Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
PLoS One ; 14(6): e0218110, 2019.
Article in English | MEDLINE | ID: mdl-31173603

ABSTRACT

PURPOSE: We previously reported that inhibitory G protein (Gi) exerts intrinsic receptor-independent inhibitory activity upon adenylyl cyclase (AC) that regulates contractile force in rat ventricle. The two major subtypes of AC in the heart are AC5 and AC6. The aim of this study was to determine if this intrinsic Gi inhibition regulating contractile force is AC subtype selective. METHODS: Wild-type (WT), AC5 knockout (AC5KO) and AC6 knockout (AC6KO) mice were injected with pertussis toxin (PTX) to inactivate Gi or saline (control).Three days after injection, we evaluated the effect of simultaneous inhibition of phosphodiesterases (PDE) 3 and 4 with cilostamide and rolipram respectively upon in vivo and ex vivo left ventricular (LV) contractile function. Also, changes in the level of cAMP were measured in left ventricular homogenates and at the membrane surface in cardiomyocytes obtained from the same mouse strains expressing the cAMP sensor pmEPAC1 using fluorescence resonance energy transfer (FRET). RESULTS: Simultaneous PDE3 and PDE4 inhibition increased in vivo and ex vivo rate of LV contractility only in PTX-treated WT and AC5KO mice but not in saline-treated controls. Likewise, Simultaneous PDE3 and PDE4 inhibition elevated total cAMP levels in PTX-treated WT and AC5KO mice compared to saline-treated controls. In contrast, simultaneous PDE3 and PDE4 inhibition did not increase in vivo or ex vivo rate of LV contractility or cAMP levels in PTX-treated AC6KO mice compared to saline-treated controls. Using FRET analysis, an increase of cAMP level was detected at the membrane of cardiomyocytes after simultaneous PDE3 and PDE4 inhibition in WT and AC5KO but not AC6KO. These FRET data are consistent with the functional data indicating that AC6 activity and PTX inhibition of Gi is necessary for simultaneous inhibition of PDE3 and PDE4 to elicit an increase in contractility. CONCLUSIONS: Together, these data suggest that AC6 is tightly regulated by intrinsic receptor-independent Gi activity, thus providing a mechanism for maintaining low basal cAMP levels in the functional compartment that regulates contractility.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Myocardial Contraction , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Female , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardium/metabolism , Pertussis Toxin/pharmacology
2.
PLoS One ; 10(3): e0115547, 2015.
Article in English | MEDLINE | ID: mdl-25738589

ABSTRACT

AIMS: We recently published that the positive inotropic response (PIR) to levosimendan can be fully accounted for by phosphodiesterase (PDE) inhibition in both failing human heart and normal rat heart. To determine if the PIR of the active metabolite OR-1896, an important mediator of the long-term clinical effects of levosimendan, also results from PDE3 inhibition, we compared the effects of OR-1896, a representative Ca2+ sensitizer EMD57033 (EMD), levosimendan and other PDE inhibitors. METHODS: Contractile force was measured in rat ventricular strips. PDE assay was conducted on rat ventricular homogenate. cAMP was measured using RII_epac FRET-based sensors. RESULTS: OR-1896 evoked a maximum PIR of 33 ± 10% above basal at 1 µM. This response was amplified in the presence of the PDE4 inhibitor rolipram (89 ± 14%) and absent in the presence of the PDE3 inhibitors cilostamide (0.5 ± 5.3%) or milrinone (3.2 ± 4.4%). The PIR was accompanied by a lusitropic response, and both were reversed by muscarinic receptor stimulation with carbachol and absent in the presence of ß-AR blockade with timolol. OR-1896 inhibited PDE activity and increased cAMP levels at concentrations giving PIRs. OR-1896 did not sensitize the concentration-response relationship to extracellular Ca2+. Levosimendan, OR-1896 and EMD all increased the sensitivity to ß-AR stimulation. The combination of either EMD and levosimendan or EMD and OR-1896 further sensitized the response, indicating at least two different mechanisms responsible for the sensitization. Only EMD sensitized the α1-AR response. CONCLUSION: The observed PIR to OR-1896 in rat ventricular strips is mediated through PDE3 inhibition, enhancing cAMP-mediated effects. These results further reinforce our previous finding that Ca2+ sensitization does not play a significant role in the inotropic (and lusitropic) effect of levosimendan, nor of its main metabolite OR-1896.


Subject(s)
Acetamides/pharmacology , Cardiotonic Agents/pharmacology , Myocardium/enzymology , Phosphodiesterase 3 Inhibitors/pharmacology , Pyridazines/pharmacology , Animals , Calcium/physiology , Cells, Cultured , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Male , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Rats, Wistar
3.
Naunyn Schmiedebergs Arch Pharmacol ; 387(12): 1177-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25216690

ABSTRACT

Studies suggest that increased activity of Gi contributes to the reduced ß-adrenoceptor-mediated inotropic response (ßAR-IR) in failing cardiomyocytes and that ß2AR-IR but not ß1AR-IR is blunted by dual coupling to Gs and Gi. We aimed to clarify the role of Gi upon the ß1AR-IR and ß2AR-IR in Sham and failing myocardium by directly measuring contractile force and cAMP accumulation. Contractility was measured ex vivo in left ventricular strips and cAMP accumulation in cardiomyocytes from rats with post-infarction heart failure (HF) or sham operates (Sham). The ß2AR-IR in Sham and HF was small and was amplified by simultaneously inhibiting phosphodiesterases 3 and 4 (PDE3&4). In HF, the inotropic response and cAMP accumulation evoked by ß1AR- or ß2AR-stimulation were reduced. Inactivation of Gi with pertussis toxin (PTX) did not restore the ß1AR-IR or ß2AR-IR in HF to Sham levels but did enhance the maximal ß2AR-IR. PTX increased both ß1AR- and ß2AR-evoked cAMP accumulation more in Sham than that in HF, and HF levels approached those in untreated Sham. The potency of agonists at ß1 and at ß2ARs (only under PDE3&4 inhibition) was increased in HF and by PTX in both HF and Sham. Without PDE3&4 inhibition, PTX increased only the maximal ß2AR-IR, not potency. We conclude that Gi regulates both ß1AR- and ß2AR-IR independent of receptor coupling with Gi. Gi together with PDE3&4 tonically restrict the ß2AR-IR. Gi inhibition did not restore the ßAR-IR in HF despite increasing cAMP levels, suggesting that the mechanism of impairment resides downstream to cAMP signalling.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heart Failure/physiopathology , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-1 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Cyclic AMP/metabolism , Disease Models, Animal , Heart Failure/etiology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Myocardial Infarction/complications , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2/drug effects
4.
PLoS One ; 9(9): e106608, 2014.
Article in English | MEDLINE | ID: mdl-25203113

ABSTRACT

BACKGROUND AND PURPOSE: Despite the view that only ß2- as opposed to ß1-adrenoceptors (ßARs) couple to G(i), some data indicate that the ß1AR-evoked inotropic response is also influenced by the inhibition of Gi. Therefore, we wanted to determine if Gi exerts tonic receptor-independent inhibition upon basal adenylyl cyclase (AC) activity in cardiomyocytes. EXPERIMENTAL APPROACH: We used the Gs-selective (R,R)- and the Gs- and G(i)-activating (R,S)-fenoterol to selectively activate ß2ARs (ß1AR blockade present) in combination with Gi inactivation with pertussis toxin (PTX). We also determined the effect of PTX upon basal and forskolin-mediated responses. Contractility was measured ex vivo in left ventricular strips and cAMP accumulation was measured in isolated ventricular cardiomyocytes from adult Wistar rats. KEY RESULTS: PTX amplified both the (R,R)- and (R,S)-fenoterol-evoked maximal inotropic response and concentration-dependent increases in cAMP accumulation. The EC50 values of fenoterol matched published binding affinities. The PTX enhancement of the Gs-selective (R,R)-fenoterol-mediated responses suggests that Gi regulates AC activity independent of receptor coupling to Gi protein. Consistent with this hypothesis, forskolin-evoked cAMP accumulation was increased and inotropic responses to forskolin were potentiated by PTX treatment. In non-PTX-treated tissue, phosphodiesterase (PDE) 3 and 4 inhibition or removal of either constitutive muscarinic receptor activation of Gi with atropine or removal of constitutive adenosine receptor activation with CGS 15943 had no effect upon contractility. However, in PTX-treated tissue, PDE3 and 4 inhibition alone increased basal levels of cAMP and accordingly evoked a large inotropic response. CONCLUSIONS AND IMPLICATIONS: Together, these data indicate that Gi exerts intrinsic receptor-independent inhibitory activity upon AC. We propose that PTX treatment shifts the balance of intrinsic G(i) and Gs activity upon AC towards Gs, enhancing the effect of all cAMP-mediated inotropic agents.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Animals , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Enzyme Activation/drug effects , Fenoterol/chemistry , Fenoterol/pharmacology , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pertussis Toxin/pharmacology , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Stereoisomerism
5.
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
6.
Basic Res Cardiol ; 107(5): 295, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22923058

ABSTRACT

Prostaglandins have displayed both beneficial and detrimental effects in clinical studies in patients with severe heart failure. Prostaglandins are known to increase cardiac output, but the mechanism is not clarified. Here, we tested the hypothesis that prostaglandins can increase contractility in human heart by amplifying cAMP-dependent inotropic responses. Contractility was measured ex vivo in isolated left ventricular strips and phosphodiesterase (PDE) and adenylyl cyclase (AC) activity was measured in homogenates or membranes from failing human left ventricles. PGE(1) (1 µM) alone did not modify contractility, but given prior, amplified maximal serotonin (5-HT)-evoked (10 µM) contractile responses mediated by 5-HT(4) receptors several fold (24 ± 7 % with PGE(1) vs. 3 ± 2 % above basal with 5-HT alone). The 5-HT(4)-mediated inotropic response was amplified by the PDE3 inhibitor cilostamide and further amplified in combination with PGE(1) (26 ± 6 vs. 56 ± 12 % above basal). PGE(1) reduced the time to reach 90 % of both the maximal 5-HT- and isoproterenol-evoked inotropic response compared to 5-HT or isoproterenol alone. PGE(1) did not modify PDE activity in the homogenate, either alone or when given simultaneously with PDE3 and/or PDE4 inhibitors. Neither 5-HT- nor isoproterenol-stimulated AC activity was significantly amplified by PGE(1). Sensitivity of ventricular strips to Ca(2+) was not enhanced in the presence of PGE(1). Our results show that PGE(1) can enhance cAMP-mediated responses in failing human left ventricle, through a mechanism independent of PDE inhibition, amplification of AC activity or increasing sensitivity to calcium. This effect of PGE(1) possibly contributes to the increase of cardiac output, independent of decreased afterload, observed after prostaglandin administration in humans.


Subject(s)
Alprostadil/pharmacology , Heart Failure/physiopathology , Myocardial Contraction/drug effects , Receptors, Adrenergic, beta/physiology , Receptors, Serotonin, 5-HT4/physiology , Adenylyl Cyclases/metabolism , Adolescent , Adult , Aged , Calcium/metabolism , Child , Cyclic AMP/physiology , Female , Humans , In Vitro Techniques , Male , Middle Aged , Phosphodiesterase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL