Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
1.
Phlebology ; 26(8): 332-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21705478

ABSTRACT

AIM: To determine whether calcium dobesilate can act in chronic venous insufficiency by similar antioxidant, anti-inflammatory mechanisms as in diabetic retinopathy. METHODS: Calcium dobesilate was tested in vitro for its protective action against oxidative/inflammatory stress in human varicose veins. Varicose greater saphenous veins were obtained from 14 patients (11 men, 3 women) aged 53-65 years. Oxidative stress was induced exogenously in the vein segments, with the phenazine methosulphate (PMS)/NADH couple. Total antioxidant status (TAS) and malondialdehyde (MDA) contents were used as markers of oxidative stress. RESULTS: Calcium dobesilate significantly prevented oxidative disturbances in the micromolar range. PMS/NADH-dependent TAS decrease was fully prevented with IC(50) = 11.4 ± 2.3 µmol/L (n = 6 veins), whereas MDA increase was fully prevented with IC(50) = (102 ± -3) µmol/L (n = 6 veins). Calcium dobesilate acted quali- and quantitatively like rutin, the reference compound. Comparison with pharmacokinetic data suggests that calcium dobesilate can act at therapeutic concentrations. CONCLUSION: Calcium dobesilate protected human varicose veins against oxidative stress in vitro at levels that correspond to therapeutic concentrations. Further studies are required to investigate whether a similar action is found in varicose veins from patients orally treated with calcium dobesilate.


Subject(s)
Antioxidants/metabolism , Calcium Dobesilate/pharmacology , Hemostatics/pharmacology , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Saphenous Vein/metabolism , Varicose Veins/metabolism , Aged , Dose-Response Relationship, Drug , Female , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/pathology , Inflammation/physiopathology , Male , Methylphenazonium Methosulfate/toxicity , Middle Aged , Organ Culture Techniques , Saphenous Vein/pathology , Saphenous Vein/physiopathology , Varicose Veins/drug therapy , Varicose Veins/pathology , Varicose Veins/physiopathology
2.
Encephale ; 37 Suppl 1: S83-9, 2011 May.
Article in French | MEDLINE | ID: mdl-21600338

ABSTRACT

INTRODUCTION: Anxiety is a major and frequent symptom of schizophrenia, which is associated with an increased risk of relapse, impaired functioning, lower quality of life and increased incidence of suicide attempts. Despite its clinical relevance, anxiety in schizophrenia remains poorly understood. In the prodromic phase, anxiety indicates a progression towards psychotic decompensation. After a first episode, it is an indicator of relapse. LITERATURE FINDINGS: Two approaches have been used to investigate anxiety in schizophrenia: (i) categorical approach (comorbidity of schizophrenia and anxiety disorders) and (ii) dimensional approach (anxiety as a major symptom of the "dysphoric" dimension). Clinical categorical studies reported an increased frequency of comorbidity between schizophrenia and obsessive-compulsive disorder, panic disorder, social phobia, post-traumatic stress disorder, generalized anxiety disorder, agoraphobia, and specific phobia. The dimensional approach proposes that five different factors contribute to the structure of the Positive and Negative Syndrome Scale (PANSS), with anxiety as a major symptom of the "dysphoria" dimension. Concerning diagnosis, it is unclear whether psychotic and neurotic anxiety differs in nature or intensity. Nevertheless, both are frequently opposed. DISCUSSION: Psychotic anxiety is intense, profound and hermetic. In contrast to neurotic anxiety, it is associated with psychomotor disturbances, such as agitation and sideration. There is no specific tool to evaluate anxiety in schizophrenia. The dimensional approach usually runs an evaluation using items or factors extracted from the most widely-used scales, i.e. PANSS or Brief Psychiatric Rating Scale (BPRS) or from anxiety scales developed in non-schizophrenic populations, such as the Hamilton Anxiety Scale (HAMA). Recently, we developed a specific scale for hetero-evaluation (Échelle Anxiété Schizophrénie [EAS scale]). The EAS scale was recently validated and the study of its sensitivity is ongoing. THERAPEUTICAL ISSUES: Several studies have examined the effects of antipsychotics on the anxious/depressive cluster extracted from the PANSS, and some other studies have specifically evaluated the effect of antipsychotics on depressive symptoms using the Montgomery and Asberg Depression Rating Scale (MADRS) and Calgary Depression Scale for Schizophrenia (CDSS), but to our knowledge, no study has reported the effect of antipsychotics or other treatment on anxiety when using a schizophrenia-specific scale. There are no specific guideline treatments for anxiety in schizophrenia. Among phenothiazines, cyamemazine is frequently prescribed in France, because of its potent anxiolytic activity and good neurological tolerance. Some authors have suggested a specific treatment with benzodiazepines. However, benzodiazepines should be used with caution, due to undesirable actions such as dependence, rebound and potentiation of certain lateral effects.


Subject(s)
Anti-Anxiety Agents/administration & dosage , Antipsychotic Agents/administration & dosage , Anxiety Disorders/drug therapy , Schizophrenia/drug therapy , Schizophrenic Psychology , Anti-Anxiety Agents/adverse effects , Antipsychotic Agents/adverse effects , Anxiety Disorders/diagnosis , Anxiety Disorders/psychology , Benzodiazepines/adverse effects , Benzodiazepines/therapeutic use , Comorbidity , Depressive Disorder/diagnosis , Depressive Disorder/drug therapy , Diagnosis, Differential , Drug Therapy, Combination , Humans , Personality Inventory , Phenothiazines/adverse effects , Phenothiazines/therapeutic use , Psychiatric Status Rating Scales , Schizophrenia/diagnosis
3.
Arch Mal Coeur Vaiss ; 97(7-8): 731-3, 2004.
Article in French | MEDLINE | ID: mdl-15506055

ABSTRACT

Renal NaCl reabsorption is increased in Dahl "salt-sensitive" (DS) rats, due to an increased activity of the Na-K-Cl cotransporter NKCC2. On the other hand, nitric oxide (NO) is an inhibitor of NKCC2 and a deficient nitric oxide synthase (NOS) seems to play an important role in salt-sensitivity of DS rats. Here, we investigated the hypothesis that NKCC2 hyperactivity in DS rats is due to a deficient NOS, via the interactions cyclic GMP (cGMP)/cyclic AMP (cAMP) at the level of the thick ascending Henle's loop (TAL). DS rats DS (males, 250-300 g) and their normotensive controls DR ("salt-resistant") are sacrificed, the kidneys removed and NKCC2 activity is measured in medullary TAL (mTAL) as previously described. Medullary contents of NO are measured with a NitroFlux analyser by heat-reduction of nitrates and nitrites to NO. AMPc levels in mTAL are measured by an EIA immunotest. Neither L-NAME (3 mM), nor L-arginine were able to modify NKCC2 activity in mTAL from DS (pre-hypertensive) or DR rats. Levels of NO in the medullary interstitium and AMPc in mTAL were not significantly different between DS and DR rats. Conversely, in DS rats charged with 2% salt (in the food) during 7 weeks, L-arginine significantly inhibited NKCC2 in DS (35.6 +/- 6.8 vs 25.3 +/- 4.9 nmoles/mg protein/min; p<0.05 non-paired Student's t-test), but not in DR rats. In conclusion, NKCC2 in our mTAL preparation of prehypertensive DS and DR rats is insensitive to L-NAME and L-arginine. This suggests the absence of a functional NOS. NKCC2 hyperactivity of prehypertensive DS is therefore not due to a deficient NOS. This was confirmed by the normal levels of interstitial NO and mTAL cAMP in prehypertensive DS rats. Finally, a salt-load seems to induce NOS expression in mTAL of DS rats. This last observation deserves further investigation.


Subject(s)
Hypertension/physiopathology , Nitric Oxide Synthase/pharmacology , Nitric Oxide/pharmacology , Sodium Chloride/metabolism , Sodium Chloride/pharmacokinetics , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/pharmacology , Animals , Disease Models, Animal , Male , Rats , Rats, Inbred Dahl , Solute Carrier Family 12, Member 1
4.
Pulm Pharmacol Ther ; 17(4): 213-8, 2004.
Article in English | MEDLINE | ID: mdl-15219266

ABSTRACT

In vitro studies in rat mastocytes and human monocytes suggested that reproterol (a selective beta(2)-adrenoceptor agonist with a theophylline moiety) exerts anti-inflammatory actions through inhibition of cyclic AMP (cAMP) PDE activity. Thus, reproterol was tested for its ability to inhibit cAMP PDE in cultured mouse mastocytoma P-815 cells. cAMP PDE activity was measured in intact cells by spectrofluorometry using the fluorescent substrate 2'-O-anthraniloyl cAMP. Reproterol was more potent than theophylline to inhibit cAMP PDE (pIC(50)=4.28+/-0.25 vs. 3.16+/-0.05). This contrasted with disrupted cells, where the PDE inhibitory potency of reproterol was low (pIC(50)=2.85+/-0.03) and similar to that of theophylline (pIC(50)=2.66+/-0.19). No cAMP PDE inhibition was found with other beta(2)-agonists tested (fenoterol, salbutamol, salmeterol and formoterol). Finally, the selective PDE inhibitors calmidazolium (100 nM), milrinone (5 microM) and rolipram (50 microM) inhibited cAMP PDE activity by approximately 20, 30 and 25% respectively. In conclusion, reproterol potently and non-specifically inhibited intracellular cAMP phosphodiesterases in intact mastocytoma cells. This can explain the previously reported beta(2)-adrenoceptor-independent anti-inflammatory actions of reproterol in vitro. Further studies are required to define the anti-inflammatory potential of reproterol in asthma.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Adrenergic beta-Agonists/pharmacology , Bronchodilator Agents/pharmacology , Cyclic AMP/antagonists & inhibitors , Metaproterenol/analogs & derivatives , Metaproterenol/pharmacology , Theophylline/analogs & derivatives , Theophylline/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adrenergic beta-Agonists/administration & dosage , Animals , Bronchodilator Agents/administration & dosage , Dose-Response Relationship, Drug , Drug Combinations , Metaproterenol/administration & dosage , Mice , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/pharmacology , Theophylline/administration & dosage , Tumor Cells, Cultured
5.
Naunyn Schmiedebergs Arch Pharmacol ; 367(2): 134-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12595954

ABSTRACT

The low incidence of extrapyramidal effects with atypical neuroleptics has been ascribed to their 5-HT(2A)- and 5-HT(2C)-serotonin receptor antagonistic properties. On the other hand, the acute increase in striatal dopamine release by submaximal dopamine D(2) autoreceptor blockade can be respectively reduced and increased by 5-HT(2A)- and 5-HT(2C)-antagonists. Cyamemazine is a neuroleptic D(2)- and 5-HT(2A)-receptor antagonist, with small antagonistic activity at 5-HT(2C) receptors and low incidence of extrapyramidal side effects. Therefore, submaximal cyamemazine was tested in rats for its acute action on the extracellular concentrations of dopamine and dopamine metabolites (DOPAC: 3,4,dihydroxyphenylacetic acid and HVA: 4-hydroxy-3-methoxy-phenyl-acetic acid) in the corpus striatum. The serotonin metabolite 5-HIAA (5-hydroxy-indole-acetic acid) was measured in parallel. Rats prepared for microdialysis (striatum) were intraperitoneally given cyamemazine 1 mg/kg, 5 mg/kg or vehicle ( n=4 in each group). Dopamine, DOPAC, HVA and 5-HIAA concentrations in perfusates under basal conditions and after stimulation by high K(+) were measured by HPLC coupled to electrochemical detection. Cyamemazine 1 mg/kg significantly reduced extracellular concentrations of basal dopamine (-77%), DOPAC (-54%), HVA (-54%) and 5-HIAA (-65%). No such effects were seen with the dose of cyamemazine 5 mg/kg or for K(+)-evoked dopamine release. In conclusion, submaximal cyamemazine can acutely reduce basal dopamine release and metabolism in the rat striatum. Such unusual action can be explained by the original pharmacological profile of cyamemazine (potent D(2)- and 5-HT(2A)-antagonist, with small antagonistic activity at 5-HT(2C) receptors). Further experiments are required to explain the low incidence of extrapyramidal side actions with cyamemazine.


Subject(s)
Corpus Striatum/drug effects , Dopamine Antagonists/pharmacology , Dopamine/metabolism , Phenothiazines/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Corpus Striatum/metabolism , Dopamine Antagonists/administration & dosage , Dose-Response Relationship, Drug , Homovanillic Acid/metabolism , Hydroxyindoleacetic Acid/metabolism , Injections, Intraperitoneal , Male , Microdialysis , Phenothiazines/administration & dosage , Rats , Rats, Wistar , Time Factors
6.
Naunyn Schmiedebergs Arch Pharmacol ; 365(3): 193-9, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11882915

ABSTRACT

It is generally assumed that bumetanide possesses some selectivity for the renal Na-K-Cl cotransporter NKCC2, although the results are scarce in the literature and comparisons were done with extra-renal NKCC1 at its basal, almost silent state. Here we investigated NKCC2/NKCC1 selectivity of loop diuretic drugs (bumetanide, piretanide and furosemide) as a function of the NKCC1 activated state (NKCC1 was activated by hypertonic media). NKCC2 activity was measured in isolated rat medullary thick ascending limb (mTAL) and NKCC1 in rat thymocytes and erythrocytes. When NKCC2 was compared with NKCC1at its activated state, all three diuretic drugs inhibited NKCC2 and NKCC1 with the same potency (bumetanide pIC50=6.48, 6.48 and 6.47; piretanide pIC50=5.97, 5.99 and 6.29; and furosemide pIC50=5.15, 5.04 and 5.21 for mTAL NKCC2, erythrocyte NKCC1 and thymocyte NKCC1, respectively). Basal NKCC1 exhibited a lower diuretic sensitivity, although with marked differences depending on the diuretic drug and the cell type in consideration and with the notable exception of furosemide in erythrocytes. Molecular modelling showed that bumetanide and piretanide possess four potentially active groups, of which three are shared with furosemide at similar intergroup distances. Of these three common groups, one should not bind to basal NKCC1 in thymocytes. The fourth (phenoxy) group (absent in furosemide) confers higher lipophilicity and should not bind to basal NKCC1 in erythrocytes. In conclusion, loop diuretics had no NKCC2/NKCC1 selectivity, when NKCC1 is measured at its activated state. Basal NKCC1 has a reduced diuretic sensitivity, of very different magnitude depending on the diuretic drug and cell type in consideration.


Subject(s)
Diuretics/pharmacology , Ion Transport/drug effects , Loop of Henle/drug effects , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Bumetanide/chemistry , Bumetanide/pharmacology , Diuretics/chemistry , Erythrocytes/drug effects , Erythrocytes/metabolism , Furosemide/chemistry , Furosemide/pharmacology , Loop of Henle/metabolism , Male , Rats , Rats, Wistar , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 2 , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/metabolism
7.
Eur J Pharmacol ; 428(2): 277-86, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11675046

ABSTRACT

Calcium dobesilate possesses antioxidant properties and protects against capillary permeability by reactive oxygen species in the rat peritoneal cavity, but whether a similar action can take place in the diabetic rat retina is unknown. We investigated the oral treatment of diabetic rats with calcium dobesilate on the prevention of free radical-mediated retinal injury induced by ischemia/reperfusion (90 min ischemia followed by 3 min and/or 24 h of reperfusion). Streptozotocin-induced diabetic rats were orally treated with 50 and 100 mg/kg of calcium dobesilate for 10 days (n=12 in each group). In the first series of studies, calcium dobesilate was found to significantly reduce the maldistribution of ion content in diabetic ischemic/reperfused rat retina. Thus, in diabetic rats treated with 100 mg/kg/day calcium dobesilate, ischemia/reperfusion provoked: (i) 27.5% increase in retinal Na(+) content compared to 51.8% in the vehicle-treated group (P<0.05), and (ii) 59.6% increase in retinal Ca(2+) content compared to 107.1% in vehicle-treated animals (P<0.05). In the second series of studies, calcium dobesilate was found to significantly protect diabetic rat retina against inhibition of Na(+)/K(+)-ATPase and Ca(2+)/Mg(2+)-ATPase activities by ischemia/reperfusion (54% and 41% reduction, respectively, with 100 mg/kg of calcium dobesilate) and also against changes in retinal ATP, reduced glutathione (GSH), and oxidized glutathione (GSSG) contents. In the third series of experiments, rats treated with 100 mg/kg of calcium dobesilate reduced the hydroxyl radical signal intensity to 41% (measured by electron paramagnetic resonance), induced by ischemia/reperfusion in diabetic rat retina. Finally, 100 mg/kg calcium dobesilate significantly reduced retinal edema (measured by the thickness of the inner plexiform layer) in diabetic rats. In conclusion, oral treatment with calcium dobesilate significantly protected diabetic rat retina against oxidative stress induced by ischemia/reperfusion. Whether the antioxidant properties of calcium dobesilate explain, at least in part, its beneficial therapeutic effects in diabetic retinopathy deserves further investigation.


Subject(s)
Antioxidants/pharmacology , Calcium Dobesilate/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetic Retinopathy/prevention & control , Reperfusion Injury/complications , Adenosine Triphosphate/metabolism , Animals , Antioxidants/therapeutic use , Ca(2+) Mg(2+)-ATPase/drug effects , Ca(2+) Mg(2+)-ATPase/metabolism , Calcium/metabolism , Calcium Dobesilate/therapeutic use , Cations/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/etiology , Diabetic Retinopathy/metabolism , Free Radicals/metabolism , Glutathione/drug effects , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , Magnesium/metabolism , Male , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/metabolism , Retina/pathology , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism
8.
Article in English | MEDLINE | ID: mdl-11244365

ABSTRACT

The H(1) antagonist azelastine is used in nasal sprays for the treatment of allergic rhinitis, but its therapeutic efficacy in vasomotor rhinitis is unknown. We performed a multicenter randomized double-blind placebo-controlled study of the efficacy and tolerance of azelastine nasal spray in 89 adult patients with vasomotor rhinitis (confirmed by negative Phadiatop). Following a washout period, patients were treated for 15 days with one puff three times daily per nostril of azelastine (n = 44) or placebo (n = 45) nasal spray. Efficacy was evaluated by the reduction in symptomatology and by rhinoscopy. Intent-to-treat analysis revealed better results in the azelastine group for all assessed symptoms; the significance level was reached for nasal obstruction on day 15 (p = 0.042). Using per protocol analysis (in 85 patients complying with the protocol), the significance level was reached for nasal obstruction on day 15 (p = 0.017) and for the percentage of success in rhinorrhea (p = 0.023). In the azelastine group, rhinoscopy examination showed a significantly higher reduction in the inflammatory level and edema of the nasal mucosa (p = 0.03 and 0.02 for VAS on day 15 respectively, per protocol analysis). General efficacy assessment by the physician and the patient was in favor of azelastine (with significance levels <0.01). No drowsiness or serious adverse event was reported, and the frequency of mouth dryness and headaches was similar in the two treatment groups. The present study demonstrates the efficacy of azelastine nasal spray in the treatment of vasomotor rhinitis. The best achieved results were a decrease in nasal obstruction and mucosal edema. Further studies are required to investigate if this therapeutic benefit results from H(1) antagonism or from another, not well-characterized pharmacological action of azelastine.


Subject(s)
Histamine H1 Antagonists/therapeutic use , Phthalazines/therapeutic use , Rhinitis, Vasomotor/drug therapy , Administration, Intranasal , Adolescent , Adult , Aged , Aged, 80 and over , Female , Histamine H1 Antagonists/administration & dosage , Histamine H1 Antagonists/adverse effects , Humans , Male , Middle Aged , Nasal Obstruction/drug therapy , Phthalazines/administration & dosage , Phthalazines/adverse effects , Treatment Outcome
9.
Rev Mal Respir ; 17(2 Pt 2): 531-3, 2000 Jun.
Article in French | MEDLINE | ID: mdl-10939109

ABSTRACT

Myosin is an ATPase, able to form filaments with actin, thus initiating smooth muscle contraction (conversion of chemical energy into mechanical energy). Myosin activity is regulated by cytosolic calcium, via a calcium-calmodulin-MLCK-dependent phosphorylation. Extrusion of cytosolic calcium via calcium pumps (in the plasma membrane and sarcoplasmic reticulum) and via a sodium-calcium exchange allow smooth muscle cells to maintain their resting state. Constrictor agonists (hormones, neurotransmitters or drugs) act at membrane receptors inducing: (i) a fast and transient calcium mobilization from the sarcoplasmic reticulum, via phospholipase C (PLC) stimulation and inositol triphosphate (IP3) production or via a "calcium-induced calcium release" mechanism and opening of calcium channels in the sarcoplasmic reticulum and (ii) a slow and maintained mobilization of extracellular calcium, via the opening of voltage-dependent calcium channels in plasma membranes. Smooth muscle relaxation is ensured by a phosphatase which hydrolyzes phosphorylated myosin and decreases the calcium sensitivity of the contractile apparatus. Calcium signal is regulated at that level by: (i) protein kinase C, tyrosine kinase and arachidonic acid which inhibit phosphatase activity and (ii) cyclic AMP (cAMP) and cyclic GMP (cGMP) which enhance phosphatase activity. A second regulatory site is situated at the level of the non-contractile calcium compartment, which buffers signal transduction and where cGMP and/or cAMP enhance calcium extrusion mechanisms.


Subject(s)
Muscle Contraction/physiology , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium-Transporting ATPases/metabolism , Humans , Muscle Proteins/metabolism , Muscle Proteins/physiology , Muscle Relaxation/physiology , Muscle, Smooth/enzymology
10.
Psychopharmacology (Berl) ; 147(4): 412-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10672635

ABSTRACT

RATIONALE: Cyamemazine is a neuroleptic compound which possesses anxiolytic properties in humans. On the other hand, 5-HT3- and 5-HT2C-receptors have been implicated in anxiety disorders and a previous binding study has shown that cyamemazine possesses high affinity for both serotonin receptor types. OBJECTIVE: The present study was undertaken to establish whether cyamemazine antagonizes 5-HT3- and/or 5-HT2C-mediated responses, and whether it compares with reference compounds. METHODS: Cyamemazine was tested for its ability to antagonize: (i) 5-HT3-dependent contraction of the isolated guinea-pig ileum and bradycardic responses in the rat and (ii) 5-HT2C-dependent phospholipase C (PLC) stimulation in rat brain membranes. RESULTS: In isolated guinea-pig ileum, cyamemazine potently and competitively antagonized 5-HT-dependent contractions (pA2 = 7.52 +/- 0.08; n = 5). In this test, cyamemazine was 5-7 times more potent (pIC50 = 6.75 +/- 0.13) than tropisetron (pIC50 = 6.02 +/- 0.04). In rats, cyamemazine i.v. antagonized 5-HT-dependent bradycardic responses with ID50% = 3.2 +/- 1.5 mg/kg (n = 4). Finally, in rat brain membranes cyamemazine antagonized 5-HT2C-dependent PLC stimulation with Ki = 424 nM (mianserin exhibits a Ki = 113 nM). CONCLUSIONS: Cyamemazine behaves as an antagonist at both 5-HT3- and 5-HT2C-receptors, which compares well with reference compounds. These 5-HT3- and 5-HT2C-antagonistic actions of cyamemazine can be involved, at least in part, in its beneficial therapeutic actions in anxiety disorders.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antipsychotic Agents/pharmacology , Phenothiazines/pharmacology , Receptors, Serotonin/drug effects , Serotonin Antagonists/pharmacology , Animals , Caudate Nucleus/enzymology , Guinea Pigs , Heart Rate/drug effects , In Vitro Techniques , Male , Membranes/metabolism , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Radioligand Assay , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2C , Receptors, Serotonin, 5-HT3 , Type C Phospholipases/metabolism
11.
Biochem Biophys Res Commun ; 268(3): 673-6, 2000 Feb 24.
Article in English | MEDLINE | ID: mdl-10679263

ABSTRACT

When incubated in 150 mM KCl, rat thymocytes exhibited a very important magnesium efflux (11.4 +/- 0.7 mmoles/liter cells/20 min, n = 29), about 90 times higher than the physiological magnesium efflux catalyzed by the Na-Mg exchanger (0.126 +/- 0.093 mmoles/liter cells/20 min). Cells remained viable (trypan blue test) and membrane integrity was shown by the absence of an increase in sodium permeability. K(+)-induced magnesium efflux exhibited the following properties: (i) it required the presence of external chloride; (ii) it was fully blocked by DIOA, a selective KCl-cotransporter inhibitor (IC(50) = 35 microm); and (iii) it was associated to a progressive increase in cell volume via the DIOA-sensitive K-Cl cotransporter. Such cell swelling seems to play a causal role, because (i) hypertonic media (+400 mM sucrose) abolished K(+)-induced magnesium efflux and (ii) hypotonic Ringer media (205 mOsm) increased both cell volume and magnesium efflux (from a basal value of 0.35 +/- 0.03 mmoles/liter cells/20 min up to 1.44 +/- 0.24 mmoles/liter cells/20 min), even in the presence of DIOA. In conclusion, high potassium induced a dramatic release of intracellular magnesium from rat thymocytes. Such a phenomenon was, at least in part, caused by cell swelling via the DIOA-sensitive K-Cl cotransporter. The nature of the magnesium transport mechanism and its role in the transduction signal of K-Cl cotransporter activation by cell swelling deserve further investigation.


Subject(s)
Magnesium/metabolism , Potassium/pharmacology , Symporters , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Animals , Antiporters/metabolism , Carboxylic Acids/pharmacology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Size/drug effects , Chlorides/metabolism , In Vitro Techniques , Indenes/pharmacology , Ion Transport/drug effects , Male , Osmolar Concentration , Potassium/metabolism , Rats , Rats, Wistar , Signal Transduction , Sodium/metabolism , K Cl- Cotransporters
12.
Arch Mal Coeur Vaiss ; 92(8): 1001-4, 1999 Aug.
Article in French | MEDLINE | ID: mdl-10486654

ABSTRACT

UNLABELLED: Although the renal receptor at which cicletanine acts is unknown, cicletanine was assumed to act like thiazide diuretics. Here we tested cicletanine and its natriuretic metabolite, cicletanine-sulfate, for inhibitory activity against the thiazide-sensitive NaCl cotransporter expressed in Xenopus oocytes. The renal thiazide-sensitive NaCl cotransporter was expressed in Xenopus laevis oocytes injected with rat cRNA TSCr (TSCr: thiazide-sensitive cotransporter from rat kidney) and both, racemic (+/-) cicletanine and its sulfoconjugated metabolite were tested for inhibitory activity against oocyte 22Na+ uptake catalyzed by this cotransporter. Polythiazide was used as reference thiazide. Polythiazide fully inhibited NaCl cotransporter function with IC50 approximately 1.2 x 10(-7) M. Conversely, neither cicletanine, nor cicletanine sulfate were able to inhibit such cotransporter, i.e.: a minimum concentration of 10(-4) M of cicletanine was necessary to induce a slight cotransporter inhibition (29.5 +/- 18.2%). Cicletanine sulfate was inactive, even at 10(-4) M. IN CONCLUSION: (i) the natriuretic metabolite of cicletanine (cicletanine sulfate) is unable to inhibit thiazide-sensitive NaCl cotransporter and (ii) inhibition of such cotransporter by cicletanine required concentrations equal or higher than 10(-4) M--concentrations much more higher than urinary therapeutic ones in humans (approximately 10(-6) M). These results clearly demonstrate that cicletanine does not act like thiazide diuretics.


Subject(s)
Benzothiadiazines , Carrier Proteins/drug effects , Diuretics/pharmacology , Oocytes/drug effects , Pyridines/pharmacology , Receptors, Drug/drug effects , Sodium Chloride Symporter Inhibitors/pharmacology , Symporters , Animals , Female , Sodium Chloride Symporters , Sodium Radioisotopes , Xenopus laevis
13.
Arch Mal Coeur Vaiss ; 92(8): 1033-8, 1999 Aug.
Article in French | MEDLINE | ID: mdl-10486660

ABSTRACT

In the 80s, erythrocyte Na-K-Cl cotransporter of essential hypertensive was reported: (i) decreased in fresh erythrocytes and (ii) increased, following repeated cell washings and incubations. This suggested to us that the manipulation of erythrocytes (from essential hypertensives) was able to dissociate a cotransport inhibitory factor, thus unmasking up-regulation of membrane cotransport units. This working hypothesis was recently confirmed in Dahl salt-sensitive rats (DS). The primary defect of DS rats seems to be hyperactivity of cotransporter Na-K-Cl BSC1 at the thick ascending limb of Henle's loop (TAL). Moreover, oral salt-loading induces an abnormally high increase in the urinary and plasmatic CIF levels of DS rats. The increase in urinary CIF excretion seems to be a compensatory mechanism, able to reduce BSC1 hyperactivity and NaCl reabsorption at the TAL. The increase in plasmatic CIF should inhibit erythrocyte BSC2, thus inducing "up-regulation" of the membrane density of cotransport proteins. Further studies are required to test this model in human with "salt-sensitive" hypertension.


Subject(s)
Carrier Proteins/metabolism , Hypertension/blood , Sodium Chloride/adverse effects , Absorption , Administration, Oral , Animals , Erythrocytes/metabolism , Humans , Hypertension/etiology , Rats , Rats, Inbred Dahl , Sodium Chloride/pharmacokinetics , Sodium-Potassium-Chloride Symporters
14.
Pulm Pharmacol Ther ; 12(1): 55-60, 1999.
Article in English | MEDLINE | ID: mdl-10208836

ABSTRACT

The beta2-adrenoceptor agonist reproterol and disodium cromoglycate (DSCG) are used in fixed combination for the treatment of asthma, because they act on bronchial smooth muscle and inflammatory cells, respectively. Here, we investigated if reproterol can also act in rat mast cells in vitro to facilitate the inhibitory action of disodium cromoglycate (DSCG) on histamine secretion induced by compound 48/80. Reproterol was as potent as DSCG to inhibit histamine release in rat mast cells (32.8+/-6.0 vs. 36.7+/-6.2% at 1 microM of each compound, n=10 and n=8 respectively). Mast cell stabilization by DSCG (1-100 microM) was strongly and significantly enhanced in the presence of a fixed saturating concentration of reproterol (100 microM). Conversely, the combination of DSCG (1-100 microM) with the beta2-agonist used as reference compound, salbutamol (100 microM) did not inhibit histamine release more than DSCG alone. In combination with a saturating concentration of DSCG (100 microM), reproterol inhibited histamine release more than reproterol alone. The potent adenylate cyclase stimulator forskolin (50 microM) was able to inhibit histamine release to a similar extent as DSCG and significantly (P<0.05) enhanced the inhibition of histamine release by DSCG. Finally, the phosphodiesterase inhibitor theophylline (100 microM) was equipotent to reproterol and DSCG in stabilizing rat mast cells. In conclusion, reproterol enhances the ability of disodium cromoglycate to stabilize rat mast cells. This effect is not shared by salbutamol and can be, at least in part, independent of beta2-adrenoceptor stimulation.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Bronchodilator Agents/pharmacology , Cromolyn Sodium/pharmacology , Histamine Release/drug effects , Mast Cells/drug effects , Metaproterenol/analogs & derivatives , Theophylline/analogs & derivatives , Animals , Cell Line , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Mast Cells/metabolism , Metaproterenol/pharmacology , Rats , Rats, Wistar , Theophylline/pharmacology
15.
Fundam Clin Pharmacol ; 13(1): 50-8, 1999.
Article in English | MEDLINE | ID: mdl-10027088

ABSTRACT

The alpha 1-adrenergic blocking activity of nicergoline was re-examined in rats, with a particular emphasis on alpha 1-adrenoceptor subtypes. In pithed rats, nicergoline and prazosin infused at a single small dose (0.5 microgram/kg/min i.v.) produced a substantial and identical shift to the right of the control dose pressor response curve to the specific alpha 1-agonist cirazoline (ED50 = 4.0 +/- 0.1, 4.0 +/- 0.1 and 0.9 +/- 0.01 microgram/kg i.v. for nicergoline, prazosin and vehicle respectively). In the isolated perfused mesenteric vascular bed, nicergoline strongly inhibited the pressor responses elicited by cirazoline, with approximately 40-fold higher potency (pA2 = 11.1 +/- 0.3) than prazosin (pA2 = 9.5 +/- 0.3). Conversely, nicergoline was 20-fold less potent than prazosin to antagonize the contractile effects of cirazoline in isolated endothelium-denuded aorta (pA2 = 8.6 +/- 0.2 and 9.9 +/- 0.2 for nicergoline and prazosin respectively). Pretreatment of mesenteric vascular beds with chloroethylclonidine did not significantly modify nicergoline antagonistic potency (pA2 = 10.6 +/- 0.2). Nicergoline displaced [3H]-prazosin bound to rat forebrain membranes pretreated with chloroethylclonidine (pKi = 9.9 +/- 0.2) at concentrations 60-fold lower than in rat liver membranes (pKi = 8.1 +/- 0.2). Finally, of the nicergoline metabolites studied, lumilysergol acted as a modest alpha 1 antagonist (bromonicotinic acid was devoid of alpha 1 antagonist activity). In conclusion, nicergoline is a potent and selective alpha 1A-adrenoceptor subtype antagonist, an alpha 1-adrenoceptor subtype which is mainly represented in resistance arteries.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Mesenteric Arteries/drug effects , Nicergoline/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Aorta/drug effects , Azepines/pharmacology , Binding, Competitive , Blood Pressure/drug effects , Clonidine/analogs & derivatives , Clonidine/pharmacology , Decerebrate State , Dose-Response Relationship, Drug , Imidazoles/pharmacology , In Vitro Techniques , Male , Prazosin/metabolism , Radioligand Assay , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1 , Sensitivity and Specificity , Tritium
16.
J Med Food ; 2(3-4): 257-60, 1999.
Article in English | MEDLINE | ID: mdl-19281395

ABSTRACT

A furosemide-sensitive Na-K-Cl cotransporter (NKCC2 isoform) accounts for almost all luminal NaCl reabsorption in the thick ascending limb of Henle's loop (TALH). The activity of this transport protein is regulated by humoral factors known as cotransport inhibitory factors. One family of these compounds is represented by the urinary phytoestrogens equol and genistein, which inhibit cotransport fluxes at concentrations similar to furosemide. Moreover, they possess salidiuretic potency similar to furosemide in the isolated perfused rat kidney, but are less potent than furosemide (in vivo). Thus, dietary phytoestrogens can be responsible, at least in part, for the low blood pressure of vegetarians.

17.
J Hypertens ; 16(10): 1499-504, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9814622

ABSTRACT

BACKGROUND: Abnormal Na,K,Cl cotransport is thought to be a pathogenic factor in Dahl salt-sensitive rat models, but the only direct evidence for this is an increased cotransport activity found in erythrocytes from salt-loaded Dahl salt-sensitive rats. OBJECTIVE: To re-examine erythrocyte cotransport fluxes and a circulating cotransport inhibitory factor (CIF) in inbred Dahl rats maintained on a low (0.2%) salt diet. Cotransport fluxes were investigated both under basal conditions and after stimulation by cell shrinking. METHODS: Blood was drawn from 12 male Dahl salt-sensitive and 12 Dahl salt-resistant rats of the inbred John Rapp strain. Erythrocyte Na,K,Cl cotransport activity was equated to the bumetanide-sensitive fluxes of sodium, rubidium or lithium. Plasma CIF activity was tested in human erythrocytes. RESULTS: In Dahl salt-sensitive rats: (1) plasma CIF activity (5.7+/-0.4 units/ml) was modestly higher than in Dahl salt-resistant rats (2.97+/-0.12 units/ml, P < 0.0001), but much lower than that previously found in salt-loaded Dahl salt-sensitive rats (16.1 units/ml), and (2) erythrocytes exhibited a similar bumetanide-sensitive sodium efflux (rate constant 0.056+/-0.008 h(-1)) as in Dahl salt-resistant rats (0.047+/-0.007 h(-1)). Following hypertonic shock, the bumetanide-sensitive rubidium influx reacted more to cell shrinkage in Dahl salt-sensitive than in Dahl salt-resistant erythrocytes (cell volume decrease required to stimulate bumetanide-sensitive rubidium influx by 4000 micromol/l cells per h=-4.04+/-0.36 versus -5.89+/-0.44 fl, respectively; P< 0.01). CONCLUSIONS: When fed a low-salt diet, Dahl salt-sensitive rats present slightly increased plasma CIF levels and normal erythrocyte cotransport fluxes under basal conditions, but an increased response to a hypertonic shock. Therefore, if there is any primary cotransport abnormality in Dahl salt-sensitive rats, it appears to be restricted to the renal Na,K,Cl cotransporter BSC1 isoform. Alternatively, any such change may be the consequence of abnormal regulation by osmolarity-dependent mechanisms.


Subject(s)
Carrier Proteins/blood , Erythrocyte Membrane/metabolism , Hypertension/blood , Membrane Proteins/blood , Natriuretic Agents/blood , Animals , Bumetanide/pharmacology , Carrier Proteins/antagonists & inhibitors , Chlorides/blood , Diuretics/pharmacology , Erythrocyte Membrane/drug effects , Humans , Hypertension/drug therapy , Ion Transport/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Potassium/blood , Rats , Rats, Inbred Dahl , Sodium/blood , Sodium Chloride/pharmacology , Sodium-Potassium-Chloride Symporters
18.
Eur J Pharmacol ; 358(3): 213-20, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9822887

ABSTRACT

Calcium dobesilate possesses antioxidant properties in vitro, but the in vivo significance and putative angioprotective role of these properties are undefined. Here, calcium dobesilate was tested in a newly developed in vivo model of microvascular permeabilization induced by reactive oxygen species in the rat peritoneal cavity. In this model, microvascular permeabilization is equated to the rate of Evans blue extravasation toward the peritoneal cavity. Basal Evans blue extravasation (rate constant values ke = 0.0176 +/- 0.0015 h-1) was markedly and significantly increased by reactive oxygen species generated in situ, with: (i) phenazine methosulfate/NADH (delta ke(phenazine methosulfate) = 0.0419 +/- 0.0043 h-1) and (ii) xanthine/xanthine oxidase (delta ke(xo) = 0.0383 +/- 0.0010x h-1). These actions of reactive oxygen species were abolished by locally injected superoxide dismutase (i.p., 300 units/kg). Intraperitoneally given calcium dobesilate (100 mg/kg) inhibited 75-100% of reactive oxygen species-induced Evans blue extravasation. By the intravenous route, calcium dobesilate i.v. (1-50 mg/kg) dose dependently inhibited phenazine methosulfate-induced Evans blue extravasation with an ID50 of 2-5 mg/kg (full inhibition was reached at 20-50 mg/kg). After single oral administration, calcium dobesilate (5-500 mg/kg) dose dependently inhibited phenazine methosulfate-dependent Evans blue extravasation with an ID50 of 50-100 mg/kg (81% inhibition at 500 mg/kg, P < 0.003). After 7 days of oral calcium dobesilate (50 mg/kg once/day) phenazine methosulfate-induced Evans blue peritoneal extravasation was significantly reduced by half. These effects of calcium dobesilate were similar to those observed with a comparative antioxidant molecule, rutin. In conclusion, rat peritoneal microvascular permeability was strongly increased by reactive oxygen species, an effect that was significantly reduced by intraperitoneal, intravenous and oral calcium dobesilate. These results support the hypothesis that the antioxidant properties of calcium dobesilate could play a role in its angioprotective properties in vivo.


Subject(s)
Calcium Dobesilate/pharmacology , Capillary Permeability/drug effects , Hemostatics/pharmacology , Reactive Oxygen Species/physiology , Administration, Oral , Animals , Calcium Dobesilate/chemistry , Calcium Dobesilate/therapeutic use , Capillary Permeability/physiology , Dose-Response Relationship, Drug , Drug Administration Routes , Evans Blue , Extravasation of Diagnostic and Therapeutic Materials/physiopathology , Extravasation of Diagnostic and Therapeutic Materials/prevention & control , Hemostatics/therapeutic use , Injections, Intraperitoneal , Injections, Intravenous , Male , Peritoneal Cavity/physiopathology , Rats , Rats, Wistar
19.
Clin Exp Hypertens ; 20(5-6): 675-82, 1998.
Article in English | MEDLINE | ID: mdl-9682923

ABSTRACT

A furosemide-sensitive Na-K-Cl cotransporter (NKCC2 isoform) accounts for almost all luminal NaCl reabsorption in the thick ascending limb of Henle's loop (TALH). The activity of this transport protein is regulated by humoral factors (CIF: cotransport inhibitory factors). One family of CIF compounds is represented by the urinary phytoestrogens equol and genistein, which inhibit cotransport fluxes at similar concentrations as furosemide. Moreover, they possess similar salidiuretic potency as furosemide in the isolated perfused rat kidney, but are less potent than furosemide in vivo. Thus, dietary phytoestrogens can be responsible, at least in part, for the low blood pressure of vegetarians. A second type of CIF is represented by a circulating and urinary factor which is evoked by salt-loading. This, which is not a "ouabain-like" factor, appears to be a new retropituitary natriuretic compound. Endogenous CIF is increased in hypertensive Dahl salt-sensitive rats, probably as a compensatory mechanism against the enhanced NaCl reabsorption in the TALH, which characterizes this model of hypertension. Finally, chronic excess of circulating CIF inhibits and induces up-regulation of erythrocyte Na-K-Cl cotransporter NKCC1.


Subject(s)
Carrier Proteins/metabolism , Hypertension/metabolism , Membrane Proteins/metabolism , Natriuretic Agents/pharmacology , Animals , Carrier Proteins/antagonists & inhibitors , Diuretics/pharmacology , Erythrocytes/drug effects , Erythrocytes/metabolism , Furosemide/pharmacology , Humans , Hypertension/drug therapy , Loop of Henle/drug effects , Loop of Henle/metabolism , Membrane Proteins/antagonists & inhibitors , Rats , Sodium-Potassium-Chloride Symporters
20.
Am J Hypertens ; 11(5): 579-84, 1998 May.
Article in English | MEDLINE | ID: mdl-9633794

ABSTRACT

In pithed rats, the levorotatory (-)-enantiomer of cicletanine reduces the pressor responses to angiotensin II (AII) and also, to a lesser extent, those to arginine-vasopressin (AVP). Here we have attempted to characterize further these inhibitory effects by studies of isolated perfused rat kidney and mesenteric vascular beds. In the isolated rat kidney, (-)-cicletanine behaves as a noncompetitive antagonist of AII- and AVP-receptor stimulation, with Ki values of 9.6 and 208 micromol/L respectively. In the isolated mesenteric vascular bed, (-)-cicletanine antagonized both AII dependent contractions with an inhibitory concentration (IC50) of 54.0 +/- 20.5 micromol/L (n = 6), and AVP dependent contractions with an IC50 of 31.6 +/- 5.0 micromol/L (n = 8). In conclusion, (-)-cicletanine antagonizes AII more effectively in rat kidney than in mesenteric vascular beds. Moreover, in rat kidney vascular beds (-)-cicletanine is more potent in blocking the pressor responses to AII than in blocking those to AVP. A selective blockade of AII induced contractions in kidney vascular beds can be one factor explaining both the greater antagonistic potency of (-)-cicletanine against AII compared with AVP in pithed rats, and the renal protective properties of cicletanine in both hypertensive and aged rats.


Subject(s)
Angiotensin II/pharmacology , Antihypertensive Agents/pharmacology , Arginine Vasopressin/pharmacology , Pyridines/pharmacology , Renal Circulation/drug effects , Splanchnic Circulation/drug effects , Angiotensin II/antagonists & inhibitors , Animals , Arginine Vasopressin/antagonists & inhibitors , In Vitro Techniques , Male , Rats , Rats, Wistar , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...