioned MEN13346 and MEN13405) in order to hinder or facilitate local absorption. MEN13346 and MEN13031 (pKi = 9.7and 9.5, pA2 = 8.2 and 7.9, pKB = 7.9 and 8.5, respectively) were 10- to 30-fold less active in vivo than MEN1 1270, without improving the discrimination between BK-induced BC and hypotension after either systemic or topical administration. It is concluded that the decreased in vivo activities of cyclic analogues of MEN11270 on BK-induced BC and hypotension following either their intratracheal or their intravenous routes of administration might be due in large part to metabolic degradation.


Subject(s)
Bradykinin Receptor Antagonists , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , Adult , Animals , Bradykinin/administration & dosage , Bronchoconstriction/drug effects , Bronchoconstriction/physiology , CHO Cells , Cricetinae , Female , Guinea Pigs , Humans , Hypotension/drug therapy , Hypotension/metabolism , In Vitro Techniques , Injections, Intravenous , Male , Oligopeptides/metabolism , Peptides, Cyclic/metabolism , Receptor, Bradykinin B2 , Receptors, Bradykinin/metabolism
13.
J Urol ; 166(6): 2237-40, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11696742

ABSTRACT

PURPOSE: Management of neurogenic incontinence is complex and available treatments are not satisfactory. Nociceptin/orphanin FQ, a recently discovered neuropeptide, has been reported to inhibit the voiding reflex in the rat. These experimental results prompted us to investigate the urodynamic and clinical effects of intravesical instillation of nociceptin/orphanin FQ in humans. MATERIAL AND METHODS: Our study involved 5 normal subjects (group 1) with a mean age of 40.4 years (range 21 to 54) and 9 patients (group 2) 40.4 years (24 to 54). All patients in group 2 presented with detrusor hyperreflexia refractory to standard therapy. They were invited to undergo a filling cystometrogram with saline solution and after 30 minutes, a new one with a solution containing 1 microM. nociceptin/orphanin FQ. The urodynamic parameters that were recorded included bladder capacity, volume threshold for the appearance of detrusor hyperreflexia and maximum bladder pressure. Clinical and urodynamic followup was performed after 15 days. The data were statistically analyzed with 1-way analysis of variance followed by the Dunnett test for multiple comparison considered statistically significant with p <0.05. RESULTS: Intravesical instillation of 1 microM. nociceptin/orphanin FQ in group 1 did not produce significant functional changes. This infusion in group 2 produced a statistically significant increase in mean bladder capacity and volume threshold for the appearance of detrusor hyperreflexia from 164 plus or minus standard deviation (SD) 84 to 301 +/- 118 and 93 plus or minus SD 41 to 231 +/- 104 ml. (p <0.05, respectively). Mean maximum bladder pressure decreased from 79 plus or minus SD 25 to 54 +/- 44 cm. water but was not statistically significant (p = 0.19). After 15 days an absence of clinical improvement was noticed in group 2, and the urodynamic control did not show any significant changes compared to the values before nociceptin/orphanin FQ treatment. No severe symptomatic reactions were observed during infusion of 1 microM. nociceptin/orphanin FQ. CONCLUSIONS: Our results demonstrate that nociceptin/orphanin FQ is able to elicit a robust inhibitory effect on voiding reflex in group 2 but not 1. The ideal dosage, route of administration of nociceptin/orphanin FQ and treatment interval are not yet established.


Subject(s)
Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Opioid Peptides/administration & dosage , Receptors, Opioid/agonists , Urinary Incontinence/physiopathology , Urodynamics/drug effects , Administration, Intravesical , Adult , Humans , Middle Aged , Pilot Projects , Nociceptin
14.
J Med Chem ; 44(23): 3956-64, 2001 Nov 08.
Article in English | MEDLINE | ID: mdl-11689082

ABSTRACT

A total of 32 compounds was prepared to investigate the functional role of Phe(4) in NC(1-13)-NH(2), the minimal sequence maintaining the same activity as the natural peptide nociceptin. These compounds could be divided into three series in which Phe(4) was replaced with residues that would (i) alter aromaticity or side chain length, (ii) introduce steric constraint, and (iii) modify the phenyl ring. Compounds were tested for biological activity as (a) inhibitors of the electrically stimulated contraction of the mouse vas deferens; (b) competitors of the binding of [(3)H]-NC-NH(2) to mouse brain membranes; and (c) inhibitors of forskolin-stimulated cAMP accumulation in CHO cells expressing the recombinant human OP(4) receptor. Results indicate that all compounds of the first and second series were inactive or very weak with the exception of [N(CH(3))Phe(4)]NC(1-13)-NH(2), which was only 3-fold less potent than NC(1-13)-NH(2). Compounds of the third series showed higher, equal, or lower potencies than NC(1-13)-NH(2). In particular, [(pF)Phe(4)]NC(1-13)-NH(2) (pF) and [(pNO(2))Phe(4)]NC(1-13)-NH(2) (pNO(2)) were more active than NC(1-13)-NH(2) by a factor of 5. In the mVD, these compounds showed the following order of potency: (pF) = (pNO(2)) > or = (pCN) > (pCl) > (pBr) > (pI) = (pCF(3)) = (pOCH(3)) > (pCH(3)) > (pNH(2)) = (pOH). (oF) and especially (mF) maintained high potencies but were less active than (pF). Similar orders of potency were observed in binding competition and cAMP accumulation studies. There was a strong (r(2) > or = 0.66) correlation between data observed in these assays. Biological activity data of compounds of the third series were plotted against some Hansch parameters that are currently used to quantify physicochemical features of the substituents. In the three biological assays agonist potency/affinity positively correlates with the electron withdrawal properties of the groups in the p-position of Phe(4) and inversely with their size.


Subject(s)
Opioid Peptides/chemical synthesis , Peptide Fragments/chemical synthesis , Phenylalanine/chemistry , Receptors, Opioid/agonists , Animals , Brain/metabolism , CHO Cells , Cricetinae , Cyclic AMP/biosynthesis , Electric Stimulation , Humans , In Vitro Techniques , Male , Mice , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Structure-Activity Relationship , Vas Deferens/drug effects , Nociceptin Receptor
15.
Neurosci Lett ; 316(1): 25-8, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11720770

ABSTRACT

Nociceptin/orphanin FQ (NC) and its receptor (OP(4)) have been implicated in the regulation of various functions including nociception. [Nphe(1)]NC(1-13)NH(2) (Nphe) is a selective OP(4) antagonist which prevents the pronociceptive effects of supraspinal NC and causes per se a naloxone-insensitive antinociceptive effect. In the present study, we tested Nphe in wild type (WT) and OP(4) receptor knock out mice and found that a clear antinociceptive effect of the antagonist was evident only in WT mice. Moreover, we evaluated, over 5 days of treatment, the antinociceptive effects of Nphe in comparison with those of DAMGO and found that tolerance develops to the effects of the opioid receptor agonist but not to Nphe. These data demonstrate that the antinociceptive action of Nphe is due to the block of OP(4) receptors and that no tolerance develops to this kind of antinociception.


Subject(s)
Analgesics/pharmacology , Narcotic Antagonists , Opioid Peptides/pharmacology , Peptide Fragments/pharmacology , Analgesics/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , In Vitro Techniques , Injections, Intraventricular , Isometric Contraction/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Opioid Peptides/administration & dosage , Pain Measurement/drug effects , Peptide Fragments/administration & dosage , Reaction Time/drug effects , Receptors, Opioid/deficiency , Nociceptin Receptor
16.
Neuroreport ; 12(14): 3009-13, 2001 Oct 08.
Article in English | MEDLINE | ID: mdl-11568627

ABSTRACT

Nociceptin/orphanin FQ (NC) and its receptor (OP4) have been implicated in pain transmission. The aim of the present study was to investigate the role of the NC/OP4 system in stress-induced analgesia (SIA). The tail-withdrawal assay was performed in mice stressed by forced swimming in water at 15 degrees C (high severity swims) or 32 degrees C (low severity swims). High severity swims produced a naloxone-insensitive antinociceptive effect which was blocked by supraspinal NC (1 nmol). The selective OP4 receptor antagonist, [Nphe1]NC(-13)NH2 (30 nmol), was inactive by itself, but prevented the effect of NC. Low severity swims produced a milder analgesic effect that was partially antagonized by naloxone, completely blocked by NC and potentiated by [Nphe1]NC(-13)NH2. These findings confirm the anti-analgesic role of supraspinal NC and suggest that endogenous NC signaling counteracts the opioid component of SIA.


Subject(s)
Analgesia , Central Nervous System/metabolism , Opioid Peptides/metabolism , Pain/metabolism , Receptors, Opioid/metabolism , Signal Transduction/physiology , Stress, Physiological/metabolism , Animals , Central Nervous System/drug effects , Dose-Response Relationship, Drug , Drug Interactions/physiology , Male , Mice , Narcotic Antagonists , Opioid Peptides/antagonists & inhibitors , Opioid Peptides/pharmacology , Pain/physiopathology , Pain Measurement , Pain Threshold/drug effects , Pain Threshold/physiology , Peptide Fragments/pharmacology , Reaction Time/drug effects , Reaction Time/physiology , Stress, Physiological/physiopathology , Swimming/physiology , Nociceptin Receptor , Nociceptin
17.
Naunyn Schmiedebergs Arch Pharmacol ; 363(5): 551-5, 2001 May.
Article in English | MEDLINE | ID: mdl-11383716

ABSTRACT

The pharmacological profile of the non-peptide OP4 receptor (ORL1, LC132) agonist, Ro 64-6198, was investigated, in three electrically stimulated nociceptin/orphanin FQ (NC)-sensitive preparations, namely the mouse and rat vas deferens and the guinea pig ileum. Ro 64-6198 mimicked the inhibitory effect of NC in the three preparations, while showing slow kinetics of action and a slowly reversible effect compared to the fast and immediately and completely reversible effect of the natural peptide. Ro 64-6198 displayed similar pEC50 and Emax values as NC in the mouse and rat vas deferens while it was 100-fold less potent but more efficacious (higher Emax) than NC in the guinea pig ileum. In the rat vas deferens the effects of Ro 64-6198 were antagonised by [Nphe1]NC(1-13)NH2 and J-113397 with pKB values (6.30 and 8.05, respectively) similar to those obtained against NC (6.20 and 7.77, respectively). Naloxone (1 microM) was inactive. In the guinea pig ileum a clear shift of the concentration response curve to Ro 64-6198 was obtained only using a cocktail of antagonists (naloxone + [Nphe1]NC(1-13)NH2 or naloxone + J-113397). In the mouse vas deferens the antagonists were inactive against Ro 64-6198 either when tested alone or in combination. Therefore, Ro 64-6198 behaved as a selective OP4 receptor agonist only in the rat tissue. These results suggest a physiological heterogeneity in OP4 receptors across tissues and species and may explain why, when tested in vivo, Ro 64-6198 mimics the potent anxiolytic effect of NC better in the rat than in the mouse.


Subject(s)
Anti-Anxiety Agents/pharmacology , Imidazoles/pharmacology , Opioid Peptides/pharmacology , Spiro Compounds/pharmacology , Vasodilator Agents/pharmacology , Animals , Dose-Response Relationship, Drug , Electric Stimulation , Guinea Pigs , Ileum/drug effects , Ileum/physiology , Male , Mice , Organ Specificity/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid/agonists , Receptors, Opioid/physiology , Species Specificity , Vas Deferens/drug effects , Vas Deferens/physiology , Nociceptin Receptor , Nociceptin
18.
J Pept Res ; 57(3): 215-22, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11298922

ABSTRACT

A series of analogs of the ORL1 receptor antagonist [Nphe1]-NC(1-13)-NH2 was prepared and tested for agonistic and antagonistic activities in the mouse vas deferens, a preparation that shows high sensitivity to nociceptin and related peptides. The purpose of this study was to determine the role of the aromatic residue at the N-terminal for antagonism and eventually identify compounds with improved potency. Results indicated that all 23 compounds are inactive as agonists, and the antagonistic potency of the initial template [Nphe1]-NC(1-13)-NH2 is high (pKB 6.43) compared with those of all other compounds except [(S)(betaMe)Nphe1]NC(1-13)-NH2 (pK(B) 6.48). The other 22 compounds can be divided into two groups: 10 show antagonistic potencies (pK(B)) ranging from 5.30 to 5.86, whereas the other 12 compounds are inactive. This study clearly shows that the aromatic ring of Nphe is very critical for the interaction with the ORL1 receptor and can not be enlarged or sterically modified without significant loss of antagonistic potency.


Subject(s)
Narcotic Antagonists , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Drug Evaluation, Preclinical , Electric Stimulation , Male , Mice , Molecular Sequence Data , Receptors, Opioid/drug effects , Structure-Activity Relationship , Vas Deferens/drug effects , Vas Deferens/physiology , Nociceptin Receptor
19.
Biol Chem ; 382(1): 31-5, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11258668

ABSTRACT

This minireview is divided into three parts: the first part refers to the characterization and classification of kinin receptors using agonists and antagonists in isolated tissues (classical pharmacology). Two kinin receptors have been considered on the basis of their distinct pharmacology, namely the B1 receptor of the rabbit aorta (rank order of potency of agonists: LysdesArg9BK > desArg9BK > or = LysBK > BK; apparent affinities of antagonists Lys[Leu8]desArg9BK (pIC50 8.4) > [Leu8]desArg9BK (pIC50 7.4) >>> HOE 140, a B2 receptor antagonist, pIC50<5.0), and the B2 receptor of the rabbit jugular vein (potency of agonists: LysBK = BK >>> LysdesArg9BK = desArg9BK and HOE 140 (pIC50 9.0) >>> Lys[Leu8]desArg9BK, pIC50<5.0). The second part describes species-related B1 receptor subtypes, demonstrated by different pharmacological profiles of agonists and antagonists: human, rabbit and pig subtypes (LysdesArg9BK >> desArg9BK and Lys[Leu8]desArg9BK > [Leu8]desArg9BK) and dog, rat, mouse and hamster B1 receptors (desArg9BK = LysdesArg9BK and [Leus]desArg9BK = Lys[Leu8]desArg9BK). Affinities of agonists and antagonists in some species (man, rabbit, pig) are significantly increased (at least 10-fold) by the presence of a Lys at their N-terminus. The last part describes species-related B2 receptor subtypes supported by results obtained with non-peptide receptor agonists (FR 190997) and antagonists (FR 173657). While BK acts as a full agonist in man, rabbit and pig, FR 190997 behaves as a full agonist on human, as partial agonist on rabbit, and as pure antagonist on pig B2 receptors. Various hypotheses are considered to interpret these findings.


Subject(s)
Kinins/metabolism , Receptors, Cell Surface/classification , Animals , Biological Assay , Humans , Peptides/pharmacology , Receptors, Cell Surface/agonists , Receptors, Cell Surface/antagonists & inhibitors , Species Specificity
20.
Am J Physiol Lung Cell Mol Physiol ; 280(4): L732-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11238014

ABSTRACT

Using monolayers of bovine aortic endothelial cells (BAEC) in modified Boyden chambers, we examined the role of prostaglandins (PGs) in the bradykinin (BK)-induced increase of albumin permeability. BK induced a concentration-dependent increase of the permeability of BAEC, which reached 49.9 +/- 1% at the concentration of 10(-8) M. Two inhibitors of the prostaglandin G/H synthase, indomethacin (2.88 microM) and ibuprofen (10 microM), potentiated BK-induced permeability 1.8- and 3.9-fold, respectively. Exogenously administered PGE2 and iloprost, a stable analog of prostacyclin, attenuated the effect of BK in a concentration-dependent manner. Butaprost equally reduced the effect of BK, suggesting the participation of the EP2 receptor in this phenomenon. However, the EP4-selective antagonist AH-23848 did not significantly inhibit the protective effect of PGE2. The inhibitory effect of PGE2 was reversed by the adenylate cyclase inhibitor MDL-12330A (10 microM). These results suggest that BK-induced increase of permeability of BAEC monolayer to (125)I-labeled albumin is negatively regulated by PGs. This postulated autocrine activity of PGs may involve an increase in the intracellular level of cAMP.


Subject(s)
Alprostadil/analogs & derivatives , Bradykinin/pharmacology , Capillary Permeability/drug effects , Endothelium, Vascular/metabolism , Prostaglandins/pharmacology , Serum Albumin/metabolism , Adrenergic beta-Agonists/pharmacology , Albuterol/pharmacology , Alprostadil/pharmacology , Animals , Cattle , Cyclic AMP/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Endothelium, Vascular/cytology , Iloprost/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...