Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 61(3): 291-8, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11172733

ABSTRACT

The tritiated bradykinin B1 receptor agonist [3H]des-Arg(10)-kallidin bound to a single class of high-affinity binding sites (K(d) = 0.5 +/- 0.16 nM; B(max) = 15,000 +/- 8,000 sites/cell) on cultured rat aortic smooth muscle cells. [3H]Des-Arg(10)-kallidin association and dissociation kinetics were monoexponential, making it possible to determine the association and dissociation rate constants (k(+1) = 1.5 10(5) M(-1) sec(-1); k(-1) = 4.2 10(-5) sec(-1)). [3H]Des-Arg(10)-kallidin binding was inhibited by specific ligands of bradykinin B1 and B2 receptors with a rank order of potency consistent with that known for bradykinin B1 receptors in other species (des-Arg(9)-[Leu(8)]bradykinin = des-Arg(10)-kallidin = des-Arg(9)-bradykinin = des-Arg(10)-[Leu(9)]kallidin > des-Arg(10)-HOE-140 >> bradykinin >> HOE-140). Bradykinin B1 receptor mRNA was also detected in these cells. Des-Arg(10)-kallidin increased cytosolic free Ca2+ levels, phosphoinositide turnover, and arachidonic acid release at nanomolar concentrations (respective EC(50) values: 16 +/- 2, 4 +/- 2.7, 6 +/- 2 nM). These functional effects of des-Arg(10)-kallidin could be blocked by the bradykinin B1 receptor antagonist des-Arg(9)-[Leu(8)]bradykinin, but were not sensitive to bradykinin B2 receptor antagonists. These results therefore show that rat aortic smooth muscle cells in culture express functional bradykinin B1 receptors.


Subject(s)
Kallidin/analogs & derivatives , Muscle, Smooth, Vascular/metabolism , Receptors, Bradykinin/analysis , Animals , Aorta/cytology , Aorta/metabolism , In Vitro Techniques , Kallidin/pharmacology , Male , RNA, Messenger/analysis , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B1 , Receptors, Bradykinin/genetics , Tritium
2.
Eur J Pharmacol ; 397(2-3): 303-10, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10844128

ABSTRACT

The effect of ¿2-[4-(4-chloro-2, 5-dimethoxy-phenyl)-5-[2-cyclohexyl-ethyl)-thiazol-2-ylcarbamoy l]-5, 7-dimethyl-indol-1-yl¿-acetic acid (SR146131), a novel non-peptide agonist of cholecystokinin (CCK) CCK(1) receptors, was compared to the effect of sulphated cholecystokinin octapeptide (CCK-8-S) on CCK(1) receptors of the human neuroblastoma cell line IMR-32. SR146131 inhibited [125I]CCK-8-S binding to IMR-32 cells at nanomolar concentrations. SR146131 and CCK-8-S increased intracellular free Ca(2+) levels ([Ca(2+)](i)) in the same concentration range (EC(50)=6+/-2.3 and 1.3+/-0.14 nM, respectively). Although the shape of the [Ca(2+)](i) increase induced by CCK-8-S and SR146131 was slightly different, extracellular Ca(2+) removal affected the response of both compounds to a similar degree, and the response of both compounds was essentially due to Ca(2+) release from intracellular stores. This was also confirmed by measuring the [Ca(2+)](i) response of single cells: both compounds induced [Ca(2+)](i) oscillations at subnanomolar concentrations and elicited a large peak increase in [Ca(2+)](i) at higher concentrations (EC(50)=0.5+/-0.04 and 5.7+/-1.9 nM for CCK-8-S and SR146131, respectively). Both CCK-8-S and SR146131 induced a sustained increase of phosphoinositide turnover in these cells, and acted at similar concentrations (EC(50)=2.7+/-0.7 and 6+/-3.1 nM, respectively), although the maximal effect of SR146131 was somewhat lower than the effect of CCK-8-S. These data show that SR146131 activates human CCK(1) receptors on IMR-32 cells in a manner and with a potency similar to that of CCK-8-S.


Subject(s)
Indoles/pharmacology , Neuroblastoma/metabolism , Receptors, Cholecystokinin/agonists , Thiazoles/pharmacology , Benzodiazepinones/pharmacology , Binding, Competitive/drug effects , Calcium/metabolism , Devazepide/pharmacology , Dose-Response Relationship, Drug , Humans , Iodine Radioisotopes , Neuroblastoma/pathology , Phenylurea Compounds/pharmacology , Phosphatidylinositols/metabolism , Radioligand Assay , Receptors, Cholecystokinin/antagonists & inhibitors , Sincalide/analogs & derivatives , Sincalide/metabolism , Sincalide/pharmacology , Tumor Cells, Cultured
3.
Anesthesiology ; 92(2): 542-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10691243

ABSTRACT

BACKGROUND: Propofol is considered to be an anesthetic agent with few or no negative inotropic effects. This study evaluated a possible direct depressant effect of propofol on sarcoplasmic reticulum Ca2+ accumulation and cardiomyocytes. METHODS: The effects of propofol on intracellular Ca2+ transients were evaluated in isolated rat cardiomyocytes using a microfluorometric technique with Indo-1. Sarcoplasmic reticulum function was also assessed by measuring the oxalate-stimulated Ca2+ uptake from homogenates of rat ventricles. RESULTS: The Ca2+ uptake capacity of the sarcoplasmic reticulum was decreased by propofol (10(-4) M). Large concentrations of propofol decreased the rate of decrease of the intracellular Ca2+ transient, which resulted in an increase of diastolic Ca2+ when the diastolic interval was decreased. The increased diastolic Ca2+ also resulted in a decrease in Ca2+ transient. This effect appeared for lower doses (10(-5) M) after a short diastolic pause rather than after a long (2- to 3-min) rest (appearing at 10(-4) M). CONCLUSIONS: For doses more than 10(-5) M, propofol induces a Ca2+ uptake capacity impairment of the sarcoplasmic reticulum. This is probably responsible for a slowing of the decrease of the Ca2+ transient, which in turn increases the diastolic Ca2+ for high heart rate. These diastolic modifications may participate in the slight negative inotropic effect of the drug.


Subject(s)
Anesthetics, Intravenous/pharmacology , Calcium Signaling/drug effects , Myocardium/metabolism , Propofol/pharmacology , Sarcoplasmic Reticulum/metabolism , Animals , Calcium/metabolism , Cell Separation , Chelating Agents , Electric Stimulation , Fat Emulsions, Intravenous/pharmacology , Fluorometry , Heart/drug effects , In Vitro Techniques , Indoles , Male , Myocardial Contraction/drug effects , Myocardium/cytology , Myocardium/ultrastructure , Rats , Rats, Wistar , Sarcoplasmic Reticulum/drug effects
4.
J Mol Cell Cardiol ; 31(9): 1697-708, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10471353

ABSTRACT

Induction of nitric oxide synthase (NOS2, also designated as iNOS) in the heart is known to occur in response to various stimuli. It is not known, however, whether in vivo hypoxia leads to cardiac NOS2 induction. We thus investigated the effects of normobaric hypoxia (10% O(2)for 8, 15 and 21 days) on NOS2 protein expression and enzyme activity in rat right ventricle (RV) and left ventricle (LV). Chronic hypoxia induced RV hypertrophy: the RV weight to body weight ratio was increased by 45% upon 15 days of exposure, with no change thereafter and no change in left ventricular (LV) weight. Treatment of hypoxic rats with l -NAME for 1 month decreased pulmonary artery pressure and RV hypertrophy compared to hypoxic non-treated rats. NOS2 activity detected by [(3)H]l -arginine to [(3)H]l -citrulline conversion increased in RV during hypoxia, with a maximum at 15 days (+161% of control rats P<0.05), whereas it increased less (by 60%) in LV. In parallel, after 15 days of hypoxia there was a three-fold increase in NOS2 protein abundance detected by Western blotting using an isoform-specific antibody in the RVs (two-fold increase in the LV). Immunochemistry with the specific antibody demonstrated the expression in cardiomyocytes isolated from both ventricles of normoxic and hypoxic rats. Protein kinase C (PKC) content and activity was unchanged in LV of hypoxic rats, but increased in RV as compared with normoxic rats. These results clearly show that, in the heart, NOS2 is upregulated by hypoxia with an expression in cardiomyocytes of both ventricles. In addition, NOS2 is more inducible in the right hypertrophied ventricle than in the left non-hypertrophied hypoxic ventricle.


Subject(s)
Hypertrophy, Right Ventricular/physiopathology , Hypoxia/physiopathology , Myocardium/enzymology , Nitric Oxide Synthase/biosynthesis , Amino Acid Sequence , Animals , Blotting, Western , Chronic Disease , Enzyme Induction , Heart Ventricles , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/etiology , Hypoxia/enzymology , Immunohistochemistry , Interleukin-6/analysis , Male , Molecular Sequence Data , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type II , Peptide Fragments/chemistry , Protein Kinase C/analysis , Rats , Rats, Wistar , Ventricular Function, Left , Ventricular Function, Right
5.
J Cardiovasc Pharmacol ; 33(3): 394-400, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10069674

ABSTRACT

The goal of this study was to evaluate, in rat cardiomyocytes, the effects on cytosolic calcium of a pure K-adenosine triphosphate (ATP) channel opener, aprikalim, and those of nicorandil, a dual-acting agent that increases cyclic guanosine monophosphate (cGMP) levels and opens K-ATP channels. These effects were compared with those of a pure NO donor, 3-morpholino-sydnonimine (Sin-1). Ventricular myocytes were isolated from the hearts of adult rats. Changes in cytosolic calcium concentration ([Ca2+]i) were measured by using a Ca2+ indicator, indo-1/AM. Alterations in indo-1 fluorescence were recorded during regular electrical stimulation. After 10 min of pacing, end-diastolic [Ca2+]i was significantly increased as compared with control without significant changes in calcium transient. For doses of 10(-7) to 10(-4) M, aprikalim and nicorandil did not affect significantly the calcium transient. Sin-1 produced a significant decrease in calcium transient (by approximately 20%), which was already maximal at 10(-7) M. When given with the potassium channel antagonist glibenclamide (10(-5) M), nicorandil induced the same effects as those observed with Sin-1. We conclude that potassium channel openers aprikalim and nicorandil do not not decrease calcium transient. Thus the NO-donor properties of nicorandil are not apparent when given alone but appear when ATP-dependent potassium channels are blocked.


Subject(s)
Adenosine Triphosphate/pharmacology , Calcium/metabolism , Myocardium/metabolism , Nitric Oxide Donors/pharmacology , Potassium Channels/drug effects , Animals , Cardiac Pacing, Artificial , Cytosol , Glyburide/pharmacology , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hypoglycemic Agents/pharmacology , Male , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Myocardium/cytology , Nicorandil/pharmacology , Picolines/pharmacology , Pyrans/pharmacology , Rats , Rats, Wistar , Vasodilator Agents/pharmacology
6.
J Cardiovasc Pharmacol ; 31(4): 545-50, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9554803

ABSTRACT

The novel compound SR142948A was compared with SR48692 as an antagonist of neurotensin-induced cardiovascular effects both in vitro and in vivo. SR142948A inhibited [125I]-neurotensin binding [median inhibitory concentration (IC50) = 0.24 +/- 0.01 nM], neurotensin-induced cytosolic free Ca2+ increase (IC50 = 19 +/- 6 nM), and prostacyclin production in human umbilical vein endothelial cells (IC50 = 17 +/- 3 nM) at much lower concentrations than did SR48692 (respective IC50 values, 14 +/- 5, 41 +/- 16, and 86 +/- 16 nM). Oral administration of SR142948A (10 microg/kg) resulted in significant inhibition of neurotensin-induced blood pressure changes, whereas SR48692 was active only at 10-fold higher doses. Furthermore, SR142948A administered i.v. in microg/kg quantities in the rat was as active as mg/kg doses of SR48692 on neurotensin-induced increase in hematocrit. SR142948A injected intradermally also significantly inhibited neurotensin-induced plasma extravasation at concentrations as low as 10 pmol/site, whereas 1,000 pmol/site of SR48692 were necessary to reach a significant inhibition. These data show that SR142948A is a novel, extremely potent antagonist of neurotensin-induced cardiovascular responses both in vitro and in vivo. SR142948A and SR48692 constitute a pair of nonpeptide neurotensin antagonists of different potency, which may be used to probe for the implication of neurotensin receptors in physiologic or pathologic phenomena.


Subject(s)
Adamantane/analogs & derivatives , Blood Pressure/drug effects , Imidazoles/pharmacology , Neurotensin/antagonists & inhibitors , Receptors, Neurotensin/antagonists & inhibitors , Umbilical Veins/drug effects , Adamantane/administration & dosage , Adamantane/pharmacology , Administration, Oral , Animals , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Epoprostenol/metabolism , Hematocrit , Humans , Imidazoles/administration & dosage , Injections, Intradermal , Iodine Radioisotopes , Male , Neurotensin/metabolism , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Quinolines/administration & dosage , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Umbilical Veins/metabolism
7.
J Mol Cell Cardiol ; 29(6): 1687-94, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9220354

ABSTRACT

Protein Kinase C (PKC) is implicated in the induction of myocardial hypertrophy. Recent studies showed an increased activity and expression of PKC in rat left ventricular hypertrophy, but we demonstrated a decreased PKC activity and content in rabbit heart failure. The present study was designed to evaluate whether these differences were due to species or model differences. PKC activity and expression were measured in a model of mild ventricular overload, induced by a 40-50% constriction of the abdominal aorta in rabbits. Left ventricular (LV) weight/body weight ratio was increased by 14, 21 and 36% after 4, 18 and 42 days of stenosis, respectively. PKC activity was significantly decreased after 18 and 42 days of stenosis in the particulate fraction of LV, but it was not modified in the cytosolic fraction leading to a significantly decreased translocation index (particulate/total activity ratio): 18.6 +/- 2.2% and 19.4 +/- 1.6% at 18 days and 42 days of aortic stenosis, respectively, compared with 25.7 +/- 2.0% and 25.8 +/- 1.2% in corresponding sham-operated rabbits (both Ps < 0.05). Similarly, PKC content, measured by immunoblotting, was not modified in the cytosolic fractions, but decreased significantly in the particulate fractions after 18 and 42 days of stenosis. These data are, thus, different from those obtained in rat LV hypertrophy showing species differences in PKC expression in hypertrophy. They also show that hypertrophy may take place without induction of PKC.


Subject(s)
Hypertrophy, Left Ventricular/enzymology , Protein Kinase C/metabolism , Animals , Aortic Valve Stenosis/enzymology , Blotting, Western , Disease Models, Animal , Female , Hemodynamics , Isoenzymes , Protein Kinase C/analysis , Rabbits , Species Specificity , Time Factors
8.
Eur J Pharmacol ; 323(2-3): 215-21, 1997 Apr 04.
Article in English | MEDLINE | ID: mdl-9128841

ABSTRACT

Human umbilical vein endothelial cells express high affinity neurotensin receptors which are coupled to phosphoinositide turnover and 45Ca2+ efflux (Schaeffer et al., 1995. J. Biol. Chem. 270, 3409-3413). In order to assess the physiological significance of neurotensin receptor activation in endothelial cells, we have compared the in vitro effect of neurotensin on prostacyclin release and cytosolic free calcium increase ([Ca2+]i) as determined by fura-2 fluorescence experiments to the in vivo effect of neurotensin on blood pressure and haematocrit. Neurotensin increased [Ca2+]i levels at low concentrations (EC50 = 4.2 +/- 0.2 nM, n = 3). At similar concentrations, neurotensin was also able to induce prostacyclin release from human umbilical vein endothelial cells (EC50 = 14 +/- 1 nM, n = 3) as determined by a 6-keto-prostaglandin F1 alpha enzyme immunoassay. The neurotensin (100 nM)-induced [Ca2+]i increase and prostacyclin release were inhibited by the specific non-peptide neurotensin receptor antagonist SR 48692 at similar concentrations (IC50 = 41 +/- 16 nM and 86 +/- 17 nM, respectively, n = 3), confirming that these responses were mediated by high affinity neurotensin receptors. Intravenous injection of neurotensin (1-4 nmol/kg i.v.) in the rat resulted in a drop of blood pressure and increased haematocrit, and nearly doubled the plasma levels of 6-keto-prostaglandin F1 alpha, the stable metabolite of prostacyclin. Whereas indomethacin (10 mg/kg i.v.) pretreatment significantly reduced the effect of neurotensin on blood pressure, it did not alter its effect on haematocrit. These results suggest that prostacyclin release plays a role in the hypotensive effects of neurotensin, but is not involved in its effects on haematocrit.


Subject(s)
Endothelium, Vascular/metabolism , Epoprostenol/metabolism , Neurotensin/pharmacology , Umbilical Veins/metabolism , 6-Ketoprostaglandin F1 alpha/blood , Animals , Blood Pressure/drug effects , Calcium/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Epoprostenol/blood , Hematocrit , Humans , Male , Rats , Rats, Sprague-Dawley
9.
Prog Urol ; 7(1): 78-80, 1997 Feb.
Article in French | MEDLINE | ID: mdl-9116743

ABSTRACT

The authors report the case of a 40-year-old woman who presented with very large right renal varices. Computed tomography and arteriography showed that these varices were situated on the convex surface of the kidney, in front of its anterior surface, and drained venous blood derived from the renal parenchyma. These varices were probably secondary to undiagnosed renal vein thrombosis.


Subject(s)
Kidney/blood supply , Varicose Veins/diagnostic imaging , Adult , Angiography , Female , Humans , Kidney/diagnostic imaging , Renal Veins , Thrombosis/complications , Thrombosis/diagnostic imaging , Tomography, X-Ray Computed , Varicose Veins/etiology
10.
J Membr Biol ; 153(3): 233-9, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8849418

ABSTRACT

In the cortical collecting duct (CCD), arginin vasopressin (AVP) has been shown to increase the number and activity of basolateral Na+-K+-ATPase by recruiting or activating a latent pool of pumps. However, the precise mechanism of this phenomenon is still unknown. The aim of this study was to investigate whether this AVP-induced increase in basolateral Na+-K+-ATPase could depend on a dephosphorylation process. To this purpose, the effect of protein serine/threonine phosphatase (PP) inhibitors was examined on both the specific 3H-ouabain binding (to evaluate the number of pumps in the basolateral membrane) and the ouabain-dependent 86Rb uptake (to evaluate pump functionality) in the presence or absence of AVP. In addition, the activity of two PP, PP1 and PP2A, was measured and the influence of AVP was examined on both enzymes. Experiments have been performed on mouse CCD isolated by microdissection. Results show that inhibition of PP2A prevents the AVP-induced increase in the number and activity of Na+-K+-ATPases, independent of an effect on the apical cell sodium entry. In addition, AVP rapidly increased the activity of PP2A without effect on PP1. These data suggest that PP2A is implied in the regulation of Na+-K+-ATPase activity by AVP in the CCD and that the AVP-dependent increase in the number of Na+-K+-ATPases is mediated by a PP2A-dependent dephosphorylation process.


Subject(s)
Kidney Cortex/metabolism , Phosphoric Monoester Hydrolases/metabolism , Renal Agents/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Vasopressins/pharmacology , Animals , Mice , Phosphorylation
11.
Kidney Int ; 50(2): 367-76, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8840262

ABSTRACT

This study describes the establishment of a rat kidney cortical collecting duct (CCD) clonal cell line (RCCD1 cells) that maintains high transepithelial resistance and specific hormonal sensitivities. Immortalized cells were obtained by infection of primary cultured CCD cells with the wild-type simian virus 40. Grown on Petri dishes, RCCD1 cells are organized as monolayers of cuboid cells separated by tight junctions and form domes. Grown on permeable filters, confluent RCCD1 cells exhibit high transepithelial resistance (Rt: 2390 +/- 140 omega. cm2), transepithelial potential difference (PD) of -10.5 +/- 1.2 mV lumen negative, an associated short-circuit current (Isc) of 4.3 +/- 0.5 microA/cm2, and generated significant Na+, K+, H+ and HCO3- gradients, reflecting Na+ and H+ reabsorption and K+ and HCO3- secretion. RCCD1 cells exhibit features of both principal (PC) and intercalated (IC) cells. Consistent with PC phenotype, about 50% of the cells were positively stained by a PC-specific agglutinin. In situ hybridization studies revealed the presence of alpha, beta and gamma subunit mRNAs of the amiloride-sensitive epithelial Na+ channel and alpha 1 and beta 1 subunits of Na(+)-K(+)-ATPase. Moreover, Na(+)-K(+)-ATPase was immunolocalized at the basolateral side of the cells. Arginine vasopressin (AVP) induced a significant increase in both cellular cAMP content and Isc. Amiloride decreased in a dose-dependent manner Isc from untreated and AVP-treated RCCD1 cells. In addition, a barium-sensitive K+ conductance was evidenced in the apical side of the cells. Consistent with IC phenotype, isoproterenol (ISO) provoked a large increase in cellular cAMP and stimulated Isc. The effect of ISO on Isc was blocked by 5 x 10(-3) M DPC, a chloride channel blocker. Finally, AVP plus ISO had additive effect on Isc. Taken together, these results provide evidence that the RCCD1 cell line has maintained many of the original properties of rat CCD from which they were derived.


Subject(s)
Cell Line , Kidney Cortex/cytology , Kidney Cortex/metabolism , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Animals , Arginine Vasopressin/pharmacology , Cell Polarity , Cyclic AMP/metabolism , Electric Impedance , Epithelial Cells , Epithelium/metabolism , In Situ Hybridization , Ion Transport , Isoproterenol/pharmacology , Kidney Cortex/drug effects , Kidney Tubules, Collecting/drug effects , Membrane Potentials , Microscopy, Electron , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sodium-Potassium-Exchanging ATPase/genetics
12.
J Urol (Paris) ; 102(3): 127-9, 1996.
Article in French | MEDLINE | ID: mdl-9091558

ABSTRACT

Authors report on a case of emphysematous pyelonephritis in a woman affected with diabetes and renal failure. In order to avoid chronic dialysis, no nephrectomy was performed and the patient was treated only with drugs. Full recovery was obtained, without worsening of the renal function.


Subject(s)
Emphysema/microbiology , Escherichia coli Infections/microbiology , Pyelonephritis/microbiology , Aged , Anti-Bacterial Agents , Drug Therapy, Combination/administration & dosage , Drug Therapy, Combination/therapeutic use , Emphysema/diagnostic imaging , Emphysema/drug therapy , Escherichia coli Infections/diagnostic imaging , Escherichia coli Infections/drug therapy , Female , Humans , Pyelonephritis/diagnostic imaging , Pyelonephritis/drug therapy , Tomography, X-Ray Computed , Urography
13.
Cardiovasc Res ; 30(6): 848-56, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8746198

ABSTRACT

OBJECTIVE: The goal was to examine left ventricular (LV) regional contraction alterations and especially, regional inotropic reserve changes in tachycardia-induced heart failure (HF). METHODS: Eleven dogs were chronically instrumented to measure LV pressure and its first time derivative (LV dP/dt), left atrial and aortic pressures and to measure antero-apical (AS), -basal (BS) and postero-apical (PS) subendocardial segmental contractions by ultrasonic crystals. Dobutamine (5-15 micrograms/kg per min) and left atrial pacing (150-240 beats/min) were performed in the control state (C) and in HF induced by chronic right ventricular pacing (240 beats/min, 3 weeks). RESULTS: In HF, as compared with in C, LV dP/dt max decreased and LV end-diastolic pressure and end-diastolic segmental lengths increased (Ps < 0.005). The percentage of systolic shortening was more depressed in PS (from 21 +/- 1% to 7 +/- 1%, P < 0.001) than in AS and BS (from 24 +/- 1% to 17 +/- 1% and from 20 +/- 2% to 13 +/- 1% respectively, Ps < 0.05). During dobutamine infusion, in HF as compared with C, the increases in LV dP/dt max were smaller (dobutamine 15 micrograms/kg per min: HF: + 36 +/- 6% vs C: + 68 +/- 11%, P < 0.01) and the increases in the systolic shortening of the three segments were also smaller. However, the responses of the three segments were similar in HF and in C. During left atrial pacing, LV dP/dt max increased less in HF than in C and the poststimulation potentiation of LV dP/dt max was impaired in HF. However, the responses of the systolic shortening during regular left atrial pacing and the increase in the percentage of systolic shortening of the first poststimulation beat were similar in all regions. CONCLUSION: In tachycardia-induced HF, although LV regional contraction is heterogeneously altered, the inotropic reserve appears to be similarly modified in all regions.


Subject(s)
Heart Failure/physiopathology , Myocardial Contraction/physiology , Ventricular Dysfunction, Left/physiopathology , Analysis of Variance , Animals , Cardiac Pacing, Artificial , Dobutamine/pharmacology , Dogs , Myocardial Contraction/drug effects , Stimulation, Chemical
14.
J Lipid Mediat Cell Signal ; 12(1): 49-57, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7551689

ABSTRACT

In order to evaluate the relative importance platelet-activating factor (PAF) in the proliferative process leading to restenosis, the effect of SR 27417, a novel highly potent PAF receptor antagonist, on PAF-induced rabbit aortic smooth muscle cell (SMC) proliferation and intimal hyperplasia in rabbit carotid arteries subjected to air-drying endothelial injury was investigated. When added to low concentrations of foetal calf serum, PAF showed a dose-dependent mitogenic effect with regard to rabbit arterial SMC. SR 27417 inhibited PAF-induced SMC growth (IC50 = 2.4 +/- 0.4 nM) but remained without effect on the mitogenic effect of foetal calf serum. A 16 day treatment of SR 27417 (10 mg/kg per day, p.o.) abrogated PAF-induced platelet aggregation ex vivo but did not affect the development of intimal thickening, therefore showing that PAF is not an essential component of the cascade leading to restenosis following vascular injury.


Subject(s)
Cell Division , Muscle, Smooth, Vascular/pathology , Platelet Activating Factor/physiology , Vascular Diseases/pathology , Animals , Aorta , Cell Division/drug effects , Endothelium, Vascular/pathology , Hyperplasia , Male , Platelet Activating Factor/antagonists & inhibitors , Platelet Activating Factor/pharmacology , Platelet Aggregation/drug effects , Rabbits , Thiazoles/pharmacology , Vascular Diseases/etiology
15.
Basic Res Cardiol ; 90(4): 305-13, 1995.
Article in English | MEDLINE | ID: mdl-8534255

ABSTRACT

OBJECTIVES: Little comparative information is available on mitochondrial function changes during experimentally-induced hypertrophy. Respiratory control mechanisms are not exactly the same in situ and in isolated mitochondria. This study assessed in situ mitochondrial function in two myocardial hypertrophy models. METHODS: Cytochrome aa3 (Cytaa3) and myoglobin (Mb) absorption changes were monitored in isolated rat hearts using dual wavelength spectrophotometry (Cytaa3: 605-630 nm, Mb: 581-592 nm). Hypertrophy was induced by creation of an aortic stenosis or of an aorto-caval fistula. Optical monitoring was performed on diastole-arrested perfused hearts using the sequence O2 perfusion, N2 perfusion during 4 min, and reoxygenation. The plateaus of the Cytaa3 and Mb curves were used to quantify oxidation-reduction and oxygenation levels. Respiratory kinetics were characterized by the slopes of transition phase curves. RESULTS: Myoglobin oxygenation was comparable in the hypertrophied and control hearts. However, Cytaa3 oxidation-reduction levels in the hypertrophied hearts showed a shift towards greater reduction in comparison with the controls (controls: 0.580 +/- 0.008 DO605/DO630 nm, n = 34; fistula: 0.530 +/- 0.023, n = 23; stenosis: 0.522 +/- 0.016, n = 20, p < 0.001). The rate of Cytaa3 reduction and the rate of myoglobin deoxygenation were significantly accelerated (p < 0.005) in the volume overload group (0.507 +/- 0.043, n = 23), whereas the respiratory rate in the pressure overload group (0.389 +/- 0.034, n = 20) was comparable to that in the control hearts (0.358 +/- 0.026 delta DO 605 nm/DO630 nm.min-1, n = 34). CONCLUSION: We found mitochondrial function alterations in both volume overload- and pressure overload-induced cardiac hypertrophy, despite adequate cytosol oxygenation. The patterns of these alterations differed: the redox state showed a shift of similar magnitude toward greater reduction in both models, but the respiratory rate was increased in the volume-overloaded hearts and unchanged in the pressure-overloaded hearts. The modification in the oxidation-reduction state suggested that overload hypertrophy may induce changes in the metabolism of the myocardium, which may, in turn, load to persistent modifications in mitochondrial function. The differences between the two models suggest that adaptation to hypertrophy-inducing events exists at the level of the mitochondrion.


Subject(s)
Cardiomegaly/metabolism , Mitochondria, Heart/metabolism , Absorption , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Cytoplasm/metabolism , Electron Transport Complex IV/metabolism , Female , Myoglobin/metabolism , Oxidation-Reduction , Oxygen Consumption , Rats , Rats, Wistar
16.
J Biol Chem ; 270(7): 3409-13, 1995 Feb 17.
Article in English | MEDLINE | ID: mdl-7852427

ABSTRACT

The binding of 125I-neurotensin (NT) to human umbilical vein endothelial cell monolayers was studied. At 20 degrees C, 125I-NT bound to a single class of binding sites with a dissociation constant of 0.23 +/- 0.08 nM and a binding site density of 5500 +/- 1300 sites/cell (n = 3). 125I-NT also bound to human aortic endothelial cells with a dissociation constant of 0.6 +/- 0.26 nM and a binding site density of 32000 +/- 1700 sites/cell. Association and dissociation kinetics were of a pseudo-first order and gave association and dissociation rate constant values of 1.6 x 10(6) M-1 s-1 and 3.5 x 10(-4) s-1, respectively. 125I-NT binding was inhibited by NT analogues with a rank order of potency similar to that characterizing brain high affinity NT binding sites (K0.5, nM): NT8-13 (0.11) > NT (0.35) > acetyl-NT8-13 (1.5) > [Phe11]NT (12) > [D-Tyr11]NT (> 1000). 125I-NT binding was also inhibited by the non-peptide NT antagonist SR 48692 (Ki = 16 nM) but was not affected by levocabastine, an inhibitor of low affinity brain NT binding sites. NT had no effect on cGMP levels in endothelial cells but NT and its analogues increased 45Ca2+ efflux from endothelial cells at nanomolar concentrations with a rank order of potency which was identical to that observed in binding experiments. This effect was inhibited by SR 48692 (IC50 = 8 nM). NT was able to increase phosphoinositide turnover in these cells, and this effect was blocked by SR 48692. The correlation between dissociation constants of NT analogues in binding experiments and IC50 values in 45Ca2+ efflux experiments was very high (r = 0.997) with a slope near unity, indicating that 125I-NT binding sites are functional NT receptors coupled to phosphoinositide hydrolysis and Ca2+ release in human umbilical vein endothelial cells.


Subject(s)
Calcium/metabolism , Endothelium, Vascular/metabolism , Gene Expression , Neurotensin/metabolism , Neurotensin/pharmacology , Receptors, Neurotensin/metabolism , Binding Sites , Binding, Competitive , Cells, Cultured , Cyclic GMP/metabolism , Endothelium, Vascular/drug effects , Humans , Kinetics , Neurotensin/analogs & derivatives , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptors, Neurotensin/biosynthesis , Umbilical Veins
17.
Biochem J ; 303 ( Pt 1): 227-31, 1994 Oct 01.
Article in English | MEDLINE | ID: mdl-7945245

ABSTRACT

Binding of 125I-thrombin to human umbilical vein endothelial cells (HUVECs) was specifically displaced by the synthetic tetradecapeptide SFLLRNPNDKYEPF, named thrombin receptor agonist peptide (TRAP), which has recently been described as a peptide mimicking the new N-terminus created by cleavage of the thrombin receptor, and F-14, a tetradecapeptide representing residues 365-378 of the human alpha-thrombin B chain. Binding of 125I-TRAP to HUVECs was time-dependent, reversible and saturable, showing high affinity (KD = 1.5 +/- 0.4 microM) and high binding capacity (Bmax. = 7.1 +/- 0.6 x 10(6) sites/cell) (n = 3). Unlabelled thrombin and TRAP competitively and selectively inhibited the specific binding of 125I-TRAP with IC50 values of 5.8 +/- 0.7 nM and 2.8 +/- 0.4 microM respectively, whereas F-14 remained ineffective at displacing 125I-TRAP from its binding sites, suggesting the presence of at least two different types of thrombin-binding sites on HUVECs. TRAP was a potent mitogen for HUVECs in culture. Both TRAP and alpha-thrombin stimulated the proliferation of HUVECs with half-maximum mitogenic responses between 1 and 10 nM. F-14 also promoted HUVEC growth. The mitogenic effects of F-14 and TRAP were additive. N alpha-(2-Naphthylsulphonylglycyl)-DL-p-amidinophenylalanylpiper idine (NAPAP) and hirudin (two specific inhibitors of the enzyme activity of thrombin) specifically inhibited thrombin-induced HUVEC growth (IC50 values 400 +/- 60 and 52 +/- 8 nM respectively) but remained without effect on the mitogenic effect of TRAP or F-14. This demonstrated that the mitogenic effect of alpha-thrombin for HUVECs was intimately linked to its esterolytic activity but also showed that thrombin can stimulate HUVEC growth via another non-enzymic pathway. This hypothesis was further reinforced by the fact that F-14-induced proliferation of HUVECs remained unaltered by two antibodies directed against TRAP or the cleavage site on the extracellular portion of the thrombin receptor, which both strongly reduced thrombin-induced proliferation of HUVECs. Thrombin-, TRAP- or F-14-induced HUVEC proliferation was strongly inhibited by a neutralizing monoclonal antibody directed against basic fibroblast growth factor (bFGF), suggesting that thrombin regulates the autocrine release of bFGF in HUVECs.


Subject(s)
Cell Division/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Receptors, Thrombin/metabolism , Thrombin/pharmacology , Amino Acid Sequence , Cells, Cultured , Endothelium, Vascular/drug effects , Heparin/pharmacology , Humans , Kinetics , Molecular Sequence Data , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Receptors, Thrombin/antagonists & inhibitors , Structure-Activity Relationship , Thrombin/metabolism , Umbilical Veins
18.
Thromb Res ; 76(2): 157-69, 1994 Oct 15.
Article in English | MEDLINE | ID: mdl-7863466

ABSTRACT

[3H]-2-Methylthio-ADP ([3H]-2-MeS-ADP), a stable analogue of ADP bound to one type of specific binding sites on rat platelets (KD = 0.77 +/- 0.07 nM, Bmax = 160 +/- 11 fmol/10(8) cells). 2-MeS-ADP and ADP antagonized [3H]-2-MeS-ADP binding, showing respective Ki values of 1.4 +/- 0.1 nM and 486 +/- 78 nM. Clopidogrel, a potent and specific inhibitor of ADP-induced platelet aggregation partially inhibited (approximately 70% inhibition) the binding of [3H]-2-MeS-ADP at the same time it abrogated 2-MeS-ADP- and ADP-induced adenylyl cyclase inhibition and aggregation. A population of clopidogrel-resistant [3H]-2-MeS-ADP binding sites was detected on platelets from treated animals. These receptor sites (KD = 0.9 +/- 0.2 nM, Bmax = 47 +/- 5 fmol/10(8) platelets) which showed high affinity for both ADP and 2-MeS-ADP (Ki values in the nanomolar range) might be involved in the ADP-induced shape change, a clopidogrel-resistant ADP-induced event. Using clopidogrel which acts via a direct and irreversible inhibition of ADP binding to its adenylyl cyclase-coupled receptor sites on platelets, we were able to discriminate between two types of ADP receptor sites. The former which was clopidogrel-sensitive represented about 70% of the total [3H]-2-MeS-ADP receptors and was responsible for ADP-induced platelet aggregation and adenylyl cyclase inhibition. The latter which was not affected by clopidogrel might be involved in ADP-induced shape-change.


Subject(s)
Adenosine Diphosphate/metabolism , Blood Platelets/metabolism , Ticlopidine/analogs & derivatives , Adenylyl Cyclases/metabolism , Animals , Binding Sites , Blood Platelets/ultrastructure , Clopidogrel , Female , Platelet Aggregation , Rats , Rats, Sprague-Dawley , Signal Transduction , Ticlopidine/metabolism
19.
Biochem Pharmacol ; 48(6): 1211-7, 1994 Sep 15.
Article in English | MEDLINE | ID: mdl-7945414

ABSTRACT

Malformin-A1, a cyclic pentapeptide of microbial origin, antagonized in a competitive manner the binding of 125I-IL1 beta (interleukin-1 beta) to human monocytes and cultured human umbilical vein endothelial cells (HUVEC) with IC50 values (doses which reduce specific binding by 50%) of 250 +/- 80 and 230 +/- 25 nM, respectively (N = 3). IL1 increased in a dose-dependent manner the expression of tissue factor, a ubiquitous membrane-anchored glycoprotein that initiates blood coagulation at the surface of HUVEC and human monocytes. Malformin-A1 strongly inhibited IL1-induced tissue factor expression in HUVEC and monocytes with IC50 values of 420 +/- 35 and 105 +/- 25 nM, respectively (N = 3), and reduced IL1-induced expression of intercellular adhesion molecule-1 (ICAM-1, CD54) on HUVEC (IC50 = 125 +/- 18 nM) (N = 4). These observations demonstrate that malformin-A1 recognizes and blocks IL1 beta binding to its receptor sites on monocytes and endothelial cells and protects these cells from IL1-induced procoagulant changes.


Subject(s)
Endothelium, Vascular/drug effects , Interleukin-1/antagonists & inhibitors , Monocytes/drug effects , Peptides, Cyclic/pharmacology , Thromboplastin/metabolism , Cells, Cultured , Endothelium, Vascular/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Iodine Radioisotopes , Monocytes/metabolism , Receptors, Interleukin-1/antagonists & inhibitors
20.
J Pharmacol Exp Ther ; 269(2): 772-7, 1994 May.
Article in English | MEDLINE | ID: mdl-8182545

ABSTRACT

Thienopyridine compounds, including ticlopidine and clopidogrel, have been found to selectively inhibit adenosine 5' diphosphate (ADP)-induced platelet aggregation and adenylyl cyclase ex vivo, but the mechanism of their antiplatelet action remains to be determined. This study was aimed at investigating the effect of clopidogrel and ticlopidine on the binding of [3H]-2-methylthio- adenosine-5'-diphosphate (2-MeS-ADP) to rat platelets. Binding of [3H]-2-MeS-ADP to rat platelets was time-dependent and saturable. Scatchard analysis of the saturation binding data indicated that [3H]-2-MeS-ADP bound to one population of specific binding sites with high affinity (KD = 0.78 +/- 0.05 nM; Bmax = 156.3 +/- 4.8 fmole/10(8) cells) (n = 3). Unlabeled 2-MeS-ADP and ADP competitively and selectively inhibited the specific binding of [3H]-2-MeS-ADP with IC50 values of 11.3 +/- 1.2 nM and 11.3 +/- 0.7 microM, respectively (n = 3). Other nucleotide analogs such as ADP-beta S, ATP and ATP-alpha S also antagonized [3H]-2-MeS-ADP binding. When administered orally at doses ranging from 1 to 25 mg/kg, clopidogrel inhibited ADP- or 2-MeS-ADP-induced platelet aggregation as well as ADP or 2-MeS-ADP-induced inhibition of intraplatelet adenylyl cyclase. When measured in parallel, clopidogrel reduced in a dose-dependent manner the binding of [3H]-2-MeS-ADP to rat platelets ex vivo. Clopidogrel administration resulted in the decrease of [3H]-2-MeS-ADP binding sites on platelets without any significant change in the affinity; this indicates noncompetitive binding. Ticlopidine (200 mg/kg a day for 3 days) behaved in the same way.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Thionucleotides/blood , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology , Adenosine Diphosphate/blood , Adenylyl Cyclases/metabolism , Animals , Binding Sites/drug effects , Blood Platelets/enzymology , Blood Platelets/metabolism , Clopidogrel , Female , In Vitro Techniques , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL