Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Curr Med Chem ; 18(3): 398-426, 2011.
Article in English | MEDLINE | ID: mdl-21143118

ABSTRACT

The intestinal epithelial monolayer constitutes a physical and functional barrier between the organism and the external environment. It regulates nutrients absorption, water and ion fluxes, and represents the first defensive barrier against toxins and enteric pathogens. Epithelial cells are linked together at the apical junctional complex by tight junctions that reduce the extracellular space and the passage of charge entities while forming a physical barrier to lipophilic molecules. Cultured intestinal epithelial cells have been extensively used to study intestinal absorption of newly synthesized drugs and the regulation of tight junctions structure and function. In vitro mild irritants, proinflammatory cytokines, toxins and pathogens, and adverse environmental conditions open tight junctions and increase paracellular permeability, an effect often accompanied by immune activation of the enterocytes. Conversely, inhibition of proinflammatory cytokines, exposure to growth factors and probiotics, among others, exert a protective effect. Impaired barrier function results from activation of signalling pathways that lead to alteration of junctional proteins expression and/or distribution. In vivo, intestinal barrier dysfunction is associated with various intestinal and non-intestinal disorders including inflammatory bowel disease, celiac disease, and diarrhoeal infection. This review will describe the current knowledge of the mechanisms regulating tight junctions and intestinal permeability, how these findings have lead to a better understanding of barrier alteration in human intestinal disorders, and what the emerging therapies to treat these pathologies are.


Subject(s)
Intercellular Junctions/chemistry , Intestines/physiopathology , Animals , Anti-Inflammatory Agents/therapeutic use , Celiac Disease/pathology , Celiac Disease/therapy , Cytokines/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Intercellular Junctions/metabolism , Intestinal Mucosa/metabolism , Permeability , Polymorphism, Genetic , Probiotics/therapeutic use
2.
Naunyn Schmiedebergs Arch Pharmacol ; 363(2): 175-81, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11218070

ABSTRACT

We have compared the effect of bradykinin (BK) and the nonpeptide kinin B2 receptor agonist, FR 190997, in producing changes in membrane potential and tension in the circular muscle of guinea-pig colon by the sucrose gap technique. In the presence of atropine (1 microM), S-ketoprofen (3 microM) and apamin (0.1 microM), BK (1 microM for 20 s) induced a transient depolarization of the membrane with superimposed action potentials (spikes) and transient contraction. Nifedipine (1 microM) eliminated the spikes and markedly inhibited the BK-induced contractions. FR 190997 (3-10 microM for 20 s) induced a slowly developing sustained small depolarization associated with a slowly developing and sustained contraction but, contrary to BK, FR 190997 was unable to trigger spikes. Nifedipine had no effect on depolarization and contraction induced by FR 190997. In the presence of 1 microM nifedipine, the combined application of a blocker of receptor-operated cation channels, SKF 96365 (50 microM for 30 min), and of an inhibitor of sarcoplasmic reticulum calcium pump, cyclopiazonic acid (CPA 10 microM for 30 min), reduced the BK-induced depolarization and contraction by about 45%-60%. The same treatment induced about 40% reduction of the sustained contraction induced by FR 190997, whereas the concomitant depolarization was not significantly affected. The tyrosine kinase inhibitor genistein (40 microM for 20 min) had no effect on the BK- and FR 190997-induced depolarization and contraction in the presence of nifedipine. In radioligand binding experiments performed in membranes of colonic smooth muscle cells, both agonists displaced the [3H]BK specific binding with a pIC50 of 9.6 and 8.5 for BK and FR 190997, respectively. These findings indicate a substantial qualitative difference in mechanisms of excitation contraction coupling activated by BK and FR 190997 via B2 receptors in guinea-pig colon.


Subject(s)
Bradykinin/pharmacology , Colon/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Quinolines/pharmacology , Receptors, Bradykinin/agonists , Animals , Cells, Cultured , Colon/metabolism , Guinea Pigs , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscle Contraction/physiology , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Receptor, Bradykinin B2 , Receptors, Bradykinin/metabolism
3.
Br J Pharmacol ; 130(2): 331-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10807670

ABSTRACT

Prostanoids, generated from cyclooxygenase (COX) isoenzymes, play a role in the physiological function of the lower urinary tract and are important mediators of inflammatory hyperalgesia. The present work evaluates the effects of the COX-1/COX-2 inhibitor dexketoprofen as well as of a selective COX-2 inhibitor, NS-398, on urodynamic function following endotoxin (LPS) or cyclophosphamide (CYP)-induced inflammation of the urinary bladder. The application of arachidonic acid (330 microgram rat(-1)) onto the serosal surface of the urinary bladder in control rats elicited bladder contractions which could be blocked in a dose-dependent manner by dexketoprofen (0.1 - 3 mg kg(-1), i.v.) but not by NS-398 (0.2 - 6 mg kg(-1), i.v. ). Dexketoprofen (3 mg kg(-1), i.v.) decreased the micturition frequency and increased the pressure threshold for triggering the micturition either when administered within 15 min or 3 h following surgery in control animals. NS-398 (6 mg kg(-1), i.v.) decreased the micturition frequency and increased the pressure threshold when administered 3 h but not 15 min following surgery. Administration of LPS (2 mg kg(-1), i.v., 90 - 120 min) increased both the micturition frequency and the pressure threshold for triggering the micturition reflex. Changes in urodynamic parameters induced by LPS were prevented by doses of either dexketoprofen (1 mg kg(-1), i.v.) or NS-398 (2 mg kg(-1), i.v.) which were ineffective in control animals. Pretreatment with CYP (150 mg kg(-1), i.p., 48 h) increased the micturition frequency, pressure threshold, and the minimal intravesical pressure but decreased the mean amplitude of micturition contractions. In CYP-treated rats, dexketoprofen (1 mg kg(-1), i.v.) or NS-398 (2 mg kg(-1), i.v.) blocked the CYP-induced urodynamic changes with exception of the micturition contraction amplitude. These results indicate that COX-1 may be involved in modulating the threshold for activating the micturition reflex in the normal rats and also demonstrates that inhibition of COX-2 prevents or reverses the urodynamic changes associated with bladder inflammation induced either by surgery, LPS or CYP treatments.


Subject(s)
Cystitis/enzymology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Arachidonic Acids/metabolism , Cyclooxygenase 1 , Cyclooxygenase 2 , Cystitis/physiopathology , Isoenzymes/drug effects , Isoenzymes/pharmacology , Isoenzymes/physiology , Male , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/pharmacology , Prostaglandin-Endoperoxide Synthases/physiology , Rats , Rats, Wistar , Urinary Bladder/enzymology , Urinary Bladder/physiopathology , Urination
4.
Naunyn Schmiedebergs Arch Pharmacol ; 361(4): 452-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10763862

ABSTRACT

In isolated strips of the hamster urinary bladder the selective tachykinin NK2 receptor agonist [betaAla8]NKA(4-10) induced a concentration-dependent (1 nM-10 microM) contraction (EC50 104 nM) associated with significant release of prostaglandin E2 (PGE2, 50+/-17 pg/mg tissue). In mucosa-free bladder strips [betaAla8]NKA(4-10) was as potent as in the presence of mucosa (EC50 46 nM), although the evoked PGE2 release was significantly less than in controls (6+/-1.7 pg/mg tissue). Dexketoprofen (10 microM) produced a significant but limited rightward shift of the concentration/response curve to [betaAla8]NKA(4-10) both in the presence and absence of the mucosal layer: the EC50 for [betaAla8]NKA(4-10) was increased five- and threefold, respectively. The evoked PGE2 release was abolished in both cases. The selective tachykinin NK2 receptor antagonist, nepadutant (10 nM-1 microM) produced a concentration-dependent and even inhibition of both contraction and PGE2 release induced by [betaAla8]NKA(4-10). The L-type calcium channel blocker, nifedipine (1 microM) and the non-selective cationic channel blocker SKF 96365 (30 microM) both inhibited the contractile response to [betaAla8]NKA(4-10) (89+/-2 and 83+/-2% inhibition, respectively). The evoked PGE2 release was not affected by nifedipine but was almost abolished by SKF 96365. Arachidonic acid (100 microM) induced a contractile response (5.9+/-0.7 mN) associated with a large production of PGE2 (383+/-78 pg/mg tissue). The contractile response to arachidonic acid was inhibited by both nifedipine (1 microM) and SKF 96365 (30 microM) (83+/-5 and 79+/-3% inhibition, respectively). The PGE2 production induced by arachidonic acid was markedly inhibited by SKF 96365 only (about 94% inhibition). Exogenous PGE2 contracted hamster bladder strips in the presence of dexketoprofen (EC50 1 microM) and strongly potentiated the contractile response to a submaximal concentration of [betaAla8]NKA(4-10). In anaesthetized hamsters the administration of [betaAla8]NKA(4-10) (10 nmol/kg, i.v.) produced a contractile response of the urinary bladder (13+/-0.4 mmHg) that was inhibited partly by dexketoprofen (25+/-3 and 35+/-4% inhibition for 0.2 and 2 mg/kg, i.v. dexketoprofen, respectively). We conclude that activation of tachykinin NK2 receptors determines prostanoid synthesis/release in the hamster urinary bladder and that this effect is largely ascribable to structures present in the bladder mucosa. Prostanoids generated following NK2 receptor activation amplify the direct contractile effect of NK2 receptor agonists. This latter response is largely due to activation of L-type calcium channels (nifedipine-sensitive) although this source of calcium apparently is not essential for activation of prostanoid synthesis.


Subject(s)
Prostaglandins/physiology , Receptors, Neurokinin-2/agonists , Urinary Bladder/drug effects , Animals , Cricetinae , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/metabolism , Dinoprostone/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Ketoprofen/chemistry , Ketoprofen/pharmacology , Male , Mesocricetus , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Neurokinin A/analogs & derivatives , Neurokinin A/pharmacology , Nifedipine/pharmacology , Peptide Fragments/pharmacology , Stereoisomerism , Urinary Bladder/metabolism , Urinary Bladder/physiology
5.
J Pharmacol Exp Ther ; 290(2): 487-95, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10411554

ABSTRACT

We used membranes from Chinese hamster ovary cells stably transfected with the human tachykinin NK(2) receptor, either wild-type or mutated, at four aromatic residues (His(198), Tyr(266), Phe(270), Tyr(289)) located in transmembrane segments V to VII, to assess the role of these residues in the binding of natural tachykinins and peptide and nonpeptide antagonists. Three radioligands, the agonist [(125)I]neurokinin A (NKA), the peptide antagonist [(3)H]MEN 11420, and the nonpeptide antagonist [(3)H]SR 48968 bound to the wild-type receptor with high affinity (K(d) = 2.4 nM, 0.3 nM, and 4.0 nM, respectively). Four of the six mutant receptors tested retained high affinity for at least one of the radioligands. H(198)A mutation abrogated the binding of NKA but not that of MEN 11420 or SR 48968 (K(d) = 4.8 and 11.5 nM, respectively); Y(266)F mutation abrogated the binding of MEN 11420 but not that of NKA or SR 48968 (K(d) = 2.8 nM and 1.2 nM, respectively); F(270)A mutation abrogated the binding of both NKA and MEN 11420 but not that of SR 48968 (K(d) = 1.6 nM); Y(289)F mutation abrogated the binding of SR 48968 but not that of NKA and MEN 11420 (K(d) = 2.0 and 2.9 nM, respectively). Y(266)A and Y(289)A mutations abrogated the binding of all radioligands. Among the unlabeled antagonists, the affinity of the nonpeptide GR 159897, at variance with SR 48968, resulted heavily compromised by H(198)A and Y(266)F mutations; the peptide antagonists R396 and MEN 10376 essentially followed the binding profile of NKA, but R396 showed markedly increased affinity for the Y(289)F mutant receptor. Taken together, these results indicate that different, partially overlapping sets of sites may be involved in the binding of agonists and diverse antagonists to the human tachykinin NK(2) receptor.


Subject(s)
Peptides/metabolism , Receptors, Neurokinin-2/antagonists & inhibitors , Receptors, Neurokinin-2/chemistry , Animals , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Binding, Competitive , CHO Cells , Cricetinae , DNA, Complementary/drug effects , DNA, Complementary/genetics , Humans , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacology , Mutagenesis, Site-Directed , Mutation , Neurokinin A/chemistry , Neurokinin A/metabolism , Neurokinin A/pharmacology , Peptides/chemistry , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Piperidines/chemistry , Piperidines/metabolism , Piperidines/pharmacology , Protein Conformation , Receptors, Neurokinin-2/genetics , Receptors, Neurokinin-2/metabolism
6.
Biochem Pharmacol ; 57(8): 899-906, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10086323

ABSTRACT

A point mutation was made at position 289 in the transmembrane segment 7 of the human tachykinin NK2 receptor to yield a tyrosine/phenylalanine (Tyr/Phe) substitution. Chinese hamster ovary cells stably transfected with the wild-type or Tyr289Phe mutant NK2 receptor both bound neurokinin A (NKA) and the synthetic NK2 receptor-selective agonists, GR 64349 and [betaAla8]NKA(4-10), with high and even affinities. Neurokinin B (NKB) and substance P (SP) also displayed sizeable binding affinities, albeit with lower affinity as compared to NKA. In a functional assay (production of inositol-1,4,5-trisphosphate, IP3), NKA, GR 64349, and [betaAla8]INKA(4-10) stimulated IP3 accumulation via the wild-type and mutant receptors with similar potencies. On the other hand, NKB and SP exhibited a dramatic reduction in their agonist efficacies at the mutant receptor, NKB acting as a partial agonist (maximum effect = 50% of the response to NKA) and SP being totally inactive. The results obtained with phenoxybenzamine inactivation experiments indicated that a large and similar receptor reserve existed for both the wild-type and the mutant receptor. SP, which displayed sizeable binding affinity for the mutant receptor but did not stimulate IP3 accumulation, antagonized the agonist effect of NKA. The antagonist action of SP at the mutant NK2 receptor cannot be ascribed to receptor internalization. The Tyr/Phe replacement at position 289 markedly reduced the binding affinity and antagonist potency of the non-peptide ligand, SR 48968, without affecting the binding affinity and antagonist potency of the bicyclic peptide antagonist MEN 11420. The results indicate that the hydroxyl radical function of Tyr289 in transmembrane segment 7 of the human NK2 receptor is, directly or indirectly, involved in stimulus transduction when the NK2 receptor is occupied by NKB or SP, but not when using NKA or NK2 receptor-selective agonists.


Subject(s)
Phenylalanine/physiology , Receptors, Neurokinin-2/physiology , Signal Transduction , Tachykinins/metabolism , Tyrosine/physiology , Animals , Binding, Competitive , CHO Cells , Cricetinae , Guanosine Triphosphate/metabolism , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Neurokinin A/antagonists & inhibitors , Neurokinin A/metabolism , Phenoxybenzamine/pharmacology , Phenylalanine/genetics , Point Mutation , Receptors, Neurokinin-2/genetics , Substance P/pharmacology , Transfection , Tyrosine/genetics
7.
Naunyn Schmiedebergs Arch Pharmacol ; 358(4): 395-403, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9826060

ABSTRACT

The human tachykinin NK2 receptor stably expressed in Chinese hamster ovary cells (CHO-hNK2R cells) was characterized by studying the effect of neurokinin A (NKA), the preferred natural ligand, and that of other agonists and antagonists in both binding experiments and functional assays. Competition experiments using [125I]NKA showed that CHO-hNK2R cells express binding sites which have high affinity for NKA (Ki=3.4+/-0.9 nM), GR 64349 (Ki=12+/-3 nM) and [betaAla8]NKA(4-10) (Ki=21+/-8 nM) and for the antagonists MEN 10627 (Ki=0.55+/-0.2 nM), and MEN 11420 (Ki=2.4+/-0.8 nM). In contrast, the tachykinin NK1 and NK3 receptor agonists [Sar9,Met(O2)11]SP and senktide, respectively, were recognized with low affinity (Ki>10 microM). NKA (EC50=68+/-18 nM) induced a rapid and concentration-dependent increase in the intracellular level of inositoltrisphosphate (IP3). The concentration-response curve to GR 64349 (EC50=155+/-14 nM) was close to that of NKA, whereas [betaAla8]NKA(4-10) (EC50=445+/-78 nM) and SP (EC50=3197+/-669 nM) were 7- and 50-fold less potent, respectively. In addition, NKA stimulated the release of arachidonic acid and the production of prostaglandin E2 (PGE2) in a concentration-dependent manner. Also in this assay, NKA was found to be more potent than the other agonists tested (the EC50 values were 3+/-0.3, 9+/-3, 7.8+/-0.9 and 217+/-37 nM for NKA, GR 64349, [betaAla8]NKA(4-10) and SP, respectively). MEN 10627 and MEN 11420 were potent and competitive antagonists in blocking NKA-induced IP3 formation and PGE2 release: MEN 10627 and MEN 11420 displayed comparable potencies in blocking the two functional responses initiated by occupancy of the NK2 receptor by NKA. Pretreatment of the cells with pertussis toxin (500 ng/ml for 18 h) did not significantly modify the basal or stimulated phosphatidylinositol turnover but reduced the basal and NKA-induced PGE2 release by about 35%. The phospholipase C inhibitor U-73122 (10 microM) prevented the NKA-induced formation of IP3 but did not affect PGE2 release. Conversely, the phospholipase A2 inhibitor quinacrine (100 microM) blocked the release of arachidonic acid and PGE2 without affecting the NKA-stimulated formation of IP3. Chelation of extracellular calcium with 3 mM EGTA inhibited the NKA-induced PGE2 release by 81% but was without effect on basal and NKA-stimulated IP3 production. The calcium channel blockers verapamil (10 microM) and omega-conotoxin GVIA (0.1 microM) did not modify the basal PGE2 production and had no significant effect on the response to tachykinins while the blocker of non-selective cation channels, SKF-96365 (10 microM), inhibited the response to NKA by about 74%. SKF-96365 did not affect the basal or the NKA-induced IP3 formation. In conclusion, our data demonstrate that the human tachykinin NK2 receptor expressed in CHO cells displays binding affinity and functional properties which are those of a native NK2 receptor. No pharmacological evidence for heterogeneity of the human NK2 receptor was obtained in this study. Our findings indicate that the human tachykinin NK2 receptor is independently coupled to both PLC and PLA2 signaling pathways. Activation of the PLA2 pathway may be linked to the opening of a voltage-independent cation channel which activates a Ca2+-dependent PLA2.


Subject(s)
Phospholipases A/physiology , Receptors, Neurokinin-2/physiology , Type C Phospholipases/physiology , Animals , Arachidonic Acid/metabolism , Binding Sites , CHO Cells , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cricetinae , Dinoprostone/metabolism , Humans , Inositol 1,4,5-Trisphosphate/biosynthesis , Pertussis Toxin , Phospholipases A2 , Transfection , Virulence Factors, Bordetella/pharmacology
8.
J Pharmacol Exp Ther ; 287(1): 137-43, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9765332

ABSTRACT

The contractile responses elicited by the selective kinin B1 and B2 receptor agonists [desArg9]-bradykinin ([desArg9]-BK) and [Hyp3, Tyr(Me)8]-bradykinin ([Hyp3, Tyr(Me)8]-BK) (1 nM-10 microM), respectively, were evaluated in control vs. inflamed (cyclophosphamide 150 mg kg-1 i.p., 48 h before the sacrifice) rat isolated urinary bladder strips. The contractile responses to the B2 receptor agonist did not differ in control vs. inflamed bladders, whereas the contractile responses to [desArg9]-BK were potentiated in inflamed bladders. The selective B1 and B2 receptor antagonists B 9858 (H-Lys-Lys-Arg-Pro-Hyp-Gly-Igl-Ser-DIgl-Oic-OH) and Hoe 140 (H-DArg-Arg-Pro-Hyp-Gly-Thi-Ser-DTic-Oic-Arg-OH), both at 1 microM, inhibited the response to the B1 and B2 receptor agonists, respectively, in both control and inflamed bladders. In addition, the concentration-response curve to [Hyp3, Tyr(Me)8]-BK was shifted to the right and depressed by B 9858 in inflamed bladders. The nonselective cyclooxygenase (COX) inhibitors S-(-)-ketoprofen (10 microM) and piroxicam (30 microM) markedly depressed the concentration-response curves to [desArg9]-BK and [Hyp3, Tyr(Me)8]-BK in control bladders, but neither drug affected the B1 or B2 receptor agonist-mediated responses in inflamed bladders. The selective inhibitor of the inducible COX-2 isoenzyme, NS-398 (1 microM), did not inhibit the contractile responses to [desArg9]-BK and [Hyp3, Tyr(Me)8]-BK in either control or inflamed bladders, whereas it significantly potentiated the response to the B1 receptor agonist in inflamed bladders. The exogenous administration of prostaglandin E2 (PGE2) induced S-(-)-ketoprofen-resistant contractile responses that were depressed in inflamed bladders. Pretreatment with S-(-)-ketoprofen restored the PGE2-mediated contractile responses of inflamed bladders to control values. PGE2 assay revealed that the basal production of PGE2 is significantly higher after inflammation than in control conditions. [desArg9]-BK and [Hyp3, Tyr(Me)8]-BK (1 microM each) both stimulated PGE2 production, and their effect was larger in inflamed than in control bladders. Piroxicam (30 microM) prevented the PGE2 production evoked by [desArg9]-BK in both control and inflamed bladders and likewise abolished that produced by [Hyp3, Tyr(Me)8]-BK. NS-398 (1 microM) reduced the PGE2 production elicited by [desArg9]-BK in control and inflamed bladders. When NS-398 was tested on the [Hyp3, Tyr(Me)8]-BK-induced PGE2 production, it inhibited PGE2 production in the inflamed bladders only, without significantly modifying the response obtained in controls. These findings demonstrate that 1) in normal bladders, the activation of B1 and B2 receptors evokes contraction that is largely mediated by COX-1 metabolites, whereas the COX-2 appears to be involved in PGE2 production after the activation of B1 receptor only, without interfering with contraction, and 2) in inflamed bladders, the activation of B1 and B2 receptors still produce PGE2, but the contractile response is not reduced by COX inhibitors, a result that indicates that additional mechanisms play a compensatory role.


Subject(s)
Bradykinin/pharmacology , Cystitis/physiopathology , Muscle Contraction/drug effects , Prostaglandin-Endoperoxide Synthases/physiology , Receptors, Bradykinin/agonists , Urinary Bladder/drug effects , Animals , Dinoprostone/biosynthesis , Dinoprostone/pharmacology , In Vitro Techniques , Ketoprofen/pharmacology , Male , Nitrobenzenes/pharmacology , Piroxicam/pharmacology , Rats , Rats, Wistar , Receptor, Bradykinin B1 , Receptor, Bradykinin B2 , Sulfonamides/pharmacology , Urinary Bladder/physiology
9.
Biochem Biophys Res Commun ; 248(1): 78-82, 1998 Jul 09.
Article in English | MEDLINE | ID: mdl-9675089

ABSTRACT

[3H]MEN 11420, a radiolabeled glycosylated peptide antagonist of the tachykinin NK2 receptor, has been investigated in ligand-receptor binding assays using membranes of CHO cells transfected with the human tachykinin NK2 receptor. [3H]MEN 11420 bound to a single class of high affinity binding sites: its binding was inhibited by natural tachykinins (potency ranking: NKA >> SP > or = NKB), as well as by peptide (MEN 11420 > MEN 10376 >> R 396) and nonpeptide (SR 48968 > GR 159897) selective NK2 receptor antagonists. These data indicate that [3H]MEN 11420 is a potent radioligand for the human tachykinin NK2 receptor that may represent a useful tool for studying ligand-receptor interactions at the molecular level.


Subject(s)
Peptides, Cyclic/metabolism , Receptors, Neurokinin-2/antagonists & inhibitors , Animals , Binding Sites , Binding, Competitive , CHO Cells , Cricetinae , Humans , Kinetics , Radioligand Assay , Receptors, Neurokinin-2/genetics , Receptors, Neurokinin-2/metabolism , Tachykinins/metabolism , Tachykinins/pharmacology , Transfection
10.
Br J Pharmacol ; 123(1): 81-91, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9484857

ABSTRACT

1. The pharmacological profile was studied of MEN 11420, or cyclo[[Asn(beta-D-GlcNAc)-Asp-Trp-Phe-Dap-Leu]cyclo(2beta-5beta )], a glycosylated derivative of the potent, selective, conformationally-constrained tachykinin NK2 receptor antagonist MEN 10627 (cyclo(Met-Asp-Trp-Phe-Dap-Leu)cyclo(2beta-5beta)). 2. MEN 11420 competitively bound with high affinity to the human NK2 receptor stably transfected in CHO cells, displacing radiolabelled [125I]-neurokinin A and [3H]-SR 48968 with Ki values of 2.5+/-0.7 nM (n = 6) and 2.6+/-0.4 nM (n = 3), respectively. 3. MEN 11420 showed negligible binding affinity (pIC50 < 6) at 50 different receptors (including tachykinin NK1 and NK3 receptors) and ion channels. 4. In the rabbit isolated pulmonary artery and rat urinary bladder MEN 11420 potently and competitively antagonized tachykinin NK2 receptor-mediated contractions (pK(B) = 8.6+/-0.07, n = 10, and 9.0+/-0.04, n = 12; Schild plot slope = -1.06 (95% c.l. = -1.3; -0.8) and -1.17 (95% c.l. = -1.3; -1.0), respectively). MEN 11420 produced an insurmountable antagonism at NK2 receptors in the hamster trachea and mouse urinary bladder. However, in both preparations, the effect of MEN 11420 was reverted by washout and an apparent pK(B) of 10.2+/-0.14, n = 9, and 9.8+/-0.15, n = 9, was calculated in the hamster trachea and mouse urinary bladder, respectively. 5. MEN 11420 showed low affinity (pK(B) < 6) at guinea-pig and rat tachykinin NK1 (guinea-pig ileum and rat urinary bladder) and NK3 (guinea-pig ileum and rat portal vein) receptors. On the whole, the affinities (potency and selectivity) showed by MEN 11420 for different tachykinin receptors, measured either in binding or in functional bioassays, were similar to those shown by the parent compound, MEN 10627. 6. The in vivo antagonism of the contractions produced by [betaAla8]neurokinin A(4-10) (1 nmol kg(-1)) was observed after intravenous (dose range: 1-10 nmol kg(-1)), intranasal (3-10 nmol kg(-1)), intrarectal (30-100 nmol kg(-1)) and intraduodenal (100-300 nmol kg(-1)) administration of MEN 11420. MEN 11420 was more potent (about 10 fold) and longer lasting than its parent compound MEN 10627, possibly due to a greater metabolic stability. 7. A dose of MEN 11420 (100 nmol kg(-1), i.v.), that produced potent and long lasting inhibition of the contraction of the rat urinary bladder induced by challenge with the NK2 selective receptor agonist [betaAla8]neurokinin A(4-10) (10-300 nmol kg(-1)), was without effect on the responses produced by the NK1 receptor selective agonist [Sar9]substance P sulphone (1-10 nmol kg(-1)). 8. These findings indicate that MEN 11420 is a potent and selective tachykinin NK2 receptor antagonist. The introduction of a sugar moiety did not produce major changes in the affinity profile of this antagonist as compared to MEN 10627, but markedly improved its in vivo potency and duration of action. With these characteristics, MEN 11420 is a suitable candidate for studying the pathophysiological significance of tachykinin NK2 receptors in humans.


Subject(s)
Peptides, Cyclic/pharmacology , Receptors, Neurokinin-2/antagonists & inhibitors , Animals , CHO Cells , Cricetinae , Female , Guinea Pigs , Humans , In Vitro Techniques , Ion Channels/drug effects , Ion Channels/metabolism , Male , Peptides, Cyclic/antagonists & inhibitors , Peptides, Cyclic/pharmacokinetics , Rabbits , Radioligand Assay , Rats , Rats, Wistar , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-2/metabolism , Receptors, Neurokinin-3/metabolism
11.
Can J Physiol Pharmacol ; 75(6): 696-703, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9276151

ABSTRACT

Substance P (SP) and neurokinin A (NKA) are synthesized by enteric cholinergic motorneurons that project to the longitudinal and circular muscle of the mammalian intestine. Thus, acetylcholine, SP, and NKA are the excitatory neuromuscular transmitters in the intestine. Tachykinin NK1 and NK2 receptors are expressed by smooth muscle cells in most regions of the intestine: the corelease of SP and NKA from nerves thus realizes paradigms of tachykininergic cotransmission. Examples have been found in which a cooperative model can be applied to account for the action of SP-NKA acting at NK1 and NK2 receptors (e.g., circular muscle of guinea-pig duodenum), as well as examples in which the message produced by activation of the two receptors diverges sharply in producing responses that have a markedly different time course and use different effector systems (e.g., circular muscle of guinea-pig colon). NK3 receptors are expressed on both excitatory and inhibitory motor neurons: indirect contractions (via release of acetylcholine and tachykinins) and relaxations (via release of nitric oxide) can be evoked in the gut by selective stimulation of NK3 receptors. Although a role of NK3 receptors in certain enteric reflexes has been evidenced, the importance of this system in mediating hexamethonium-resistant enteric transmission appears less important than previously speculated.


Subject(s)
Gastrointestinal Motility/physiology , Receptors, Tachykinin/physiology , Animals , Guinea Pigs , Humans , Rats
12.
Thromb Haemost ; 76(4): 603-9, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8903003

ABSTRACT

The aim of the present work was to study how human umbilical vein smooth muscle cells (HUVSMC) can initiate the coagulation process and to investigate the responses of these cells to thrombin. Exposure of HUVSMC to recalcified human plasma led to a time-dependent production of thrombin, measured both as amidolytic activity and as release of fibrinopeptide A. Thrombin activity was dose-dependently reduced by an anti-human tissue factor antibody (76 +/- 3% at 10 micrograms/ml) and by inhibitors like heparin, rec-hirudin, hirulog 1, Napap and hirunorm, a novel hirudin-like thrombin inhibitor (IC50 = 2 +/- 0.4, 8 +/- 1, 130 +/- 22, 199 +/- 29 and 68 +/- 8 nM, respectively). The release of fibrinopeptide A was similarly prevented (IC50 = 14 +/- 1, 132 +/- 25 and 50 +/- 8 nM for rec-hirudin, Napap and hirunorm, respectively). Exogenously added thrombin increased thymidine incorporation into HUVSMC to 240 +/- 30% of basal (EC50 = 0.49 +/- 0.09 nM) and thrombin inhibitors blocked this effect (IC50 = 10 +/- 3, 37 +/- 17, 343 +/- 165 and 1402 +/- 758 nM for rec-hirudin, hirunorm, Napap and hirulog-1, respectively). Also recalcified human plasma was mitogenic for HUVSMC and its effect was mainly due to endogenously generated thrombin, as shown by the use of thrombin inhibitors. In conclusion, HUVSMC are capable of initiating the extrinsic coagulation cascade, leading to the formation of thrombin which promotes clotting and stimulates DNA synthesis. Thrombin inhibitors prevent both coagulative and cellular effects of thrombin.


Subject(s)
Blood Coagulation Factors/physiology , Muscle, Smooth, Vascular/drug effects , Serine Proteinase Inhibitors/pharmacology , Thrombin/physiology , Amino Acid Sequence , Cells, Cultured , DNA/biosynthesis , Evaluation Studies as Topic , Fibrinopeptide A/metabolism , Humans , Logistic Models , Models, Cardiovascular , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Peptides/pharmacology , Thrombin/antagonists & inhibitors , Thrombin/biosynthesis , Umbilical Veins/drug effects , Umbilical Veins/metabolism
13.
J Cardiovasc Pharmacol ; 27(2): 195-200, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8720417

ABSTRACT

Cellular calcium modulates enzyme activity, cell proliferation, and differentiation. In vascular smooth muscle cells (VSMC), calcium may contribute to increased vascular contractility and structural alterations in both hypertension and atherosclerosis. We investigated the role of calcium in angiotensin II (AII)-induced prostaglandin release and DNA synthesis in VSMC. Prostaglandin levels were determined by radioimmunoassay, and DNA synthesis was determined by the incorporation of [3H]thymidine. AII dose-dependently stimulated the release of prostaglandin E2 and prostaglandin I2, and this effect was synergistically enhanced by the Ca2+ ionophore A23187. Conversely, the AII response was inhibited by EGTA, a chelator of Ca2+ ions and by verapamil and nifedipine, two Ca2+ channel blockers or by incubation of the cells without exogenous Ca2+. TMB-8, an inhibitor of calcium mobilization, also strongly reduced angiotensin response. Similar results were obtained for angiotensin III (AIII) and vasopressin, two other agonists of prostaglandin production. AII- or serum-stimulated DNA synthesis was almost abolished by EGTA, whereas TMB-8, verapamil, and nifedipine had little or no effect. The production of prostaglandins triggered by angiotensins and vasopressin in VSMC is dependent on both intracellular and extracellular calcium, with calcium entering through L-type Ca2+ channels. Extracellular calcium is important for AII and serum mitogenic activity, but L-type Ca2+ channels do not appear to be implicated.


Subject(s)
Angiotensin II/pharmacology , Calcimycin/pharmacology , Calcium/physiology , DNA/biosynthesis , Dinoprostone/metabolism , Epoprostenol/metabolism , Ionophores/pharmacology , Muscle, Smooth, Vascular/drug effects , Animals , Calcium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Male , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley
14.
Eur J Pharmacol ; 280(3): 285-92, 1995 Jul 14.
Article in English | MEDLINE | ID: mdl-8566096

ABSTRACT

The present study investigates the effect of angiotensin II and LR-B/081 (-methyl 2-[[4-butyl-2-methyl-6-oxo-5-[[2'-(1H-tetra-zol-5-yl) [1,1'-biphenyl]-4-yl] methyl]-1(6H)-pyrimidinyl] methyl]-3-thiophenecarboxylate), a novel non-peptide angiotensin II receptor antagonist, on both early and late responses in rat vascular smooth muscle cells. Angiotensin II induced a rapid and transient elevation of inositol trisphosphate intracellular levels, triggered the release of both prostaglandin E2 and prostaglandin I2 (EC50 = 21 +/- 3 and 16 +/- 2 nM, respectively), and, in long-term studies, increased leucine and thymidine incorporation. All angiotensin II effects were antagonized by LR-B/081 and losartan, the reference non-peptide angiotensin AT1-selective receptor antagonist, whereas they were unaffected by PD123177 (1-(4-amino-3-methylphenyl)methyl-5-diphenylacetyl-4,5,6,7-tetr ahy dro-1H- imidazo[4,5-c]pyridine carboxylic acid), a non-peptide angiotensin AT2-selective receptor antagonist. LR-B/081 displayed a much higher potency than losartan in inhibiting angiotensin II-induced prostaglandin E2 (IC50 = 0.15 +/- 0.02 and 39 +/- 9 nM, respectively) and prostaglandin I2 release (IC50 = 0.18 +/- 0.04 and 134 +/- 40 nM, respectively) and was also more potent in blocking the increase in protein synthesis (IC50 = 242 +/- 119 nM and 1221 +/- 687 nM, respectively). Moreover, LR-B/081 and losartan blocked the response to angiotensin III but failed to inhibit the prostaglandin release stimulated by vasopressin or the mitogenic effect of serum. LR-B/081 and losartan were devoid of intrinsic properties in the experimental conditions employed.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Inositol 1,4,5-Trisphosphate/metabolism , Muscle, Smooth, Vascular/drug effects , Pyrimidinones/pharmacology , Thiophenes/pharmacology , Angiotensin II/antagonists & inhibitors , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Biphenyl Compounds/pharmacology , Cells, Cultured , DNA/biosynthesis , Dinoprostone/metabolism , Epoprostenol/metabolism , Imidazoles/pharmacology , Leucine/metabolism , Losartan , Male , Muscle, Smooth, Vascular/cytology , Protein Biosynthesis , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Tetrazoles/pharmacology , Thymidine/metabolism
15.
Regul Pept ; 56(2-3): 175-84, 1995 Apr 14.
Article in English | MEDLINE | ID: mdl-7652193

ABSTRACT

The presence of specific AII receptors in 6 different human neuroblastoma cell lines was investigated using binding, cAMP and [Ca2+]i studies. We found high affinity (0.1 nM), low capacity ((1-2).10(3) sites/cell) binding sites for [125I](Sar-1,Ile-8)AII in one half of the cell lines studied. In the positive cell lines mathematical modeling of multiple competition curves among AII and analogs strongly indicated the presence of a homogenous class of sites, i.e., AT1 receptors. The presence of AT1 receptors was further substantiated by AII-induced inhibition of VIP-stimulated cAMP levels and by AII-evoked [Ca2+]i transient. The density of AT1 receptors in neuroblastoma cells was not affected by treatment with pertussis toxin and retinoic acid but was significantly increased by subacute treatment with VIP. In neuroblastoma cells, AII does not stimulate DNA synthesis, suggesting other roles rather than mitogenesis. Neuroblastoma cells represents an interesting model to investigate the function of AII in neural crest derived tissues.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin II/metabolism , Calcium/metabolism , Cyclic AMP/metabolism , Neuroblastoma/metabolism , Receptors, Angiotensin/metabolism , 1-Sarcosine-8-Isoleucine Angiotensin II/metabolism , Angiotensin II/analogs & derivatives , Angiotensin III/metabolism , Angiotensin Receptor Antagonists , Binding, Competitive , Biphenyl Compounds/metabolism , Dose-Response Relationship, Drug , Humans , Imidazoles/metabolism , Kinetics , Losartan , Oligopeptides/metabolism , Pyridines/metabolism , Tetrazoles/metabolism , Tumor Cells, Cultured , Vasoactive Intestinal Peptide/pharmacology
16.
Naunyn Schmiedebergs Arch Pharmacol ; 350(4): 410-5, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7845478

ABSTRACT

The effects of an i.v. administration of endothelin-1, -2 and -3 (0.25-3 nmol kg-1) or their corresponding proendothelins (1-20 nmol kg-1) on blood pressure and 6 keto-prostaglandin F1 alpha (6 keto-PGF1 alpha) release in the anaesthetized ganglion-blocked rat were evaluated. The same peptides were tested for their ability to release 6 keto-PGF1 alpha from the rat vas deferens in vitro. Endothelins and proendothelins showed a transient hypotensive effect followed by a potent, long lasting vasopressor response. Blood pressure increase induced by endothelins was found to be dose-dependently correlated with 6 keto-PGF1 alpha plasma level increases. On the other hand proendothelins produced similar pressor responses, but their effect on 6 keto-PGF1 alpha plasma levels was much less intense at equipressor doses. The effects of endothelins on arterial pressure and 6 keto-PGF1 alpha release were phosphoramidon-insensitive, while the activities of proendothelins were reduced by phosphoramidon (10 mg kg-1 i.v.). Both endothelins (5-15 nmol/l) and proendothelins (100-300 nmol/l) were able to increase to a similar extent 6 keto-PGF1 alpha levels in the rat vas deferens incubation buffer. The releasing activity of endothelins was not modified by the pretreatment with phosphoramidon (50 mumol/l). This pretreatment strongly inhibited proendothelin-1 and -2 effects, but not that of proendothelin-3.


Subject(s)
Endothelins/physiology , Epoprostenol/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Aspartic Acid Endopeptidases/pharmacology , Dose-Response Relationship, Drug , Endothelin-Converting Enzymes , Endothelins/administration & dosage , Male , Metalloendopeptidases , Protein Precursors/physiology , Rats , Rats, Sprague-Dawley , Vas Deferens/drug effects , Vas Deferens/metabolism
17.
Eur J Pharmacol ; 256(1): 93-7, 1994 Apr 11.
Article in English | MEDLINE | ID: mdl-8026565

ABSTRACT

Angiotensin II and angiotensin III stimulated prostacyclin release in a time- and dose-dependent manner in both the prostatic and the non-prostatic part of the rabbit vas deferens. Also, angiotensin I enhanced the production of prostacyclin and its effect was blocked by captopril. Losartan, a type 1 (angiotensin AT1)-selective receptor antagonist, prevented the angiotensin II-induced prostacyclin release. The agonist peptide, p-aminophenylalanine angiotensin II, and the type 2 (angiotensin AT2)-selective receptor antagonist, PD123319, were found active only in the prostatic portion, suggesting heterogeneity of the receptor population. In conclusion, an angiotensin AT1 receptor mostly mediates the angiotensin-induced release of prostacyclin in the rabbit vas deferens.


Subject(s)
Angiotensin III/pharmacology , Angiotensin II/pharmacology , Epoprostenol/metabolism , Muscle, Smooth/metabolism , Receptors, Angiotensin/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Angiotensin I/metabolism , Angiotensin I/pharmacology , Angiotensin II/analogs & derivatives , Angiotensin II/metabolism , Angiotensin III/metabolism , Angiotensin Receptor Antagonists , Animals , Biphenyl Compounds/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Losartan , Male , Muscle, Smooth/drug effects , Pyridines/pharmacology , Rabbits , Receptors, Angiotensin/drug effects , Tetrazoles/pharmacology , Vas Deferens/drug effects , Vas Deferens/metabolism
18.
Peptides ; 14(5): 1037-41, 1993.
Article in English | MEDLINE | ID: mdl-8284253

ABSTRACT

Angiotensin II (AII) receptor subtypes were investigated in bovine cerebellar cortical membranes (BCCM) using agonists and antagonists. Differences were observed whether the tissue for the characterization was used fresh or frozen. In fresh BCCM, AII and angiotensin III (AIII) (not selective), p -aminophenylalanine6 -AII ([pNH2Phe6]AII) (AT2 selective), and DuP 753 (AT1 selective) interacted with two sites, at high and low affinity. In the presence of 10 mM dithiothreitol (DTT), the low-affinity site for AIII and [pNH2Phe6]AII and the high-affinity site for DuP 753 were no longer detectable. The same effect was obtained with 1 microM DuP 753, with only the low-affinity component of [pNH2Phe6]AII being resistant to this blocker. In frozen BCCM, DuP 753, PD 123319 (AT2 selective), and sarcosine1, isoleucine8-AII ([Sar1Ile8] AII) yielded monophasic curves. These data confirm the presence of multiple receptors (AT1 and AT2) for AII on BCCM and suggest the possibility that within the AT2 receptors, one subtype, DTT sensitive, may exist. Alternatively, a heterogeneity in the AT1 class, with respect to the sensitivity to DuP753, may be considered.


Subject(s)
Angiotensin Receptor Antagonists , Biphenyl Compounds/pharmacology , Cerebellar Cortex/drug effects , Dithiothreitol/pharmacology , Imidazoles/pharmacology , Tetrazoles/pharmacology , Animals , Cattle , Losartan , Radioligand Assay
19.
Growth Factors ; 6(3): 255-64, 1992.
Article in English | MEDLINE | ID: mdl-1389231

ABSTRACT

The role of growth hormone (GH) in the differentiation process of Ob1771 mouse preadipocyte cells has been studied under culture conditions that were serum-free and hormone-supplemented and which were previously shown to lead to terminal differentiation. In the absence of GH, a dramatic decrease in the adipogenic activity of the culture medium could be observed, as indicated 12 days after confluence by the low levels of glycerol-3-phosphate dehydrogenase activity and the sharp reduction of the number of triacylglycerol-containing cells. This decrease in adipogenic activity was accompanied by a parallel loss of the mitogenic potency of the culture medium. Determination of the half-maximal and maximal concentrations of GH required for the restoration of growth and differentiation were identical, 0.5 and 2 nM, respectively. Despite the presence of insulin-like growth factor-I (IGF-I) to substitute for supraphysiological concentrations of insulin and to saturate IGF-I receptor, GH was still required to induce terminal differentiation of a maximal number of cells. However, protein kinase C activators such as prostaglandin F2 alpha, phorbol esters and diacylglycerol were able to mimic GH in promoting a maximal mitogenic-adipogenic response, indicating that the ability of GH to induce diacylglycerol production (Doglio et al., 1989; Catalioto et al., 1990) plays a prominent role in this process. Furthermore, in agreement with the fact that the mitoses which precede terminal differentiation of Ob1771 preadipocytes are strictly controlled by cAMP and only modulated by protein kinase C, terminal differentiation of Ob1771 preadipocytes occurred in the absence of GH upon supplementation with high concentrations of carbaprostacyclin, added as a cAMP-elevating agent or with 8-Br-cAMP, added as a cAMP analogue. It is concluded that the control exerted by GH on terminal differentiation of mouse preadipocytes corresponds to a modulating mitogenic effect mediated through protein kinase C activation and leading to a potentiation of the cAMP and IGF-I mitogenic signalling pathways.


Subject(s)
Adipose Tissue/cytology , Growth Hormone/physiology , Protein Kinase C/physiology , Signal Transduction/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Corticosterone/pharmacology , Culture Media, Serum-Free , DNA/biosynthesis , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Glycerolphosphate Dehydrogenase/biosynthesis , Growth Hormone/antagonists & inhibitors , Mice , Mitosis/drug effects , Protein Kinase C/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...