1
|
Chen Q, Xia Y, Liu HN, Chi Y, Li X, Shan LS, Dai B, Zhu Y, Wang YT, Miao X, Sun Q. Synthetic approaches and clinical application of representative small-molecule inhibitors of phosphodiesterase. Eur J Med Chem 2024; 277:116769. [PMID: 39163778 DOI: 10.1016/j.ejmech.2024.116769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024]
Abstract
Phosphodiesterases (PDEs) constitute a family of enzymes that play a pivotal role in the regulation of intracellular levels of cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Dysregulation of PDE activity has been implicated in diverse pathological conditions encompassing cardiovascular disorders, pulmonary diseases, and neurological disorders. Small-molecule inhibitors targeting PDEs have emerged as promising therapeutic agents for the treatment of these ailments, some of which have been approved for their clinical use. Despite their success, challenges such as resistance mechanisms and off-target effects persist, urging continuous research for the development of next-generation PDE inhibitors. The objective of this review is to provide an overview of the synthesis and clinical application of representative approved small-molecule PDE inhibitors, with the aim of offering guidance for further advancements in the development of novel PDE inhibitors.
Collapse
Affiliation(s)
- Qingqing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - He-Nan Liu
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan Chi
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Xun Li
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Li-Shen Shan
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Dai
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Ya-Tao Wang
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Xinxin Miao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qian Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Numata G, Takimoto E. Cyclic GMP and PKG Signaling in Heart Failure. Front Pharmacol 2022; 13:792798. [PMID: 35479330 PMCID: PMC9036358 DOI: 10.3389/fphar.2022.792798] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP), produced by guanylate cyclase (GC), activates protein kinase G (PKG) and regulates cardiac remodeling. cGMP/PKG signal is activated by two intrinsic pathways: nitric oxide (NO)-soluble GC and natriuretic peptide (NP)-particulate GC (pGC) pathways. Activation of these pathways has emerged as a potent therapeutic strategy to treat patients with heart failure, given cGMP-PKG signaling is impaired in heart failure with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF). Large scale clinical trials in patients with HFrEF have shown positive results with agents that activate cGMP-PKG pathways. In patients with HFpEF, however, benefits were observed only in a subgroup of patients. Further investigation for cGMP-PKG pathway is needed to develop better targeting strategies for HFpEF. This review outlines cGMP-PKG pathway and its modulation in heart failure.
Collapse
Affiliation(s)
- Genri Numata
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Department of Advanced Translational Research and Medicine in Management of Pulmonary Hypertension, The University of Tokyo Hospital, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, United States
| |
Collapse
|
3
|
Calamera G, Moltzau LR, Levy FO, Andressen KW. Phosphodiesterases and Compartmentation of cAMP and cGMP Signaling in Regulation of Cardiac Contractility in Normal and Failing Hearts. Int J Mol Sci 2022; 23:2145. [PMID: 35216259 PMCID: PMC8880502 DOI: 10.3390/ijms23042145] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Cardiac contractility is regulated by several neural, hormonal, paracrine, and autocrine factors. Amongst these, signaling through β-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.
Collapse
Affiliation(s)
| | | | | | - Kjetil Wessel Andressen
- Department of Pharmacology, Institute of Clinical Medicine, Oslo University Hospital, University of Oslo, P.O. Box 1057 Blindern, 0316 Oslo, Norway; (G.C.); (L.R.M.); (F.O.L.)
| |
Collapse
|
4
|
Koziol-White CJ, Ghosh A, Sandner P, Erzurum SE, Stuehr DJ, Panettieri RA. Soluble Guanylate Cyclase Agonists Induce Bronchodilation in Human Small Airways. Am J Respir Cell Mol Biol 2020; 62:43-48. [PMID: 31340135 PMCID: PMC6938135 DOI: 10.1165/rcmb.2019-0001oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 07/24/2019] [Indexed: 01/11/2023] Open
Abstract
The soluble guanylyl cyclase (sGC)-cyclic guanosine monophosphate signaling pathway evokes vascular smooth muscle relaxation; whether this pathway mediates airway smooth muscle relaxation remains controversial. We posit that sGC activators are equi-effective as β-agonists in reversing contractile agonist-induced airway smooth muscle shortening. To provide clarity, we tested the efficacy of sGC stimulator and activator drugs, BAY 41-2272 and BAY 60-2270, respectively, in reversing bronchoconstriction of human small airways using human precision-cut lung slices (hPCLS). Both BAY drugs reversed carbachol-induced bronchoconstriction to a maximal degree comparable to that of formoterol. Moreover, the sGC drugs remained effective bronchodilators despite formoterol-induced desensitization of the airways. Analysis of the hPCLS after their activation by sGC or β2-adrenergic receptor agonist showed distinct cyclic nucleotide accumulation in the hPCLS. Collectively, these data suggest that cAMP and cyclic guanosine monophosphate pathways are equi-effective for reversing carbachol-induced bronchoconstriction in the human airway via separate and distinct second messenger pathways. This should open the door for future studies to test whether sGC-targeted drugs alone or in combination can serve as effective bronchodilators in asthma and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Cynthia J. Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Arnab Ghosh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, Wuppertal, Germany
| | - Serpil E. Erzurum
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - Dennis J. Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - Reynold A. Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
5
|
Menges L, Krawutschke C, Füchtbauer EM, Füchtbauer A, Sandner P, Koesling D, Russwurm M. Mind the gap (junction): cGMP induced by nitric oxide in cardiac myocytes originates from cardiac fibroblasts. Br J Pharmacol 2019; 176:4696-4707. [PMID: 31423565 PMCID: PMC6965686 DOI: 10.1111/bph.14835] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 07/23/2019] [Accepted: 08/05/2019] [Indexed: 01/02/2023] Open
Abstract
Background and Purpose The intracellular signalling molecule cGMP, formed by NO‐sensitive GC (NO–GC), has an established function in the vascular system. Despite numerous reports about NO‐induced cGMP effects in the heart, the underlying cGMP signals are poorly characterized. Experimental Approach Therefore, we analysed cGMP signals in cardiac myocytes and fibroblasts isolated from knock‐in mice expressing a FRET‐based cGMP indicator. Key Results Whereas in cardiac myocytes, none of the known NO–GC‐activating substances (NO, GC activators, and GC stimulators) increased cGMP even in the presence of PDE inhibitors, they induced substantial cGMP increases in cardiac fibroblasts. As cardiac myocytes and fibroblasts are electrically connected via gap junctions, we asked whether cGMP can take the same route. Indeed, in cardiomyocytes co‐cultured on cardiac fibroblasts, NO‐induced cGMP signals were detectable, and two groups of unrelated gap junction inhibitors abolished these signals. Conclusion and Implication We conclude that NO‐induced cGMP formed in cardiac fibroblasts enters cardiac myocytes via gap junctions thereby turning cGMP into an intercellular signalling molecule. The findings shed new light on NO/cGMP signalling in the heart and will potentially broaden therapeutic opportunities for cardiac disease.
Collapse
Affiliation(s)
- Lukas Menges
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | | | - Ernst-Martin Füchtbauer
- Department of Molecular Biology and Genetics, Molecular Cell and Developmental Biology Aarhus University, Aarhus C, Denmark
| | - Annette Füchtbauer
- Department of Molecular Biology and Genetics, Molecular Cell and Developmental Biology Aarhus University, Aarhus C, Denmark
| | - Peter Sandner
- Drug Discovery, Cardiovascular Research, Bayer AG, Pharmaceuticals, Wuppertal, Germany
| | - Doris Koesling
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | - Michael Russwurm
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
6
|
Calamera G, Li D, Ulsund AH, Kim JJ, Neely OC, Moltzau LR, Bjørnerem M, Paterson D, Kim C, Levy FO, Andressen KW. FRET-based cyclic GMP biosensors measure low cGMP concentrations in cardiomyocytes and neurons. Commun Biol 2019; 2:394. [PMID: 31701023 PMCID: PMC6820734 DOI: 10.1038/s42003-019-0641-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 10/02/2019] [Indexed: 01/13/2023] Open
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.
Collapse
Affiliation(s)
- Gaia Calamera
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Dan Li
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Andrea Hembre Ulsund
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jeong Joo Kim
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX USA
| | - Oliver C. Neely
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Lise Román Moltzau
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Marianne Bjørnerem
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - David Paterson
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Choel Kim
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX USA
| | - Finn Olav Levy
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Kjetil Wessel Andressen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Center for Heart Failure Research, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
7
|
Wu R, Yao PA, Wang HL, Gao Y, Yu HL, Wang L, Cui XH, Xu X, Gao JP. Effect of fermented Cordyceps sinensis on doxorubicin‑induced cardiotoxicity in rats. Mol Med Rep 2018; 18:3229-3241. [PMID: 30066944 PMCID: PMC6102656 DOI: 10.3892/mmr.2018.9310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 11/17/2017] [Indexed: 01/08/2023] Open
Abstract
Cordyceps sinensis (CS) is a prominent medicinal herb in traditional Chinese medicine, and fermented CS is frequently used as a substitute for natural CS. Doxorubicin (DOX), an antitumor drug used in chemotherapy, is limited by its poor cardiotoxicity. The aim of the present study was to evaluate the protective effect of fermented CS against DOX-induced cardiotoxicity and the potential underlying mechanisms. Male Sprague-Dawley rats (180–200 g) were randomly assigned to seven different treatment groups: Normal control, DOX control, DOX+captopril (0.05 g/kg), 0.75, 1.5 and 3 g/kg DOX+CS, and the CS (1.5 g/kg) control. Histopathological changes, cardiac energy metabolism, cyclic adenosine monophosphate (cAMP) signaling and the associated mRNA expression of AMP-activated protein kinase (AMPK) were then evaluated. Fermented CS decreased the left ventricular weight index, heart weight index and mortality; however, it increased diastolic blood pressure and mean arterial pressure. In addition, it shortened the duration of the QRS complex and Sα-T segment, decreased serum creatine kinase (CK) and aspartate aminotransferase activity, inhibited histopathological changes and reduced brain natriuretic peptide content. Treatment with fermented CS also increased the activities of superoxide dismutase and glutathione peroxidase, reduced malondialdehyde content, increased the mitochondrial activities of Na+K+-adenosine 5′-triphosphate (ATP) ase, Ca2+Mg2+-ATPase and CK, and increased the creatine phosphate/ATP ratio and AMP/ATP ratio. Furthermore, it decreased the ATP/adenosine 5′-diphosphate (ADP) ratio, upregulated AMPKα2 expression, reduced the activity of serum phosphodiesterases (PDEs) and increased myocardial cAMP content. The results of the present study demonstrated that fermented CS attenuated DOX-induced cardiotoxicity by inhibiting myocardial hypertrophy and myocardial damage, ameliorating systolic function and the antioxidant enzyme system, improving cardiac energy metabolism, depressing the activities of PDEs, and by upregulating the cAMP and AMPK signaling pathways. Thus, fermented CS may be a candidate for the prevention of DOX-induced cardiotoxicity, cardiac energy impairment and against a number of cardiac diseases.
Collapse
Affiliation(s)
- Rong Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Ping-An Yao
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Hui-Lin Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yan Gao
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Hai-Lun Yu
- Department of Chemical and Environmental Engineering, School of Shanghai Institute of Technology, Shanghai 201418, P.R. China
| | - Lei Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Xiao-Hua Cui
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Xu Xu
- Department of Chemical and Environmental Engineering, School of Shanghai Institute of Technology, Shanghai 201418, P.R. China
| | - Jian-Ping Gao
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
8
|
Modification of levosimendan-induced suppression of atrial natriuretic peptide secretion in hypertrophied rat atria. Eur J Pharmacol 2018; 829:54-62. [PMID: 29653089 DOI: 10.1016/j.ejphar.2018.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 10/17/2022]
Abstract
This study aimed to determine the effects of levosimendan, a calcium sensitizer, on atrial contractility and atrial natriuretic peptide (ANP) secretion and its modification in hypertrophied atria. Isolated perfused beating rat atria were used from control and isoproterenol-treated rats. Levosimendan and its metabolite OR-1896 caused a positive inotropic effect and suppressed ANP secretion in rat atria. Similar to levosimendan, the selective phosphodiesterase 3 (PDE3) or PDE4 inhibitor also suppressed ANP secretion. Suppression of ANP secretion by 1 µM levosimendan was abolished by PDE3 inhibitor, but reversed by PDE4 inhibitor. Levosimendan-induced suppression of ANP secretion was potentiated by KATP channel blocker, but blocked by KATP channel opener. Levosimendan alone did not significantly change cyclic adenosine monophosphate (cAMP) efflux in the perfusate; however, levosimendan combined with PDE4 inhibitor markedly increased this efflux. The stimulation of ANP secretion induced by levosimendan combined with PDE4 inhibitor was blocked by the protein kinase A (PKA) inhibitor. In isoproterenol-treated atria, levosimendan augmented the positive inotropic effect and ANP secretion in response to an increased extracellular calcium concentration ([Ca+]o). These results suggests that levosimendan suppresses ANP secretion by both inhibiting PDE3 and opening KATP channels and that levosimendan combined with PDE4 inhibitor stimulates ANP secretion by activating the cAMP-PKA pathway. Modification of the effects of levosimendan on [Ca+]o-induced positive inotropic effects and ANP secretion in isoproterenol-treated rat atria might be related to a disturbance in calcium metabolism.
Collapse
|
9
|
Does Neprilysin Inhibition Potentiate or Minimize the Adverse Effects of Glucagon-Like Peptide-1 Receptor Agonists in Chronic Heart Failure? J Card Fail 2018; 24:109-111. [DOI: 10.1016/j.cardfail.2017.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/12/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023]
|
10
|
Meier S, Andressen KW, Aronsen JM, Sjaastad I, Hougen K, Skomedal T, Osnes JB, Qvigstad E, Levy FO, Moltzau LR. PDE3 inhibition by C-type natriuretic peptide-induced cGMP enhances cAMP-mediated signaling in both non-failing and failing hearts. Eur J Pharmacol 2017; 812:174-183. [PMID: 28697992 DOI: 10.1016/j.ejphar.2017.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/28/2022]
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 β1- and β2-adrenoceptor-mediated inotropic and β1-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 β1-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.
Collapse
Affiliation(s)
- Silja Meier
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Kjetil Wessel Andressen
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Magnus Aronsen
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Institute for Experimental Medical Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Bjørknes College, Oslo, Norway
| | - Ivar Sjaastad
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Institute for Experimental Medical Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Karina Hougen
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Institute for Experimental Medical Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Tor Skomedal
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan-Bjørn Osnes
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Eirik Qvigstad
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Finn Olav Levy
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway.
| | - Lise Román Moltzau
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; Center for Heart Failure Research, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
11
|
Weber S, Zeller M, Guan K, Wunder F, Wagner M, El-Armouche A. PDE2 at the crossway between cAMP and cGMP signalling in the heart. Cell Signal 2017; 38:76-84. [PMID: 28668721 DOI: 10.1016/j.cellsig.2017.06.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 11/26/2022]
Abstract
The cyclic nucleotides cAMP and cGMP are central second messengers in cardiac cells and critical regulators of cardiac physiology as well as pathophysiology. Consequently, subcellular compartmentalization allows for spatiotemporal control of cAMP/cGMP metabolism and subsequent regulation of their respective effector kinases PKA or PKG is most important for cardiac function in health and disease. While acute cAMP-mediated signalling is a mandatory prerequisite for the physiological fight-or-flight response, sustained activation of this pathway may lead to the progression of heart failure. In contrast, acute as well as sustained cGMP-mediated signalling can foster beneficial features, e.g. anti-hypertrophic and vasodilatory effects. These two signalling pathways seem to be intuitively counteracting and there is increasing evidence for a functionally relevant crosstalk between cAMP and cGMP signalling pathways on the level of cyclic nucleotide hydrolysing phosphodiesterases (PDEs). Among this diverse group of enzymes, PDE2 may fulfill a unique integrator role. Equipped with dual substrate specificity for cAMP as well as for cGMP, it is the only cAMP hydrolysing PDE, which is allosterically activated by cGMP. Recent studies have revealed strongly remodelled cAMP/cGMP microdomains and subcellular concentration profiles in different cardiac pathologies, leading to a putatively enhanced involvement of PDE2 in cAMP/cGMP breakdown and crosstalk compared to the other cardiac PDEs. This review sums up the current knowledge about molecular properties and regulation of PDE2 and explains the complex signalling network encompassing PDE2 in order to better understand the functional role of PDE2 in distinct cell types in cardiac health and disease. Moreover, this review gives an outlook in which way PDE2 may serve as a therapeutic target to treat cardiac disease.
Collapse
Affiliation(s)
- Silvio Weber
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany.
| | - Miriam Zeller
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany
| | - Kaomei Guan
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany
| | - Frank Wunder
- Drug Discovery, Bayer AG, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Michael Wagner
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany.
| |
Collapse
|
12
|
MOUBARAK M, MAGAUD C, SALIBA Y, CHATELIER A, BOIS P, FAIVRE JF, FARÈS N. Effects of Atrial Natriuretic Peptide on Rat Ventricular Fibroblasts During Differentiation Into Myofibroblasts. Physiol Res 2015; 64:495-503. [DOI: 10.33549/physiolres.932839] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Atrial natriuretic peptide antifibrotic properties are mainly described in cardiac myocytes or in induced cardiac myofibroblasts (Angiotensin II or TGF-β induced differentiation). In the present work, we investigate the effects of ANP/NPRA/cGMP system in modulating rat cardiac fibroblasts function. Cardiac fibroblasts were isolated from adult Wistar male rats and cultured in the presence of serum in order to induce fibroblasts differentiation. Cultures were then treated with ANP (1 µM), 8-Br-cGMP (100 µM) or IBMX (100 µM), a non-specific phosphodiesterases inhibitor. ANP significantly decreased proliferation rate and collagen secretion. Its effect was mimicked by the cGMP analog, while combining ANP with 8-Br-cGMP did not lead to additional effects. Moreover intracellular cGMP levels were elevated when cells were incubated with ANP confirming that ANP intracellular pathway is mediated by cGMP. Additionally, immunoblotting and immunofluorescence were used to confirm the presence of guanylyl cyclase specific natriuretic peptide receptors A and B. Finally we scanned specific cGMP dependent PDEs via RT-qPCR, and noticed that inhibiting all PDEs led to an important decrease in proliferation rate. Effect of ANP became more prominent after 10 culture days, confirming the importance of ANP in fibroblasts to myofibroblasts differentiation. Uncovering cellular aspects of ANP/NPRA/cGMP signaling system provided more elements to help understand cardiac fibrotic process.
Collapse
Affiliation(s)
| | | | | | | | | | | | - N. FARÈS
- Laboratoire de Recherche en Physiologie et Physiopathologie, Pôle Technologie Santé, Faculté de Médecine, Université Saint Joseph, Beyrouth, Liban
| |
Collapse
|
13
|
Orstavik O, Ata SH, Riise J, Dahl CP, Andersen GØ, Levy FO, Skomedal T, Osnes JB, Qvigstad E. Inhibition of phosphodiesterase-3 by levosimendan is sufficient to account for its inotropic effect in failing human heart. Br J Pharmacol 2015; 171:5169-81. [PMID: 24547784 DOI: 10.1111/bph.12647] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 10/28/2013] [Accepted: 11/10/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Levosimendan is known as a calcium sensitizer, although it is also known to inhibit PDE3. We aimed to isolate each component and estimate their contribution to the increased cardiac contractility induced by levosimendan. EXPERIMENTAL APPROACH Contractile force was measured in electrically stimulated ventricular strips from explanted failing human hearts and left ventricular strips from normal male Wistar rats. PDE activity was measured in a two-step PDE activity assay on failing human ventricle. KEY RESULTS Levosimendan exerted a positive inotropic effect (PIE) reaching maximum at 10(-5) M in ventricular strips from failing human hearts. In the presence of the selective PDE3 inhibitor cilostamide, the PIE of levosimendan was abolished. During treatment with a PDE4 inhibitor and a supra-threshold concentration of isoprenaline, levosimendan generated an amplified inotropic response. This effect was reversed by β-adrenoceptor blockade and undetectable in strips pretreated with cilostamide. Levosimendan (10(-6) M) increased the potency of β-adrenoceptor agonists by 0.5 log units in failing human myocardium, but not in the presence of cilostamide. Every inotropic response to levosimendan was associated with a lusitropic response. Levosimendan did not affect the concentration-response curve to calcium in rat ventricular strips, in contrast to the effects of a known calcium sensitizer, EMD57033 [5-(1-(3,4-dimethoxybenzoyl)-1,2,3,4-tetrahydroquinolin-6-yl)-6-methyl-3,6-dihydro-2H-1,3,4-thiadiazin-2-one]. PDE activity assays confirmed that levosimendan inhibited PDE3 as effectively as cilostamide. CONCLUSIONS AND IMPLICATIONS Our results indicate that the PDE3-inhibitory property of levosimendan was enough to account for its inotropic effect, leaving a minor, if any, effect to a calcium-sensitizing component.
Collapse
Affiliation(s)
- O Orstavik
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway; K.G. Jebsen Cardiac Research Centre, Faculty of Medicine, University of Oslo, Oslo, Norway; Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Ørstavik Ø, Manfra O, Andressen KW, Andersen GØ, Skomedal T, Osnes JB, Levy FO, Krobert KA. The inotropic effect of the active metabolite of levosimendan, OR-1896, is mediated through inhibition of PDE3 in rat ventricular myocardium. PLoS One 2015; 10:e0115547. [PMID: 25738589 PMCID: PMC4349697 DOI: 10.1371/journal.pone.0115547] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/25/2014] [Indexed: 11/25/2022] Open
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 β-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 β-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.
Collapse
Affiliation(s)
- Øivind Ørstavik
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ornella Manfra
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjetil Wessel Andressen
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Geir Øystein Andersen
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Tor Skomedal
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jan-Bjørn Osnes
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Finn Olav Levy
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- * E-mail:
| | - Kurt Allen Krobert
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
15
|
Melsom CB, Hussain RI, Ørstavik Ø, Aronsen JM, Sjaastad I, Skomedal T, Osnes JB, Levy FO, Krobert KA. Non-classical regulation of β1- and β 2-adrenoceptor-mediated inotropic responses in rat heart ventricle by the G protein Gi. Naunyn Schmiedebergs Arch Pharmacol 2014; 387:1177-86. [PMID: 25216690 DOI: 10.1007/s00210-014-1036-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/05/2014] [Indexed: 01/08/2023]
Abstract
Studies suggest that increased activity of Gi contributes to the reduced β-adrenoceptor-mediated inotropic response (βAR-IR) in failing cardiomyocytes and that β2AR-IR but not β1AR-IR is blunted by dual coupling to Gs and Gi. We aimed to clarify the role of Gi upon the β1AR-IR and β2AR-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 β2AR-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 β1AR- or β2AR-stimulation were reduced. Inactivation of Gi with pertussis toxin (PTX) did not restore the β1AR-IR or β2AR-IR in HF to Sham levels but did enhance the maximal β2AR-IR. PTX increased both β1AR- and β2AR-evoked cAMP accumulation more in Sham than that in HF, and HF levels approached those in untreated Sham. The potency of agonists at β1 and at β2ARs (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 β2AR-IR, not potency. We conclude that Gi regulates both β1AR- and β2AR-IR independent of receptor coupling with Gi. Gi together with PDE3&4 tonically restrict the β2AR-IR. Gi inhibition did not restore the βAR-IR in HF despite increasing cAMP levels, suggesting that the mechanism of impairment resides downstream to cAMP signalling.
Collapse
Affiliation(s)
- Caroline Bull Melsom
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Melsom CB, Ørstavik Ø, Osnes JB, Skomedal T, Levy FO, Krobert KA. Gi proteins regulate adenylyl cyclase activity independent of receptor activation. PLoS One 2014; 9:e106608. [PMID: 25203113 PMCID: PMC4159282 DOI: 10.1371/journal.pone.0106608] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 07/30/2014] [Indexed: 11/19/2022] Open
Abstract
Background and purpose Despite the view that only β2- as opposed to β1-adrenoceptors (βARs) couple to Gi, some data indicate that the β1AR-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 Gi-activating (R,S)-fenoterol to selectively activate β2ARs (β1AR 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 Gi and Gs activity upon AC towards Gs, enhancing the effect of all cAMP-mediated inotropic agents.
Collapse
Affiliation(s)
- Caroline Bull Melsom
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Øivind Ørstavik
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jan-Bjørn Osnes
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tor Skomedal
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Finn Olav Levy
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- * E-mail:
| | - Kurt Allen Krobert
- Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Moltzau LR, Aronsen JM, Meier S, Skogestad J, Ørstavik Ø, Lothe GB, Sjaastad I, Skomedal T, Osnes JB, Levy FO, Qvigstad E. Different compartmentation of responses to brain natriuretic peptide and C-type natriuretic peptide in failing rat ventricle. J Pharmacol Exp Ther 2014; 350:681-90. [PMID: 25022512 DOI: 10.1124/jpet.114.214882] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
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.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Heart Failure/drug therapy
- Heart Failure/metabolism
- Heart Failure/pathology
- Male
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Natriuretic Peptide, Brain/metabolism
- Natriuretic Peptide, Brain/pharmacology
- Natriuretic Peptide, Brain/therapeutic use
- Natriuretic Peptide, C-Type/metabolism
- Natriuretic Peptide, C-Type/pharmacology
- Natriuretic Peptide, C-Type/therapeutic use
- Organ Culture Techniques
- Rats
- Rats, Wistar
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
Collapse
Affiliation(s)
- Lise Román Moltzau
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Jan Magnus Aronsen
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Silja Meier
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Jonas Skogestad
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Øivind Ørstavik
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Gustav B Lothe
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Ivar Sjaastad
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Tor Skomedal
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Jan-Bjørn Osnes
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Finn Olav Levy
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| | - Eirik Qvigstad
- Department of Pharmacology (L.R.M., S.M., Ø.Ø., T.S., J.-B.O., F.O.L., E.Q.) and Institute for Experimental Medical Research (J.M.A., J.S., G.B.L., I.S.), Institute of Clinical Medicine, University of Oslo and Oslo University Hospital; K. G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway (L.R.M., S.M., J.S., Ø.Ø., I.S., T.S., J.-B.O., F.O.L., E.Q.); and Bjørknes College, Oslo, Norway (J.M.A., G.B.L.)
| |
Collapse
|
18
|
Seifert R. Is cIMP a second messenger with functions opposite to those of cGMP? Naunyn Schmiedebergs Arch Pharmacol 2014; 387:897-9. [PMID: 25017018 DOI: 10.1007/s00210-014-1013-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 06/18/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Roland Seifert
- Institute of Pharmacology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany,
| |
Collapse
|
19
|
Moltzau LR, Meier S, Aronsen JM, Afzal F, Sjaastad I, Skomedal T, Osnes JB, Levy FO, Qvigstad E. Differential regulation of C-type natriuretic peptide-induced cGMP and functional responses by PDE2 and PDE3 in failing myocardium. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2014; 387:407-17. [PMID: 24424715 DOI: 10.1007/s00210-013-0953-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 12/19/2013] [Indexed: 11/29/2022]
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 β1-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.
Collapse
Affiliation(s)
- Lise Román Moltzau
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, P.O. Box 1057 Blindern, N-0316, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Bach T, Bergholtz S, Riise J, Qvigstad E, Skomedal T, Osnes JB, Levy FO. Identification of small molecule NPR-B antagonists by high throughput screening — potential use in heart failure. Naunyn Schmiedebergs Arch Pharmacol 2013; 387:5-14. [DOI: 10.1007/s00210-013-0940-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 11/12/2013] [Indexed: 02/06/2023]
|