51
|
cAMP/PKA signaling compartmentalization in cardiomyocytes: Lessons from FRET-based biosensors. J Mol Cell Cardiol 2019; 131:112-121. [PMID: 31028775 DOI: 10.1016/j.yjmcc.2019.04.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/19/2019] [Accepted: 04/19/2019] [Indexed: 12/29/2022]
Abstract
3',5'-cyclic adenosine monophosphate (cAMP) is a ubiquitous second messenger produced in response to the stimulation of G protein-coupled receptors (GPCRs). It regulates a plethora of pathophysiological processes in different organs, including the cardiovascular system. It is now clear that cAMP is not uniformly distributed within cardiac myocytes but confined in specific subcellular compartments where it modulates key players of the excitation-contraction coupling as well as other processes including gene transcription, mitochondrial homeostasis and cell death. This review will cover the major cAMP microdomains in cardiac myocytes. We will describe recent work using pioneering tools developed for investigating the organization and the function of the major cAMP microdomains in cardiomyocytes, including the plasma membrane, the sarcoplasmic reticulum, the myofilaments, the nucleus and the mitochondria.
Collapse
|
52
|
Rabal O, Sánchez-Arias JA, Cuadrado-Tejedor M, de Miguel I, Pérez-González M, García-Barroso C, Ugarte A, Estella-Hermoso de Mendoza A, Sáez E, Espelosin M, Ursua S, Haizhong T, Wei W, Musheng X, Garcia-Osta A, Oyarzabal J. Discovery of in Vivo Chemical Probes for Treating Alzheimer's Disease: Dual Phosphodiesterase 5 (PDE5) and Class I Histone Deacetylase Selective Inhibitors. ACS Chem Neurosci 2019; 10:1765-1782. [PMID: 30525452 DOI: 10.1021/acschemneuro.8b00648] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In order to determine the contributions of histone deacetylase (HDAC) isoforms to the beneficial effects of dual phosphodiesterase 5 (PDE5) and pan-HDAC inhibitors on in vivo models of Alzheimer's disease (AD), we have designed, synthesized, and tested novel chemical probes with the desired target compound profile of PDE5 and class I HDAC selective inhibitors. Compared to previous hydroxamate-based series, these molecules exhibit longer residence times on HDACs. In this scenario, shorter or longer preincubation times may have a significant impact on the IC50 values of these compounds and therefore on their corresponding selectivity profiles on the different HDAC isoforms. On the other hand, different chemical series have been explored and, as expected, some pairwise comparisons show a clear impact of the scaffold on biological responses (e.g., 35a vs 40a). The lead identification process led to compound 29a, which shows an adequate ADME-Tox profile and in vivo target engagement (histone acetylation and cAMP/cGMP response element-binding (CREB) phosphorylation) in the central nervous system (CNS), suggesting that this compound represents an optimized chemical probe; thus, 29a has been assayed in a mouse model of AD (Tg2576).
Collapse
Affiliation(s)
| | | | - Mar Cuadrado-Tejedor
- Anatomy Department, School of Medicine, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | | | | | | | | | | | | | | | | | - Tan Haizhong
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, Fourth Avenue, Tianjin 300456, PR China
| | - Wu Wei
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, Fourth Avenue, Tianjin 300456, PR China
| | - Xu Musheng
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, Fourth Avenue, Tianjin 300456, PR China
| | | | | |
Collapse
|
53
|
Li D, Paterson DJ. Pre-synaptic sympathetic calcium channels, cyclic nucleotide-coupled phosphodiesterases and cardiac excitability. Semin Cell Dev Biol 2019; 94:20-27. [PMID: 30658154 DOI: 10.1016/j.semcdb.2019.01.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/07/2019] [Accepted: 01/14/2019] [Indexed: 12/20/2022]
Abstract
In sympathetic neurons innervating the heart, action potentials activate voltage-gated Ca2+ channels and evoke Ca2+ entry into presynaptic terminals triggering neurotransmitter release. Binding of transmitters to specific receptors stimulates signal transduction pathways that cause changes in cardiac function. The mechanisms contributing to presynaptic Ca2+ dynamics involve regulation of endogenous Ca2+ buffers, in particular the endoplasmic reticulum, mitochondria and cyclic nucleotide targeted pathways. The purpose of this review is to summarize and highlight recent findings about Ca2+ homeostasis in cardiac sympathetic neurons and how modulation of second messengers can drive neurotransmission and affect myocyte excitability in cardiovascular disease. Moreover, we discuss the underlying mechanism of abnormal intracellular Ca2+ homeostasis and signaling in these neurons, and speculate on the role of phosphodiesterases as a therapeutic target to restore normal autonomic transmission in disease states of overactivity.
Collapse
Affiliation(s)
- Dan Li
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, UK.
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, UK.
| |
Collapse
|
54
|
Chan SHH, Chan JYH. Phosphodiesterase 2 as a Therapeutic Target for Heart Failure: Is Upregulation an Option? Circ Res 2018; 120:13-16. [PMID: 28057782 DOI: 10.1161/circresaha.116.310250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Samuel H H Chan
- From the Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Taiwan, Republic of China
| | - Julie Y H Chan
- From the Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Taiwan, Republic of China.
| |
Collapse
|
55
|
Liu Z, Hua J, Cai W, Zhan Q, Lai W, Zeng Q, Ren H, Xu D. N‑terminal truncated peroxisome proliferator‑activated receptor‑γ coactivator‑1α alleviates phenylephrine‑induced mitochondrial dysfunction and decreases lipid droplet accumulation in neonatal rat cardiomyocytes. Mol Med Rep 2018; 18:2142-2152. [PMID: 29901150 PMCID: PMC6072228 DOI: 10.3892/mmr.2018.9158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 04/30/2018] [Indexed: 11/05/2022] Open
Abstract
N‑terminal truncated peroxisome proliferator‑activated receptor‑γ coactivator‑1α (NT‑PGC‑1α) is an alternative splice variant of PGC‑1α. NT‑PGC‑1α exhibits stronger anti‑obesity effects in adipose tissue than PGC‑1α; however, NT‑PGC‑1α has not yet been investigated in neonatal rat cardiomyocytes (NRCMs). The present study aimed to investigate the role of NT‑PGC‑1α in mitochondrial fatty acid metabolism and its possible regulatory mechanism in NRCMs. NRCMs were exposed to phenylephrine (PE) or angiotensin II (Ang II) to induce cardiac hypertrophy. Following this, NRCMs were infected with adenovirus expressing NT‑PGC‑1α, and adenosine 5'‑triphsophate (ATP) levels, reactive oxygen species (ROS) generation and mitochondrial membrane potential were subsequently detected. In addition, western blotting, lipid droplet staining and oxygen consumption assays were performed to examine the function of NT‑PGC‑1α in fatty acid metabolism. NT‑PGC‑1α was demonstrated to be primarily expressed in the cytoplasm, which differed from full‑length PGC‑1α, which was predominantly expressed in the nucleus. NT‑PGC‑1α overexpression alleviated mitochondrial function impairment, including ATP generation, ROS production and mitochondrial membrane potential integrity. Furthermore, NT‑PGC‑1α overexpression alleviated the PE‑induced suppression of fatty acid metabolism‑associated protein expression, increased extracellular oxygen consumption and decreased lipid droplet accumulation in NRCMs. Taken together, the present study demonstrated that NT‑PGC‑1α alleviated PE‑induced mitochondrial impairment and decreased lipid droplet accumulation in NRCMs, indicating that NT‑PGC‑1α may have ameliorated mitochondrial energy defects in NRCMs, and may be considered as a potential target for the treatment of heart failure.
Collapse
Affiliation(s)
- Zuheng Liu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jinghai Hua
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wanqiang Cai
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Zhan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wenyan Lai
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of The People's Republic of China, Guangzhou, Guangdong 510515, P.R. China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
56
|
Røe ÅT, Aronsen JM, Skårdal K, Hamdani N, Linke WA, Danielsen HE, Sejersted OM, Sjaastad I, Louch WE. Increased passive stiffness promotes diastolic dysfunction despite improved Ca2+ handling during left ventricular concentric hypertrophy. Cardiovasc Res 2018; 113:1161-1172. [PMID: 28472418 PMCID: PMC5852536 DOI: 10.1093/cvr/cvx087] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/03/2017] [Indexed: 02/06/2023] Open
Abstract
Aims Concentric hypertrophy following pressure-overload is linked to preserved systolic function but impaired diastolic function, and is an important substrate for heart failure with preserved ejection fraction. While increased passive stiffness of the myocardium is a suggested mechanism underlying diastolic dysfunction in these hearts, the contribution of active diastolic Ca2+ cycling in cardiomyocytes remains unclear. In this study, we sought to dissect contributions of passive and active mechanisms to diastolic dysfunction in the concentrically hypertrophied heart following pressure-overload. Methods and results Rats were subjected to aortic banding (AB), and experiments were performed 6 weeks after surgery using sham-operated rats as controls. In vivo ejection fraction and fractional shortening were normal, confirming preservation of systolic function. Left ventricular concentric hypertrophy and diastolic dysfunction following AB were indicated by thickening of the ventricular wall, reduced peak early diastolic tissue velocity, and higher E/e' values. Slowed relaxation was also observed in left ventricular muscle strips isolated from AB hearts, during both isometric and isotonic stimulation, and accompanied by increases in passive tension, viscosity, and extracellular collagen. An altered titin phosphorylation profile was observed with hypophosphorylation of the phosphosites S4080 and S3991 sites within the N2Bus, and S12884 within the PEVK region. Increased titin-based stiffness was confirmed by salt-extraction experiments. In contrast, isolated, unloaded cardiomyocytes exhibited accelerated relaxation in AB compared to sham, and less contracture at high pacing frequencies. Parallel enhancement of diastolic Ca2+ handling was observed, with augmented NCX and SERCA2 activity and lowered resting cytosolic [Ca2+]. Conclusion In the hypertrophied heart with preserved systolic function, in vivo diastolic dysfunction develops as cardiac fibrosis and alterations in titin phosphorylation compromise left ventricular compliance, and despite compensatory changes in cardiomyocyte Ca2+ homeostasis.
Collapse
MESH Headings
- Adaptation, Physiological
- Animals
- Aorta/physiopathology
- Aorta/surgery
- Arterial Pressure
- Calcium/metabolism
- Calcium Signaling
- Collagen/metabolism
- Compliance
- Connectin/metabolism
- Constriction
- Diastole
- Disease Models, Animal
- Fibrosis
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Isolated Heart Preparation
- Male
- Myocardium/metabolism
- Myocardium/pathology
- Phosphorylation
- Rats, Wistar
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Sodium-Calcium Exchanger/metabolism
- Systole
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Ventricular Remodeling
Collapse
Affiliation(s)
- Åsmund T. Røe
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
- Corresponding author. Tel: +47 23 01 68 00; fax: +47 23 01 67 99, E-mail:
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Kristine Skårdal
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Wolfgang A. Linke
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Håvard E. Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| | - Ole M. Sejersted
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
| |
Collapse
|
57
|
Bhogal NK, Hasan A, Gorelik J. The Development of Compartmentation of cAMP Signaling in Cardiomyocytes: The Role of T-Tubules and Caveolae Microdomains. J Cardiovasc Dev Dis 2018; 5:jcdd5020025. [PMID: 29751502 PMCID: PMC6023514 DOI: 10.3390/jcdd5020025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/18/2018] [Accepted: 04/28/2018] [Indexed: 12/26/2022] Open
Abstract
3′-5′-cyclic adenosine monophosphate (cAMP) is a signaling messenger produced in response to the stimulation of cellular receptors, and has a myriad of functional applications depending on the cell type. In the heart, cAMP is responsible for regulating the contraction rate and force; however, cAMP is also involved in multiple other functions. Compartmentation of cAMP production may explain the specificity of signaling following a stimulus. In particular, transverse tubules (T-tubules) and caveolae have been found to be critical structural components for the spatial confinement of cAMP in cardiomyocytes, as exemplified by beta-adrenergic receptor (β-ARs) signaling. Pathological alterations in cardiomyocyte microdomain architecture led to a disruption in compartmentation of the cAMP signal. In this review, we discuss the difference between atrial and ventricular cardiomyocytes in respect to microdomain organization, and the pathological changes of atrial and ventricular cAMP signaling in response to myocyte dedifferentiation. In addition, we review the role of localized phosphodiesterase (PDE) activity in constraining the cAMP signal. Finally, we discuss microdomain biogenesis and maturation of cAMP signaling with the help of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Understanding these mechanisms may help to overcome the detrimental effects of pathological structural remodeling.
Collapse
Affiliation(s)
- Navneet K Bhogal
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| | - Alveera Hasan
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| | - Julia Gorelik
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
58
|
Vandenberghe P, Hagué P, Hockman SC, Manganiello VC, Demetter P, Erneux C, Vanderwinden JM. Phosphodiesterase 3A: a new player in development of interstitial cells of Cajal and a prospective target in gastrointestinal stromal tumors (GIST). Oncotarget 2018; 8:41026-41043. [PMID: 28454120 PMCID: PMC5522287 DOI: 10.18632/oncotarget.17010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 03/25/2017] [Indexed: 12/19/2022] Open
Abstract
We previously identified phosphodiesterase 3A (PDE3A) as a marker for interstitial cells of Cajal (ICC) in adult mouse gut. However, PDE3A expression and function during gut development and in ICC-derived gastrointestinal stromal tumors (GIST) remained unknown. Here we found that PDE3A was expressed throughout ICC development and that ICC density was halved in PDE3A-deficient mice. In the human imatinib-sensitive GIST882 cell line, the PDE3 inhibitor cilostazol halved cell viability (IC50 0.35 μM) and this effect synergized with imatinib (Chou-Talalay's CI50 0.15). Recently the compound 6-(4-(diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one, or DNMDP was found to be cytotoxic selectively for cells expressing both PDE3A and Schlafen12 (SLFN12) (de Waal L et al. Nat Chem Bio 2016), identifying a new, non-catalytic, role for PDE3A. 108 out of 117 (92%) of our human GIST samples displayed both PDE3A and SLFN12 immunoreactivity. GIST882 cells express both PDE3A and SLFN12 and DNMDP decreased their viability by 90%. Our results suggest a role for PDE3A during ICC development and open novel perspectives for PDE3A in targeted GIST therapy, on one hand by the synergism between imatinib and cilostazol, a PDE3 inhibitor already in clinical use for other indications, and, on the other hand, by the neomorphic, druggable, PDE3A-SLFN12 cytotoxic interplay.
Collapse
Affiliation(s)
- Pierre Vandenberghe
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Perrine Hagué
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Steven C Hockman
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vincent C Manganiello
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pieter Demetter
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Christophe Erneux
- IRIBHM, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Marie Vanderwinden
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
59
|
Makary S, Abdo M, Fekry E. Oxidative stress burden inhibits spermatogenesis in adult male rats: testosterone protective effect. Can J Physiol Pharmacol 2018; 96:372-381. [DOI: 10.1139/cjpp-2017-0459] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In this study, we aimed to investigate the protective effects of androgens, using letrozole (LET; an aromatase inhibitor), grape seed extract (GSE; a naturally occurring aromatase inhibitor and antioxidant), and testosterone propionate (Tp), against methotrexate (MTX)-induced testicular toxicity in adult male rats. MTX has been shown to induce oxidative stress and exhibit antiproliferative effects in the testes. Adult male rats received oral saline gavage (control group with no treatment), the potential protective agents (LET, GSE, or Tp) alone, MTX alone, or a combination of one of the potential protective agents and MTX. The testicular levels of oxidative stress markers and cytokines (tumor necrosis factor-α and interleukin-1β) were measured. Spermatogenesis and sperm viability were microscopically evaluated. Administration of LET and GSE 7 days before MTX improved spermatogenesis and sperm viability, as well as reduced the levels of oxidative stress markers and cellular cytokines. Exogenous testosterone exhibited anti-inflammatory and antioxidant activities, similar to GSE and LET. We also showed that enhancing the endogenous androgenic activity by LET and GSE protected spermatogenesis against MTX-induced testicular toxicity via reduction of inflammation and oxidative stress in the testes. Our data suggest that testosterone protected spermatogenesis owing to its antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Samy Makary
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Mohamed Abdo
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ereny Fekry
- Department of Histology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
60
|
Wobst J, Schunkert H, Kessler T. Genetic alterations in the NO-cGMP pathway and cardiovascular risk. Nitric Oxide 2018; 76:105-112. [PMID: 29601927 DOI: 10.1016/j.niox.2018.03.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/18/2018] [Accepted: 03/26/2018] [Indexed: 12/18/2022]
Abstract
In the past ten years, several chromosomal loci have been identified by genome-wide association studies to influence the risk of coronary artery disease (CAD) and its risk factors. The GUCY1A3 gene encoding the α1 subunit of the soluble guanylyl cyclase (sGC) resides at one of these loci and has been strongly associated with blood pressure and CAD risk. More recently, further genes in the pathway encoding the endothelial nitric oxide synthase, the phosphodiesterases 3A and 5A, and the inositol 1,4,5-trisphosphate receptor I-associated protein (IRAG), i.e., NOS3, PDE3A, PDE5A, and MRVI1, respectively, were likewise identified as CAD risk genes. In this review, we highlight the genetic findings linking variants in NO-cGMP signaling and cardiovascular disease, discuss the potential underlying mechanisms which might propagate the development of atherosclerosis, and speculate about therapeutic implications.
Collapse
Affiliation(s)
- Jana Wobst
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
61
|
Rabal O, Sánchez-Arias JA, Cuadrado-Tejedor M, de Miguel I, Pérez-González M, García-Barroso C, Ugarte A, Estella-Hermoso de Mendoza A, Sáez E, Espelosin M, Ursua S, Haizhong T, Wei W, Musheng X, Garcia-Osta A, Oyarzabal J. Design, synthesis, biological evaluation and in vivo testing of dual phosphodiesterase 5 (PDE5) and histone deacetylase 6 (HDAC6)-selective inhibitors for the treatment of Alzheimer's disease. Eur J Med Chem 2018; 150:506-524. [PMID: 29549837 DOI: 10.1016/j.ejmech.2018.03.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 02/06/2023]
Abstract
We have identified chemical probes that act as dual phosphodiesterase 5 (PDE5) and histone deacetylase 6 (HDAC6)-selective inhibitors (>1 log unit difference versus class I HDACs) to decipher the contribution of HDAC isoforms to the positive impact of dual-acting PDE5 and HDAC inhibitors on mouse models of Alzheimer's disease (AD) and fine-tune this systems therapeutics approach. Structure- and knowledge-based approaches led to the design of first-in-class molecules with the desired target compound profile: dual PDE5 and HDAC6-selective inhibitors. Compound 44b, which fulfilled the biochemical, functional and ADME-Tox profiling requirements and exhibited adequate pharmacokinetic properties, was selected as pharmacological tool compound and tested in a mouse model of AD (Tg2576) in vivo.
Collapse
Affiliation(s)
- Obdulia Rabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Juan A Sánchez-Arias
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Mar Cuadrado-Tejedor
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain; Anatomy Department, School of Medicine, University of Navarra, Irunlarrea 1, E-31008, Pamplona, Spain
| | - Irene de Miguel
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Marta Pérez-González
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Carolina García-Barroso
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Ana Ugarte
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Ander Estella-Hermoso de Mendoza
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Elena Sáez
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Maria Espelosin
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Susana Ursua
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Tan Haizhong
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, 4th Avenue, Tianjin, 300456, PR China
| | - Wu Wei
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, 4th Avenue, Tianjin, 300456, PR China
| | - Xu Musheng
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, 4th Avenue, Tianjin, 300456, PR China
| | - Ana Garcia-Osta
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, E-31008, Pamplona, Spain.
| |
Collapse
|
62
|
Peng T, Gong J, Jin Y, Zhou Y, Tong R, Wei X, Bai L, Shi J. Inhibitors of phosphodiesterase as cancer therapeutics. Eur J Med Chem 2018; 150:742-756. [PMID: 29574203 DOI: 10.1016/j.ejmech.2018.03.046] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/26/2018] [Accepted: 03/16/2018] [Indexed: 01/05/2023]
Abstract
Phosphodiesterases (PDEs) are a class of enzymes that hydrolyze cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) which is involved in many physiological processes including visual transduction, cell proliferation and differentiation, cell-cycle regulation, gene expression, inflammation, apoptosis, and metabolic function. PDEs are composed of 11 different families and each family contains different subtypes. The distribution, expression, regulation mode and sensitivity to inhibitors of each subtype are different, and they are involved in cancer, inflammation, asthma, depression, erectile dysfunction and other pathological processes of development. A large number of studies have shown that PDEs play an important role in the development of tumors by affecting the intracellular level of cAMP and/or cGMP and PDEs could become diagnostic markers or therapeutic targets. This review will give a brief overview of the expression and regulation of PDE families in the process of tumorigenesis and their anti-tumor inhibitors, which may guide the design of novel therapeutic drugs targeting PDEs for anticancer agent.
Collapse
Affiliation(s)
- Ting Peng
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jun Gong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yongzhe Jin
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yanping Zhou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xin Wei
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lan Bai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
63
|
Movsesian M, Ahmad F, Hirsch E. Functions of PDE3 Isoforms in Cardiac Muscle. J Cardiovasc Dev Dis 2018; 5:jcdd5010010. [PMID: 29415428 PMCID: PMC5872358 DOI: 10.3390/jcdd5010010] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/21/2022] Open
Abstract
Isoforms in the PDE3 family of cyclic nucleotide phosphodiesterases have important roles in cyclic nucleotide-mediated signalling in cardiac myocytes. These enzymes are targeted by inhibitors used to increase contractility in patients with heart failure, with a combination of beneficial and adverse effects on clinical outcomes. This review covers relevant aspects of the molecular biology of the isoforms that have been identified in cardiac myocytes; the roles of these enzymes in modulating cAMP-mediated signalling and the processes mediated thereby; and the potential for targeting these enzymes to improve the profile of clinical responses.
Collapse
Affiliation(s)
- Matthew Movsesian
- Department of Internal Medicine/Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT 841132, USA.
| | - Faiyaz Ahmad
- Vascular Biology and Hypertension Branch, Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA.
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Center for Molecular Biotechnology, University of Turin, 10126 Turin, Italy.
| |
Collapse
|
64
|
Leroy J, Vandecasteele G, Fischmeister R. Cyclic AMP signaling in cardiac myocytes. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
65
|
Fertig BA, Baillie GS. PDE4-Mediated cAMP Signalling. J Cardiovasc Dev Dis 2018; 5:jcdd5010008. [PMID: 29385021 PMCID: PMC5872356 DOI: 10.3390/jcdd5010008] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/18/2018] [Accepted: 01/23/2018] [Indexed: 12/20/2022] Open
Abstract
cAMP is the archetypal and ubiquitous second messenger utilised for the fine control of many cardiovascular cell signalling systems. The ability of cAMP to elicit cell surface receptor-specific responses relies on its compartmentalisation by cAMP hydrolysing enzymes known as phosphodiesterases. One family of these enzymes, PDE4, is particularly important in the cardiovascular system, where it has been extensively studied and shown to orchestrate complex, localised signalling that underpins many crucial functions of the heart. In the cardiac myocyte, cAMP activates PKA, which phosphorylates a small subset of mostly sarcoplasmic substrate proteins that drive β-adrenergic enhancement of cardiac function. The phosphorylation of these substrates, many of which are involved in cardiac excitation-contraction coupling, has been shown to be tightly regulated by highly localised pools of individual PDE4 isoforms. The spatial and temporal regulation of cardiac signalling is made possible by the formation of macromolecular “signalosomes”, which often include a cAMP effector, such as PKA, its substrate, PDE4 and an anchoring protein such as an AKAP. Studies described in the present review highlight the importance of this relationship for individual cardiac PKA substrates and we provide an overview of how this signalling paradigm is coordinated to promote efficient adrenergic enhancement of cardiac function. The role of PDE4 also extends to the vascular endothelium, where it regulates vascular permeability and barrier function. In this distinct location, PDE4 interacts with adherens junctions to regulate their stability. These highly specific, non-redundant roles for PDE4 isoforms have far reaching therapeutic potential. PDE inhibitors in the clinic have been plagued with problems due to the active site-directed nature of the compounds which concomitantly attenuate PDE activity in all highly localised “signalosomes”.
Collapse
|
66
|
Pavlaki N, Nikolaev VO. Imaging of PDE2- and PDE3-Mediated cGMP-to-cAMP Cross-Talk in Cardiomyocytes. J Cardiovasc Dev Dis 2018; 5:jcdd5010004. [PMID: 29367582 PMCID: PMC5872352 DOI: 10.3390/jcdd5010004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/13/2022] Open
Abstract
Cyclic nucleotides 3′,5′-cyclic adenosine monophosphate (cAMP) and 3′,5′-cyclic guanosine monophosphate (cGMP) are important second messengers that regulate cardiovascular function and disease by acting in discrete subcellular microdomains. Signaling compartmentation at these locations is often regulated by phosphodiesterases (PDEs). Some PDEs are also involved in the cross-talk between the two second messengers. The purpose of this review is to summarize and highlight recent findings about the role of PDE2 and PDE3 in cardiomyocyte cyclic nucleotide compartmentation and visualization of this process using live cell imaging techniques.
Collapse
Affiliation(s)
- Nikoleta Pavlaki
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany.
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany.
| |
Collapse
|
67
|
Cardiac Phosphodiesterases and Their Modulation for Treating Heart Disease. Handb Exp Pharmacol 2017; 243:249-269. [PMID: 27787716 DOI: 10.1007/164_2016_82] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
An important hallmark of cardiac failure is abnormal second messenger signaling due to impaired synthesis and catabolism of cyclic adenosine 3',5'- monophosphate (cAMP) and cyclic guanosine 3',5'- monophosphate (cGMP). Their dysregulation, altered intracellular targeting, and blunted responsiveness to stimulating pathways all contribute to pathological remodeling, muscle dysfunction, reduced cell survival and metabolism, and other abnormalities. Therapeutic enhancement of either cyclic nucleotides can be achieved by stimulating their synthesis and/or by suppressing members of the family of cyclic nucleotide phosphodiesterases (PDEs). The heart expresses seven of the eleven major PDE subtypes - PDE1, 2, 3, 4, 5, 8, and 9. Their differential control over cAMP and cGMP signaling in various cell types, including cardiomyocytes, provides intriguing therapeutic opportunities to counter heart disease. This review examines the roles of these PDEs in the failing and hypertrophied heart and summarizes experimental and clinical data that have explored the utility of targeted PDE inhibition.
Collapse
|
68
|
Barone I, Giordano C, Bonofiglio D, Andò S, Catalano S. Phosphodiesterase type 5 and cancers: progress and challenges. Oncotarget 2017; 8:99179-99202. [PMID: 29228762 PMCID: PMC5716802 DOI: 10.18632/oncotarget.21837] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/23/2017] [Indexed: 01/05/2023] Open
Abstract
Cancers are an extraordinarily heterogeneous collection of diseases with distinct genetic profiles and biological features that directly influence response patterns to various treatment strategies as well as clinical outcomes. Nevertheless, our growing understanding of cancer cell biology and tumor progression is gradually leading towards rational, tailored medical treatments designed to destroy cancer cells by exploiting the unique cellular pathways that distinguish them from normal healthy counterparts. Recently, inhibition of the activity of phosphodiesterase type 5 (PDE5) is emerging as a promising approach to restore normal intracellular cyclic guanosine monophosphate (cGMP) signalling, and thereby resulting into the activation of various downstream molecules to inhibit proliferation, motility and invasion of certain cancer cells. In this review, we present an overview of the experimental and clinical evidences highlighting the role of PDE5 in the pathogenesis and prevention of various malignancies. Current data are still not sufficient to draw conclusive statements for cancer patient management, but could provide further rational for testing PDE5-targeting drugs as anticancer agents in clinical settings.
Collapse
Affiliation(s)
- Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Cinzia Giordano
- Centro Sanitario, University of Calabria, Arcavacata di Rende, CS, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
69
|
Shafiee-Nick R, Afshari AR, Mousavi SH, Rafighdoust A, Askari VR, Mollazadeh H, Fanoudi S, Mohtashami E, Rahimi VB, Mohebbi M, Vahedi MM. A comprehensive review on the potential therapeutic benefits of phosphodiesterase inhibitors on cardiovascular diseases. Biomed Pharmacother 2017; 94:541-556. [PMID: 28779712 DOI: 10.1016/j.biopha.2017.07.084] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/02/2017] [Accepted: 07/19/2017] [Indexed: 12/18/2022] Open
Abstract
Phosphodiesterases are a group of enzymes that hydrolyze cyclic nucleotides, which assume a key role in directing intracellular levels of the second messengers' cAMP and cGMP, and consequently cell function. The disclosure of 11 isoenzyme families and our expanded knowledge of their functions at the cell and molecular level stimulate the improvement of isoenzyme selective inhibitors for the treatment of various diseases, particularly cardiovascular diseases. Hence, future and new mechanistic investigations and carefully designed clinical trials could help reap additional benefits of natural/synthetic PDE inhibitors for cardiovascular disease in patients. This review has concentrated on the potential therapeutic benefits of phosphodiesterase inhibitors on cardiovascular diseases.
Collapse
Affiliation(s)
- Reza Shafiee-Nick
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbasali Rafighdoust
- Department of Cardiology, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Sahar Fanoudi
- Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Mohtashami
- Department of Pharmacodynamic and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Mohebbi
- Department of Internal Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mahdi Vahedi
- Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Health Promotion Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
70
|
Parks RJ, Bogachev O, Mackasey M, Ray G, Rose RA, Howlett SE. The impact of ovariectomy on cardiac excitation-contraction coupling is mediated through cAMP/PKA-dependent mechanisms. J Mol Cell Cardiol 2017; 111:51-60. [PMID: 28778766 DOI: 10.1016/j.yjmcc.2017.07.118] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/11/2017] [Accepted: 07/28/2017] [Indexed: 12/18/2022]
Abstract
Ovariectomy (OVX) promotes sarcoplasmic reticulum (SR) Ca2+ overload in ventricular myocytes. We hypothesized that the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway contributes to this Ca2+ dysregulation. Myocytes were isolated from adult female C57BL/6 mice following either OVX or sham surgery (surgery at ≈1mos). Contractions, Ca2+ concentrations (fura-2) and ionic currents were measured simultaneously (37°C, 2Hz) in voltage-clamped myocytes. Intracellular cAMP levels were determined with an enzyme immunoassay; phosphodiesterase (PDE) and adenylyl cyclase (AC) isoform expression was examined with qPCR. Ca2+ currents were similar in myocytes from sham and OVX mice but Ca2+ transients, excitation-contraction (EC)-coupling gain, SR content and contractions were larger in OVX than sham cells. To determine if the cAMP/PKA pathway mediated OVX-induced alterations in EC-coupling, cardiomyocytes were incubated with the PKA inhibitor H-89 (2μM), which abolished baseline differences. While basal intracellular cAMP did not differ, levels were higher in OVX than sham in the presence of a non-selective PDE inhibitor (300μM IBMX), or an AC activator (10μM forskolin). This suggests the production of cAMP by AC and its breakdown by PDE were enhanced by OVX. Consistent with this, mRNA levels for both AC5 and PDE4A were higher in OVX in comparison to sham. Differences in Ca2+ homeostasis and contractions were abolished when sham and OVX cells were dialyzed with patch pipettes containing the same concentration of 8-bromoadenosine-cAMP (50μM). Interestingly, selective inhibition of PDE4 increased Ca2+ current only in OVX cells. Together, these findings suggest that estrogen suppresses SR Ca2+ release and that this is regulated, at least in part, by the cAMP/PKA pathway. These changes in the cAMP/PKA pathway may promote Ca2+ dysregulation and cardiovascular disease when ovarian estrogen levels fall. These results advance our understanding of female-specific cardiomyocyte mechanisms that may affect responses to therapeutic interventions in older women.
Collapse
Affiliation(s)
- Randi J Parks
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, 5850 College Street, P.O. Box 15000, Halifax B3H 4R2, Nova Scotia, Canada.
| | - Oleg Bogachev
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, 5850 College Street, P.O. Box 15000, Halifax B3H 4R2, Nova Scotia, Canada.
| | - Martin Mackasey
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, 5850 College Street, P.O. Box 15000, Halifax B3H 4R2, Nova Scotia, Canada.
| | - Gibanananda Ray
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, 5850 College Street, P.O. Box 15000, Halifax B3H 4R2, Nova Scotia, Canada.
| | - Robert A Rose
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, 5850 College Street, P.O. Box 15000, Halifax B3H 4R2, Nova Scotia, Canada.
| | - Susan E Howlett
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, 5850 College Street, P.O. Box 15000, Halifax B3H 4R2, Nova Scotia, Canada; Department of Medicine (Geriatric Medicine), Faculty of Medicine, Dalhousie University, 5850 College Street, P.O. Box 15000, Halifax B3H 4R2, Nova Scotia, Canada.
| |
Collapse
|
71
|
Components of the mitochondrial cAMP signalosome. Biochem Soc Trans 2017; 45:269-274. [PMID: 28202681 DOI: 10.1042/bst20160394] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 12/25/2022]
Abstract
3'-5'-Cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signalling is activated by different extracellular stimuli and mediates many diverse processes within the same cell. It is now well established that in order to translate into the appropriate cellular function multiple extracellular inputs, which may act simultaneously on the same cell, the cAMP/PKA signalling pathway is compartmentalised. Multimolecular complexes are organised at specific subcellular sites to generate spatially confined signalosomes, which include effectors, modulators and targets of the pathway. In recent years, it has become evident that mitochondria represent sites of compartmentalised cAMP signalling. However, the exact location and the molecular composition of distinct mitochondria signalosomes and their function remain largely unknown. In this review, we focus on individual components of the cAMP/PKA signalling pathway at distinct mitochondria subdomains represented by the outer and inner mitochondrial membranes, the intermembrane space and the matrix, highlighting some of the questions that remain unanswered.
Collapse
|
72
|
Sánchez-Arias JA, Rabal O, Cuadrado-Tejedor M, de Miguel I, Pérez-González M, Ugarte A, Sáez E, Espelosin M, Ursua S, Haizhong T, Wei W, Musheng X, Garcia-Osta A, Oyarzabal J. Impact of Scaffold Exploration on Novel Dual-Acting Histone Deacetylases and Phosphodiesterase 5 Inhibitors for the Treatment of Alzheimer's Disease. ACS Chem Neurosci 2017; 8:638-661. [PMID: 27936591 DOI: 10.1021/acschemneuro.6b00370] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
A novel systems therapeutics approach, involving simultaneous inhibition of phosphodiesterase 5 (PDE5) and histone deacetylase (HDAC), has been validated as a potentially novel therapeutic strategy for the treatment of Alzheimer's disease (AD). First-in-class dual inhibitors bearing a sildenafil core have been very recently reported, and the lead molecule 7 has proven this strategy in AD animal models. Because scaffolds may play a critical role in primary activities and ADME-Tox profiling as well as on intellectual property, we have explored alternative scaffolds (vardenafil- and tadalafil-based cores) and evaluated their impact on critical parameters such as primary activities, permeability, toxicity, and in vivo (pharmacokinetics and functional response in hippocampus) to identify a potential alternative lead molecule bearing a different chemotype for in vivo testing.
Collapse
Affiliation(s)
| | | | - Mar Cuadrado-Tejedor
- Anatomy Department,
School of Medicine, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | | | | | | | | | | | | | - Tan Haizhong
- WuXi Apptec (Tianjin) Co. Ltd., TEDA,
No. 111 HuangHai Road, fourth Avenue, Tianjin 300456, PR China
| | - Wu Wei
- WuXi Apptec (Tianjin) Co. Ltd., TEDA,
No. 111 HuangHai Road, fourth Avenue, Tianjin 300456, PR China
| | - Xu Musheng
- WuXi Apptec (Tianjin) Co. Ltd., TEDA,
No. 111 HuangHai Road, fourth Avenue, Tianjin 300456, PR China
| | | | | |
Collapse
|
73
|
Johnson R, Dludla PV, Muller CJF, Huisamen B, Essop MF, Louw J. The Transcription Profile Unveils the Cardioprotective Effect of Aspalathin against Lipid Toxicity in an In Vitro H9c2 Model. Molecules 2017; 22:molecules22020219. [PMID: 28146135 PMCID: PMC6155936 DOI: 10.3390/molecules22020219] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/25/2017] [Indexed: 01/14/2023] Open
Abstract
Aspalathin, a C-glucosyl dihydrochalcone, has previously been shown to protect cardiomyocytes against hyperglycemia-induced shifts in substrate preference and subsequent apoptosis. However, the precise gene regulatory network remains to be elucidated. To unravel the mechanism and provide insight into this supposition, the direct effect of aspalathin in an isolated cell-based system, without the influence of any variables, was tested using an H9c2 cardiomyocyte model. Cardiomyocytes were exposed to high glucose (33 mM) for 48 h before post-treatment with or without aspalathin. Thereafter, RNA was extracted and RT2 PCR Profiler Arrays were used to profile the expression of 336 genes. Results showed that, 57 genes were differentially regulated in the high glucose or high glucose and aspalathin treated groups. Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) analysis revealed lipid metabolism and molecular transport as the biological processes altered after high glucose treatment, followed by inflammation and apoptosis. Aspalathin was able to modulate key regulators associated with lipid metabolism (Adipoq, Apob, CD36, Cpt1, Pparγ, Srebf1/2, Scd1 and Vldlr), insulin resistance (Igf1, Akt1, Pde3 and Map2k1), inflammation (Il3, Il6, Jak2, Lepr, Socs3, and Tnf13) and apoptosis (Bcl2 and Chuk). Collectively, our results suggest that aspalathin could reverse metabolic abnormalities by activating Adipoq while modulating the expression of Pparγ and Srebf1/2, decreasing inflammation via Il6/Jak2 pathway, which together with an observed increased expression of Bcl2 prevents myocardium apoptosis.
Collapse
Affiliation(s)
- Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council (MRC), Tygerberg 7505, South Africa.
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.
| | - Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council (MRC), Tygerberg 7505, South Africa.
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council (MRC), Tygerberg 7505, South Africa.
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.
- Department of Biochemistry and Microbiology, University of Zululand, Kwadlangezwa 3886, South Africa.
| | - Barbara Huisamen
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council (MRC), Tygerberg 7505, South Africa.
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.
| | - M Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7599, South Africa.
| | - Johan Louw
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council (MRC), Tygerberg 7505, South Africa.
- Department of Biochemistry and Microbiology, University of Zululand, Kwadlangezwa 3886, South Africa.
| |
Collapse
|
74
|
Bedioune I, Bobin P, Leroy J, Fischmeister R, Vandecasteele G. Cyclic Nucleotide Phosphodiesterases and Compartmentation in Normal and Diseased Heart. MICRODOMAINS IN THE CARDIOVASCULAR SYSTEM 2017. [DOI: 10.1007/978-3-319-54579-0_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
75
|
Subcellular Targeting of PDE4 in Cardiac Myocytes and Generation of Signaling Compartments. MICRODOMAINS IN THE CARDIOVASCULAR SYSTEM 2017. [DOI: 10.1007/978-3-319-54579-0_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
76
|
Kokkonen K, Kass DA. Nanodomain Regulation of Cardiac Cyclic Nucleotide Signaling by Phosphodiesterases. Annu Rev Pharmacol Toxicol 2016; 57:455-479. [PMID: 27732797 DOI: 10.1146/annurev-pharmtox-010716-104756] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) form an 11-member superfamily comprising 100 different isoforms that regulate the second messengers cyclic adenosine or guanosine 3',5'-monophosphate (cAMP or cGMP). These PDE isoforms differ with respect to substrate selectivity and their localized control of cAMP and cGMP within nanodomains that target specific cellular pools and synthesis pathways for the cyclic nucleotides. Seven PDE family members are physiologically relevant to regulating cardiac function, disease remodeling of the heart, or both: PDE1 and PDE2, both dual-substrate (cAMP and cGMP) esterases; PDE3, PDE4, and PDE8, which principally hydrolyze cAMP; and PDE5A and PDE9A, which target cGMP. New insights regarding the different roles of PDEs in health and disease and their local signaling control are broadening the potential therapeutic utility for PDE-selective inhibitors. In this review, we discuss these PDEs, focusing on the different mechanisms by which they control cardiac function in health and disease by regulating intracellular nanodomains.
Collapse
Affiliation(s)
- Kristen Kokkonen
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; .,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
77
|
Lin YK, Chen YC, Chen YA, Yeh YH, Chen SA, Chen YJ. B-Type Natriuretic Peptide Modulates Pulmonary Vein Arrhythmogenesis: A Novel Potential Contributor to the Genesis of Atrial Tachyarrhythmia in Heart Failure. J Cardiovasc Electrophysiol 2016; 27:1462-1471. [PMID: 27571932 DOI: 10.1111/jce.13093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 08/11/2016] [Accepted: 08/25/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Heart failure (HF) plays a critical role in the genesis of atrial fibrillation (AF). A high B-type natriuretic peptide (BNP) level occurs in patients with HF and in patients with AF. However, the role of BNP in the pathophysiology of AF is not clear. The purposes of this study were to evaluate the effects of BNP on pulmonary vein (PV) arrhythmogenesis. METHODS AND RESULTS Whole-cell patch clamp and fluorescence were used to study the action potential, ionic currents, and calcium homeostasis in isolated single rabbit PV cardiomyocytes before and after a BNP infusion, with or without ODQ (10 μM), milrinone (50 μM), or ouabain (1 μM). BNP increased PV spontaneous activity by 28.2 ± 7.5% at 100 nM and by 23.8 ± 9.1% at 300 nM. Similar to those with BNP, milrinone 50 μM increased the PV beating rate from 3.0 ± 0.2 to 3.6 ± 0.3 Hz (P < 0.0005, n = 7). In the presence of ODQ application, BNP didn't change PV spontaneous activity. BNP (100 nM) increased calcium transients (F/F0 from 1.6 ± 0.1 to 1.9 ± 0.2, n = 20, P < 0.05) and increased the pacemaker current (0.4 ± 0.1 to 1.0 ± 0.2 pA/pF, n = 17, P < 0.0005) in PV cardiomyocytes. Moreover, BNP (100 nM) increased the transient inward current, sodium currents, sodium-calcium exchanger currents, and L-type calcium current; but reduced late sodium currents and the Na-K pump in PV cardiomyocytes. CONCLUSION BNP increases PV arrhythmogenesis, which may contribute to the genesis of atrial tachyarrhythmogenesis in HF. Cyclic GMP activation, phosphodiesterase 3 inhibition and Na+ /K+ -ATPase inhibition might participate in the BNP modulation of PV electrophysiology.
Collapse
Affiliation(s)
- Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan.,Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Ann Chen
- Division of Nephrology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Yung-Hsin Yeh
- The First Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University, Taoyuan, Taiwan
| | - Shih-Ann Chen
- School of Medicine, National Yang-Ming University; Division of Cardiology and Cardiovascular Research Center, Veterans General Hospital-Taipei, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
78
|
Rabal O, Sánchez-Arias JA, Cuadrado-Tejedor M, de Miguel I, Pérez-González M, García-Barroso C, Ugarte A, Estella-Hermoso de Mendoza A, Sáez E, Espelosin M, Ursua S, Haizhong T, Wei W, Musheng X, Garcia-Osta A, Oyarzabal J. Design, Synthesis, and Biological Evaluation of First-in-Class Dual Acting Histone Deacetylases (HDACs) and Phosphodiesterase 5 (PDE5) Inhibitors for the Treatment of Alzheimer’s Disease. J Med Chem 2016; 59:8967-9004. [DOI: 10.1021/acs.jmedchem.6b00908] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - Mar Cuadrado-Tejedor
- Anatomy Department, School of Medicine, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | | | | | | | | | | | | | | | | | - Tan Haizhong
- WuXi Apptec (Tianjin) Co. Ltd.,
TEDA, No. 111 HuangHai Road, Fourth
Avenue, Tianjin 300456, PR China
| | - Wu Wei
- WuXi Apptec (Tianjin) Co. Ltd.,
TEDA, No. 111 HuangHai Road, Fourth
Avenue, Tianjin 300456, PR China
| | - Xu Musheng
- WuXi Apptec (Tianjin) Co. Ltd.,
TEDA, No. 111 HuangHai Road, Fourth
Avenue, Tianjin 300456, PR China
| | | | | |
Collapse
|
79
|
Effects of heterologous expression of human cyclic nucleotide phosphodiesterase 3A (hPDE3A) on redox regulation in yeast. Biochem J 2016; 473:4205-4225. [PMID: 27647936 DOI: 10.1042/bcj20160572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/07/2016] [Accepted: 09/19/2016] [Indexed: 01/11/2023]
Abstract
Oxidative stress plays a pivotal role in pathogenesis of cardiovascular diseases and diabetes; however, the roles of protein kinase A (PKA) and human phosphodiesterase 3A (hPDE3A) remain unknown. Here, we show that yeast expressing wild-type (WT) hPDE3A or K13R hPDE3A (putative ubiquitinylation site mutant) exhibited resistance or sensitivity to exogenous hydrogen peroxide (H2O2), respectively. H2O2-stimulated ROS production was markedly increased in yeast expressing K13R hPDE3A (Oxidative stress Sensitive 1, OxiS1), compared with yeast expressing WT hPDE3A (Oxidative stress Resistant 1, OxiR1). In OxiR1, YAP1 and YAP1-dependent antioxidant genes were up-regulated, accompanied by a reduction in thioredoxin peroxidase. In OxiS1, expression of YAP1 and YAP1-dependent genes was impaired, and the thioredoxin system malfunctioned. H2O2 increased cyclic adenosine monophosphate (cAMP)-hydrolyzing activity of WT hPDE3A, but not K13R hPDE3A, through PKA-dependent phosphorylation of hPDE3A, which was correlated with its ubiquitinylation. The changes in antioxidant gene expression did not directly correlate with differences in cAMP-PKA signaling. Despite differences in their capacities to hydrolyze cAMP, total cAMP levels among OxiR1, OxiS1, and mock were similar; PKA activity, however, was lower in OxiS1 than in OxiR1 or mock. During exposure to H2O2, however, Sch9p activity, a target of Rapamycin complex 1-regulated Rps6 kinase and negative-regulator of PKA, was rapidly reduced in OxiR1, and Tpk1p, a PKA catalytic subunit, was diffusely spread throughout the cytosol, with PKA activation. In OxiS1, Sch9p activity was unchanged during exposure to H2O2, consistent with reduced activation of PKA. These results suggest that, during oxidative stress, TOR-Sch9 signaling might regulate PKA activity, and that post-translational modifications of hPDE3A are critical in its regulation of cellular recovery from oxidative stress.
Collapse
|
80
|
Corradini E, Klaasse G, Leurs U, Heck AJR, Martin NI, Scholten A. Charting the interactome of PDE3A in human cells using an IBMX based chemical proteomics approach. MOLECULAR BIOSYSTEMS 2016. [PMID: 26205238 DOI: 10.1039/c5mb00142k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In the cell the second messenger cyclic nucleotides cAMP and cGMP mediate a wide variety of external signals. Both signaling molecules are degraded by the superfamily of phosphodiesterases (PDEs) consisting of more than 50 different isoforms. Several of these PDEs are implicated in disease processes inspiring the quest for and synthesis of selective PDE inhibitors, that unfortunately have led to very mixed successes in clinical trials. This may be partially caused by their pharmacological action. Accumulating data suggests that small differences between different PDE isoforms may already result in specific tissue distributions, cellular localization and different involvement in higher order signal protein complexes. The role of PDEs in these higher order signal protein complexes has only been marginally addressed, as no screening methodology is available to address this in a more comprehensive way. Affinity based chemical proteomics is a relatively new tool to identify specific protein-protein interactions. Here, to study the interactome of PDEs, we synthesized a broad spectrum PDE-capturing resin based on the non-selective PDE inhibitor 3-isobutyl-1-methylxanthine (IBMX). Chemical proteomics characterization of this resin in HeLa cell lysates led to the capture of several different PDEs. Combining the IBMX-resin with in-solution competition with the available more selective PDE inhibitors, cilostamide and papaverine, allowed us to selectively probe the interactome of PDE3A in HeLa cells. Besides known interactors such as the family of 14-3-3 proteins, PDE3A was found to associate with a PP2A complex composed of a regulatory, scaffold and catalytic subunit.
Collapse
Affiliation(s)
- Eleonora Corradini
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science Faculty, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
81
|
Bobin P, Belacel-Ouari M, Bedioune I, Zhang L, Leroy J, Leblais V, Fischmeister R, Vandecasteele G. Cyclic nucleotide phosphodiesterases in heart and vessels: A therapeutic perspective. Arch Cardiovasc Dis 2016; 109:431-43. [PMID: 27184830 DOI: 10.1016/j.acvd.2016.02.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/28/2016] [Accepted: 02/02/2016] [Indexed: 01/21/2023]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) degrade the second messengers cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), thereby regulating multiple aspects of cardiac and vascular muscle functions. This highly diverse class of enzymes encoded by 21 genes encompasses 11 families that are not only responsible for the termination of cyclic nucleotide signalling, but are also involved in the generation of dynamic microdomains of cAMP and cGMP, controlling specific cell functions in response to various neurohormonal stimuli. In the myocardium and vascular smooth muscle, the PDE3 and PDE4 families predominate, degrading cAMP and thereby regulating cardiac excitation-contraction coupling and smooth muscle contractile tone. PDE3 inhibitors are positive inotropes and vasodilators in humans, but their use is limited to acute heart failure and intermittent claudication. PDE5 is particularly important for the degradation of cGMP in vascular smooth muscle, and PDE5 inhibitors are used to treat erectile dysfunction and pulmonary hypertension. There is experimental evidence that these PDEs, as well as other PDE families, including PDE1, PDE2 and PDE9, may play important roles in cardiac diseases, such as hypertrophy and heart failure, as well as several vascular diseases. After a brief presentation of the cyclic nucleotide pathways in cardiac and vascular cells, and the major characteristics of the PDE superfamily, this review will focus on the current use of PDE inhibitors in cardiovascular diseases, and the recent research developments that could lead to better exploitation of the therapeutic potential of these enzymes in the future.
Collapse
Affiliation(s)
- Pierre Bobin
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Milia Belacel-Ouari
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Ibrahim Bedioune
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Liang Zhang
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Jérôme Leroy
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Véronique Leblais
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Rodolphe Fischmeister
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France.
| | - Grégoire Vandecasteele
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France.
| |
Collapse
|
82
|
Movsesian M. Novel approaches to targeting PDE3 in cardiovascular disease. Pharmacol Ther 2016; 163:74-81. [PMID: 27108947 DOI: 10.1016/j.pharmthera.2016.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 03/18/2016] [Indexed: 10/24/2022]
Abstract
Inhibitors of PDE3, a family of dual-specificity cyclic nucleotide phosphodiesterases, are used clinically to increase cardiac contractility by raising intracellular cAMP content in cardiac myocytes and to reduce vascular resistance by increasing intracellular cGMP content in vascular smooth muscle myocytes. When used in the treatment of patients with heart failure, PDE3 inhibitors are effective in the acute setting but increase sudden cardiac death with long-term administration, possibly reflecting pro-apoptotic and pro-hypertrophic consequences of increased cAMP-mediated signaling in cardiac myocytes. cAMP-mediated signaling in cardiac myocytes is highly compartmentalized, and different phosphodiesterases, by controlling cAMP content in functionally discrete intracellular microcompartments, regulate different cAMP-mediated pathways. Four variants/isoforms of PDE3 (PDE3A1, PDE3A2, PDE3A3, and PDE3B) are expressed in cardiac myocytes, and new experimental results have demonstrated that these isoforms, which are differentially localized intracellularly through unique protein-protein interactions, control different physiologic responses. While the catalytic regions of these isoforms may be too similar to allow the catalytic activity of each isoform to be selectively inhibited, targeting their unique protein-protein interactions may allow desired responses to be elicited without the adverse consequences that limit the usefulness of existing PDE3 inhibitors.
Collapse
Affiliation(s)
- Matthew Movsesian
- VA Salt Lake City Health Care System, Salt Lake City, UT, USA; University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
83
|
Abstract
In the heart, Ca(2+) signals regulate a variety of biological functions ranging from contractility to gene expression, cellular hypertrophy and death. In this review, we summarize the role of local Ca(2+) homeostasis in these processes in healthy cardiac muscle cells, and highlight how mismanaged Ca(2+) handling contributes to the pathophysiology of conditions such as cardiac arrhythmia, ischemic heart disease, cardiac hypertrophy and heart failure. Aiming to provide an introduction to the field with a clinical perspective, we also indicate how current and future therapies may modulate cardiomyocytes Ca(2+) handling for the treatment of patients.
Collapse
Affiliation(s)
| | - William E Louch
- b Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway
| | - Ivar Sjaastad
- b Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo , Oslo , Norway
| |
Collapse
|
84
|
Bobin P, Varin A, Lefebvre F, Fischmeister R, Vandecasteele G, Leroy J. Calmodulin kinase II inhibition limits the pro-arrhythmic Ca2+ waves induced by cAMP-phosphodiesterase inhibitors. Cardiovasc Res 2016; 110:151-61. [PMID: 26851245 DOI: 10.1093/cvr/cvw027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 01/22/2016] [Indexed: 01/29/2023] Open
Abstract
AIMS A major concern of using phosphodiesterase (PDE) inhibitors in heart failure is their potential to increase mortality by inducing arrhythmias. By diminishing cyclic adenosine monophosphate (cAMP) hydrolysis, they promote protein kinase A (PKA) activity under β-adrenergic receptor (β-AR) stimulation, hence enhancing Ca(2+) cycling and contraction. Yet, cAMP also activates CaMKII via PKA or the exchange protein Epac, but it remains unknown whether these pathways are involved in the pro-arrhythmic effect of PDE inhibitors. METHODS AND RESULTS Excitation-contraction coupling was investigated in isolated adult rat ventricular myocytes loaded with Fura-2 and paced at 1 Hz allowing coincident measurement of intracellular Ca(2+) and sarcomere shortening. The PDE4 inhibitor Ro 20-1724 (Ro) promoted the inotropic effects of the non-selective β-AR agonist isoprenaline (Iso) and also spontaneous diastolic Ca(2+) waves (SCWs). PDE4 inhibition potentiated RyR2 and PLB phosphorylation at specific PKA and CaMKII sites increasing sarcoplasmic reticulum (SR) Ca(2+) load and SR Ca(2+) leak measured in a 0Na(+)/0Ca(2+) solution ± tetracaine. PKA inhibition suppressed all the effects of Iso ± Ro, whereas CaMKII inhibition prevented SR Ca(2+) leak and diminished SCW incidence without affecting the inotropic effects of Ro. Inhibition of Epac2 but not Epac1 diminished the occurrence of SCWs. PDE3 inhibition with cilostamide induced an SR Ca(2+) leak, which was also blocked by CaMKII inhibition. CONCLUSION Our results show that PDE inhibitors exert inotropic effects via PKA but lead to SCWs via both PKA and CaMKII activation partly via Epac2, suggesting the potential use of CaMKII inhibitors as adjuncts to PDE inhibition to limit their pro-arrhythmic effects.
Collapse
Affiliation(s)
- Pierre Bobin
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Audrey Varin
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Florence Lefebvre
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Rodolphe Fischmeister
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Grégoire Vandecasteele
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| | - Jérôme Leroy
- Inserm, UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, F-92296, Châtenay-Malabry, France
| |
Collapse
|
85
|
New pharmacologic interventions to increase cardiac contractility: challenges and opportunities. Curr Opin Cardiol 2015; 30:285-91. [PMID: 25807221 DOI: 10.1097/hco.0000000000000165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The most extensively studied inotropic agents in patients with heart failure are phosphodiesterase (PDE) 3 inhibitors, which increase contractility by raising intracellular cyclic adenosine monophosphate content. In clinical trials, the inotropic benefits of these agents have been outweighed by an increase in sudden cardiac death. Here, I review recent findings that help explain what are likely to be distinct mechanisms involved in the beneficial and adverse effects of PDE3 inhibition. RECENT FINDINGS The proapoptotic consequences of PDE3 inhibition are becoming more apparent. Moreover, it has also become clear that individual PDE3 isoforms in cardiac myocytes are selectively regulated to interact with different proteins in different intracellular compartments. The beneficial and adverse effects of PDE3 inhibition may thus be attributable to the inhibition of different isoforms in different intracellular domains. In particular, PDE3A1 has been shown to interact directly with sarcoplasmic/endoplasmic reticulum Ca ATPase (SERCA2) in the sarcoplasmic reticulum through a phosphorylation of a site in its unique N-terminal domain, making it possible that this isoform can be selectively targeted to increase intracellular Ca cycling. SUMMARY Conventional PDE3 inhibitors target several functionally distinct isoforms of these enzymes. Isoform-selective and/or compartment-selective targeting of PDE3, through its protein-protein interactions, may produce the inotropic benefits of PDE3 inhibition without the adverse consequences.
Collapse
|
86
|
Identification of cancer-cytotoxic modulators of PDE3A by predictive chemogenomics. Nat Chem Biol 2015; 12:102-8. [PMID: 26656089 PMCID: PMC4718766 DOI: 10.1038/nchembio.1984] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/28/2015] [Indexed: 12/28/2022]
Abstract
High cancer death rates indicate the need for new anticancer therapeutic agents. Approaches to discovering new cancer drugs include target-based drug discovery and phenotypic screening. Here, we identified phosphodiesterase 3A modulators as cell-selective cancer cytotoxic compounds through phenotypic compound library screening and target deconvolution by predictive chemogenomics. We found that sensitivity to 6-(4-(diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one, or DNMDP, across 766 cancer cell lines correlates with expression of the gene PDE3A, encoding phosphodiesterase 3A. Like DNMDP, a subset of known PDE3A inhibitors kill selected cancer cells, whereas others do not. Furthermore, PDE3A depletion leads to DNMDP resistance. We demonstrated that DNMDP binding to PDE3A promotes an interaction between PDE3A and Schlafen 12 (SLFN12), suggestive of a neomorphic activity. Coexpression of SLFN12 with PDE3A correlates with DNMDP sensitivity, whereas depletion of SLFN12 results in decreased DNMDP sensitivity. Our results implicate PDE3A modulators as candidate cancer therapeutic agents and demonstrate the power of predictive chemogenomics in small-molecule discovery.
Collapse
|
87
|
Kato N, Loh M, Takeuchi F, Verweij N, Wang X, Zhang W, Kelly TN, Saleheen D, Lehne B, Leach IM, Drong AW, Abbott J, Wahl S, Tan ST, Scott WR, Campanella G, Chadeau-Hyam M, Afzal U, Ahluwalia TS, Bonder MJ, Chen P, Dehghan A, Edwards TL, Esko T, Go MJ, Harris SE, Hartiala J, Kasela S, Kasturiratne A, Khor CC, Kleber ME, Li H, Yu Mok Z, Nakatochi M, Sapari NS, Saxena R, Stewart AFR, Stolk L, Tabara Y, Teh AL, Wu Y, Wu JY, Zhang Y, Aits I, Da Silva Couto Alves A, Das S, Dorajoo R, Hopewell JC, Kim YK, Koivula RW, Luan J, Lyytikäinen LP, Nguyen QN, Pereira MA, Postmus I, Raitakari OT, Bryan MS, Scott RA, Sorice R, Tragante V, Traglia M, White J, Yamamoto K, Zhang Y, Adair LS, Ahmed A, Akiyama K, Asif R, Aung T, Barroso I, Bjonnes A, Braun TR, Cai H, Chang LC, Chen CH, Cheng CY, Chong YS, Collins R, Courtney R, Davies G, Delgado G, Do LD, Doevendans PA, Gansevoort RT, Gao YT, Grammer TB, Grarup N, Grewal J, Gu D, Wander GS, Hartikainen AL, Hazen SL, He J, Heng CK, Hixson JE, Hofman A, Hsu C, Huang W, Husemoen LLN, Hwang JY, et alKato N, Loh M, Takeuchi F, Verweij N, Wang X, Zhang W, Kelly TN, Saleheen D, Lehne B, Leach IM, Drong AW, Abbott J, Wahl S, Tan ST, Scott WR, Campanella G, Chadeau-Hyam M, Afzal U, Ahluwalia TS, Bonder MJ, Chen P, Dehghan A, Edwards TL, Esko T, Go MJ, Harris SE, Hartiala J, Kasela S, Kasturiratne A, Khor CC, Kleber ME, Li H, Yu Mok Z, Nakatochi M, Sapari NS, Saxena R, Stewart AFR, Stolk L, Tabara Y, Teh AL, Wu Y, Wu JY, Zhang Y, Aits I, Da Silva Couto Alves A, Das S, Dorajoo R, Hopewell JC, Kim YK, Koivula RW, Luan J, Lyytikäinen LP, Nguyen QN, Pereira MA, Postmus I, Raitakari OT, Bryan MS, Scott RA, Sorice R, Tragante V, Traglia M, White J, Yamamoto K, Zhang Y, Adair LS, Ahmed A, Akiyama K, Asif R, Aung T, Barroso I, Bjonnes A, Braun TR, Cai H, Chang LC, Chen CH, Cheng CY, Chong YS, Collins R, Courtney R, Davies G, Delgado G, Do LD, Doevendans PA, Gansevoort RT, Gao YT, Grammer TB, Grarup N, Grewal J, Gu D, Wander GS, Hartikainen AL, Hazen SL, He J, Heng CK, Hixson JE, Hofman A, Hsu C, Huang W, Husemoen LLN, Hwang JY, Ichihara S, Igase M, Isono M, Justesen JM, Katsuya T, Kibriya MG, Kim YJ, Kishimoto M, Koh WP, Kohara K, Kumari M, Kwek K, Lee NR, Lee J, Liao J, Lieb W, Liewald DCM, Matsubara T, Matsushita Y, Meitinger T, Mihailov E, Milani L, Mills R, Mononen N, Müller-Nurasyid M, Nabika T, Nakashima E, Ng HK, Nikus K, Nutile T, Ohkubo T, Ohnaka K, Parish S, Paternoster L, Peng H, Peters A, Pham ST, Pinidiyapathirage MJ, Rahman M, Rakugi H, Rolandsson O, Ann Rozario M, Ruggiero D, Sala CF, Sarju R, Shimokawa K, Snieder H, Sparsø T, Spiering W, Starr JM, Stott DJ, Stram DO, Sugiyama T, Szymczak S, Tang WHW, Tong L, Trompet S, Turjanmaa V, Ueshima H, Uitterlinden AG, Umemura S, Vaarasmaki M, van Dam RM, van Gilst WH, van Veldhuisen DJ, Viikari JS, Waldenberger M, Wang Y, Wang A, Wilson R, Wong TY, Xiang YB, Yamaguchi S, Ye X, Young RD, Young TL, Yuan JM, Zhou X, Asselbergs FW, Ciullo M, Clarke R, Deloukas P, Franke A, Franks PW, Franks S, Friedlander Y, Gross MD, Guo Z, Hansen T, Jarvelin MR, Jørgensen T, Jukema JW, kähönen M, Kajio H, Kivimaki M, Lee JY, Lehtimäki T, Linneberg A, Miki T, Pedersen O, Samani NJ, Sørensen TIA, Takayanagi R, Toniolo D, BIOS-consortium, CARDIo GRAMplusCD, LifeLines Cohort Study, The InterAct Consortium, Ahsan H, Allayee H, Chen YT, Danesh J, Deary IJ, Franco OH, Franke L, Heijman BT, Holbrook JD, Isaacs A, Kim BJ, Lin X, Liu J, März W, Metspalu A, Mohlke KL, Sanghera DK, Shu XO, van Meurs JBJ, Vithana E, Wickremasinghe AR, Wijmenga C, Wolffenbuttel BHW, Yokota M, Zheng W, Zhu D, Vineis P, Kyrtopoulos SA, Kleinjans JCS, McCarthy MI, Soong R, Gieger C, Scott J, Teo YY, He J, Elliott P, Tai ES, van der Harst P, Kooner JS, Chambers JC. Trans-ancestry genome-wide association study identifies 12 genetic loci influencing blood pressure and implicates a role for DNA methylation. Nat Genet 2015; 47:1282-1293. [PMID: 26390057 PMCID: PMC4719169 DOI: 10.1038/ng.3405] [Show More Authors] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 08/21/2015] [Indexed: 12/17/2022]
Abstract
We carried out a trans-ancestry genome-wide association and replication study of blood pressure phenotypes among up to 320,251 individuals of East Asian, European and South Asian ancestry. We find genetic variants at 12 new loci to be associated with blood pressure (P = 3.9 × 10(-11) to 5.0 × 10(-21)). The sentinel blood pressure SNPs are enriched for association with DNA methylation at multiple nearby CpG sites, suggesting that, at some of the loci identified, DNA methylation may lie on the regulatory pathway linking sequence variation to blood pressure. The sentinel SNPs at the 12 new loci point to genes involved in vascular smooth muscle (IGFBP3, KCNK3, PDE3A and PRDM6) and renal (ARHGAP24, OSR1, SLC22A7 and TBX2) function. The new and known genetic variants predict increased left ventricular mass, circulating levels of NT-proBNP, and cardiovascular and all-cause mortality (P = 0.04 to 8.6 × 10(-6)). Our results provide new evidence for the role of DNA methylation in blood pressure regulation.
Collapse
Affiliation(s)
- Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Marie Loh
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Xu Wang
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Danish Saleheen
- Center for Non-Communicable Diseases, Karachi, Pakistan
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Irene Mateo Leach
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander W Drong
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - James Abbott
- Bioinformatics Support Service, Imperial College London, London, UK
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sian-Tsung Tan
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - William R Scott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Gianluca Campanella
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Marc Chadeau-Hyam
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Uzma Afzal
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
| | - Tarunveer S Ahluwalia
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood (COSPAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Marc Jan Bonder
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peng Chen
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Todd L Edwards
- Vanderbilt Epidemiology Center, Center for Human Genetics Research, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Division of Endocrinology, Children’s Hospital Boston, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Min Jin Go
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Sarah E Harris
- Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine and Medical Research Council (MRC) Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Jaana Hartiala
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Institute for Genetic Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Silva Kasela
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | | | - Chiea-Chuen Khor
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Genome Institute of Singapore, A*STAR, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore
- Department of Paediatrics, National University of Singapore, Singapore
| | - Marcus E Kleber
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Huaixing Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zuan Yu Mok
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Masahiro Nakatochi
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Nur Sabrina Sapari
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Richa Saxena
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandre F R Stewart
- University of Ottawa Heart Institute, Cardiovascular Research Methods Centre, Ottawa, Ontario, Canada
- Ruddy Canadian Cardiovascular Genetics Centre, Ottawa, Ontario, Canada
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ai Ling Teh
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Singapore
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yi Zhang
- State Key Laboratory of Medical Genetics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Hypertension, Shanghai, China
| | - Imke Aits
- Institute of Epidemiology and Biobank popgen, Christian Albrechts University of Kiel, Kiel, Germany
| | - Alexessander Da Silva Couto Alves
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (PHE) Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Shikta Das
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (PHE) Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | | | - Jemma C Hopewell
- Clinical Trials Support Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yun Kyoung Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Robert W Koivula
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Malmö, Sweden
| | - Jian’an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland
| | - Quang N Nguyen
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam
| | - Mark A Pereira
- School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Iris Postmus
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Molly Scannell Bryan
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Rossella Sorice
- Institute of Genetics and Biophysics A Buzzati-Traverso, CNR, Naples, Italy
| | - Vinicius Tragante
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) ‘Burlo Garofolo’, Trieste, Italy
| | - Jon White
- University College London Genetics Institute, Department of Genetics, Environment and Evolution, University College London, London, UK
| | - Ken Yamamoto
- Division of Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Linda S Adair
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Koichi Akiyama
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Rasheed Asif
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Inês Barroso
- Metabolic Disease Group, Wellcome Trust Sanger Institute, Cambridge, UK
- National Institute for Health Research (NIHR) Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Andrew Bjonnes
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy R Braun
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Hui Cai
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Li-Ching Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Yu Cheng
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Centre for Quantitative Medicine, Office of Clinical Sciences, Duke–National University of Singapore Graduate Medical School, Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Rory Collins
- Clinical Trials Support Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Regina Courtney
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Gail Davies
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Graciela Delgado
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Loi D Do
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam
| | - Pieter A Doevendans
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ron T Gansevoort
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Tanja B Grammer
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Niels Grarup
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jagvir Grewal
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
| | - Dongfeng Gu
- Fu Wai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gurpreet S Wander
- Dayanand Medical College and Hospital Unit, Hero DMC Heart Institute, Ludhiana, India
| | - Anna-Liisa Hartikainen
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- Medical Research Center, University of Oulu, Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Stanley L Hazen
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jing He
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, USA
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, Singapore
| | - James E Hixson
- Human Genetics Center, University of Texas School of Public Health at Houston, Houston, Texas, USA
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Chris Hsu
- University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Wei Huang
- Department of Genetics, Chinese National Human Genomic Center, Shanghai, China
| | - Lise L N Husemoen
- Research Centre for Prevention and Health, Glostrup University Hospital, Glostrup, Denmark
| | - Joo-Yeon Hwang
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Sahoko Ichihara
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Michiya Igase
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masato Isono
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Johanne M Justesen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tomohiro Katsuya
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Muhammad G Kibriya
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Young Jin Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | | | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Duke–National University of Singapore Graduate Medical School, Singapore
| | - Katsuhiko Kohara
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Meena Kumari
- Department of Epidemiology and Public Health, University College London, London, UK
| | | | - Nanette R Lee
- University of San Carlos Office of Population Studies Foundation, University of San Carlos, Cebu City, Philippines
- Department of Anthropology, Sociology and History, University of San Carlos, Cebu City, Philippines
| | - Jeannette Lee
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Jiemin Liao
- Department of Ophthalmology, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank popgen, Christian Albrechts University of Kiel, Kiel, Germany
| | - David C M Liewald
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Tatsuaki Matsubara
- Department of Internal Medicine, Aichi-Gakuin University School of Dentistry, Nagoya, Japan
| | - Yumi Matsushita
- National Center for Global Health and Medicine, Toyama, Japan
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | | | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Rebecca Mills
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, Ludwig Maximilians University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Toru Nabika
- Department of Functional Pathology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Eitaro Nakashima
- Division of Endocrinology and Diabetes, Department of Internal Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Diabetes and Endocrinology, Chubu Rosai Hospital, Nagoya, Japan
| | - Hong Kiat Ng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Kjell Nikus
- Heart Centre, Department of Cardiology, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Teresa Nutile
- Institute of Genetics and Biophysics A Buzzati-Traverso, CNR, Naples, Italy
| | - Takayoshi Ohkubo
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo, Japan
| | - Keizo Ohnaka
- Department of Geriatric Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sarah Parish
- Clinical Trials Support Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | - Hao Peng
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Son T Pham
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam
| | | | - Mahfuzar Rahman
- UChicago Research Bangladesh, Uttara, Dhaka, Bangladesh
- Research and Evaluation Division, Bangladesh Rehabilitation Assistance Committee (BRAC), Dhaka, Bangladesh
| | - Hiromi Rakugi
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Olov Rolandsson
- Department of Public Health and Clinical Medicine, Section for Family Medicine, Umeå Universitet, Umeå, Sweden
| | - Michelle Ann Rozario
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics A Buzzati-Traverso, CNR, Naples, Italy
| | - Cinzia F Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Ralhan Sarju
- Dayanand Medical College and Hospital Unit, Hero DMC Heart Institute, Ludhiana, India
| | - Kazuro Shimokawa
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Thomas Sparsø
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Wilko Spiering
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - John M Starr
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - David J Stott
- Academic Section of Geriatric Medicine, Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, Glasgow, UK
| | - Daniel O Stram
- University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Takao Sugiyama
- Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Silke Szymczak
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lin Tong
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Väinö Turjanmaa
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, Finland
| | - Hirotsugu Ueshima
- Department of Health Science, Shiga University of Medical Science, Otsu, Japan
- Center for Epidemiologic Research in Asia, Shiga University of Medical Science, Otsu, Japan
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Satoshi Umemura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Marja Vaarasmaki
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- Medical Research Center, University of Oulu, Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Wiek H van Gilst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jorma S Viikari
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Medicine, University of Turku, Turku, Finland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Yiqin Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Aili Wang
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Rory Wilson
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Tien-Yin Wong
- Department of Ophthalmology, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Yong-Bing Xiang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Shuhei Yamaguchi
- Third Department of Internal Medicine, Shimane University Faculty of Medicine, Izumo, Japan
| | - Xingwang Ye
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Robin D Young
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Terri L Young
- Neuroscience and Behavioural Disorders (NBD) Program, Duke–National University of Singapore Graduate Medical School, Singapore
- Duke Eye Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Jian-Min Yuan
- Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Xueya Zhou
- Bioinformatics Division, Tsinghua National Laboratory for Informatics Science and Technology (TNLIST), Ministry of Education Key Laboratory of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
- Center for Synthetic and Systems Biology, TNLIST, Ministry of Education Key Laboratory of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
- Department of Psychiatry, University of Hong Kong, Hong Kong
| | - Folkert W Asselbergs
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute of the Netherlands (ICIN)–Netherlands Heart Institute, Utrecht, the Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
| | - Marina Ciullo
- Institute of Genetics and Biophysics A Buzzati-Traverso, CNR, Naples, Italy
| | - Robert Clarke
- Clinical Trials Support Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- King Abdulaziz University, Jeddah, Saudi Arabia
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Public Health and Clinical Medicine, Section for Family Medicine, Umeå Universitet, Umeå, Sweden
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Steve Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, UK
| | | | - Myron D Gross
- School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zhirong Guo
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Torben Hansen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marjo-Riitta Jarvelin
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (PHE) Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Center for Life Course Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Torben Jørgensen
- Research Centre for Prevention and Health, Glostrup University Hospital, Glostrup, Denmark
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute of the Netherlands (ICIN)–Netherlands Heart Institute, Utrecht, the Netherlands
- ICIN, Utrecht, the Netherlands
| | - Mika kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, Finland
| | - Hiroshi Kajio
- National Center for Global Health and Medicine, Toyama, Japan
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Jong-Young Lee
- Ministry of Health and Welfare, Seoul, Republic of Korea
- THERAGEN ETEX Bio Institute, Suwon, Republic of Korea
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup University Hospital, Glostrup, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tetsuro Miki
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Oluf Pedersen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Ryoichi Takayanagi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Kyushu, Japan
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- Institute of Molecular Genetics, National Research Council (CNR), Pavia, Italy
| | | | | | | | | | - Habibul Ahsan
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Hooman Allayee
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Institute for Genetic Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - John Danesh
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, UK
| | - Ian J Deary
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bastiaan T Heijman
- Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Aaron Isaacs
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bong-Jo Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianjun Liu
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Genome Institute of Singapore, A*STAR, Singapore
| | - Winfried März
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Synlab Academy, Synlab Services, Mannheim, Germany
| | | | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Dharambir K Sanghera
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Xiao-Ou Shu
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Eranga Vithana
- Department of Ophthalmology, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Neuroscience and Behavioural Disorders (NBD) Program, Duke–National University of Singapore Graduate Medical School, Singapore
| | | | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bruce H W Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mitsuhiro Yokota
- Department of Genome Science, Aichi-Gakuin University School of Dentistry, Nagoya, Japan
| | - Wei Zheng
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Dingliang Zhu
- State Key Laboratory of Medical Genetics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Hypertension, Shanghai, China
| | - Paolo Vineis
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Soterios A Kyrtopoulos
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, Athens, Greece
| | - Jos C S Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, the Netherlands
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pathology, National University of Singapore, Singapore
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - James Scott
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Yik-Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Genome Institute of Singapore, A*STAR, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- National University of Singapore Graduate School for Integrative Science and Engineering, National University of Singapore, Singapore
- Life Sciences Institute, National University of Singapore, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute of the Netherlands (ICIN)–Netherlands Heart Institute, Utrecht, the Netherlands
| | - Jaspal S Kooner
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
88
|
Soler F, Fernández-Belda F, Pérez-Schindler J, Hernández-Cascales J. Single inhibition of either PDE3 or PDE4 unmasks β2-adrenoceptor-mediated inotropic and lusitropic effects in the left but not right ventricular myocardium of rat. Eur J Pharmacol 2015; 765:429-36. [DOI: 10.1016/j.ejphar.2015.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/18/2015] [Accepted: 09/01/2015] [Indexed: 02/05/2023]
|
89
|
Dema A, Perets E, Schulz MS, Deák VA, Klussmann E. Pharmacological targeting of AKAP-directed compartmentalized cAMP signalling. Cell Signal 2015; 27:2474-87. [PMID: 26386412 DOI: 10.1016/j.cellsig.2015.09.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/08/2015] [Accepted: 09/14/2015] [Indexed: 01/26/2023]
Abstract
The second messenger cyclic adenosine monophosphate (cAMP) can bind and activate protein kinase A (PKA). The cAMP/PKA system is ubiquitous and involved in a wide array of biological processes and therefore requires tight spatial and temporal regulation. Important components of the safeguard system are the A-kinase anchoring proteins (AKAPs), a heterogeneous family of scaffolding proteins defined by its ability to directly bind PKA. AKAPs tether PKA to specific subcellular compartments, and they bind further interaction partners to create local signalling hubs. The recent discovery of new AKAPs and advances in the field that shed light on the relevance of these hubs for human disease highlight unique opportunities for pharmacological modulation. This review exemplifies how interference with signalling, particularly cAMP signalling, at such hubs can reshape signalling responses and discusses how this could lead to novel pharmacological concepts for the treatment of disease with an unmet medical need such as cardiovascular disease and cancer.
Collapse
Affiliation(s)
- Alessandro Dema
- Max Delbrück Center for Molecular Medicine Berlin in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Ekaterina Perets
- Max Delbrück Center for Molecular Medicine Berlin in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Maike Svenja Schulz
- Max Delbrück Center for Molecular Medicine Berlin in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Veronika Anita Deák
- Max Delbrück Center for Molecular Medicine Berlin in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine Berlin in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany; DZHK, German Centre for Cardiovascular Research, Oudenarder Straße 16, 13347 Berlin, Germany.
| |
Collapse
|
90
|
Froese A, Nikolaev VO. Imaging alterations of cardiomyocyte cAMP microdomains in disease. Front Pharmacol 2015; 6:172. [PMID: 26379548 PMCID: PMC4548481 DOI: 10.3389/fphar.2015.00172] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/03/2015] [Indexed: 11/13/2022] Open
Abstract
3',5'-cyclic adenosine monophosphate (cAMP) is an important second messenger which regulates heart function by acting in distinct subcellular microdomains. Recent years have provided deeper mechanistic insights into compartmentalized cAMP signaling and its link to cardiac disease. In this mini review, we summarize newest developments in this field achieved by cutting-edge biochemical and biophysical techniques. We further compile the data from different studies into a bigger picture of so far uncovered alterations in cardiomyocyte cAMP microdomains which occur in compensated cardiac hypertrophy and chronic heart failure. Finally, future research directions and translational perspectives are briefly discussed.
Collapse
Affiliation(s)
- Alexander Froese
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, Georg August University Medical Center , Göttingen, Germany
| | - Viacheslav O Nikolaev
- German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf , Hamburg, Germany ; Institute of Experimental Cardiovascular Research , Hamburg, Germany
| |
Collapse
|
91
|
Gosal K, Dunlop K, Dhaliwal R, Ivanovska J, Kantores C, Desjardins JF, Connelly KA, McNamara PJ, Jain A, Jankov RP. Rho Kinase Mediates Right Ventricular Systolic Dysfunction in Rats with Chronic Neonatal Pulmonary Hypertension. Am J Respir Cell Mol Biol 2015; 52:717-27. [DOI: 10.1165/rcmb.2014-0201oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
92
|
Maass PG, Aydin A, Luft FC, Schächterle C, Weise A, Stricker S, Lindschau C, Vaegler M, Qadri F, Toka HR, Schulz H, Krawitz PM, Parkhomchuk D, Hecht J, Hollfinger I, Wefeld-Neuenfeld Y, Bartels-Klein E, Mühl A, Kann M, Schuster H, Chitayat D, Bialer MG, Wienker TF, Ott J, Rittscher K, Liehr T, Jordan J, Plessis G, Tank J, Mai K, Naraghi R, Hodge R, Hopp M, Hattenbach LO, Busjahn A, Rauch A, Vandeput F, Gong M, Rüschendorf F, Hübner N, Haller H, Mundlos S, Bilginturan N, Movsesian MA, Klussmann E, Toka O, Bähring S. PDE3A mutations cause autosomal dominant hypertension with brachydactyly. Nat Genet 2015; 47:647-53. [PMID: 25961942 DOI: 10.1038/ng.3302] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 04/17/2015] [Indexed: 01/02/2023]
Abstract
Cardiovascular disease is the most common cause of death worldwide, and hypertension is the major risk factor. Mendelian hypertension elucidates mechanisms of blood pressure regulation. Here we report six missense mutations in PDE3A (encoding phosphodiesterase 3A) in six unrelated families with mendelian hypertension and brachydactyly type E (HTNB). The syndrome features brachydactyly type E (BDE), severe salt-independent but age-dependent hypertension, an increased fibroblast growth rate, neurovascular contact at the rostral-ventrolateral medulla, altered baroreflex blood pressure regulation and death from stroke before age 50 years when untreated. In vitro analyses of mesenchymal stem cell-derived vascular smooth muscle cells (VSMCs) and chondrocytes provided insights into molecular pathogenesis. The mutations increased protein kinase A-mediated PDE3A phosphorylation and resulted in gain of function, with increased cAMP-hydrolytic activity and enhanced cell proliferation. Levels of phosphorylated VASP were diminished, and PTHrP levels were dysregulated. We suggest that the identified PDE3A mutations cause the syndrome. VSMC-expressed PDE3A deserves scrutiny as a therapeutic target for the treatment of hypertension.
Collapse
Affiliation(s)
- Philipp G Maass
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Atakan Aydin
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Friedrich C Luft
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [3] Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Carolin Schächterle
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anja Weise
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Sigmar Stricker
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Carsten Lindschau
- 1] Department of Nephrology, Hannover University Medical School, Hannover, Germany. [2] Staatliche Technikerschule Berlin, Berlin, Germany
| | - Martin Vaegler
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Department of Urology, Laboratory of Tissue Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Fatimunnisa Qadri
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Hakan R Toka
- 1] Division of Nephrology and Hypertension, Eastern Virginia Medical School, Norfolk, Virginia, USA. [2] Division of Nephrology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Herbert Schulz
- 1] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Peter M Krawitz
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany. [3] Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Dmitri Parkhomchuk
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany. [3] Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jochen Hecht
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Irene Hollfinger
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Yvette Wefeld-Neuenfeld
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Eireen Bartels-Klein
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Astrid Mühl
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Martin Kann
- 1] Department II of Medicine, University of Cologne, Cologne, Germany. [2] Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | - David Chitayat
- 1] Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada. [2] Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Martin G Bialer
- 1] Division of Medical Genetics, North Shore/LIJ Health System, Manhasset, New York, USA. [2] Department of Pediatrics, North Shore/LIJ Health System, Manhasset, New York, USA
| | - Thomas F Wienker
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Jürg Ott
- 1] Institute of Psychology, Chinese Academy of Sciences, Beijing, China. [2] Statistical Genetics, Rockefeller University, New York, New York, USA
| | - Katharina Rittscher
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Thomas Liehr
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Jens Jordan
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Ghislaine Plessis
- Centre Hospitalier Universitaire de Caen, Cytogénétique Postnatale et Génétique Clinique, Caen, France
| | - Jens Tank
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Knut Mai
- Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ramin Naraghi
- Department of Neurosurgery, Bundeswehrkrankenhaus Ulm, Ulm, Germany
| | - Russell Hodge
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Maxwell Hopp
- Department of Pediatrics, Griffith Base Hospital, Griffith, New South Wales, Australia
| | - Lars O Hattenbach
- Department of Ophthalmology, Hospital Ludwigshafen, Ludwigshafen, Germany
| | | | - Anita Rauch
- Institute for Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Fabrice Vandeput
- 1] Cardiology Section, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA. [2] Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA. [3] Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Maolian Gong
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Franz Rüschendorf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Norbert Hübner
- 1] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] DZHK (German Centre for Cardiovascular Research), Berlin, Germany. [3] Charité Universitätsmedizin, Berlin, Germany
| | - Hermann Haller
- Department of Nephrology, Hannover University Medical School, Hannover, Germany
| | - Stefan Mundlos
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany. [3] Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nihat Bilginturan
- Department of Pediatric Oncology, Hacettepe University, Ankara, Turkey
| | - Matthew A Movsesian
- 1] Cardiology Section, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA. [2] Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA. [3] Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Enno Klussmann
- 1] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Okan Toka
- Department of Pediatric Cardiology, Children's Hospital, Friedrich Alexander University Erlangen, Erlangen, Germany
| | - Sylvia Bähring
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| |
Collapse
|
93
|
Sprenger JU, Perera RK, Steinbrecher JH, Lehnart SE, Maier LS, Hasenfuss G, Nikolaev VO. In vivo model with targeted cAMP biosensor reveals changes in receptor-microdomain communication in cardiac disease. Nat Commun 2015; 6:6965. [PMID: 25917898 DOI: 10.1038/ncomms7965] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/18/2015] [Indexed: 11/09/2022] Open
Abstract
3',5'-cyclic adenosine monophosphate (cAMP) is an ubiquitous second messenger that regulates physiological functions by acting in distinct subcellular microdomains. Although several targeted cAMP biosensors are developed and used in single cells, it is unclear whether such biosensors can be successfully applied in vivo, especially in the context of disease. Here, we describe a transgenic mouse model expressing a targeted cAMP sensor and analyse microdomain-specific second messenger dynamics in the vicinity of the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA). We demonstrate the biocompatibility of this targeted sensor and its potential for real-time monitoring of compartmentalized cAMP signalling in adult cardiomyocytes isolated from a healthy mouse heart and from an in vivo cardiac disease model. In particular, we uncover the existence of a phosphodiesterase-dependent receptor-microdomain communication, which is affected in hypertrophy, resulting in reduced β-adrenergic receptor-cAMP signalling to SERCA.
Collapse
Affiliation(s)
- Julia U Sprenger
- 1] Emmy Noether Group of the DFG, European Heart Research Institute Göttingen, University Medical Center Göttingen, D-37075 Göttingen, Germany [2] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [3] Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Ruwan K Perera
- 1] Emmy Noether Group of the DFG, European Heart Research Institute Göttingen, University Medical Center Göttingen, D-37075 Göttingen, Germany [2] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [3] Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Julia H Steinbrecher
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany
| | - Stephan E Lehnart
- 1] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [2] German Center for Cardiovascular Research (DZHK), D-93053 Regensburg, Germany
| | - Lars S Maier
- Department of Internal Medicine II, University Hospital Regensburg, D-93053 Regensburg, Germany
| | - Gerd Hasenfuss
- 1] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [2] German Center for Cardiovascular Research (DZHK), D-93053 Regensburg, Germany
| | - Viacheslav O Nikolaev
- 1] Emmy Noether Group of the DFG, European Heart Research Institute Göttingen, University Medical Center Göttingen, D-37075 Göttingen, Germany [2] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [3] Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany [4] German Center for Cardiovascular Research (DZHK), D-93053 Regensburg, Germany
| |
Collapse
|
94
|
Targeted disruption of PDE3B, but not PDE3A, protects murine heart from ischemia/reperfusion injury. Proc Natl Acad Sci U S A 2015; 112:E2253-62. [PMID: 25877153 DOI: 10.1073/pnas.1416230112] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although inhibition of cyclic nucleotide phosphodiesterase type 3 (PDE3) has been reported to protect rodent heart against ischemia/reperfusion (I/R) injury, neither the specific PDE3 isoform involved nor the underlying mechanisms have been identified. Targeted disruption of PDE3 subfamily B (PDE3B), but not of PDE3 subfamily A (PDE3A), protected mouse heart from I/R injury in vivo and in vitro, with reduced infarct size and improved cardiac function. The cardioprotective effect in PDE3B(-/-) heart was reversed by blocking cAMP-dependent PKA and by paxilline, an inhibitor of mitochondrial calcium-activated K channels, the opening of which is potentiated by cAMP/PKA signaling. Compared with WT mitochondria, PDE3B(-/-) mitochondria were enriched in antiapoptotic Bcl-2, produced less reactive oxygen species, and more frequently contacted transverse tubules where PDE3B was localized with caveolin-3. Moreover, a PDE3B(-/-) mitochondrial fraction containing connexin-43 and caveolin-3 was more resistant to Ca(2+)-induced opening of the mitochondrial permeability transition pore. Proteomics analyses indicated that PDE3B(-/-) heart mitochondria fractions were enriched in buoyant ischemia-induced caveolin-3-enriched fractions (ICEFs) containing cardioprotective proteins. Accumulation of proteins into ICEFs was PKA dependent and was achieved by ischemic preconditioning or treatment of WT heart with the PDE3 inhibitor cilostamide. Taken together, these findings indicate that PDE3B deletion confers cardioprotective effects because of cAMP/PKA-induced preconditioning, which is associated with the accumulation of proteins with cardioprotective function in ICEFs. To our knowledge, our study is the first to define a role for PDE3B in cardioprotection against I/R injury and suggests PDE3B as a target for cardiovascular therapies.
Collapse
|
95
|
Isidori AM, Cornacchione M, Barbagallo F, Di Grazia A, Barrios F, Fassina L, Monaco L, Giannetta E, Gianfrilli D, Garofalo S, Zhang X, Chen X, Xiang YK, Lenzi A, Pellegrini M, Naro F. Inhibition of type 5 phosphodiesterase counteracts β2-adrenergic signalling in beating cardiomyocytes. Cardiovasc Res 2015; 106:408-20. [PMID: 25852085 DOI: 10.1093/cvr/cvv123] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 03/20/2015] [Indexed: 12/31/2022] Open
Abstract
AIMS Compartmentalization of cAMP and PKA activity in cardiac muscle cells plays a key role in maintaining basal and enhanced contractility stimulated by sympathetic nerve activity. In cardiomyocytes, activation of adrenergic receptor increases cAMP production, which is countered by the hydrolytic activity of selective phosphodiesterases (PDEs). The intracellular regional dynamics of cAMP production and hydrolysis modulate downstream signals resulting in different biological responses. The interplay between beta receptors (βARs) signalling and phosphodiesterase 5 (PDE5) activity remains to be addressed. METHODS AND RESULTS Using combined strategies with pharmacological inhibitors and genetic deletion of PDEs and βAR isoforms, we revealed a specific pool of cAMP that is under dual regulation by PDE2 and, indirectly, PDE5 activity. Inhibition of PDE5 with sildenafil produces a cGMP-dependent activation of PDE2 that attenuates cAMP generation induced by βAR agonists, with concomitant modulation of stimulated contraction rate and calcium transients. PDE2 haploinsufficiency abolished the effects of sildenafil. The negative chronotropic effect of PDE5 inhibition through PDE2 activation was also observed in sinoatrial node tissue from adult mice. PDE5 inhibition selectively lowered contraction rate stimulated by β2AR, but not β1AR activation, supporting a compartmentalization of the cGMP-modulated pool of cAMP. CONCLUSION These data identify a new effect of PDE5 inhibitors on the modulation of cardiomyocyte response to adrenergic stimulation via PDE5-PDE2-mediated cross-talk.
Collapse
Affiliation(s)
- Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marisa Cornacchione
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Federica Barbagallo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonio Di Grazia
- Department of Anatomical, Histological, Forensic, and Orthopaedic Sciences, Sapienza University, Rome, Italy
| | - Florencia Barrios
- Department of Anatomical, Histological, Forensic, and Orthopaedic Sciences, Sapienza University, Rome, Italy
| | - Lorenzo Fassina
- Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvio Garofalo
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Xiaoxiao Zhang
- Department of Physiology, Temple University, Philadelphia, PA, USA
| | - Xiongwen Chen
- Department of Physiology, Temple University, Philadelphia, PA, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Manuela Pellegrini
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic, and Orthopaedic Sciences, Sapienza University, Rome, Italy
| |
Collapse
|
96
|
Ahmad F, Shen W, Vandeput F, Szabo-Fresnais N, Krall J, Degerman E, Goetz F, Klussmann E, Movsesian M, Manganiello V. Regulation of sarcoplasmic reticulum Ca2+ ATPase 2 (SERCA2) activity by phosphodiesterase 3A (PDE3A) in human myocardium: phosphorylation-dependent interaction of PDE3A1 with SERCA2. J Biol Chem 2015; 290:6763-76. [PMID: 25593322 DOI: 10.1074/jbc.m115.638585] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclic nucleotide phosphodiesterase 3A (PDE3) regulates cAMP-mediated signaling in the heart, and PDE3 inhibitors augment contractility in patients with heart failure. Studies in mice showed that PDE3A, not PDE3B, is the subfamily responsible for these inotropic effects and that murine PDE3A1 associates with sarcoplasmic reticulum Ca(2+) ATPase 2 (SERCA2), phospholamban (PLB), and AKAP18 in a multiprotein signalosome in human sarcoplasmic reticulum (SR). Immunohistochemical staining demonstrated that PDE3A co-localizes in Z-bands of human cardiac myocytes with desmin, SERCA2, PLB, and AKAP18. In human SR fractions, cAMP increased PLB phosphorylation and SERCA2 activity; this was potentiated by PDE3 inhibition but not by PDE4 inhibition. During gel filtration chromatography of solubilized SR membranes, PDE3 activity was recovered in distinct high molecular weight (HMW) and low molecular weight (LMW) peaks. HMW peaks contained PDE3A1 and PDE3A2, whereas LMW peaks contained PDE3A1, PDE3A2, and PDE3A3. Western blotting showed that endogenous HMW PDE3A1 was the principal PKA-phosphorylated isoform. Phosphorylation of endogenous PDE3A by rPKAc increased cAMP-hydrolytic activity, correlated with shift of PDE3A from LMW to HMW peaks, and increased co-immunoprecipitation of SERCA2, cav3, PKA regulatory subunit (PKARII), PP2A, and AKAP18 with PDE3A. In experiments with recombinant proteins, phosphorylation of recombinant human PDE3A isoforms by recombinant PKA catalytic subunit increased co-immunoprecipitation with rSERCA2 and rat rAKAP18 (recombinant AKAP18). Deletion of the recombinant human PDE3A1/PDE3A2 N terminus blocked interactions with recombinant SERCA2. Serine-to-alanine substitutions identified Ser-292/Ser-293, a site unique to human PDE3A1, as the principal site regulating its interaction with SERCA2. These results indicate that phosphorylation of human PDE3A1 at a PKA site in its unique N-terminal extension promotes its incorporation into SERCA2/AKAP18 signalosomes, where it regulates a discrete cAMP pool that controls contractility by modulating phosphorylation-dependent protein-protein interactions, PLB phosphorylation, and SERCA2 activity.
Collapse
Affiliation(s)
- Faiyaz Ahmad
- From the Cardiovascular Pulmonary Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892,
| | - Weixing Shen
- From the Cardiovascular Pulmonary Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Fabrice Vandeput
- VA Salt Lake City Health Care System and University of Utah, Salt Lake City, Utah
| | | | - Judith Krall
- VA Salt Lake City Health Care System and University of Utah, Salt Lake City, Utah
| | - Eva Degerman
- Department of Experimental Medical Science, Division for Diabetes, Metabolism, and Endocrinology, Lund University, Lund, Sweden
| | - Frank Goetz
- Max Delbrueck Center for Molecular Medicine Berlin-Buch (MDC), 13125 Germany, and
| | - Enno Klussmann
- Max Delbrueck Center for Molecular Medicine Berlin-Buch (MDC), 13125 Germany, and DZHK, German Centre for Cardiovascular Research, 13347 Berlin, Germany
| | - Matthew Movsesian
- VA Salt Lake City Health Care System and University of Utah, Salt Lake City, Utah
| | - Vincent Manganiello
- From the Cardiovascular Pulmonary Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
97
|
Weninger S, Van Craenenbroeck K, Cameron RT, Vandeput F, Movsesian MA, Baillie GS, Lefebvre RA. Phosphodiesterase 4 interacts with the 5-HT4(b) receptor to regulate cAMP signaling. Cell Signal 2014; 26:2573-82. [PMID: 25101859 DOI: 10.1016/j.cellsig.2014.07.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/28/2014] [Accepted: 07/28/2014] [Indexed: 12/14/2022]
Abstract
Phosphodiesterase (PDE) 3 and PDE4, which degrade cyclic adenosine monophosphate (cAMP), are important regulators of 5-hydroxytryptamine (5-HT) 4 receptor signaling in cardiac tissue. Therefore, we investigated whether they interact with the 5-HT4(b) receptor, and whether A-kinase anchoring proteins (AKAPs), scaffolding proteins that bind to the regulatory subunit of protein kinase A (PKA) and contribute to the spacial-temporal control of cAMP signaling, are involved in the regulation of 5-HT4(b) receptor signaling. By measuring PKA activity in the absence and presence of PDE3 and PDE4 inhibitiors, we found that constitutive signaling of the overexpressed HA-tagged 5-HT4(b) receptor in HEK293 cells is regulated predominantly by PDE4, with a secondary role for PDE3 that is unmasked in the presence of PDE4 inhibition. Overexpressed PDE4D3 and PDE3A1, and to a smaller extent PDE4D5 co-immunoprecipitate constitutively with the 5-HT4(b) receptor. PDE activity measurements in immunoprecipitates of the 5-HT4(b) receptor confirm the association of PDE4D3 with the receptor and provide evidence that the activity of this PDE may be increased upon receptor stimulation with 5-HT. A possible involvement of AKAPs in 5-HT4(b) receptor signaling was uncovered in experiments using the St-Ht31 inhibitor peptide, which disrupts the interaction of AKAPs with PKA. However, St-Ht31 did not influence 5-HT4(b) receptor-stimulated PKA activity, and endogenous AKAP79 and gravin were not found in immunoprecipitates of the 5-HT4(b) receptor. In conclusion, we found that both PDE3A1 and PDE4D3 are integrated into complexes that contain the 5-HT4(b) receptor and may thereby regulate 5-HT4(b) receptor-mediated signaling.
Collapse
Affiliation(s)
- S Weninger
- Heymans Institute of Pharmacology, Ghent University, De Pintelaan 185, Ghent 9000, Belgium
| | - K Van Craenenbroeck
- Laboratory for Eukaryotic Gene Expression and Signal Transduction, Ghent University, Proeftuinstraat 86, Ghent 9000, Belgium
| | - R T Cameron
- Institute of Cardiovascular and Medical Science, University of Glasgow, Office 534, Wolfson-Link Building, Glasgow G12 8QQ, UK
| | - F Vandeput
- Cardiovascular Medicine Division, VA Salt Lake City Health Care System and University of Utah School of Medicine, Salt Lake City, UT 84148, USA
| | - M A Movsesian
- Cardiovascular Medicine Division, VA Salt Lake City Health Care System and University of Utah School of Medicine, Salt Lake City, UT 84148, USA
| | - G S Baillie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Office 534, Wolfson-Link Building, Glasgow G12 8QQ, UK
| | - R A Lefebvre
- Heymans Institute of Pharmacology, Ghent University, De Pintelaan 185, Ghent 9000, Belgium.
| |
Collapse
|
98
|
Xin W, Li N, Cheng Q, Fernandes VS, Petkov GV. Constitutive PKA activity is essential for maintaining the excitability and contractility in guinea pig urinary bladder smooth muscle: role of the BK channel. Am J Physiol Cell Physiol 2014; 307:C1142-50. [PMID: 25318105 DOI: 10.1152/ajpcell.00167.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The elevation of protein kinase A (PKA) activity activates the large-conductance voltage- and Ca(2+)-activated K(+) (BK) channels in urinary bladder smooth muscle (UBSM) cells and consequently attenuates spontaneous phasic contractions of UBSM. However, the role of constitutive PKA activity in UBSM function has not been studied. Here, we tested the hypothesis that constitutive PKA activity is essential for controlling the excitability and contractility of UBSM. We used patch clamp electrophysiology, line-scanning confocal and ratiometric fluorescence microscopy on freshly isolated guinea pig UBSM cells, and isometric tension recordings on freshly isolated UBSM strips. Pharmacological inhibition of the constitutive PKA activity with H-89 or PKI 14-22 significantly reduced the frequency and amplitude of spontaneous transient BK channel currents (TBKCs) in UBSM cells. Confocal and ratiometric fluorescence microscopy studies revealed that inhibition of constitutive PKA activity with H-89 reduced the frequency and amplitude of the localized Ca(2+) sparks but increased global Ca(2+) levels and the magnitude of Ca(2+) oscillations in UBSM cells. H-89 abolished the spontaneous transient membrane hyperpolarizations and depolarized the membrane potential in UBSM cells. Inhibition of PKA with H-89 or KT-5720 also increased the amplitude and muscle force of UBSM spontaneous phasic contractions. This study reveals the novel concept that constitutive PKA activity is essential for controlling localized Ca(2+) signals generated by intracellular Ca(2+) stores and cytosolic Ca(2+) levels. Furthermore, constitutive PKA activity is critical for mediating the spontaneous TBKCs in UBSM cells, where it plays a key role in regulating spontaneous phasic contractions in UBSM.
Collapse
Affiliation(s)
- Wenkuan Xin
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and
| | - Ning Li
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and Department of Urology, Fourth Hospital of China Medical University, Shenyang, China
| | - Qiuping Cheng
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and
| | - Vitor S Fernandes
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and
| | - Georgi V Petkov
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and
| |
Collapse
|
99
|
Nakano SJ, Miyamoto SD, Movsesian M, Nelson P, Stauffer BL, Sucharov CC. Age-related differences in phosphodiesterase activity and effects of chronic phosphodiesterase inhibition in idiopathic dilated cardiomyopathy. Circ Heart Fail 2014; 8:57-63. [PMID: 25278000 DOI: 10.1161/circheartfailure.114.001218] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Despite the application of proven adult heart failure therapies to children with idiopathic dilated cardiomyopathy (IDC), prognosis remains poor. Clinical experience with phosphodiesterase 3 inhibitors (PDE3i) in pediatric patients with IDC, however, demonstrates improved heart failure symptoms without the increased incidence of sudden death seen in adults treated with PDE3i. We sought to determine age-related differences in PDE activity and associated intracellular signaling responsible for the efficacy and relative safety of chronic PDE3i in pediatric heart failure. METHODS AND RESULTS cAMP levels, PDE activity, and phospholamban phosphorylation (pPLB) were determined in explanted human left ventricular myocardium (pediatric n=41; adult n=88). Adults and children with IDC (not treated with PDE3i) had lower cAMP and pPLB compared with nonfailing controls. In contrast to their adult counterparts, pediatric IDC patients chronically treated with PDE3i had elevated cAMP (P=0.0403) and pPLB (P=0.0119). In addition, total PDE- and PDE3-specific activities were not altered in pediatric IDC patients on PDE3i, whereas adult IDC patients on PDE3i demonstrated higher total PDE-specific (74.6±13.8 pmol/mg per minute) and PDE3-specific (48.2±15.9 pmol/mg per minute) activities in comparison with those of nonfailing controls (59.5±14.4 and 35.5±12.8 pmol/mg per minute, respectively). CONCLUSIONS Elevated cAMP and higher pPLB may contribute to sustained hemodynamic benefits in pediatric IDC patients treated with PDE3i. In contrast, higher total PDE and PDE3 activities in adult IDC patients treated with PDE3i may perpetuate lower myocardial cAMP and pPLB levels, limiting the potential benefits of PDE3i therapy.
Collapse
Affiliation(s)
- Stephanie J Nakano
- From the Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (S.J.N., S.D.M.); Cardiology Section, Veterans Affairs Salt Lake City Health Care System and the Departments of Internal Medicine (Cardiology) and Pharmacology, University of Utah School of Medicine (M.M.); Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora (P.N., B.L.S., C.C.S.); and Division of Cardiology, Department of Medicine, Denver Health and Hospital Authority, CO (B.L.S.)
| | - Shelley D Miyamoto
- From the Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (S.J.N., S.D.M.); Cardiology Section, Veterans Affairs Salt Lake City Health Care System and the Departments of Internal Medicine (Cardiology) and Pharmacology, University of Utah School of Medicine (M.M.); Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora (P.N., B.L.S., C.C.S.); and Division of Cardiology, Department of Medicine, Denver Health and Hospital Authority, CO (B.L.S.)
| | - Matthew Movsesian
- From the Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (S.J.N., S.D.M.); Cardiology Section, Veterans Affairs Salt Lake City Health Care System and the Departments of Internal Medicine (Cardiology) and Pharmacology, University of Utah School of Medicine (M.M.); Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora (P.N., B.L.S., C.C.S.); and Division of Cardiology, Department of Medicine, Denver Health and Hospital Authority, CO (B.L.S.)
| | - Penny Nelson
- From the Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (S.J.N., S.D.M.); Cardiology Section, Veterans Affairs Salt Lake City Health Care System and the Departments of Internal Medicine (Cardiology) and Pharmacology, University of Utah School of Medicine (M.M.); Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora (P.N., B.L.S., C.C.S.); and Division of Cardiology, Department of Medicine, Denver Health and Hospital Authority, CO (B.L.S.)
| | - Brian L Stauffer
- From the Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (S.J.N., S.D.M.); Cardiology Section, Veterans Affairs Salt Lake City Health Care System and the Departments of Internal Medicine (Cardiology) and Pharmacology, University of Utah School of Medicine (M.M.); Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora (P.N., B.L.S., C.C.S.); and Division of Cardiology, Department of Medicine, Denver Health and Hospital Authority, CO (B.L.S.)
| | - Carmen C Sucharov
- From the Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora (S.J.N., S.D.M.); Cardiology Section, Veterans Affairs Salt Lake City Health Care System and the Departments of Internal Medicine (Cardiology) and Pharmacology, University of Utah School of Medicine (M.M.); Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora (P.N., B.L.S., C.C.S.); and Division of Cardiology, Department of Medicine, Denver Health and Hospital Authority, CO (B.L.S.).
| |
Collapse
|
100
|
Ahmad F, Murata T, Shimizu K, Degerman E, Maurice D, Manganiello V. Cyclic nucleotide phosphodiesterases: important signaling modulators and therapeutic targets. Oral Dis 2014; 21:e25-50. [PMID: 25056711 DOI: 10.1111/odi.12275] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 07/09/2014] [Indexed: 02/06/2023]
Abstract
By catalyzing hydrolysis of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), cyclic nucleotide phosphodiesterases are critical regulators of their intracellular concentrations and their biological effects. As these intracellular second messengers control many cellular homeostatic processes, dysregulation of their signals and signaling pathways initiate or modulate pathophysiological pathways related to various disease states, including erectile dysfunction, pulmonary hypertension, acute refractory cardiac failure, intermittent claudication, chronic obstructive pulmonary disease, and psoriasis. Alterations in expression of PDEs and PDE-gene mutations (especially mutations in PDE6, PDE8B, PDE11A, and PDE4) have been implicated in various diseases and cancer pathologies. PDEs also play important role in formation and function of multimolecular signaling/regulatory complexes, called signalosomes. At specific intracellular locations, individual PDEs, together with pathway-specific signaling molecules, regulators, and effectors, are incorporated into specific signalosomes, where they facilitate and regulate compartmentalization of cyclic nucleotide signaling pathways and specific cellular functions. Currently, only a limited number of PDE inhibitors (PDE3, PDE4, PDE5 inhibitors) are used in clinical practice. Future paths to novel drug discovery include the crystal structure-based design approach, which has resulted in generation of more effective family-selective inhibitors, as well as burgeoning development of strategies to alter compartmentalized cyclic nucleotide signaling pathways by selectively targeting individual PDEs and their signalosome partners.
Collapse
Affiliation(s)
- F Ahmad
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|