Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 320(1): 365-75, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17005916

ABSTRACT

Histamine H3 receptor inverse agonists are known to enhance the activity of histaminergic neurons in brain and thereby promote vigilance and cognition. 1-{3-[3-(4-Chlorophenyl)propoxy]propyl}piperidine, hydrochloride (BF2.649) is a novel, potent, and selective nonimidazole inverse agonist at the recombinant human H3 receptor. On the stimulation of guanosine 5'-O-(3-[35S]thio)triphosphate binding to this receptor, BF2.649 behaved as a competitive antagonist with a Ki value of 0.16 nM and as an inverse agonist with an EC50 value of 1.5 nM and an intrinsic activity approximately 50% higher than that of ciproxifan. Its in vitro potency was approximately 6 times lower at the rodent receptor. In mice, the oral bioavailability coefficient, i.e., the ratio of plasma areas under the curve after oral and i.v. administrations, respectively, was 84%. BF2.649 dose dependently enhanced tele-methylhistamine levels in mouse brain, an index of histaminergic neuron activity, with an ED50 value of 1.6 mg/kg p.o., a response that persisted after repeated administrations for 17 days. In rats, the drug enhanced dopamine and acetylcholine levels in microdialysates of the prefrontal cortex. In cats, it markedly enhanced wakefulness at the expense of sleep states and also enhanced fast cortical rhythms of the electroencephalogram, known to be associated with improved vigilance. On the two-trial object recognition test in mice, a promnesiant effect was shown regarding either scopolamine-induced or natural forgetting. These preclinical data suggest that BF2.649 is a valuable drug candidate to be developed in wakefulness or memory deficits and other cognitive disorders.


Subject(s)
Histamine Agonists/pharmacology , Histamine Antagonists/pharmacology , Piperidines/pharmacology , Receptors, Histamine H3/drug effects , Acetylcholine/metabolism , Animals , Cats , Dopamine/metabolism , Electroencephalography/drug effects , Guinea Pigs , Histamine Release/drug effects , Humans , Imidazoles/metabolism , Male , Methylhistamines/pharmacology , Mice , Mice, Inbred C57BL , Piperidines/pharmacokinetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Histamine H3/physiology , Scopolamine/pharmacology
2.
Cardiovasc Res ; 52(3): 387-96, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11738055

ABSTRACT

OBJECTIVE: Although it is believed that sodium-driven acid-base transport plays a central role in the development of the reperfusion injury that follows cardiac ischemia, research to date has demonstrated only a role for Na(+)/H(+) exchange (NHE). However, Na(+)-driven HCO(-)(3) transport, which is quantitatively as important as NHE in cardiac cells, has not been examined. METHODS AND RESULTS: Here the results show that a neutralizing antibody raised against the human heart electrogenic Na(+)/HCO(3)(-) cotransporter (hhNBC) blocked the recovery of pH after acidic pulse both in HEK-293 cells expressing hhNBC and in rat cardiac myocytes demonstrating the presence of an electrogenic NBC in rat cardiac myocytes similar to hhNBC. Administration of anti-NBC antibody to ischemic-reperfused rat hearts markedly protects systolic and diastolic functions of the heart during reperfusion. Furthermore, using a quantitative real-time RT-PCR (TaqMan) and Western blot analysis we demonstrated that in human cardiomyopathic hearts, mRNA and protein levels of hhNBC increase, whereas mRNA levels of the electroneutral Na(+)/HCO(3)(-) cotransporter (NBCn1) remain unchanged. CONCLUSION: Our data provide evidence that inhibition of hhNBC, whose role in cardiac pathologies could be amplified by overexpression, represents a novel therapeutic approach for ischemic heart disease.


Subject(s)
Antibodies, Monoclonal/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Sodium-Bicarbonate Symporters/physiology , Animals , Blotting, Western , Cell Line , Cells, Cultured , Gene Expression , Humans , Hydrogen-Ion Concentration , L-Lactate Dehydrogenase/analysis , Male , Perfusion , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Bicarbonate Symporters/immunology , Sodium-Bicarbonate Symporters/metabolism , Sodium-Hydrogen Exchangers/metabolism
3.
J Mol Cell Cardiol ; 33(9): 1589-606, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549339

ABSTRACT

Sphingosine 1 phosphate (S1P), an aminophospholipid, acts extracellularly as a ligand via the specific G protein-coupled receptors of the endothelial differentiation gene (EDG) 1, 3, 5, 6 and 8 receptors family and intracellularly as a second messenger in various cellular types. The aim of this work was to investigate biological activity of S1P in cardiomyocytes with respect to related sphingolipids. S1P was applied for 48 h on rat neonatal cardiomyocytes at 10 nM, 100 nM and 1 microM. S1P induced a concentration-dependent cellular hypertrophy evidenced by an increase in cell size, [3H]-phenylalanine incorporation, protein content and Brain Natriuretic Peptide (BNP) secretion. Among the lipids tested S1P exhibits the lower EC50 (67 nM) followed by dihydro-S1P (107 nM) and sphingosylphosphorylcholine (1.6 microM). The effect of S1P could be related to a stimulation of the EDG1 receptor since we showed that the EDG1 receptor is predominantly expressed at the mRNA and protein levels in rat cardiomyocytes and that specific anti-EDG1 antibodies inhibited the hypertrophic effect induced by S1P. Furthermore the expression level of most other EDG receptors for S1P appeared very low in cardiac myocytes. S1P (100 nM) increased the phosphorylation of p42/44MAPK, p38MAPK, JNK, Akt and p70(S6K), this effect being reversed by inhibitors of their respective phosphorylation which also rescue the hypertrophic phenotype. Finally, S1P stimulated actin stress fibre formation reverted by the Rho inhibitor, the C3 exoenzyme. Altogether, our results show that S1P induces cardiomyocyte hypertrophy mainly via the EDG1 receptor and subsequently via Gi through ERKs, p38 MAPK, JNK, PI3K and via Rho pathway.


Subject(s)
Cardiomegaly/pathology , Heart/drug effects , Immediate-Early Proteins/metabolism , Lysophospholipids , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cells, Cultured , Culture Media, Serum-Free , Dose-Response Relationship, Drug , Humans , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Myocardium/metabolism , Peptides/immunology , Peptides/metabolism , Phenylephrine/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Structure, Tertiary , Rats , Rats, Wistar , Receptors, Cell Surface/metabolism , Receptors, Lysophospholipid , Sphingosine/chemistry , Sphingosine/metabolism , Stress Fibers/metabolism , Tissue Extracts/chemistry , Virulence Factors, Bordetella/pharmacology
4.
Life Sci ; 68(7): 799-814, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11205871

ABSTRACT

Myocardial infarction (MI), leads to cardiac remodeling, thinning of the ventricle wall, ventricular dilation, and heart failure, and is a leading cause of death. Interactions between the contractile elements of the cardiac myocytes and the extracellular matrix (ECM) help maintain myocyte alignment required for the structural and functional integrity of the heart. Following MI, reorganization of the ECM and the myocytes occurs, contributing to loss of heart function. In certain pathological circumstances, the ECM is modulated such that the structure of the tissue becomes damaged. The matrix metalloproteinases (MMPs) are a family of enzymes that degrade molecules of the ECM. The present experiments were performed to define the time-course, isozyme subtypes, and cellular source of increased MMP expression that occurs following MI in an experimental rabbit model. Heart tissue samples from infarcted and sham animals were analyzed over a time-course of 1-14 days. By zymography, it was demonstrated that, unlike the sham controls, MMP-9 expression was induced within 24 hours following MI. MMP-3 expression, also absent in sham controls, was induced 2 days after MI. MMP-2 expression was detected in both the sham and infarcted samples and was modestly up-regulated following MI. Tissue inhibitor of metalloproteinase-1 (TIMP-1) expression was evaluated and shown to be down-regulated following MI, inverse of MMP-9 and MMP-3 expression. Further, MMP-9 and MMP-3 expression was detected by immunohistochemistry in myocytes within the infarct. Additional studies were conducted in which cultured rat cardiac myocytes were exposed to a hypoxic environment (2% O2) for 24 hours and the media analyzed for MMP expression. MMP-9 and MMP-3 were induced following exposure to hypoxia. It is speculated that the net increase in proteolytic activity by myocytes is a contributing factor leading to myocyte misalignment and slippage. Additional studies with a MMP inhibitor would elucidate this hypothesis.


Subject(s)
Matrix Metalloproteinases/biosynthesis , Myocardial Infarction/enzymology , Myocardium/metabolism , Animals , Blotting, Western , Cell Hypoxia , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression Regulation, Enzymologic , Immunohistochemistry , Matrix Metalloproteinase 3/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinases/genetics , Myocardial Infarction/pathology , Myocardial Ischemia/metabolism , Myocardium/cytology , Proteins/chemistry , Rabbits , Tissue Inhibitor of Metalloproteinase-1/metabolism
5.
J Mol Biol ; 294(2): 443-56, 1999 Nov 26.
Article in English | MEDLINE | ID: mdl-10610770

ABSTRACT

Mutations in human cardiac myosin-binding protein C (cMyBP-C) gene are associated with familial hypertrophic cardiomyopathy (FHC), and most of them are predicted to produce COOH-truncated proteins. To understand the molecular mechanism(s) by which such mutations cause FHC, we analyzed (i) the accumulation of human cMyBP-C mutants in fetal rat cardiomyocytes, and (ii) the protein sequence of the human wild-type (wt) cMyBP-C by hydrophobic cluster analysis with the aim of identifying new putative myosin-binding site(s). Accumulation and sarcomeric localization of the wt protein and of four FHC-mutant cMyBP-Cs (E542Q and three COOH-truncated proteins) were studied in cardiomyocytes by immunostaining and confocal microscopy after transfection with myc-tagged constructs. We found that: (i) 10 % of the cells expressing COOH-truncated mutants exhibit an incorporation into the A-band of the sarcomere without any alteration of the myofibrillar architecture versus 76 % of those expressing the wt or E542Q mutant cMyBP-Cs (p<0.001); (ii) 90 % of the cells expressing the truncated mutants show a diffuse localization of these proteins in the cardiomyocytes, out of which 45 % exhibit a significant alteration of the sarcomeric structure (p<0.0001 versus wt); and (iii) the two shortest mutant cMyBP-Cs accumulate at very low levels in fetal rat cardiomyocytes as compared to the wt (p<0.008). Protein sequence analysis indicated that a 45-residue sequence in the NH2-terminal C0 domain of cMyBP-C exhibits a consistent homology (sequence similarity score of 42 %) with a segment of the NH2-terminal domain of myomesin, another myosin-binding protein. This result suggests that the C0 domain of human cMyBP-C contains a novel putative myosin-binding site that could account for the A-band incorporation of the truncated mutants. In addition, the faint accumulation and the diffuse localization of truncated mutants could probably be explained by a low affinity of the C0 domain for myosin. We conclude that COOH-truncated cMyBP-Cs may act as poison polypeptides that disrupt the myofibrillar architecture and result in the defects observed in FHC.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Myocardium/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , COS Cells/metabolism , Cells, Cultured , Connectin , Gene Expression , Heart/embryology , Humans , Molecular Sequence Data , Muscle Proteins/metabolism , Mutation , Myocardium/cytology , Myosins/metabolism , Rats , Rats, Wistar , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sarcomeres/metabolism , Sequence Analysis, Protein
6.
J Mol Cell Cardiol ; 31(4): 705-19, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10329199

ABSTRACT

The predominant response of the heart to sustained increased work load is development of ventricular hypertrophy, principally as a result of hypertrophy of cardiomyocytes. The molecular mechanisms and factors involved in cardiomyocyte hypertrophy are poorly understood. Myotrophin is a novel 12-kilodalton protein recently implicated as a factor associated with and able to induce cardiac hypertrophy. Cloning of rat myotrophin revealed that this protein is identical to the functionally undefined rat, murine and chicken V-1 proteins. Although human myotrophin has been purified to homogeneity, its gene has not been characterized. In this report we describe the cloning, expression, purification and characterization of the human homolog of myotrophin/V-1 protein. Sequence analysis indicators high homology (>90%) between all species at both the nucleotide and amino acid levels, and Southern blot analysis of genomic DNA from diverse species verifies that myotrophin/V-1 is a highly conserved gene. Northern analysis indicates wide-spread expression of a single human transcript, and examination of mRNA distribution in 50 human tissues by dot blot analysis indicates ubiquitous expression with relatively high expressioon in adult and fetal heart. We verify that recombinant human myotrophin produces cardiomyocyte hypertrophy, and we demonstrate for the first time that elevated levels of myotrophin/V-1 protein mRNA are expressed in human dilated cardiomyopathic hearts. We report the novel findings that myotrophin expression is elevated in ischemic hearts, and that myotrophin expression correlates positively with ventricular mass in a hypoxic rat model of induced right ventricular hypertrophy.


Subject(s)
Cardiomegaly/genetics , DNA, Complementary/genetics , Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Amino Acid Sequence , Animals , Base Sequence , Cardiomegaly/metabolism , Cell Size/drug effects , Cells, Cultured , Cloning, Molecular , DNA Primers/genetics , Disease Models, Animal , Gene Expression , Growth Substances/pharmacology , Humans , Hypertrophy, Right Ventricular/genetics , Male , Mice , Molecular Sequence Data , Myocardium/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tissue Distribution
7.
Protein Sci ; 8(12): 2570-9, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10631972

ABSTRACT

Protein phosphatase 1 (PP1) is widely distributed among tissues and species and acts as a regulator of many important cellular processes. By targeting the catalytic part of PP1 (PP1C) toward particular loci and substrates, regulatory subunits constitute key elements conferring specificity to the holoenzyme. Here, we report the identification of an (alpha/beta)8-barrel-like structure within the N-ter stretch of the human PP1 regulatory subunit hGM, which is part of the family of diverse proteins associated with glycogen metabolism. Protein homology modeling gave rise to a three-dimensional (3D) model for the 381 N-ter residue stretch of hGM, based on sequence similarity with Streptomyces olivochromogenes xylose isomerase, identified by using FASTA. The alignment was subsequently extended by using hydrophobic cluster analysis. The homology-derived model includes the putative glycogen binding area located within the 142-230 domain of hGM as well as a structural characterization of the PP1C interacting domain (segment 51-67). Refinement of the latter by molecular dynamics afforded a topology that is in agreement with previous X-ray studies (Egloff et al., 1997). Finite difference Poisson-Boltzmann calculations performed on the interacting domains of PP1C and hGM confirm the complementarity of the local electrostatic potentials of the two partners. This work highlights the presence of a conserved fold among distant species (mammalian, Caenorhabditis elegans, yeast) and, thus, emphasizes the involvement of PP1 in crucial basic cellular functions.


Subject(s)
Glycogen/metabolism , Phosphoprotein Phosphatases/chemistry , Amino Acid Motifs , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Protein Phosphatase 1 , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis, Protein , Static Electricity
8.
FEBS Lett ; 439(3): 224-30, 1998 Nov 20.
Article in English | MEDLINE | ID: mdl-9845327

ABSTRACT

Regulation of the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA 2a) depends on the phosphorylation state of phospholamban (PLB). When PLB is phosphorylated, its inhibitory effect towards SERCA 2a is relieved, leading to an enhanced myocardial performance. This process is reversed by a sarcoplasmic reticulum (SR)-associated type 1 protein phosphatase (PP1) composed of a catalytic subunit PP1C and a regulatory subunit GM. Human GM and PLB have been produced in an in vitro transcription/translation system and used for co-immunoprecipitation and biosensor experiments. The detected interaction between the two partners suggests that cardiac PPI is targeted to PLB via GM and we believe that this process occurs with the identified transmembrane domains of the two proteins. Thus, the interaction between PLB and GM may represent a specific way to modulate the SR function in human cardiac muscle.


Subject(s)
Calcium-Binding Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Amino Acid Sequence , Binding Sites , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Calcium-Transporting ATPases/metabolism , Humans , Membrane Proteins/metabolism , Molecular Sequence Data , Phosphorylation , Precipitin Tests , Protein Binding , Protein Conformation , Protein Phosphatase 1 , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/physiology
9.
J Mol Cell Cardiol ; 30(10): 1945-54, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9799649

ABSTRACT

Tautomycin (TT) and calyculin A (CyA) are inhibitors of protein phosphatases type 1 and 2 (PP1, PP2). Inhibitors 1 and 2 are specific for PP1, which is the major phosphatase functionally relevant in heart and able to dephosphorylate phospholamban (PLB). TT and CyA maintain PLB in its phosphorylated state, thereby increasing calcium uptake. Rabbit saponin skinned fibers (SF) are used to assess calcium load of the sarcoplasmic reticulum (SR). The present investigation aimed to examine the effects of PP1 inhibitors on SR calcium load assessed by caffeine-induced tension transient (CITT), and to correlate this activity with the PLB phosphorylation state. TT and CyA (100 nm) applied during the uptake phase increased the amplitude of CITT by 10 and 20%, respectively,P<0.05 without effect on the release phase. Both CyA and TT were devoid of calcium sensitizing effect when studied on Triton X-100 SF. After skinning procedure, SF were grinded for biochemical studies. SDS-PAGE electrophoresis and immunoblots using a monoclonal PLB antibody showed that cAMP or Ca2+/calmodulin-dependent protein kinases phosphorylated PLB in an additive fashion. Inhibition of PP1 by inhibitor 1, CyA and TT maintained PLB in its phosphorylated state in a dose-dependent manner. The results of this study in which functional and biochemical experiments in cardiac SF were combined demonstrate that strong correlation exists between the phosphorylation-dephosphorylation cycle of PLB and calcium uptake.


Subject(s)
Enzyme Inhibitors/pharmacology , Muscle Fibers, Skeletal/physiology , Myocardial Contraction/physiology , Oxazoles/pharmacology , Papillary Muscles/physiology , Phosphoprotein Phosphatases/antagonists & inhibitors , Pyrans , Spiro Compounds , Animals , Antifungal Agents/pharmacology , Caffeine/pharmacology , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Contractile Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Egtazic Acid/pharmacology , In Vitro Techniques , Macromolecular Substances , Marine Toxins , Muscle Fibers, Skeletal/drug effects , Myocardial Contraction/drug effects , Papillary Muscles/drug effects , Protein Phosphatase 1 , Rabbits , Structure-Activity Relationship
10.
Cardiovasc Drugs Ther ; 12(6): 585-94, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10410828

ABSTRACT

LND-623 is a new aminosteroid analog of ouabain, with a greater separation between efficacy and toxicity than ouabain. To determine its mechanism of action, we studied its biochemical and physiological effects on human red blood cell sodium transports on different cellular structures regarded as sites of contractile control, and we compared its relative efficacy to ouabain in rat heart preparations and membrane-bound Na, K-ATPase isoenzymes. The response to ouabain was evaluated in Langendorff-perfused hearts and on purified membrane-bound Na, K-ATPase. LND-623 is 6.8-fold more efficient than ouabain in inhibiting the human Na+ pump (IC50 = 0.098 +/- 0.001 microM vs. 0.67 +/- 0.02 microM); (P < 0.0001). LND-623 had no effect on the following cellular functions: Na-Ca exchange, Na-K cotransport, Ca-ATPase, slow calcium channels, adenylate cyclase system, phosphodiesterase, and calcium sensitivity of the contractile protein system. The dose-response curve for the positive inotropic and inhibitory effects on rat cardiac isoenzymes produced by LND-623 were clearly biphasic. The amplitude of the maximum inotropic effect, without any toxic effect, was up to three-fold higher with LND-623 than with the same maximum dose of ouabain used. The strong positive inotropic effect of LND-623 in rats could be related to a specific inhibition of the two rat cardiac isoforms of the Na, K-ATPase.


Subject(s)
Cardiotonic Agents/pharmacology , Isoenzymes/metabolism , Mannosides/pharmacology , Myocardial Contraction/drug effects , Pregnanes/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Adenylyl Cyclases/metabolism , Animals , Dogs , Erythrocytes/drug effects , Erythrocytes/enzymology , Humans , In Vitro Techniques , Male , Membranes/drug effects , Membranes/enzymology , Myofibrils/enzymology , Ouabain/pharmacology , Phosphoric Diester Hydrolases/metabolism , Rats , Rats, Wistar , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/blood , Sodium-Potassium-Exchanging ATPase/drug effects
11.
Eur J Biochem ; 247(3): 801-9, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9288900

ABSTRACT

The Ca2+ uptake by the sarcoplasmic reticulum (SR) can be affected by direct modulation of the Ca2+ pump or by removing the inhibitory effect of dephosphorylated phospholamban. The effect of these mechanisms was assessed using ellagic acid and 1-(3,4-dimethoxyphenyl)-3-dodecanone. Both compounds (30 micromol/l) enhanced SR-Ca2+ uptake in rabbit cardiomyocytes by 65.3 +/- 13% and 44.3 +/- 6.7% for 1-(3,4-dimethoxyphenyl)-3-dodecanone and ellagic acid, respectively (at pCa 6.2). A similar effect was observed in cardiac SR microsomes (59.5 +/- 7.4% and 45.1 +/- 6.7) with 30 micromol/l 1-(3,4-dimethodoxyphenyl)-3-dodecanone and ellagic acid, respectively. 1-(3,4-Dimethoxyphenyl)-3-dodecanone increased Ca2+ storage by cardiac SR microsomes mainly at high [Ca2+] with a 57% increase of Vmax, whereas ellagic acid increased Vmax to a smaller extent (22%) and stimulated Ca2+ uptake at lower [Ca2+] with a leftward-shift of the pCa/ATPase relationship by pCa 0.24. Ellagic acid also differed from 1-(3,4-dimethoxylphenyl)-3-dodecanone in that it produced a Ca2+ sensitizing effect only in cardiac SR microsomes (by pCa 0.3) whereas 1-(3,4-dimethoxyphenyl)-3-dodecanone stimulated the ATPase, at saturating Ca2+, in both cardiac and skeletal muscle SR vesicles. It is suggested that 1-(3,4-dimethoxyphenyl)-3-dodecanone stimulates directly the Ca2+-ATPase activity, in contrast to ellagic acid which enhances the cardiac SR-Ca2+ uptake by interacting with phospholamban, as confirmed by the lack of additive effect between ellagic acid and monoclonal antibodies raised against phospholamban. 1-(3,4-dimethoxyphenyl)-3-dodecanone and ellagic acid constitute attractive pharmacological tools to investigate the functional consequences of enhancing SR Ca2+, uptake by affecting different mechanisms.


Subject(s)
Anisoles/pharmacology , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Ellagic Acid/pharmacology , Ketones/pharmacology , Sarcoplasmic Reticulum/drug effects , Animals , Dogs , In Vitro Techniques , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Rabbits , Sarcoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum/metabolism
12.
J Recept Signal Transduct Res ; 17(1-3): 137-47, 1997.
Article in English | MEDLINE | ID: mdl-9029486

ABSTRACT

Three isoenzymes of digitalis receptors (alpha 1, alpha 2, alpha 3) in the brain and only one in the kidney (alpha 1) can be distinguished by their ouabain affinities and their responsiveness to sodium. Since we have reported modulations for these digitalis receptors by their fatty acid membrane environment, anaesthesics could bind on and modulate either directly these receptors or indirectly by disturbing membrane lipids. The aim of this study was to evaluate this anaesthetic action on apparent ouabain affinities and sodium dependence of cerebral and renal Na+, K(+)-ATPase isoenzymes activities. Rat brain and kidney membrane fractions with pentobarbital-induced anaesthetized state were compared to an unanaesthetized state for their (1) fatty acid composition of total membrane phospholipids, (2) responsiveness to ouabain and (3) Na+ dependence of digitalis receptors. An anaesthesia period of 10 minutes induced (1) a fatty acid modification of brain membranes and (2) a significant sensibilization to ouabain for the alpha 2 and alpha 3 isoforms of digitalis receptors (alpha 2, IC50; 8.2 +/- 0.5 x 10(-7) mol/l vs 4.5 +/- 0.2 x 10(-7) mol/l; alpha 3, IC50; 6.0 +/- 0.3 x 10(-8) mol/l vs 2.5 +/- 0.1 x 10(-8). In contrast, the ouabain affinity of the alpha 1 subunit expressed in kidney and brain membranes was unaltered. No anaesthetic effect was observed on the Na+ dependence of the alpha 1 isoenzyme in the brain (4 mmol/l) and the kidney (8 mmol/l). Pentobarbital induced a desensibilization for alpha 2-receptors (8.3 +/- 0.5 vs 16.0 +/- 1.4 mmol/l Na+) and a sensibilization for alpha 3-receptors (14.4 +/- 0.8 vs 10 +/- 1.3 mmol/l Na+). These altered properties could be related to a selective modification of the fatty acid composition and/or to the presence of a specific binding site for pentobarbital on these two neuronal digitalis receptors.


Subject(s)
Anesthesia , Neurons/enzymology , Pentobarbital/pharmacology , Sodium-Potassium-Exchanging ATPase/drug effects , Animals , Isoenzymes/metabolism , Male , Neurons/drug effects , Ouabain/metabolism , Rats , Rats, Wistar , Sodium/metabolism
13.
FEBS Lett ; 348(1): 55-60, 1994 Jul 04.
Article in English | MEDLINE | ID: mdl-8026584

ABSTRACT

The expression of the canine alpha 2 and 3 subunit isoenzymes of NA,K-ATPase has been investigated in plasma membranes isolated from dog heart, brain and kidney by immunoblotting, employing polyclonal anti rat fusion protein, and enzymological techniques. Western blot analysis revealed with purified dog membrane Na,K-ATPase preparations, one immunoreactive signal with rat specific alpha 3 antisera in cardiac tissues, and two immunoreactive signals with rat alpha 2 and alpha 3 antisera in cerebral tissues. These findings suggested the specific expression of alpha 3 polypeptide in dog heart (99 kDa), whereas dog brain expressed the alpha 2 and 3 polypeptides. The stained bands were superimposed. The antibody to rat brain alpha 1 fusion protein did not cross-react with dog antigens whatever the three tissues tested. Expression of the alpha 3-subunit isoform in dog heart membranes was consistent with a high affinity digitoxigenin-sensitive class of Na,K-ATPase (IC50 = 7 +/- 2 nM). A single component with low affinity to digitoxigenin (IC50 = 110 +/- 10 nM) characterized the alpha 1 kidney form. The mixture of alpha 2 and alpha 3 isoforms in dog brain exhibited an apparent affinity for digitoxigenin (IC50 = 17 +/- 5 nM) lower than the heart. The sodium dependences of the high affinity digitoxigenin sites were for the cardiac alpha 3 form (K0.5 = 10 +/- 1.9 mM) and for the cerebral alpha 2 and alpha 3 mixture (K0.5 19.6 +/- 4.9 mM). The sensitivities for Na+ of the low affinity sites (alpha 1) were: 6.7 +/- 1.4 mM, 6.3 +/- 1.2 mM and 11.6 +/- 2.9 mM in heart, brain and kidney respectively. This is the first report of the catalytic characteristics of the alpha 3 subunit isoenzyme in canine cardiac plasma membranes.


Subject(s)
Isoenzymes/analysis , Myocardium/enzymology , Sodium-Potassium-Exchanging ATPase/analysis , Animals , Blotting, Western , Brain/enzymology , Digitoxigenin/pharmacology , Dogs , Female , Kidney/enzymology , Male , Microsomes/enzymology , Rats , Sodium/pharmacology , Sodium-Potassium-Exchanging ATPase/drug effects
14.
FEBS Lett ; 330(3): 297-301, 1993 Sep 20.
Article in English | MEDLINE | ID: mdl-8397103

ABSTRACT

Two functional isoforms alpha (alpha 1) and alpha+ (alpha 3) of the Na,K-ATPase catalytic subunit coexist in canine cardiac myocytes [J. Biol. Chem. (1987) 262, 8941-8943]. The in vitro turnover rates of ATP hydrolysis have been determined in sarcolemma preparations by comparing [3H]ouabain-binding and Na,K-ATPase activity at various doses of ouabain (0.3-300 nM). The correlation between the occupancy of the ouabain-binding sites and the degree of Na,K-ATPase inhibition was not linear. The results showed that the form of low-affinity for ouabain (Kd = 300-700 nM) exhibited a lower turnover rate (88 +/- 10 vs. 147 +/- 15 molecules of ATP hydrolyzed per second per ouabain-binding site) than the high affinity form (Kd = 1-8 nM). Thus our results indicate this specific isoform kinetic difference could contribute to differences in the cardiac cellular function.


Subject(s)
Isoenzymes/metabolism , Myocardium/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Dogs , Humans , Hydrolysis , Ouabain/metabolism , Phosphorylation
15.
J Recept Res ; 13(7): 1083-92, 1993.
Article in English | MEDLINE | ID: mdl-8396181

ABSTRACT

Cordil-LND796 is a new cardiotonic glycoside under development. In rat brain microsomes where three isoforms of the Na,K-ATPase with differential affinities for cardiac glycosides have been identified, Cordil had higher affinity for the alpha 3 (IC50 = 0.02 microM) than for the alpha 2 (IC50 = 0.6 microM) and the alpha 1 (IC50 = 30 microM) isozymes. Cordil is potentially a selective inhibitor for both alpha 2 and alpha 3 Na,K-ATPase isoforms. Using inside out vesicles we have shown that Cordil binds to and inhibits Na,K-ATPase at an extracellular site. The dissociation kinetic rates (k-1) from the ATPase and the phosphatase activity (K-dependent dephosphorylation) of the Na,K-ATPase were similar for Cordil. Despite these similarities to ouabain comparison of the kinetics of the Na,K-ATPase inhibition by ouabain and Cordil revealed marked differences in their association rates (k+1 = 0.7 l mol-1 min-1 and k+1 = 6 x 10(-3) l mol-1 min-1 respectively) and their dissociation rates (k-1 = 1.3 +/- 0.2 x 10(-4) s-1 and k-1 = 69 +/- 7 x 10(-4) s-1 respectively). Both binding association and dissociation rates were enhanced for Cordil. These data are compatible with a stabilizing effect of Cordil on the E2P conformational state of Na,K-ATPase.


Subject(s)
Cardiotonic Agents/pharmacology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Animals , Cardiotonic Agents/metabolism , Dogs , Ouabain/metabolism , Phosphorylation , Potassium/physiology , Protein Conformation , Rats , Steroids
16.
Arzneimittelforschung ; 42(11): 1301-5, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1337248

ABSTRACT

20R 14 beta-amino 3 beta-rhamnosyl 5 beta-pregnan 20 beta-ol (LND 623, CAS 90520-42-6) was investigated and compared to digoxin in anesthetized dogs. The hemodynamic profiles showed: a) a pure positive inotropic action of LND-623; b) its potency was four-fold higher than that of digoxin and more marked in heart failure; c) its duration of action was maintained for at least 6 h. The onset and reversal of the inotropic effects of a single dose (3.3 nmol.kg-1.min-1) were faster with LND-623 than those of digoxin. This reversal is consistent with the faster dissociation profile observed at the level of the high affinity cardiac Na+,K(+)-ATPase receptor form. The advantage of LND-623 over digoxin resides in its larger therapeutic index (ratio of arrhythmogenic to inotropic responses) in anesthetized dogs with propranolol-induced heart failure. This index was 6 for LND-623 and 2 for digoxin.


Subject(s)
Heart Failure/drug therapy , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Anesthesia , Animals , Cardiotonic Agents/pharmacology , Digoxin/pharmacology , Dogs , Female , Heart Failure/physiopathology , Hemodynamics/drug effects , Male , Mannosides/pharmacology , Myocardial Contraction/drug effects , Pregnanes/pharmacology , Propranolol/pharmacology
18.
FEBS Lett ; 281(1-2): 145-8, 1991 Apr 09.
Article in English | MEDLINE | ID: mdl-1849836

ABSTRACT

Serine-specific reagents, anticholinesterase organophosphorus compounds like Vx provoke, in the micromolar range, digitalis-like ventricular arrhythmias of non-cholinergic origin in rodent hearts. The sensitivities of the two rat cardiac Na+,K(+)-ATPase isoforms (alpha 1 and alpha 2) to Vx (0.1-100 microM) were measured in sarcolemma vesicles. At 1 microM Vx, the inhibition of the total activity averaged 18% but never exceeded 75% with 100 microM. When the alpha 2 isoform activity was inhibited by 0.1 microM ouabain, alpha 1 was 35% inhibited by 1 microM Vx, i.e. a 16 +/- 4% inhibition of the total activity. The cardiac alpha 1 being related to the digitalis-induced toxicity, its selective inhibition by a micromolar dose of Vx fully accounts for the cardiotoxicity of Vx. Inasmuch as Vx had no effect on the rat kidney alpha 1, differentially inactivated the cardiac isozymes and specifically reacted with serine residues, the putative binding-site(s) of the organophosphorus compound on the Na+-K(+)-ATPase molecules has been considered.


Subject(s)
Heart/drug effects , Isoenzymes/isolation & purification , Organothiophosphorus Compounds/pharmacology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Animals , Cholinesterase Inhibitors/pharmacology , Kinetics , Myocardium/pathology , Organothiophosphorus Compounds/toxicity , Rats , Rats, Inbred Strains
19.
Eur J Biochem ; 196(1): 129-33, 1991 Feb 26.
Article in English | MEDLINE | ID: mdl-1848182

ABSTRACT

Guinea pig left ventricular muscle contains two distinct molecular forms of the Na+/K(+)-ATPase catalytic alpha subunit. Sarcolemmal vesicles highly enriched in Na+/K(+)-ATPase were isolated by a new procedure that yielded specific activities of 60-100 mumol Pi.h-1.mg-1. SDS/PAGE of isolated sarcolemma after reduction and alkylation of the sulfhydryl groups and identification on immunoblots with specific anti-(alpha subunit) antibodies indicated the presence of two major polypeptides of 100 kDa and 103 kDa, respectively. The two alpha subunits were functional: the dose/response curves of Na+/K(+)-ATPase activity with ouabain, dihydroouabain and digitoxigenin were biphasic, revealing the presence of high-affinity [concentration of drug causing 50% inhibition (IC50) = 10 nM] and low-affinity (IC50 = 2 microM) forms with proportional contributions of 55% and 45%, respectively. The involvement of the high-affinity form in the positive inotropic effect of digitalis and of the low-affinity sites in both inotropy and toxicity are consistent with the literature data on rodents.


Subject(s)
Isoenzymes/isolation & purification , Myocardium/enzymology , Sodium-Potassium-Exchanging ATPase/isolation & purification , Animals , Electrophoresis, Polyacrylamide Gel , Guinea Pigs , Sodium-Potassium-Exchanging ATPase/analysis , Sodium-Potassium-Exchanging ATPase/immunology
20.
Pflugers Arch ; 417(3): 247-54, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2177181

ABSTRACT

The contribution of the heterogeneous digitalis receptors to the inotropic effect of ouabain was studied in hypertrophied rat hearts (aortic stenosis) by using isolated Langendorff heart preparations. Development and washout of the biological effects as well as the dose/response curves revealed two inotropic components of high and low drug sensitivity. Maximal inotropy was observed with 100 microM ouabain in both control and hypertrophied rat hearts. The high-sensitivity component accounted for only one-third of the response in control hearts but for two-thirds in hypertrophied hearts. The respective apparent affinities (10-20 nM and 10-20 microM) of the two inotropic components found in isolated hearts were similar to those of the high- and low-affinity Na+,K(+)-ATPase activities detected in isolated cardiac sarcolemma. Onset and reversion of the pharmacological effects of ouabain occurred at respective rates that were similar to those of the association and dissociation of ouabain to the Na+,K(+)-ATPase level. In hypertrophied heart, the high- and low-sensitivity components (or receptors) reacted seven- and threefold, respectively, more slowly than the corresponding sites in normal hearts. These alterations in inotropic responsiveness and properties of the digitalis receptors in cardiac hypertrophy suggest that new regulations should be taken into account in the adaptation to pressure overload.


Subject(s)
Cardiomegaly/physiopathology , Myocardial Contraction/drug effects , Ouabain/pharmacology , Animals , Dose-Response Relationship, Drug , In Vitro Techniques , Kinetics , Male , Ouabain/administration & dosage , Rats , Rats, Inbred Strains , Receptors, Drug/physiology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Stimulation, Chemical
SELECTION OF CITATIONS
SEARCH DETAIL
...