Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 180(24): 3254-3270, 2023 12.
Article in English | MEDLINE | ID: mdl-37522273

ABSTRACT

BACKGROUND AND PURPOSE: Guanylyl cyclase-A (GC-A), activated by endogenous atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), plays an important role in the regulation of cardiovascular and renal homeostasis and is an attractive drug target. Even though small molecule modulators allow oral administration and longer half-life, drug targeting of GC-A has so far been limited to peptides. Thus, in this study we aimed to develop small molecular activators of GC-A. EXPERIMENTAL APPROACH: Hits were identified through high-throughput screening and optimized by in silico design. Cyclic GMP was measured in QBIHEK293A cells expressing GC-A, GC-B or chimerae of the two receptors using AlphaScreen technology. Binding assays were performed in membrane preparations or whole cells using 125 I-ANP. Vasorelaxation was measured in aortic rings isolated from Wistar rats. KEY RESULTS: We have identified small molecular allosteric enhancers of GC-A, which enhanced ANP or BNP effects in cellular systems and ANP-induced vasorelaxation in rat aortic rings. The mechanism of action appears novel and not mediated through previously described allosteric binding sites. In addition, the selectivity and activity depend on a single amino acid residue that differs between the two similar receptors GC-A and GC-B. CONCLUSION AND IMPLICATIONS: We describe a novel allosteric binding site on GC-A, which can be targeted by small molecules to enhance ANP and BNP effects. These compounds will be valuable tools in further development and proof-of-concept of GC-A enhancement for the potential use in cardiovascular therapy.


Subject(s)
Atrial Natriuretic Factor , Guanylate Cyclase , Rats , Animals , Atrial Natriuretic Factor/pharmacology , Atrial Natriuretic Factor/metabolism , Guanylate Cyclase/metabolism , Rats, Wistar , Receptors, Atrial Natriuretic Factor/metabolism , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, Brain/pharmacology , Cyclic GMP/metabolism
2.
Int J Mol Sci ; 23(4)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35216259

ABSTRACT

Cardiac contractility is regulated by several neural, hormonal, paracrine, and autocrine factors. Amongst these, signaling through ß-adrenergic and serotonin receptors generates the second messenger cyclic AMP (cAMP), whereas activation of natriuretic peptide receptors and soluble guanylyl cyclases generates cyclic GMP (cGMP). Both cyclic nucleotides regulate cardiac contractility through several mechanisms. Phosphodiesterases (PDEs) are enzymes that degrade cAMP and cGMP and therefore determine the dynamics of their downstream effects. In addition, the intracellular localization of the different PDEs may contribute to regulation of compartmented signaling of cAMP and cGMP. In this review, we will focus on the role of PDEs in regulating contractility and evaluate changes in heart failure.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Heart Failure/metabolism , Signal Transduction/physiology , Animals , Humans , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Phosphoric Diester Hydrolases/metabolism , Second Messenger Systems/physiology
3.
Cardiovasc Res ; 118(6): 1506-1519, 2022 05 06.
Article in English | MEDLINE | ID: mdl-33970224

ABSTRACT

AIMS: Guanylyl cyclase-B (GC-B; natriuretic peptide receptor-B, NPR-B) stimulation by C-type natriuretic peptide (CNP) increases cGMP and causes a lusitropic and negative inotropic response in adult myocardium. These effects are not mimicked by NPR-A (GC-A) stimulation by brain natriuretic peptide (BNP), despite similar cGMP increase. More refined methods are needed to better understand the mechanisms of the differential cGMP signalling and compartmentation. The aim of this work was to measure cGMP near proteins involved in regulating contractility to understand compartmentation of cGMP signalling in adult cardiomyocytes. METHODS AND RESULTS: We constructed several fluorescence resonance energy transfer (FRET)-based biosensors for cGMP subcellularly targeted to phospholamban (PLB) and troponin I (TnI). CNP stimulation of adult rat cardiomyocytes increased cGMP near PLB and TnI, whereas BNP stimulation increased cGMP near PLB, but not TnI. The phosphodiesterases PDE2 and PDE3 constrained cGMP in both compartments. Local receptor stimulation aided by scanning ion conductance microscopy (SICM) combined with FRET revealed that CNP stimulation both in the t-tubules and on the cell crest increases cGMP similarly near both TnI and PLB. In ventricular strips, CNP stimulation, but not BNP, induced a lusitropic response, enhanced by inhibition of either PDE2 or PDE3, and a negative inotropic response. In cardiomyocytes from heart failure rats, CNP increased cGMP near PLB and TnI more pronounced than in cells from sham-operated animals. CONCLUSION: These targeted biosensors demonstrate that CNP, but not BNP, increases cGMP near TnI in addition to PLB, explaining how CNP, but not BNP, is able to induce lusitropic and negative inotropic responses.


Subject(s)
Biosensing Techniques , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain , Natriuretic Peptide, C-Type , Animals , Atrial Natriuretic Factor/pharmacology , Cyclic GMP/metabolism , Endoplasmic Reticulum/metabolism , Guanylate Cyclase/metabolism , Myocardial Contraction , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, C-Type/metabolism , Rats , Receptors, Atrial Natriuretic Factor/metabolism , Troponin I
4.
Commun Biol ; 2: 394, 2019.
Article in English | MEDLINE | ID: mdl-31701023

ABSTRACT

Several FRET (fluorescence resonance energy transfer)-based biosensors for intracellular detection of cyclic nucleotides have been designed in the past decade. However, few such biosensors are available for cGMP, and even fewer that detect low nanomolar cGMP concentrations. Our aim was to develop a FRET-based cGMP biosensor with high affinity for cGMP as a tool for intracellular signaling studies. We used the carboxyl-terminal cyclic nucleotide binding domain of Plasmodium falciparum cGMP-dependent protein kinase (PKG) flanked by different FRET pairs to generate two cGMP biosensors (Yellow PfPKG and Red PfPKG). Here, we report that these cGMP biosensors display high affinity for cGMP (EC50 of 23 ± 3 nM) and detect cGMP produced through soluble guanylyl cyclase and guanylyl cyclase A in stellate ganglion neurons and guanylyl cyclase B in cardiomyocytes. These biosensors are therefore optimal tools for real-time measurements of low concentrations of cGMP in living cells.


Subject(s)
Biosensing Techniques/methods , Cyclic GMP/analysis , Myocytes, Cardiac/metabolism , Neurons/metabolism , Animals , Computer Systems , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/chemistry , Cyclic GMP-Dependent Protein Kinases/metabolism , Fluorescence Resonance Energy Transfer/methods , Guanylate Cyclase/metabolism , HEK293 Cells , Humans , Male , Models, Molecular , Plasmodium falciparum/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Rats, Wistar , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis , Soluble Guanylyl Cyclase/metabolism
5.
J Mol Cell Cardiol ; 131: 132-145, 2019 06.
Article in English | MEDLINE | ID: mdl-31009605

ABSTRACT

Although only ß2-adrenergic receptors (ßAR) dually couple with stimulatory G protein (Gs) and inhibitory G protein (Gi), inactivation of Gi enhances both ß1AR and ß2AR responsiveness. We hypothesize that Gi restrains spontaneous adenylyl cyclase (AC) activity independent of receptor activation. Subcellular localization of the AC5/6 subtypes varies contributing to the compartmentation of ßAR signaling. The primary objectives were to determine: (1) if ß1AR-mediated inotropic responses were dependent upon either AC5 or AC6; (2) if intrinsic Gi inhibition is AC subtype selective and (3) the role of phosphodiesterases (PDE) 3/4 to regulate ß1AR responsiveness. ß1AR-mediated increases in contractile force and cAMP accumulation in cardiomyocytes were measured from wild type, AC5 and AC6 knockout (KO) mice, with or without pertussis toxin (PTX) pretreatment to inactivate Gi and/or after selective inhibition of PDEs 3/4. Noradrenaline potency at ß1ARs was increased in AC6 KO. PDE4 inhibition increased noradrenaline potency in wild type and AC5 KO, but not AC6 KO. PTX increased noradrenaline potency only in wild type but increased the maximal ß1AR response in all mouse strains. PDE3 inhibition increased noradrenaline potency only in AC5 KO that was treated prior with PTX. ß1AR-evoked cAMP accumulation was increased more by PDE4 inhibition than PDE3 inhibition in wild type and AC5 KO that was amplified by Gi inhibition. These data indicate that ß1AR-mediated inotropic responses are not dependent upon either AC5 or AC6 alone. Inactivation of Gi enhanced ß1AR-mediated inotropic responses despite not coupling to Gi, consistent with Gi exerting a tonic receptor independent inhibition upon AC5/6. PDE4 seems the primary regulator of ß1AR signaling through AC6 in wild type. AC6 KO results in a reorganization of ß1AR compartmentation characterized by signaling through AC5 regulated by Gi, PDE3 and PDE4 that maintains normal contractile function.


Subject(s)
Adenylyl Cyclases/metabolism , Protein Isoforms/metabolism , Receptors, Adrenergic, beta-1/metabolism , Animals , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Norepinephrine/pharmacology , Pertussis Toxin/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Eur J Pharmacol ; 812: 174-183, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28697992

ABSTRACT

We have previously shown that the natriuretic peptide receptor B (NPR-B) agonist C-type natriuretic peptide (CNP) enhances cyclic adenosine 3´,5´-monophosphate (cAMP)-mediated signaling in failing hearts, through cyclic guanosine 3´,5´-monophosphate (cGMP)-mediated phosphodiesterase (PDE) 3 inhibition. As several signaling pathways are importantly changed in failing hearts, it could not be taken for granted that this crosstalk would be the same in non-failing hearts. Thus, we wanted to clarify to which extent this effect of CNP occurred also in non-failing hearts. Inotropic and lusitropic responses were measured in muscle strips and cGMP levels, localized cAMP levels, cAMP-PDE activity and mRNA levels were analyzed in isolated cardiomyocytes from left ventricles of non-failing and failing rat hearts. CNP increased cGMP and enhanced ß1- and ß2-adrenoceptor-mediated inotropic and ß1-adrenoceptor-mediated lusitropic responses, in non-failing and failing hearts. The NPR-A agonist brain natriuretic peptide (BNP) increased cGMP, but did not affect inotropic or lusitropic responses, indicating different compartmentation of cGMP from the two natriuretic peptide receptors. cAMP-PDE activity of PDE3 was concentration-dependently inhibited by cGMP with the same potency and to the same extent in non-failing and failing cardiomyocytes. CNP enhanced ß1-adrenoceptor-induced cAMP increase in living cardiomyocytes in the absence, but not in the presence of a PDE3 inhibitor indicating involvement of PDE3. In summary, CNP sensitizes cAMP-mediated signaling in non-failing as in failing hearts, via NPR-B-mediated increase of cGMP that inhibits the cAMP-PDE activity of PDE3.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Heart Failure/pathology , Natriuretic Peptide, C-Type/pharmacology , Phosphodiesterase 3 Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Heart Failure/metabolism , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism
7.
Biochem Biophys Res Commun ; 477(1): 14-19, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27237977

ABSTRACT

In cardiac tissue, regulatory light chain (RLC, myosin light chain 2) phosphorylation (Ser(15)) leads to modulation of muscle contraction through Ca(2+)-sensitization. To elucidate which kinases that are involved in the basal (diastolic phase) RLC phosphorylation, we studied non-contracting adult rat cardiomyocytes. RLC kinase activities in situ were unmasked by maximally inhibiting myosin light chain phosphatase (MLCP) by calyculin A in the absence and presence of various protein kinase inhibitors. Surprisingly MLCK did not contribute to the phosphorylation of RLC in the non-contracting cardiomyocytes. Two kinase activity groups were revealed by different sensitivities to staurosporine. The fraction with the highest sensitivity to staurosporine was inhibited by KN-93, a selective CaMKII inhibitor, producing a 23% ± 7% reduction in RLC phosphorylation. Calmodulin antagonism (W7) and reduction in Ca(2+) (EGTA) combined with low concentration of staurosporine caused a larger decrease in RLC phosphorylation than staurosporine alone. These data strongly suggest that in addition to CaMKII, there is another Ca(2+)/calmodulin-dependent kinase and a Ca(2+)/calmodulin-independent kinase phosphorylating RLC. Thus the RLC phosphorylation seems to be ensured by redundant kinase activities.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocytes, Cardiac/enzymology , Protein Kinases/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calmodulin/metabolism , Male , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar
8.
Biochem Biophys Res Commun ; 471(1): 219-25, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26809094

ABSTRACT

The aim was to identify kinase activities involved in the phosphorylation of regulatory light chain (RLC) in situ in cardiomyocytes. In electrically stimulated rat cardiomyocytes, phosphatase inhibition by calyculin A unmasked kinase activities evoking an increase of phosphorylated RLC (P-RLC) from about 16% to about 80% after 80 min. The phosphorylation rate in cardiomyocytes was reduced by about 40% by the myosin light chain kinase (MLCK) inhibitor, ML-7. In rat ventricular muscle strips, calyculin A induced a positive inotropic effect that correlated with P-RLC levels. The inotropic effect and P-RLC elevation were abolished by ML-7 treatment. The kinase activities phosphorylating RLC in cardiomyocytes were reduced by about 60% by the non-selective kinase inhibitor staurosporine and by about 50% by the calmodulin antagonist W7. W7 eliminated the inhibitory effect of ML-7, suggesting that the cardiac MLCK is Ca(2+)/calmodulin (CaM)-dependent. The CaM-dependent kinase II (CaMKII) inhibitor KN-93 attenuated the calyculin A-induced RLC phosphorylation by about 40%, indicating a contribution from CaMKII. The residual phosphorylation in the presence of W7 indicated that also CaM-independent kinase activities might contribute. RLC phosphorylation was insensitive to protein kinase C inhibition. In conclusion, in addition to MLCK, CaMKII phosphorylates RLC in cardiomyocytes. Involvement of other kinases cannot be excluded.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Animals , Cells, Cultured , Enzyme Activation , Gene Expression Regulation/physiology , Male , Phosphorylation/physiology , Rats , Rats, Wistar
10.
J Pharmacol Exp Ther ; 350(3): 681-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25022512

ABSTRACT

We previously found a negative inotropic (NIR) and positive lusitropic response (LR) to C-type natriuretic peptide (CNP) in the failing heart ventricle. In this study, we investigated and compared the functional responses to the natriuretic peptides (NPs), brain (BNP) and C-type natriuretic peptide (CNP), and relate them to cGMP regulation and effects on downstream targets. Experiments were conducted in left ventricular muscle strips and ventricular cardiomyocytes from Wistar rats with heart failure 6 weeks after myocardial infarction. As opposed to CNP, BNP did not cause an NIR or LR, despite increasing cGMP levels. The BNP-induced cGMP elevation was mainly and markedly regulated by phosphodiesterase (PDE) 2 and was only marginally increased by PDE3 or PDE5 inhibition. Combined PDE2, -3, and -5 inhibition failed to reveal any functional responses to BNP, despite an extensive cGMP elevation. BNP decreased, whereas CNP increased, the amplitude of the Ca(2+) transient. BNP did not increase phospholamban (PLB) or troponin I (TnI) phosphorylation, Ca(2+) extrusion rate constant, or sarcoplasmatic reticulum Ca(2+) load, whereas CNP did. Both BNP and CNP reduced the peak of the L-type Ca(2+) current. Cyclic GMP elevations by BNP and CNP in cardiomyocytes were additive, and the presence of BNP did not alter the NIR to CNP or the CNP-induced PLB and TnI phosphorylation. However, a small increase in the LR to maximal CNP was observed in the presence of BNP. In conclusion, different responses to cGMP generated by BNP and CNP suggest different compartmentation of the cGMP signal and different roles of the two NPs in the failing heart.


Subject(s)
Heart Failure/metabolism , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, Brain/pharmacology , Natriuretic Peptide, C-Type/metabolism , Natriuretic Peptide, C-Type/pharmacology , Ventricular Dysfunction, Left/metabolism , Animals , Cells, Cultured , Heart Failure/drug therapy , Heart Failure/pathology , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/therapeutic use , Natriuretic Peptide, C-Type/therapeutic use , Organ Culture Techniques , Rats , Rats, Wistar , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/pathology
11.
Naunyn Schmiedebergs Arch Pharmacol ; 387(5): 407-17, 2014 May.
Article in English | MEDLINE | ID: mdl-24424715

ABSTRACT

Recently, we showed C-type natriuretic peptide (CNP)-induced negative inotropic (NIR) and positive lusitropic response (LR) in failing rat heart. We wanted to study whether, and if so, how phosphodiesterases (PDEs) regulate CNP-induced cyclic 3',5'-guanosine monophosphate (cGMP) elevation and functional responses. Inotropic and lusitropic responses were measured in left ventricular muscle strips and cyclic nucleotide levels, PDE activity and phospholamban (PLB) and troponin I (TnI) phosphorylation were measured in ventricular cardiomyocytes from Wistar rats with heart failure 6 weeks after myocardial infarction. CNP-mediated increase in global cGMP was mainly regulated by PDE2, as reflected by a marked amplification of the cGMP increase during PDE2 inhibition and by a high PDE2 activity in cardiomyocytes. PDE3 inhibition, on the other hand, caused no significant cGMP increase by CNP. The functional consequences did not correspond to the changes of cGMP. PDE3 inhibition increased the potency of the CNP-induced NIR and LR, while PDE2 inhibition desensitized the CNP-induced NIR, but not LR. A role for PDE2 on the maximal LR and PDE5 on the maximal NIR to CNP was revealed in the presence of PDE3 inhibition. CNP increased PLB phosphorylation about 25- to 30-fold and tended to increase TnI phosphorylation about twofold. As a whole, CNP-induced functional responses were only modestly regulated by PDEs compared to the cAMP-mediated functional responses to ß1-adrenoceptor stimulation, which are highly regulated by PDEs. There is a mismatch between the CNP-induced cGMP increase and functional responses. Global cGMP levels are mainly regulated by PDE2 after CNP stimulation, whereas the functional responses are modestly regulated by both PDE2 and PDE3, indicating cGMP compartmentation by PDEs affecting CNP-induced responses in failing hearts.


Subject(s)
Cyclic GMP/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 2/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Heart Failure/physiopathology , Myocardial Contraction/drug effects , Natriuretic Peptide, C-Type/pharmacology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , In Vitro Techniques , Male , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Rats , Rats, Wistar , Signal Transduction/physiology
12.
Bioorg Med Chem ; 21(22): 7134-45, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24113240

ABSTRACT

Serotonin (5-hydroxytryptamine, 5-HT) is an important signaling molecule in the central nervous system (CNS) and in non-neuronal tissues and organs. Serotonin mediates a positive chronotropic and inotropic response through 5-HT4 receptors in the atrium and ventricle of the heart. Recent investigations have revealed increased expression of the 5-HT4(b) isoform in cardiomyocytes of chronic arrhythmic and failing hearts, and that the use of 5-HT4 receptor antagonists may be beneficial for treating these conditions. The 5-HT4 receptor possesses a transmembrane (TM) binding site important for ligand affinity and recognition, as well as a capacity to accommodate bulky ligands. A new series of peripherally-acting 5-HT4 receptor antagonists were prepared by combining the acidic biphenyl group from the class of angiotensin II receptor blockers (ARBs) with the SB207266 (piboserod) scaffold. The new compounds were pharmacologically evaluated and carboxylic acid 21 was identified as a potent and promising 5-HT4 receptor antagonist with moderate affinity for the AT1 receptor. The permeability of carboxylic acid 21 in a Caco-2 assay was low and the corresponding prodrug esters 23a-f were therefore prepared. The pharmacokinetics of methyl ester 20 and n-butyl ester 23c were evaluated in a rat model, revealing incomplete metabolism to carboxylic acid 21. However, methyl ester 20 is a potent 5-HT4 receptor antagonist with binding affinities in the low picomolar range. Methyl ester 20 has promising oral bioavailability and pharmacokinetics and may target 5-HT4 receptors in both CNS and peripheral organs.


Subject(s)
Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Receptors, Serotonin, 5-HT4/chemistry , Recombinant Proteins/chemistry , Serotonin 5-HT4 Receptor Agonists/chemical synthesis , Serotonin 5-HT4 Receptor Agonists/pharmacology , Administration, Oral , Animals , Caco-2 Cells , Cell Line , Cell Survival/drug effects , Guinea Pigs , HEK293 Cells , Half-Life , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT4/genetics , Receptors, Serotonin, 5-HT4/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Serotonin 5-HT4 Receptor Agonists/pharmacokinetics
13.
Eur J Med Chem ; 64: 629-37, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23711770

ABSTRACT

5-HT4 receptor antagonists have been suggested to have clinical potential in treatment of atrial fibrillation, diarrhea-prone irritable bowel syndrome and urinary incontinence. Recently, the use of 5-HT4 antagonists has been suggested to have a therapeutic benefit in heart failure. Affinity for the hERG potassium ion channel and increased risk for prolonged QT intervals and arrhythmias has been observed for several 5-HT4 ligands. Serotonin may also have beneficial effects in the central nervous system (CNS) through stimulation of the 5-HT4 receptor, and reduced distribution of 5-HT4 antagonists to the CNS may therefore be an advantage. Replacing the amide and N-butyl side chain of the 5-HT4 receptor antagonist SB207266 with an ester and a benzyl dimethyl acetic acid group led to compound 9; a hydrophilic 5-HT4 antagonist with excellent receptor binding and low affinity for the hERG potassium ion channel. To increase oral bioavailability of carboxylic acid 9, two different prodrug approaches were applied. The tert-butyl prodrug 11 did not improve bioavailability, and LC-MS analysis revealed unmetabolized prodrug in the systemic circulation. The medoxomil ester prodrug 10 showed complete conversion and sufficient bioavailability of 9 to advance into further preclinical testing for treatment of heart failure.


Subject(s)
Receptors, Serotonin, 5-HT4/metabolism , Serotonin 5-HT4 Receptor Antagonists/pharmacology , Administration, Oral , Animals , Humans , Hydrophobic and Hydrophilic Interactions , Male , Molecular Structure , Rats , Rats, Sprague-Dawley , Serotonin 5-HT4 Receptor Antagonists/administration & dosage , Serotonin 5-HT4 Receptor Antagonists/blood , Structure-Activity Relationship
14.
Naunyn Schmiedebergs Arch Pharmacol ; 384(6): 543-53, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21901315

ABSTRACT

Activation of 5-HT(4) receptors in failing ventricles elicits a cAMP-dependent positive inotropic response which is mainly limited by the cGMP-inhibitable phosphodiesterase (PDE) 3. However, PDE4 plays an additional role which is demasked by PDE3 inhibition. The objective of this study was to evaluate the effect of cGMP generated by particulate and soluble guanylyl cyclase (GC) on the 5-HT(4)-mediated inotropic response. Extensive myocardial infarctions were induced by coronary artery ligation in Wistar rats, exhibiting heart failure 6 weeks after surgery. Contractility was measured in left ventricular preparations. Cyclic GMP was measured by EIA. In ventricular preparations, ANP or BNP displayed no impact on 5-HT(4)-mediated inotropic response. However, CNP increased the 5-HT(4)-mediated inotropic response as well as the ß(1)-adrenoceptor (ß(1)-AR)-mediated response to a similar extent as PDE3 inhibition by cilostamide. Pretreatment with cilostamide eliminated the effect of CNP. Inhibition of nitric oxide (NO) synthase and soluble GC by L-NAME and ODQ, respectively, attenuated the 5-HT(4)-mediated inotropic response, whereas the NO donor Sin-1 increased this response. The effects were absent during PDE3 inhibition, suggesting cGMP-dependent inhibition of PDE3. However, in contrast to the effects on the 5-HT(4) response, Sin-1 inhibited whereas L-NAME and ODQ enhanced the ß(1)-AR-mediated inotropic response. cGMP generated both by particulate (NPR-B) and soluble GC increases the 5-HT(4)-mediated inotropic response in failing hearts, probably through inhibition of PDE3. ß(1)-AR and 5-HT(4) receptor signalling are subject to opposite regulatory control by cGMP generated by soluble GC in failing hearts. Thus, cGMP from different sources is functionally compartmented, giving differential regulation of different G(s)-coupled receptors.


Subject(s)
Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Heart Failure/physiopathology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Animals , Disease Models, Animal , GTP-Binding Protein alpha Subunits, Gs/metabolism , Male , Myocardial Contraction , Myocardial Infarction/physiopathology , Natriuretic Peptide, C-Type/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Soluble Guanylyl Cyclase
15.
J Med Chem ; 54(8): 3086-90, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21469694

ABSTRACT

We report benzo[b]thiophene derivatives synthesized according to a dual strategy. 8j, 9c, and 9e with affinity values toward 5-HT(7)R and 5-HTT were selected to probe their antidepressant activity in vivo using the forced swimming text (FST). The results showed significant antidepressant activity after chronic treatment. 9c was effective in reducing the immobility time in FST even after acute treatment. These findings identify these compounds as a new class of antidepressants with a rapid onset of action.


Subject(s)
Antidepressive Agents/therapeutic use , Thiophenes/therapeutic use , Magnetic Resonance Spectroscopy , Spectrophotometry, Infrared
16.
Br J Pharmacol ; 162(1): 54-71, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21133887

ABSTRACT

BACKGROUND AND PURPOSE: ß-Adrenoceptors play a major role in regulating myocardial function through cAMP-dependent pathways. Different phosphodiesterases (PDEs) regulate intracellular cAMP-pools and thereby contribute to the compartmentalization of cAMP-dependent effects. We explored the involvement of PDEs in limiting the ß(2) adrenoceptor-mediated positive inotropic (PIR) and lusitropic (LR) responses in sham-operated (Sham) and failing rat hearts. EXPERIMENTAL APPROACH: Extensive myocardial infarctions were induced by coronary artery ligation in Wistar rats. Rats developing heart failure were studied 6 weeks after surgery. Contractility was measured in left ventricular strips from failing and Sham hearts. cAMP was quantified by RIA. KEY RESULTS: In ventricular strips, stimulation of ß(2) -adrenoceptors with (-)-adrenaline (300 nM CGP20712A present) exerted a small PIR and LR. In Sham hearts, ß(2) -adrenoceptor-mediated as well as ß(1) -adrenoceptor-mediated PIR and LR were increased by selective inhibition of either PDE3 (1 µM cilostamide) or PDE4 (10 µM rolipram). In failing rat hearts, PDE3 inhibition enhanced PIR and LR to both ß(1) - and ß(2) -adrenoceptor stimulation while PDE4 inhibition had no effect on these responses despite a significant increase in cAMP levels. Combined PDE3/4 inhibition further enhanced the PIR and LR of ß(2) - and ß(1) -adrenoceptor activation both in Sham and failing hearts, compared with PDE3 inhibition alone. PDE4 enzyme activity was reduced in failing hearts. CONCLUSIONS AND IMPLICATIONS: Both PDE3 and PDE4 attenuated ß(2) - and ß(1) -adrenoceptor-mediated contractile responses in Sham hearts. In failing hearts, these responses are attenuated solely by PDE3 and thus even selective PDE3 inhibitors may provide a profound enhancement of ß-adrenoceptor-mediated responses in heart failure.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Heart Ventricles/physiopathology , Receptors, Adrenergic, beta-2/physiology , Animals , Cyclic AMP/metabolism , Heart Ventricles/enzymology , Heart Ventricles/metabolism , In Vitro Techniques , Male , Phosphodiesterase Inhibitors/pharmacology , Radioimmunoassay , Rats , Rats, Wistar
17.
Bioorg Med Chem ; 18(24): 8600-13, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21075638

ABSTRACT

Serotonin (5-hydroxytryptamine, 5-HT) is an important signalling molecule in the human body. The 5-HT(4) serotonin receptor, coupled to the G protein G(s), plays important physiological and pathophysiological roles in the heart, urinary bladder, gastrointestinal tract and the adrenal gland. Both 5-HT(4) antagonists and agonists have been developed in the aim to treat diseases in these organs. 5-HT(4) agonists might have beneficial effects in the central nervous system (CNS) and therefore, 5-HT(4) antagonists might cause CNS side effects. In this study, we have developed new amphoteric 5-HT(4) antagonists. A series of cyclic indole amide derivatives possessing an oxazine ring and a piperidine alkane carboxylic acid side chain and the corresponding prodrug esters were synthesized and their binding to 5-HT(4) receptors and antagonist properties were evaluated. In addition, an indole ester without the oxazine ring and the corresponding indole amide derivatives were also tested. Octanol-water distribution (LogD(Oct7.4)) was tested for some of the synthesized ligands. The main structure-affinity characteristics of the 5-HT(4) compounds tested were that the prodrug esters show higher affinity than their corresponding free acids, indole esters show higher affinity than the corresponding amides and ligands containing the oxazine ring in the indole skeleton show higher affinity than indole derivatives not containing the ring. One representative prodrug ester and its corresponding free acid were tested for binding on a panel of receptors and showed preserved selectivity for the 5-HT(4) receptor. These new molecules may be useful to target peripheral 5-HT(4) receptors.


Subject(s)
Receptors, Serotonin, 5-HT4/metabolism , Serotonin 5-HT4 Receptor Antagonists/chemical synthesis , Amides , Esters , Humans , Indoles , Ligands , Oxazines , Piperidines , Prodrugs/chemical synthesis , Serotonin 5-HT4 Receptor Antagonists/chemistry , Serotonin 5-HT4 Receptor Antagonists/pharmacology , Structure-Activity Relationship
18.
Cell Signal ; 20(6): 1169-78, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18406106

ABSTRACT

Cyclic AMP (cAMP) is an important physiological growth inhibitor of lymphoid cells, and the cAMP/protein kinase A (PKA) pathway is disrupted in several immunological disorders and cancers. Epstein Barr virus (EBV) infection of B lymphocytes is responsible for the development of lymphoproliferative disease as well as certain B-lymphoid malignancies. Here we hypothesized that EBV infection might render B lymphocytes resistant to cAMP/PKA-mediated growth inhibition. To test this, we assessed the growth-inhibitory response of cAMP-elevating compounds such as forskolin and isoproterenol, as well as the PKA activator 8-CPT-cAMP in normal B lymphocytes, EBV-infected B cells and in the EBV-negative B lymphoid cell line Reh. We could demonstrate that EBV infection indeed abolished cAMP-mediated growth inhibition of B cells. The defect was pinpointed to defective adenylyl cyclase (AC) activation by forskolin and isoproterenol, resulting in reduced formation of cAMP and lack of PKA activation and CREB phosphorylation. In contrast, 8-CPT-cAMP which directly activates PKA was able to inhibit EBV-infected B cell growth. The physiological implications of these results were underlined by the observation that the ability of forskolin to inhibit camptothecin-induced apoptosis was abolished in EBV-infected B cells. We conclude that EBV infection of B cells abrogates the activation of AC and thereby cAMP formation, and that this dysfunction renders the cells resistant to growth inhibition via the cAMP/PKA pathway.


Subject(s)
Adenylyl Cyclases/metabolism , B-Lymphocytes/virology , Herpesvirus 4, Human/physiology , Apoptosis/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , Camptothecin/toxicity , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Colforsin/antagonists & inhibitors , Cyclic AMP/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Isoproterenol/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...