1
|
Bompierre S, Yapo C, Blaise R, Vincent P, Castro LRV. Activation of PDE2A moderates pathologically high cAMP/PKA responses to dopamine in dyskinetic mice. Neurobiol Dis 2025:106968. [PMID: 40412662 DOI: 10.1016/j.nbd.2025.106968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/29/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Dopamine depletion in Parkinson's disease (PD) leads to severe motor and cognitive disturbances. While L-DOPA replacement therapy efficiently alleviates the motor symptoms, it leads to long-term complications, notably dyskinesia. These deregulations are closely associated with an exacerbated cAMP/PKA signaling and alterations in striatal plasticity. In this study we used genetically encoded biosensors to monitor cAMP and PKA signals in the 6-OHDA mouse model of Parkinson's disease. In these mice cAMP levels and PKA signaling were markedly up-regulated. We observed that stimulation of the cGMP signaling by a NO donor, DEANO, efficiently down-regulated the amplitude of these hypersensitive responses and this regulatory effect was mediated by PDE2A. Indeed, the stimulation of PDE2A by cGMP was found to efficiently reduce the excessive cAMP/PKA signaling triggered by D1 receptor stimulation, and this, despite unaltered PDE2A expression. These findings strongly suggest that boosting PDE2A activity in the striatum would be of therapeutic value to moderate the excessive cAMP/PKA responses and mitigate the long-term changes in striatal neurons associated with the adverse effects of L-DOPA treatment.
Collapse
Affiliation(s)
- Ségolène Bompierre
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| | - Cédric Yapo
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Régis Blaise
- Centre de recherche Saint-Antoine, Inserm UMR_S938, Sorbonne University, Paris, France.
| | - Pierre Vincent
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.
| | - Liliana R V Castro
- Institut de Biologie Paris Seine (IBPS), CNRS, Sorbonne University, Paris, France.
| |
Collapse
|
2
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Kamel R, Bourcier A, Margaria JP, Jin V, Varin A, Hivonnait A, Mercier‐Nomé F, Mika D, Ghigo A, Charpentier F, Algalarrondo V, Hirsch E, Fischmeister R, Vandecasteele G, Leroy J. Cardiac Gene Therapy With Phosphodiesterase 2A Limits Remodeling and Arrhythmias in Mouse Models of Heart Failure. J Am Heart Assoc 2025; 14:e037343. [PMID: 39895533 PMCID: PMC12074716 DOI: 10.1161/jaha.124.037343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND PDE2 (phosphodiesterase 2) is upregulated in human heart failure. Cardiac PDE2-transgenic mice are protected against contractile dysfunction and arrhythmias in heart failure but whether an acute elevation of PDE2 could be of therapeutic value remains elusive. This hypothesis was tested using cardiac PDE2 gene transfer in preclinical models of heart failure. METHODS AND RESULTS C57BL/6 male mice were injected with serotype 9 adeno-associated viruses encoding for PDE2A. This led to a ≈10-fold rise of PDE2A protein levels that affected neither cardiac structure nor function in healthy mice. Two weeks after inoculation with serotype 9 adeno-associated viruses, mice were implanted with minipumps delivering either NaCl, isoproterenol (60 mg/kg per day), or isoproterenol and phenylephrine (30 mg/kg per day each) for 2 weeks. In mice injected with serotype 9 adeno-associated viruses encoding for LUC (luciferase), isoproterenol or isoproterenol+phenylephrine infusion induced left ventricular hypertrophy, decreased ejection fraction unveiled by echocardiography, and promoted fibrosis and apoptosis assessed by Masson's trichrome and Tunel, respectively. Furthermore, inotropic responses to isoproterenol of ventricular cardiomyocytes isolated from isoproterenol+phenylephrine-LUC mice loaded with 1 μmol/L Fura-2AM and stimulated at 1 Hz to record calcium transients and sarcomere shortening were dampened. Spontaneous calcium waves at the cellular level were promoted as well as ventricular arrhythmias evoked in vivo by catheter-mediated ventricular pacing after isoproterenol (1.5 mg/kg) and atropine (1 mg/kg) injection. However, increased PDE2A blunted these adverse outcomes evoked by sympathomimetic amines. CONCLUSIONS Cardiac gene therapy with PDE2A limits left ventricle remodeling, dysfunction, and arrhythmias evoked by catecholamines, providing evidence that increasing PDE2A activity acutely could prevent progression toward heart failure.
Collapse
MESH Headings
- Animals
- Heart Failure/therapy
- Heart Failure/physiopathology
- Heart Failure/genetics
- Heart Failure/enzymology
- Genetic Therapy/methods
- Disease Models, Animal
- Mice, Inbred C57BL
- Male
- Ventricular Remodeling
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/enzymology
- Arrhythmias, Cardiac/prevention & control
- Arrhythmias, Cardiac/therapy
- Cyclic Nucleotide Phosphodiesterases, Type 2/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 2/biosynthesis
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Dependovirus/genetics
- Mice
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/enzymology
- Isoproterenol
- Apoptosis
- Ventricular Function, Left
- Fibrosis
Collapse
Affiliation(s)
- Rima Kamel
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Aurélia Bourcier
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Jean Piero Margaria
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health SciencesUniversity of TorinoItaly
- Cystic Kidney Disorders Unit, Division of Genetics and Cell BiologyUniversità Vita‐Salute San RaffaeleMilanItaly
| | - Valentin Jin
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Audrey Varin
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Agnès Hivonnait
- Nantes Université, CNRS, INSERM, l’institut du thoraxNantesFrance
| | | | - Delphine Mika
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Alessandra Ghigo
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health SciencesUniversity of TorinoItaly
| | | | - Vincent Algalarrondo
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Emilio Hirsch
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health SciencesUniversity of TorinoItaly
| | - Rodolphe Fischmeister
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Grégoire Vandecasteele
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Jérôme Leroy
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| |
Collapse
|
4
|
Zhang B, Jiang MY, Luo WH, Zhang C, Wu Y. Phosphodiesterase 2 (PDE2) inhibitors: an updated patent review (2017-present). Expert Opin Ther Pat 2024; 34:1105-1119. [PMID: 39508521 DOI: 10.1080/13543776.2024.2412577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 09/18/2024] [Indexed: 11/15/2024]
Abstract
INTRODUCTION PDE2 is a dual-specific enzyme that hydrolyzes two intracellular substrates, cAMP and cGMP. PDE2 is mainly distributed in the brain, which indicates that PDE2 can serve as a potential target for central nervous system diseases without causing other peripheral side effects. Discovery of new mechanisms of PDE2 inhibitors is expected to bring new opportunities for the treatment of central nervous system diseases. AREA COVERED This review aims to provide an overview of PDE2 inhibitors reported in patents from 2017 to present. EXPERT OPINION In recent years, the development of PDE2 inhibitors and their application in the treatment of brain diseases have received much attention. The main reason is the high expression of PDE2 in the brain, which gives PDE2 a natural advantage as a research target for central nervous system diseases. This review summarizes the scaffolds of PDE2 inhibitors reported in various patents since 2017, as well as the scientific issues that need to be addressed in terms of subtype selectivity and metabolic stability, intending to provide insights for the discovery of highly active and selective PDE2 inhibitors in the future.
Collapse
Affiliation(s)
- Bei Zhang
- Jiangmen Central Hospital, Jiangmen, China
| | - Mei-Yan Jiang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Wei-Hao Luo
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, China
| | - Chen Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, China
| | - Yinuo Wu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
5
|
Ariyeloye S, Kämmerer S, Klapproth E, Wielockx B, El-Armouche A. Intertwined regulators: hypoxia pathway proteins, microRNAs, and phosphodiesterases in the control of steroidogenesis. Pflugers Arch 2024; 476:1383-1398. [PMID: 38355819 PMCID: PMC11310285 DOI: 10.1007/s00424-024-02921-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
Oxygen sensing is of paramount importance for maintaining cellular and systemic homeostasis. In response to diminished oxygen levels, the hypoxia-inducible factors (HIFs) orchestrate various biological processes. These pivotal transcription factors have been identified as key regulators of several biological events. Notably, extensive research from our group and others has demonstrated that HIF1α exerts an inverse regulatory effect on steroidogenesis, leading to the suppression of crucial steroidogenic enzyme expression and a subsequent decrease in steroid levels. These steroid hormones occupy pivotal roles in governing a myriad of physiological processes. Substantial or prolonged fluctuations in steroid levels carry detrimental consequences across multiple organ systems and underlie various pathological conditions, including metabolic and immune disorders. MicroRNAs serve as potent mediators of multifaceted gene regulatory mechanisms, acting as influential epigenetic regulators that modulate a broad spectrum of gene expressions. Concomitantly, phosphodiesterases (PDEs) play a crucial role in governing signal transduction. PDEs meticulously manage intracellular levels of both cAMP and cGMP, along with their respective signaling pathways and downstream targets. Intriguingly, an intricate interplay seems to exist between hypoxia signaling, microRNAs, and PDEs in the regulation of steroidogenesis. This review highlights recent advances in our understanding of the role of microRNAs during hypoxia-driven processes, including steroidogenesis, as well as the possibilities that exist in the application of HIF prolyl hydroxylase (PHD) inhibitors for the modulation of steroidogenesis.
Collapse
Affiliation(s)
- Stephen Ariyeloye
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Susanne Kämmerer
- Department of Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Erik Klapproth
- Department of Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
6
|
Liu YB, Wang Q, Song YL, Song XM, Fan YC, Kong L, Zhang JS, Li S, Lv YJ, Li ZY, Dai JY, Qiu ZK. Abnormal phosphorylation / dephosphorylation and Ca 2+ dysfunction in heart failure. Heart Fail Rev 2024; 29:751-768. [PMID: 38498262 DOI: 10.1007/s10741-024-10395-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 03/20/2024]
Abstract
Heart failure (HF) can be caused by a variety of causes characterized by abnormal myocardial systole and diastole. Ca2+ current through the L-type calcium channel (LTCC) on the membrane is the initial trigger signal for a cardiac cycle. Declined systole and diastole in HF are associated with dysfunction of myocardial Ca2+ function. This disorder can be correlated with unbalanced levels of phosphorylation / dephosphorylation of LTCC, endoplasmic reticulum (ER), and myofilament. Kinase and phosphatase activity changes along with HF progress, resulting in phased changes in the degree of phosphorylation / dephosphorylation. It is important to realize the phosphorylation / dephosphorylation differences between a normal and a failing heart. This review focuses on phosphorylation / dephosphorylation changes in the progression of HF and summarizes the effects of phosphorylation / dephosphorylation of LTCC, ER function, and myofilament function in normal conditions and HF based on previous experiments and clinical research. Also, we summarize current therapeutic methods based on abnormal phosphorylation / dephosphorylation and clarify potential therapeutic directions.
Collapse
Affiliation(s)
- Yan-Bing Liu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China
- Medical College, Qingdao University, Qingdao, China
| | - Qian Wang
- Medical College, Qingdao University, Qingdao, China
| | - Yu-Ling Song
- Department of Pediatrics, Huantai County Hospital of Traditional Chinese Medicine, Zibo, China
| | | | - Yu-Chen Fan
- Medical College, Qingdao University, Qingdao, China
| | - Lin Kong
- Medical College, Qingdao University, Qingdao, China
| | | | - Sheng Li
- Medical College, Qingdao University, Qingdao, China
| | - Yi-Ju Lv
- Medical College, Qingdao University, Qingdao, China
| | - Ze-Yang Li
- Medical College, Qingdao University, Qingdao, China
| | - Jing-Yu Dai
- Department of Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China.
| | - Zhen-Kang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China.
| |
Collapse
|
7
|
Barthou A, Kamel R, Leroy J, Vandecasteele G, Fischmeister R. [Cyclic nucleotide phosphodiesterases: therapeutic targets in cardiac hypertrophy and failure]. Med Sci (Paris) 2024; 40:534-543. [PMID: 38986098 DOI: 10.1051/medsci/2024083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) modulate neurohormonal regulation of cardiac function by degrading cAMP and cGMP. In cardiomyocytes, multiple isoforms of PDEs with different enzymatic properties and subcellular locally regulate cyclic nucleotide levels and associated cellular functions. This organisation is severely disrupted during hypertrophy and heart failure (HF), which may contribute to disease progression. Clinically, PDE inhibition has been seen as a promising approach to compensate for the catecholamine desensitisation that accompanies heart failure. Although PDE3 inhibitors such as milrinone or enoximone can be used clinically to improve systolic function and relieve the symptoms of acute CHF, their chronic use has proved detrimental. Other PDEs, such as PDE1, PDE2, PDE4, PDE5, PDE9 and PDE10, have emerged as potential new targets for the treatment of HF, each with a unique role in local cyclic nucleotide signalling pathways. In this review, we describe cAMP and cGMP signalling in cardiomyocytes and present the different families of PDEs expressed in the heart and their modifications in pathological cardiac hypertrophy and HF. We also review results from preclinical models and clinical data indicating the use of specific PDE inhibitors or activators that may have therapeutic potential in CI.
Collapse
Affiliation(s)
| | - Rima Kamel
- Université Paris-Saclay, Inserm UMR-S 1180, Orsay, France
| | - Jérôme Leroy
- Université Paris-Saclay, Inserm UMR-S 1180, Orsay, France
| | | | | |
Collapse
|
8
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
9
|
Bai Y, Zhang X, Li Y, Qi F, Liu C, Ai X, Tang M, Szeto C, Gao E, Hua X, Xie M, Wang X, Tian Y, Chen Y, Huang G, Zhang J, Xiao W, Zhang L, Liu X, Yang Q, Houser SR, Chen X. Protein Kinase A Is a Master Regulator of Physiological and Pathological Cardiac Hypertrophy. Circ Res 2024; 134:393-410. [PMID: 38275112 PMCID: PMC10923071 DOI: 10.1161/circresaha.123.322729] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 01/12/2024] [Indexed: 01/27/2024]
Abstract
BACKGROUND The sympathoadrenergic system and its major effector PKA (protein kinase A) are activated to maintain cardiac output coping with physiological or pathological stressors. If and how PKA plays a role in physiological cardiac hypertrophy (PhCH) and pathological CH (PaCH) are not clear. METHODS Transgenic mouse models expressing the PKA inhibition domain (PKAi) of PKA inhibition peptide alpha (PKIalpha)-green fluorescence protein (GFP) fusion protein (PKAi-GFP) in a cardiac-specific and inducible manner (cPKAi) were used to determine the roles of PKA in physiological CH during postnatal growth or induced by swimming, and in PaCH induced by transaortic constriction (TAC) or augmented Ca2+ influx. Kinase profiling was used to determine cPKAi specificity. Echocardiography was used to determine cardiac morphology and function. Western blotting and immunostaining were used to measure protein abundance and phosphorylation. Protein synthesis was assessed by puromycin incorporation and protein degradation by measuring protein ubiquitination and proteasome activity. Neonatal rat cardiomyocytes (NRCMs) infected with AdGFP (GFP adenovirus) or AdPKAi-GFP (PKAi-GFP adenovirus) were used to determine the effects and mechanisms of cPKAi on myocyte hypertrophy. rAAV9.PKAi-GFP was used to treat TAC mice. RESULTS (1) cPKAi delayed postnatal cardiac growth and blunted exercise-induced PhCH; (2) PKA was activated in hearts after TAC due to activated sympathoadrenergic system, the loss of endogenous PKIα (PKA inhibition peptide α), and the stimulation by noncanonical PKA activators; (3) cPKAi ameliorated PaCH induced by TAC and increased Ca2+ influxes and blunted neonatal rat cardiomyocyte hypertrophy by isoproterenol and phenylephrine; (4) cPKAi prevented TAC-induced protein synthesis by inhibiting mTOR (mammalian target of rapamycin) signaling through reducing Akt (protein kinase B) activity, but enhancing inhibitory GSK-3α (glycogen synthase kinase-3α) and GSK-3β signals; (5) cPKAi reduced protein degradation by the ubiquitin-proteasome system via decreasing RPN6 phosphorylation; (6) cPKAi increased the expression of antihypertrophic atrial natriuretic peptide (ANP); (7) cPKAi ameliorated established PaCH and improved animal survival. CONCLUSIONS Cardiomyocyte PKA is a master regulator of PhCH and PaCH through regulating protein synthesis and degradation. cPKAi can be a novel approach to treat PaCH.
Collapse
Affiliation(s)
- Yingyu Bai
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Heping District, Tianjin, China
| | - Xiaoying Zhang
- Department of Physiology & Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ying Li
- The Second Artillery General Hospital, Beijing, China
| | - Fei Qi
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Heping District, Tianjin, China
| | - Chong Liu
- Department of Physiology & Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Xiaojie Ai
- Department of Physiology & Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Mingxin Tang
- Department of Physiology & Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Christopher Szeto
- Department of Physiology & Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Erhe Gao
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Xiang Hua
- Fox Chase Cancer Center, Temple University, Philadelphia, PA 19111, USA
| | - Mingxing Xie
- Department of Ultrasound, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xuejun Wang
- Division of Basic Biomedical Science, University of S Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Ying Tian
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Yongjie Chen
- Department of Epidemiology and Statistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Guowei Huang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Junping Zhang
- Herman B Wells Center for Pediatric Research, Indiana University IUSM, Indianapolis, IN 46202, USA
| | - Weidong Xiao
- Herman B Wells Center for Pediatric Research, Indiana University IUSM, Indianapolis, IN 46202, USA
| | - Lili Zhang
- Research Vector Core, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xueyuan Liu
- Research Vector Core, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Steven R. Houser
- Department of Physiology & Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xiongwen Chen
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Heping District, Tianjin, China
- Department of Physiology & Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| |
Collapse
|
10
|
Dorey TW, Liu Y, Jansen HJ, Bohne LJ, Mackasey M, Atkinson L, Prasai S, Belke DD, Fatehi-Hassanabad A, Fedak PWM, Rose RA. Natriuretic Peptide Receptor B Protects Against Atrial Fibrillation by Controlling Atrial cAMP Via Phosphodiesterase 2. Circ Arrhythm Electrophysiol 2023; 16:e012199. [PMID: 37933567 DOI: 10.1161/circep.123.012199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND β-AR (β-adrenergic receptor) stimulation regulates atrial electrophysiology and Ca2+ homeostasis via cAMP-dependent mechanisms; however, enhanced β-AR signaling can promote atrial fibrillation (AF). CNP (C-type natriuretic peptide) can also regulate atrial electrophysiology through the activation of NPR-B (natriuretic peptide receptor B) and cGMP-dependent signaling. Nevertheless, the role of NPR-B in regulating atrial electrophysiology, Ca2+ homeostasis, and atrial arrhythmogenesis is incompletely understood. METHODS Studies were performed using atrial samples from human patients with AF or sinus rhythm and in wild-type and NPR-B-deficient (NPR-B+/-) mice. Studies were conducted in anesthetized mice by intracardiac electrophysiology, in isolated mouse atrial preparations using high-resolution optical mapping, in isolated mouse and human atrial myocytes using patch-clamping and Ca2+ imaging, and in mouse and human atrial tissues using molecular biology. RESULTS Atrial NPR-B protein levels were reduced in patients with AF, and NPR-B+/- mice were more susceptible to AF. Atrial cGMP levels and PDE2 (phosphodiesterase 2) activity were reduced in NPR-B+/- mice leading to larger increases in atrial cAMP in the presence of the β-AR agonist isoproterenol. NPR-B+/- mice displayed larger increases in action potential duration and L-type Ca2+ current in the presence of isoproterenol. This resulted in the occurrence of spontaneous sarcoplasmic reticulum Ca2+ release events and delayed afterdepolarizations in NPR-B+/- atrial myocytes. Phosphorylation of the RyR2 (ryanodine receptor) and phospholamban was increased in NPR-B+/- atria in the presence of isoproterenol compared with the wildtypes. C-type natriuretic peptide inhibited isoproterenol-stimulated L-type Ca2+ current through PDE2 in mouse and human atrial myocytes. CONCLUSIONS NPR-B protects against AF by preventing enhanced atrial responses to β-adrenergic receptor agonists.
Collapse
Affiliation(s)
- Tristan W Dorey
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
- Department of Physiology and Pharmacology (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Yingjie Liu
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
- Department of Physiology and Pharmacology (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Hailey J Jansen
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
- Department of Physiology and Pharmacology (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Loryn J Bohne
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
- Department of Physiology and Pharmacology (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Martin Mackasey
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
- Department of Physiology and Pharmacology (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Logan Atkinson
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada (L.A.)
| | - Shuvam Prasai
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
- Department of Physiology and Pharmacology (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Darrell D Belke
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Ali Fatehi-Hassanabad
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Paul W M Fedak
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| | - Robert A Rose
- Department of Cardiac Sciences (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., D.D.B, A.F.-H., P.W.M.F., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
- Department of Physiology and Pharmacology (T.W.D., Y.L., H.J.J., L.J.B., M.M., S.P., R.A.R.), Libin Cardiovascular Institute, Cumming School of Medicine University of Calgary, Alberta, Canada
| |
Collapse
|
11
|
Ferron M, Merlet N, Mihalache-Avram T, Mecteau M, Brand G, Gillis MA, Shi Y, Nozza A, Cossette M, Guertin MC, Rhéaume E, Tardif JC. Adcy9 Gene Inactivation Improves Cardiac Function After Myocardial Infarction in Mice. Can J Cardiol 2023; 39:952-962. [PMID: 37054880 DOI: 10.1016/j.cjca.2023.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Polymorphisms in the adenylate cyclase 9 (ADCY9) gene influence the benefits of the cholesteryl ester transfer protein (CETP) modulator dalcetrapib on cardiovascular events after acute coronary syndrome. We hypothesized that Adcy9 inactivation could improve cardiac function and remodelling following myocardial infarction (MI) in absence of CETP activity. METHODS Wild-type (WT) and Adcy9-inactivated (Adcy9Gt/Gt) male mice, transgenic or not for human CETP (tgCETP+/-), were subjected to MI by permanent left anterior descending coronary artery ligation and studied for 4 weeks. Left ventricular (LV) function was assessed by echocardiography at baseline, 1, and 4 weeks after MI. At sacrifice, blood, spleen and bone marrow cells were collected for flow cytometry analysis, and hearts were harvested for histologic analyses. RESULTS All mice developed LV hypertrophy, dilation, and systolic dysfunction, but Adcy9Gt/Gt mice exhibited reduced pathologic LV remodelling and better LV function compared with WT mice. There were no differences between tgCETP+/- and Adcy9Gt/Gt tgCETP+/- mice, which both exhibited intermediate responses. Histologic analyses showed smaller cardiomyocyte size, reduced infarct size, and preserved myocardial capillary density in the infarct border zone in Adcy9Gt/Gt vs WT mice. Count of bone marrow T cells and B cells were significantly increased in Adcy9Gt/Gt mice compared with the other genotypes. CONCLUSIONS Adcy9 inactivation reduced infarct size, pathologic remodelling, and cardiac dysfunction. These changes were accompanied by preserved myocardial capillary density and increased adaptive immune response. Most of the benefits of Adcy9 inactivation were only observed in the absence of CETP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanfen Shi
- Montréal Heart Institute, Montréal, Québec, Canada
| | - Anna Nozza
- Montréal Health Innovations Coordinating Centre (MHICC), Montréal, Québec, Canada
| | - Mariève Cossette
- Montréal Health Innovations Coordinating Centre (MHICC), Montréal, Québec, Canada
| | - Marie-Claude Guertin
- Montréal Health Innovations Coordinating Centre (MHICC), Montréal, Québec, Canada
| | - Eric Rhéaume
- Montréal Heart Institute, Montréal, Québec, Canada; Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Claude Tardif
- Montréal Heart Institute, Montréal, Québec, Canada; Department of Medicine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
12
|
Skryabin EB, De Jong KA, Subramanian H, Bork NI, Froese A, Skryabin BV, Nikolaev VO. CRISPR/Cas9 Knock-Out in Primary Neonatal and Adult Cardiomyocytes Reveals Distinct cAMP Dynamics Regulation by Various PDE2A and PDE3A Isoforms. Cells 2023; 12:1543. [PMID: 37296663 PMCID: PMC10253201 DOI: 10.3390/cells12111543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/15/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Cyclic nucleotide phosphodiesterases 2A (PDE2A) and PDE3A play an important role in the regulation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP)-to-cAMP crosstalk. Each of these PDEs has up to three distinct isoforms. However, their specific contributions to cAMP dynamics are difficult to explore because it has been challenging to generate isoform-specific knock-out mice or cells using conventional methods. Here, we studied whether the CRISPR/Cas9 approach for precise genome editing can be used to knock out Pde2a and Pde3a genes and their distinct isoforms using adenoviral gene transfer in neonatal and adult rat cardiomyocytes. Cas9 and several specific gRNA constructs were cloned and introduced into adenoviral vectors. Primary adult and neonatal rat ventricular cardiomyocytes were transduced with different amounts of Cas9 adenovirus in combination with PDE2A or PDE3A gRNA constructs and cultured for up to 6 (adult) or 14 (neonatal) days to analyze PDE expression and live cell cAMP dynamics. A decline in mRNA expression for PDE2A (~80%) and PDE3A (~45%) was detected as soon as 3 days post transduction, with both PDEs being reduced at the protein level by >50-60% in neonatal cardiomyocytes (after 14 days) and >95% in adult cardiomyocytes (after 6 days). This correlated with the abrogated effects of selective PDE inhibitors in the live cell imaging experiments based on using cAMP biosensor measurements. Reverse transcription PCR analysis revealed that only the PDE2A2 isoform was expressed in neonatal myocytes, while adult cardiomyocytes expressed all three PDE2A isoforms (A1, A2, and A3) which contributed to the regulation of cAMP dynamics as detected by live cell imaging. In conclusion, CRISPR/Cas9 is an effective tool for the in vitro knock-out of PDEs and their specific isoforms in primary somatic cells. This novel approach suggests distinct regulation of live cell cAMP dynamics by various PDE2A and PDE3A isoforms in neonatal vs. adult cardiomyocytes.
Collapse
Affiliation(s)
- Egor B. Skryabin
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (E.B.S.); (K.A.D.J.); (H.S.); (N.I.B.); (A.F.)
| | - Kirstie A. De Jong
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (E.B.S.); (K.A.D.J.); (H.S.); (N.I.B.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Hariharan Subramanian
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (E.B.S.); (K.A.D.J.); (H.S.); (N.I.B.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Nadja I. Bork
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (E.B.S.); (K.A.D.J.); (H.S.); (N.I.B.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Alexander Froese
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (E.B.S.); (K.A.D.J.); (H.S.); (N.I.B.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Boris V. Skryabin
- Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), University of Münster, 48149 Münster, Germany;
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (E.B.S.); (K.A.D.J.); (H.S.); (N.I.B.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
13
|
Cachorro E, Günscht M, Schubert M, Sadek MS, Siegert J, Dutt F, Bauermeister C, Quickert S, Berning H, Nowakowski F, Lämmle S, Firneburg R, Luo X, Künzel SR, Klapproth E, Mirtschink P, Mayr M, Dewenter M, Vettel C, Heijman J, Lorenz K, Guan K, El-Armouche A, Wagner M, Kämmerer S. CNP Promotes Antiarrhythmic Effects via Phosphodiesterase 2. Circ Res 2023; 132:400-414. [PMID: 36715019 PMCID: PMC9930893 DOI: 10.1161/circresaha.122.322031] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Ventricular arrhythmia and sudden cardiac death are the most common lethal complications after myocardial infarction. Antiarrhythmic pharmacotherapy remains a clinical challenge and novel concepts are highly desired. Here, we focus on the cardioprotective CNP (C-type natriuretic peptide) as a novel antiarrhythmic principle. We hypothesize that antiarrhythmic effects of CNP are mediated by PDE2 (phosphodiesterase 2), which has the unique property to be stimulated by cGMP to primarily hydrolyze cAMP. Thus, CNP might promote beneficial effects of PDE2-mediated negative crosstalk between cAMP and cGMP signaling pathways. METHODS To determine antiarrhythmic effects of cGMP-mediated PDE2 stimulation by CNP, we analyzed arrhythmic events and intracellular trigger mechanisms in mice in vivo, at organ level and in isolated cardiomyocytes as well as in human-induced pluripotent stem cell-derived cardiomyocytes. RESULTS In ex vivo perfused mouse hearts, CNP abrogated arrhythmia after ischemia/reperfusion injury. Upon high-dose catecholamine injections in mice, PDE2 inhibition prevented the antiarrhythmic effect of CNP. In mouse ventricular cardiomyocytes, CNP blunted the catecholamine-mediated increase in arrhythmogenic events as well as in ICaL, INaL, and Ca2+ spark frequency. Mechanistically, this was driven by reduced cellular cAMP levels and decreased phosphorylation of Ca2+ handling proteins. Key experiments were confirmed in human iPSC-derived cardiomyocytes. Accordingly, the protective CNP effects were reversed by either specific pharmacological PDE2 inhibition or cardiomyocyte-specific PDE2 deletion. CONCLUSIONS CNP shows strong PDE2-dependent antiarrhythmic effects. Consequently, the CNP-PDE2 axis represents a novel and attractive target for future antiarrhythmic strategies.
Collapse
Affiliation(s)
- Eleder Cachorro
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mario Günscht
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mario Schubert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mirna S. Sadek
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Johanna Siegert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Fabian Dutt
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Carla Bauermeister
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Susann Quickert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Henrik Berning
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Felix Nowakowski
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Simon Lämmle
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Rebecca Firneburg
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Xiaojing Luo
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Stephan R. Künzel
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Erik Klapproth
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Germany (P.M.)
| | - Manuel Mayr
- The James Black Centre, King’s College, University of London, United Kingdom (M.M.)
- Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany (M.M.)
| | - Matthias Dewenter
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Germany (M.D.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany (M.D., C.V.)
| | - Christiane Vettel
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany (M.D., C.V.)
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center Mannheim, Germany (C.V.)
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.)
| | - Kristina Lorenz
- Institut für Pharmakologie und Toxikologie, Julius-Maximilians-Universität Würzburg, Germany (K.L.)
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (K.L.)
| | - Kaomei Guan
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Ali El-Armouche
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Michael Wagner
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
- Bereich Rhythmologie, Klinik für Innere Medizin und Kardiologie, Herzzentrum Dresden, Dresden University of Technology, Germany (M.W.)
| | - Susanne Kämmerer
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| |
Collapse
|
14
|
Cyclic nucleotide phosphodiesterases as therapeutic targets in cardiac hypertrophy and heart failure. Nat Rev Cardiol 2023; 20:90-108. [PMID: 36050457 DOI: 10.1038/s41569-022-00756-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) modulate the neurohormonal regulation of cardiac function by degrading cAMP and cGMP. In cardiomyocytes, multiple PDE isozymes with different enzymatic properties and subcellular localization regulate local pools of cyclic nucleotides and specific functions. This organization is heavily perturbed during cardiac hypertrophy and heart failure (HF), which can contribute to disease progression. Clinically, PDE inhibition has been considered a promising approach to compensate for the catecholamine desensitization that accompanies HF. Although PDE3 inhibitors, such as milrinone or enoximone, have been used clinically to improve systolic function and alleviate the symptoms of acute HF, their chronic use has proved to be detrimental. Other PDEs, such as PDE1, PDE2, PDE4, PDE5, PDE9 and PDE10, have emerged as new potential targets to treat HF, each having a unique role in local cyclic nucleotide signalling pathways. In this Review, we describe cAMP and cGMP signalling in cardiomyocytes and present the various PDE families expressed in the heart as well as their modifications in pathological cardiac hypertrophy and HF. We also appraise the evidence from preclinical models as well as clinical data pointing to the use of inhibitors or activators of specific PDEs that could have therapeutic potential in HF.
Collapse
|
15
|
Klapproth E, Witt A, Klose P, Wiedemann J, Vavilthota N, Künzel SR, Kämmerer S, Günscht M, Sprott D, Lesche M, Rost F, Dahl A, Rauch E, Kattner L, Weber S, Mirtschink P, Kopaliani I, Guan K, Lorenz K, Saftig P, Wagner M, El-Armouche A. Targeting cardiomyocyte ADAM10 ectodomain shedding promotes survival early after myocardial infarction. Nat Commun 2022; 13:7648. [PMID: 36496449 PMCID: PMC9741599 DOI: 10.1038/s41467-022-35331-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
After myocardial infarction the innate immune response is pivotal in clearing of tissue debris as well as scar formation, but exaggerated cytokine and chemokine secretion with subsequent leukocyte infiltration also leads to further tissue damage. Here, we address the value of targeting a previously unknown a disintegrin and metalloprotease 10 (ADAM10)/CX3CL1 axis in the regulation of neutrophil recruitment early after MI. We show that myocardial ADAM10 is distinctly upregulated in myocardial biopsies from patients with ischemia-driven cardiomyopathy. Intriguingly, upon MI in mice, pharmacological ADAM10 inhibition as well as genetic cardiomycyte-specific ADAM10 deletion improves survival with markedly enhanced heart function and reduced scar size. Mechanistically, abolished ADAM10-mediated CX3CL1 ectodomain shedding leads to diminished IL-1β-dependent inflammation, reduced neutrophil bone marrow egress as well as myocardial tissue infiltration. Thus, our data shows a conceptual insight into how acute MI induces chemotactic signaling via ectodomain shedding in cardiomyocytes.
Collapse
Affiliation(s)
- Erik Klapproth
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anke Witt
- grid.4488.00000 0001 2111 7257Department of Physiology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Pauline Klose
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Johanna Wiedemann
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nikitha Vavilthota
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stephan R. Künzel
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Susanne Kämmerer
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mario Günscht
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - David Sprott
- grid.4488.00000 0001 2111 7257Department of Physiology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mathias Lesche
- grid.4488.00000 0001 2111 7257DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Fabian Rost
- grid.4488.00000 0001 2111 7257DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Andreas Dahl
- grid.4488.00000 0001 2111 7257DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | | | | | - Silvio Weber
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Peter Mirtschink
- grid.4488.00000 0001 2111 7257Institute of Clinical Chemistry and Laboratory Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Irakli Kopaliani
- grid.4488.00000 0001 2111 7257Department of Physiology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kaomei Guan
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kristina Lorenz
- grid.8379.50000 0001 1958 8658Institute of Pharmacology and Toxicology, Julius-Maximilians-University of Würzburg, Würzburg, Germany ,grid.419243.90000 0004 0492 9407Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Paul Saftig
- grid.9764.c0000 0001 2153 9986Biochemical Institute, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Michael Wagner
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany ,grid.4488.00000 0001 2111 7257Rhythmology, Clinic of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ali El-Armouche
- grid.4488.00000 0001 2111 7257Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
16
|
Chiong M, Houslay MD, Lavandero S. Activation of Phosphodiesterase 3A: New Hope for Cardioprotection. Circulation 2022; 146:1779-1782. [PMID: 36469592 DOI: 10.1161/circulationaha.122.062215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mario Chiong
- From Advanced Center for Chronic Diseases (ACCDiS), Faculty Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago (M.C., S.L.)
| | | | - Sergio Lavandero
- From Advanced Center for Chronic Diseases (ACCDiS), Faculty Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago (M.C., S.L.).,Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas (S.L.)
| |
Collapse
|
17
|
Rozenfeld M, Azoulay IS, Ben Kasus Nissim T, Stavsky A, Melamed M, Stutzmann G, Hershfinkel M, Kofman O, Sekler I. Essential role of the mitochondrial Na +/Ca 2+ exchanger NCLX in mediating PDE2-dependent neuronal survival and learning. Cell Rep 2022; 41:111772. [PMID: 36476859 PMCID: PMC10521900 DOI: 10.1016/j.celrep.2022.111772] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 07/06/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
Impaired phosphodiesterase (PDE) function and mitochondrial Ca2+ (i.e., [Ca2+]m) lead to multiple health syndromes by an unknown pathway. Here, we fluorescently monitor robust [Ca2+]m efflux mediated by the mitochondrial Na+/Ca2+ exchanger NCLX in hippocampal neurons sequentially evoked by caffeine and depolarization. Surprisingly, neuronal depolarization-induced Ca2+ transients alone fail to evoke strong [Ca2+]m efflux in wild-type (WT) neurons. However, pre-treatment with the selective PDE2 inhibitor Bay 60-7550 effectively rescues [Ca2+]m efflux similarly to caffeine. Moreover, PDE2 acts by diminishing mitochondrial cAMP, thus promoting NCLX phosphorylation at its PKA site. We find that the protection of neurons against excitotoxic insults, conferred by PDE2 inhibition in WT neurons, is NCLX dependent. Finally, the administration of Bay 60-7550 enhances new object recognition in WT, but not in NCLX knockout (KO), mice. Our results identify a link between PDE and [Ca2+]m signaling that may provide effective therapy for cognitive and ischemic syndromes.
Collapse
Affiliation(s)
- Maya Rozenfeld
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ivana Savic Azoulay
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tsipi Ben Kasus Nissim
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alexandra Stavsky
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Moran Melamed
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Grace Stutzmann
- Rosalind Franklin University of Medicine and Science, Chicago Medical School, Center for Neurodegenerative Disease and Therapeutics, Chicago, IL, USA
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ora Kofman
- Department of Psychology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
18
|
Ercu M, Mücke MB, Pallien T, Markó L, Sholokh A, Schächterle C, Aydin A, Kidd A, Walter S, Esmati Y, McMurray BJ, Lato DF, Yumi Sunaga-Franze D, Dierks PH, Flores BIM, Walker-Gray R, Gong M, Merticariu C, Zühlke K, Russwurm M, Liu T, Batolomaeus TUP, Pautz S, Schelenz S, Taube M, Napieczynska H, Heuser A, Eichhorst J, Lehmann M, Miller DC, Diecke S, Qadri F, Popova E, Langanki R, Movsesian MA, Herberg FW, Forslund SK, Müller DN, Borodina T, Maass PG, Bähring S, Hübner N, Bader M, Klussmann E. Mutant Phosphodiesterase 3A Protects From Hypertension-Induced Cardiac Damage. Circulation 2022; 146:1758-1778. [PMID: 36259389 DOI: 10.1161/circulationaha.122.060210] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Phosphodiesterase 3A (PDE3A) gain-of-function mutations cause hypertension with brachydactyly (HTNB) and lead to stroke. Increased peripheral vascular resistance, rather than salt retention, is responsible. It is surprising that the few patients with HTNB examined so far did not develop cardiac hypertrophy or heart failure. We hypothesized that, in the heart, PDE3A mutations could be protective. METHODS We studied new patients. CRISPR-Cas9-engineered rat HTNB models were phenotyped by telemetric blood pressure measurements, echocardiography, microcomputed tomography, RNA-sequencing, and single nuclei RNA-sequencing. Human induced pluripotent stem cells carrying PDE3A mutations were established, differentiated to cardiomyocytes, and analyzed by Ca2+ imaging. We used Förster resonance energy transfer and biochemical assays. RESULTS We identified a new PDE3A mutation in a family with HTNB. It maps to exon 13 encoding the enzyme's catalytic domain. All hitherto identified HTNB PDE3A mutations cluster in exon 4 encoding a region N-terminally from the catalytic domain of the enzyme. The mutations were recapitulated in rat models. Both exon 4 and 13 mutations led to aberrant phosphorylation, hyperactivity, and increased PDE3A enzyme self-assembly. The left ventricles of our patients with HTNB and the rat models were normal despite preexisting hypertension. A catecholamine challenge elicited cardiac hypertrophy in HTNB rats only to the level of wild-type rats and improved the contractility of the mutant hearts, compared with wild-type rats. The β-adrenergic system, phosphodiesterase activity, and cAMP levels in the mutant hearts resembled wild-type hearts, whereas phospholamban phosphorylation was decreased in the mutants. In our induced pluripotent stem cell cardiomyocyte models, the PDE3A mutations caused adaptive changes of Ca2+ cycling. RNA-sequencing and single nuclei RNA-sequencing identified differences in mRNA expression between wild-type and mutants, affecting, among others, metabolism and protein folding. CONCLUSIONS Although in vascular smooth muscle, PDE3A mutations cause hypertension, they confer protection against hypertension-induced cardiac damage in hearts. Nonselective PDE3A inhibition is a final, short-term option in heart failure treatment to increase cardiac cAMP and improve contractility. Our data argue that mimicking the effect of PDE3A mutations in the heart rather than nonselective PDE3 inhibition is cardioprotective in the long term. Our findings could facilitate the search for new treatments to prevent hypertension-induced cardiac damage.
Collapse
Affiliation(s)
- Maria Ercu
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
| | - Michael B Mücke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
| | - Tamara Pallien
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
| | - Lajos Markó
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Germany (L.M., Y.E., M.G., T.U.P.B., S.K.F., D.N.M., S.B.)
| | - Anastasiia Sholokh
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
| | - Carolin Schächterle
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Atakan Aydin
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Alexa Kidd
- Clinical Genetics Ltd, Christchurch, New Zealand (A.K.)
| | | | - Yasmin Esmati
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Germany (L.M., Y.E., M.G., T.U.P.B., S.K.F., D.N.M., S.B.)
| | - Brandon J McMurray
- Genetics and Genome Biology Program, SickKids Research Institute, Toronto, ON, Canada (B.J.M., D.F.L., P.G.M.)
| | - Daniella F Lato
- Genetics and Genome Biology Program, SickKids Research Institute, Toronto, ON, Canada (B.J.M., D.F.L., P.G.M.)
| | - Daniele Yumi Sunaga-Franze
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Philip H Dierks
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Barbara Isabel Montesinos Flores
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Ryan Walker-Gray
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Maolian Gong
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Germany (L.M., Y.E., M.G., T.U.P.B., S.K.F., D.N.M., S.B.)
| | - Claudia Merticariu
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Kerstin Zühlke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Michael Russwurm
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät MA N1, Ruhr-Universität Bochum, Germany (M.R.)
| | - Tiannan Liu
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Theda U P Batolomaeus
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Germany (L.M., Y.E., M.G., T.U.P.B., S.K.F., D.N.M., S.B.)
| | - Sabine Pautz
- Department of Biochemistry, University of Kassel, Germany (S.P., F.W.H.)
| | - Stefanie Schelenz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Martin Taube
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Hanna Napieczynska
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Arnd Heuser
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Jenny Eichhorst
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany (J.E., M.L.)
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany (J.E., M.L.)
| | - Duncan C Miller
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
| | - Sebastian Diecke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Berlin Institute of Health (BIH), Germany (S.D., S.K.F.)
| | - Fatimunnisa Qadri
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Elena Popova
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Reika Langanki
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | | | | | - Sofia K Forslund
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Germany (L.M., Y.E., M.G., T.U.P.B., S.K.F., D.N.M., S.B.)
- Berlin Institute of Health (BIH), Germany (S.D., S.K.F.)
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany (S.K.F.)
| | - Dominik N Müller
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Germany (L.M., Y.E., M.G., T.U.P.B., S.K.F., D.N.M., S.B.)
| | - Tatiana Borodina
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
| | - Philipp G Maass
- Genetics and Genome Biology Program, SickKids Research Institute, Toronto, ON, Canada (B.J.M., D.F.L., P.G.M.)
- Department of Molecular Genetics, University of Toronto, ON, Canada (P.G.M.)
| | - Sylvia Bähring
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Germany (L.M., Y.E., M.G., T.U.P.B., S.K.F., D.N.M., S.B.)
| | - Norbert Hübner
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (M.B.M., L.M., A.S., Y.E., T.U.P.B., S.K.F., S.B., N.H., M.B.)
- Institute for Biology, University of Lübeck, Germany (M.B.)
| | - Enno Klussmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (M.E., M.B.M., T.P., A.S., C.S., A.A., D.Y.S.-F., P.H.D., B.I.M.F., R.W.-G., M.G., C.M., K.Z., T.L., S.S., M.T., H.N., A.H., D.C.M., S.D., F.Q., E.P., R.L., S.K.F., D.N.M., T.B., S.B., N.H., M.B., E.K.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (M.E., M.B.M., T.P., L.M., A.S., Y.E., T.U.P.B., D.C.M., S.D., S.K.F., D.N.M., N.H., M.B., E.K.)
| |
Collapse
|
19
|
Grange RMH, Preedy MEJ, Renukanthan A, Dignam JP, Lowe VJ, Moyes AJ, Pérez-Ternero C, Aubdool AA, Baliga RS, Hobbs AJ. Multidrug resistance proteins preferentially regulate natriuretic peptide-driven cGMP signalling in the heart and vasculature. Br J Pharmacol 2022; 179:2443-2459. [PMID: 34131904 DOI: 10.1111/bph.15593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/07/2021] [Accepted: 05/14/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE cGMP underpins the bioactivity of NO and natriuretic peptides and is key to cardiovascular homeostasis. cGMP-driven responses are terminated primarily by PDEs, but cellular efflux via multidrug resistance proteins (MRPs) might contribute. Herein, the effect of pharmacological blockade of MRPs on cGMP signalling in the heart and vasculature was investigated in vitro and in vivo. EXPERIMENTAL APPROACH Proliferation of human coronary artery smooth muscle cells (hCASMCs), vasorelaxation of murine aorta and reductions in mean arterial BP (MABP) in response to NO donors or natriuretic peptides were determined in the absence and presence of the MRP inhibitor MK571. The ability of MRP inhibition to reverse morphological and contractile deficits in a murine model of pressure overload-induced heart failure was also explored. KEY RESULTS MK571 attenuated hCASMC growth and enhanced the anti-proliferative effects of NO and atrial natriuretic peptide (ANP). MRP blockade caused concentration-dependent relaxations of murine aorta and augmented responses to ANP (and to a lesser extent NO). MK571 did not decrease MABP per se but enhanced the hypotensive actions of ANP and improved structural and functional indices of disease severity in experimental heart failure. These beneficial actions of MRP inhibition were associated with a greater intracellular:extracellular cGMP ratio in vitro and in vivo. CONCLUSIONS AND IMPLICATIONS MRP blockade promotes the cardiovascular functions of natriuretic peptides in vitro and in vivo, with more modest effects on NO. MRP inhibition may have therapeutic utility in cardiovascular diseases triggered by dysfunctional cGMP signalling, particularly those associated with altered natriuretic peptide bioactivity. LINKED ARTICLES This article is part of a themed issue on cGMP Signalling in Cell Growth and Survival. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.11/issuetoc.
Collapse
Affiliation(s)
- Robert M H Grange
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael E J Preedy
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Aniruthan Renukanthan
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Joshua P Dignam
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vanessa J Lowe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Amie J Moyes
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Cristina Pérez-Ternero
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Aisah A Aubdool
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Reshma S Baliga
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
20
|
Calamera G, Moltzau LR, Levy FO, Andressen KW. Phosphodiesterases and Compartmentation of cAMP and cGMP Signaling in Regulation of Cardiac Contractility in Normal and Failing Hearts. Int J Mol Sci 2022; 23:2145. [PMID: 35216259 PMCID: PMC8880502 DOI: 10.3390/ijms23042145] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Cardiac contractility is regulated by several neural, hormonal, paracrine, and autocrine factors. Amongst these, signaling through β-adrenergic and serotonin receptors generates the second messenger cyclic AMP (cAMP), whereas activation of natriuretic peptide receptors and soluble guanylyl cyclases generates cyclic GMP (cGMP). Both cyclic nucleotides regulate cardiac contractility through several mechanisms. Phosphodiesterases (PDEs) are enzymes that degrade cAMP and cGMP and therefore determine the dynamics of their downstream effects. In addition, the intracellular localization of the different PDEs may contribute to regulation of compartmented signaling of cAMP and cGMP. In this review, we will focus on the role of PDEs in regulating contractility and evaluate changes in heart failure.
Collapse
Affiliation(s)
| | | | | | - Kjetil Wessel Andressen
- Department of Pharmacology, Institute of Clinical Medicine, Oslo University Hospital, University of Oslo, P.O. Box 1057 Blindern, 0316 Oslo, Norway; (G.C.); (L.R.M.); (F.O.L.)
| |
Collapse
|
21
|
Mollenhauer M, Bokredenghel S, Geißen S, Klinke A, Morstadt T, Torun M, Strauch S, Schumacher W, Maass M, Konradi J, Peters VBM, Berghausen E, Vantler M, Rosenkranz S, Mehrkens D, Braumann S, Nettersheim F, Hof A, Simsekyilmaz S, Winkels H, Rudolph V, Baldus S, Adam M, Freyhaus HT. Stamp2 Protects From Maladaptive Structural Remodeling and Systolic Dysfunction in Post-Ischemic Hearts by Attenuating Neutrophil Activation. Front Immunol 2021; 12:701721. [PMID: 34691017 PMCID: PMC8527169 DOI: 10.3389/fimmu.2021.701721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
The six-transmembrane protein of prostate 2 (Stamp2) acts as an anti-inflammatory protein in macrophages by protecting from overt inflammatory signaling and Stamp2 deficiency accelerates atherosclerosis in mice. Herein, we describe an unexpected role of Stamp2 in polymorphonuclear neutrophils (PMN) and characterize Stamp2’s protective effects in myocardial ischemic injury. In a murine model of ischemia and reperfusion (I/R), echocardiography and histological analyses revealed a pronounced impairment of cardiac function in hearts of Stamp2-deficient- (Stamp2-/-) mice as compared to wild-type (WT) animals. This difference was driven by aggravated cardiac fibrosis, as augmented fibroblast-to-myofibroblast transdifferentiation was observed which was mediated by activation of the redox-sensitive p38 mitogen-activated protein kinase (p38 MAPK). Furthermore, we observed increased production of reactive oxygen species (ROS) in Stamp2-/- hearts after I/R, which is the likely cause for p38 MAPK activation. Although myocardial macrophage numbers were not affected by Stamp2 deficiency after I/R, augmented myocardial infiltration by polymorphonuclear neutrophils (PMN) was observed, which coincided with enhanced myeloperoxidase (MPO) plasma levels. Primary PMN isolated from Stamp2-/- animals exhibited a proinflammatory phenotype characterized by enhanced nuclear factor (NF)-κB activity and MPO secretion. To prove the critical role of PMN for the observed phenotype after I/R, antibody-mediated PMN depletion was performed in Stamp2-/- mice which reduced deterioration of LV function and adverse structural remodeling to WT levels. These data indicate a novel role of Stamp2 as an anti-inflammatory regulator of PMN and fibroblast-to-myofibroblast transdifferentiation in myocardial I/R injury.
Collapse
Affiliation(s)
- Martin Mollenhauer
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Senai Bokredenghel
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Simon Geißen
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Anna Klinke
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany.,Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum Nordrhein-Westfalen, University Hospital Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Tobias Morstadt
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Merve Torun
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Sabrina Strauch
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Wibke Schumacher
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Martina Maass
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Jürgen Konradi
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Vera B M Peters
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Eva Berghausen
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Marius Vantler
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Stephan Rosenkranz
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Dennis Mehrkens
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Simon Braumann
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Felix Nettersheim
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Alexander Hof
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Sakine Simsekyilmaz
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Holger Winkels
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Volker Rudolph
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany.,Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum Nordrhein-Westfalen, University Hospital Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Stephan Baldus
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Matti Adam
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Henrik Ten Freyhaus
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| |
Collapse
|
22
|
Künzel SR, Hoffmann M, Weber S, Künzel K, Kämmerer S, Günscht M, Klapproth E, Rausch JS, Sadek MS, Kolanowski T, Meyer-Roxlau S, Piorkowski C, Tugtekin SM, Rose-John S, Yin X, Mayr M, Kuhlmann JD, Wimberger P, Grützmann K, Herzog N, Küpper JH, O’Reilly M, Kabir SN, Sommerfeld LC, Guan K, Wielockx B, Fabritz L, Nattel S, Ravens U, Dobrev D, Wagner M, El-Armouche A. Diminished PLK2 Induces Cardiac Fibrosis and Promotes Atrial Fibrillation. Circ Res 2021; 129:804-820. [PMID: 34433292 PMCID: PMC8487716 DOI: 10.1161/circresaha.121.319425] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Stephan R. Künzel
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
- Department of Dermatology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K.)
| | - Maximilian Hoffmann
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Silvio Weber
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Karolina Künzel
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Susanne Kämmerer
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Mario Günscht
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Erik Klapproth
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Johanna S.E. Rausch
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Mirna S. Sadek
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Tomasz Kolanowski
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Stefanie Meyer-Roxlau
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| | - Christopher Piorkowski
- Department of Rhythmology (C.P., M.W.), Clinic for Internal Medicine and Cardiology, Heart Center Dresden GmbH, Dresden, Technische Universität Dresden
| | - Sems M. Tugtekin
- Department of Cardiac Surgery (S.M.T.), Clinic for Internal Medicine and Cardiology, Heart Center Dresden GmbH, Dresden, Technische Universität Dresden
| | - Stefan Rose-John
- Unit for Degradomics of the Protease Web, Institute of Biochemistry, University of Kiel (S.R.-J.)
| | - Xiaoke Yin
- The James Black Centre, King’s College, University of London (X.Y., M.M.)
| | - Manuel Mayr
- The James Black Centre, King’s College, University of London (X.Y., M.M.)
- Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden (M.M.)
| | - Jan Dominik Kuhlmann
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden (J.D.K., P.W.)
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg (J.D.K., P.W.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden (J.D.K., P.W., K.G.)
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden (J.D.K., P.W.)
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg (J.D.K., P.W.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden (J.D.K., P.W., K.G.)
| | - Konrad Grützmann
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden (J.D.K., P.W., K.G.)
| | - Natalie Herzog
- Brandenburg University of Technology, Senftenberg (N.H., J.-H.K.)
| | | | - Molly O’Reilly
- Institute of Cardiovascular Sciences, University of Birmingham (M.O., S.N.K., L.C.S.)
| | - S. Nashitha Kabir
- Institute of Cardiovascular Sciences, University of Birmingham (M.O., S.N.K., L.C.S.)
| | - Laura C. Sommerfeld
- Institute of Cardiovascular Sciences, University of Birmingham (M.O., S.N.K., L.C.S.)
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center Hamburg (L.F., L.C.S.)
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
- Department of Rhythmology (C.P., M.W.), Clinic for Internal Medicine and Cardiology, Heart Center Dresden GmbH, Dresden, Technische Universität Dresden
- Department of Cardiac Surgery (S.M.T.), Clinic for Internal Medicine and Cardiology, Heart Center Dresden GmbH, Dresden, Technische Universität Dresden
- Unit for Degradomics of the Protease Web, Institute of Biochemistry, University of Kiel (S.R.-J.)
- The James Black Centre, King’s College, University of London (X.Y., M.M.)
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden (J.D.K., P.W.)
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg (J.D.K., P.W.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden (J.D.K., P.W., K.G.)
- Brandenburg University of Technology, Senftenberg (N.H., J.-H.K.)
- Institute of Cardiovascular Sciences, University of Birmingham (M.O., S.N.K., L.C.S.)
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden (B.W.)
- Department of Cardiology, University Hospitals Birmingham (L.F.)
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Quebec, Canada (S.N., D.D.)
- Institut für Experimentelle Kardiovaskuläre Medizin, Universitäts Herzzentrum, Freiburg Bad Krotzingen, Freiburg im Breisgau (U.R.)
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen (S.N., D.D.)
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université (S.N.)
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine (D.D.)
- Department of Dermatology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K.)
- Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden (M.M.)
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center Hamburg (L.F., L.C.S.)
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden (B.W.)
| | - Larissa Fabritz
- Department of Cardiology, University Hospitals Birmingham (L.F.)
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center Hamburg (L.F., L.C.S.)
| | - Stanley Nattel
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Quebec, Canada (S.N., D.D.)
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen (S.N., D.D.)
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université (S.N.)
| | - Ursula Ravens
- Institut für Experimentelle Kardiovaskuläre Medizin, Universitäts Herzzentrum, Freiburg Bad Krotzingen, Freiburg im Breisgau (U.R.)
| | - Dobromir Dobrev
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Quebec, Canada (S.N., D.D.)
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen (S.N., D.D.)
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine (D.D.)
| | - Michael Wagner
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
- Department of Rhythmology (C.P., M.W.), Clinic for Internal Medicine and Cardiology, Heart Center Dresden GmbH, Dresden, Technische Universität Dresden
| | - Ali El-Armouche
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden (S.R.K., M.H., S.W., K.K., S.K., M.G., E.K., J.S.E.R., M.S.S., T.K., S.M.-R., K.G., M.W., A.E.-A.)
| |
Collapse
|
23
|
Mika D, Fischmeister R. Cyclic nucleotide signaling and pacemaker activity. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:29-38. [PMID: 34298001 DOI: 10.1016/j.pbiomolbio.2021.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/21/2021] [Accepted: 07/13/2021] [Indexed: 01/01/2023]
Abstract
The sinoatrial node (SAN) is the natural pacemaker of the heart, producing the electrical impulse that initiates every heart beat. Its activity is tightly controlled by the autonomic nervous system, and by circulating and locally released factors. Neurohumoral regulation of heart rate plays a crucial role in the integration of vital functions and influences behavior and ability to respond to changing environmental conditions. At the cellular level, modulation of SAN activity occurs through intracellular signaling pathways involving cyclic nucleotides: cyclic AMP (cAMP) and cyclic GMP (cGMP). In this Review, dedicated to Professor Dario DiFrancesco and his accomplishements in the field of cardiac pacemaking, we summarize all findings on the role of cyclic nucleotides signaling in regulating the key actors of cardiac automatism, and we provide an up-to-date review on cAMP- and cGMP-phosphodiesterases (PDEs), compellingly involved in this modulation.
Collapse
Affiliation(s)
- Delphine Mika
- Université Paris-Saclay, Inserm, UMR-S, 1180, Châtenay-Malabry, France.
| | | |
Collapse
|
24
|
Rathjens FS, Blenkle A, Iyer LM, Renger A, Syeda F, Noack C, Jungmann A, Dewenter M, Toischer K, El-Armouche A, Müller OJ, Fabritz L, Zimmermann WH, Zelarayan LC, Zafeiriou MP. Preclinical evidence for the therapeutic value of TBX5 normalization in arrhythmia control. Cardiovasc Res 2021; 117:1908-1922. [PMID: 32777030 PMCID: PMC8262635 DOI: 10.1093/cvr/cvaa239] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 06/26/2020] [Accepted: 07/29/2020] [Indexed: 11/12/2022] Open
Abstract
AIMS Arrhythmias and sudden cardiac death (SCD) occur commonly in patients with heart failure. We found T-box 5 (TBX5) dysregulated in ventricular myocardium from heart failure patients and thus we hypothesized that TBX5 reduction contributes to arrhythmia development in these patients. To understand the underlying mechanisms, we aimed to reveal the ventricular TBX5-dependent transcriptional network and further test the therapeutic potential of TBX5 level normalization in mice with documented arrhythmias. METHODS AND RESULTS We used a mouse model of TBX5 conditional deletion in ventricular cardiomyocytes. Ventricular (v) TBX5 loss in mice resulted in mild cardiac dysfunction and arrhythmias and was associated with a high mortality rate (60%) due to SCD. Upon angiotensin stimulation, vTbx5KO mice showed exacerbated cardiac remodelling and dysfunction suggesting a cardioprotective role of TBX5. RNA-sequencing of a ventricular-specific TBX5KO mouse and TBX5 chromatin immunoprecipitation was used to dissect TBX5 transcriptional network in cardiac ventricular tissue. Overall, we identified 47 transcripts expressed under the control of TBX5, which may have contributed to the fatal arrhythmias in vTbx5KO mice. These included transcripts encoding for proteins implicated in cardiac conduction and contraction (Gja1, Kcnj5, Kcng2, Cacna1g, Chrm2), in cytoskeleton organization (Fstl4, Pdlim4, Emilin2, Cmya5), and cardiac protection upon stress (Fhl2, Gpr22, Fgf16). Interestingly, after TBX5 loss and arrhythmia development in vTbx5KO mice, TBX5 protein-level normalization by systemic adeno-associated-virus (AAV) 9 application, re-established TBX5-dependent transcriptome. Consequently, cardiac dysfunction was ameliorated and the propensity of arrhythmia occurrence was reduced. CONCLUSIONS This study uncovers a novel cardioprotective role of TBX5 in the adult heart and provides preclinical evidence for the therapeutic value of TBX5 protein normalization in the control of arrhythmia.
Collapse
MESH Headings
- Animals
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/prevention & control
- Chromatin Immunoprecipitation Sequencing
- Death, Sudden, Cardiac/etiology
- Death, Sudden, Cardiac/prevention & control
- Disease Models, Animal
- Gene Expression Profiling
- Genetic Therapy
- Heart Rate
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/therapy
- Isolated Heart Preparation
- Mice, Inbred C57BL
- Mice, Knockout
- RNA-Seq
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/metabolism
- Transcription, Genetic
- Transcriptome
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/therapy
- Ventricular Function, Left
- Ventricular Remodeling
- Mice
Collapse
Affiliation(s)
- Franziska S Rathjens
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
- DZHK (German Center for Cardiovascular Disease), partner site, Goettingen, Germany
| | - Alica Blenkle
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
| | - Lavanya M Iyer
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
- DZHK (German Center for Cardiovascular Disease), partner site, Goettingen, Germany
| | - Anke Renger
- Institut für Erziehungswissenschaften, Humboldt University, Berlin, Germany
| | - Fahima Syeda
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
| | - Claudia Noack
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
- DZHK (German Center for Cardiovascular Disease), partner site, Goettingen, Germany
| | - Andreas Jungmann
- Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Disease), partner site Heidelberg/Mannheim, Germany
| | - Matthias Dewenter
- DZHK (German Center for Cardiovascular Disease), partner site Heidelberg/Mannheim, Germany
- Department of Molecular Cardiology and Epigenetics, University of Heidelberg, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center, Goettingen, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Technology-Dresden, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
- Division of Rhythmology, Department of Cardiovascular Medicine, Hospital of the University of Münster, Münster, Germany
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
- DZHK (German Center for Cardiovascular Disease), partner site, Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Goettingen, Germany
| | - Laura C Zelarayan
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
- DZHK (German Center for Cardiovascular Disease), partner site, Goettingen, Germany
| | - Maria-Patapia Zafeiriou
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
- DZHK (German Center for Cardiovascular Disease), partner site, Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Goettingen, Germany
| |
Collapse
|
25
|
Cellular Mechanisms of the Anti-Arrhythmic Effect of Cardiac PDE2 Overexpression. Int J Mol Sci 2021; 22:ijms22094816. [PMID: 34062838 PMCID: PMC8125727 DOI: 10.3390/ijms22094816] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Phosphodiesterases (PDE) critically regulate myocardial cAMP and cGMP levels. PDE2 is stimulated by cGMP to hydrolyze cAMP, mediating a negative crosstalk between both pathways. PDE2 upregulation in heart failure contributes to desensitization to β-adrenergic overstimulation. After isoprenaline (ISO) injections, PDE2 overexpressing mice (PDE2 OE) were protected against ventricular arrhythmia. Here, we investigate the mechanisms underlying the effects of PDE2 OE on susceptibility to arrhythmias. Methods: Cellular arrhythmia, ion currents, and Ca2+-sparks were assessed in ventricular cardiomyocytes from PDE2 OE and WT littermates. Results: Under basal conditions, action potential (AP) morphology were similar in PDE2 OE and WT. ISO stimulation significantly increased the incidence of afterdepolarizations and spontaneous APs in WT, which was markedly reduced in PDE2 OE. The ISO-induced increase in ICaL seen in WT was prevented in PDE2 OE. Moreover, the ISO-induced, Epac- and CaMKII-dependent increase in INaL and Ca2+-spark frequency was blunted in PDE2 OE, while the effect of direct Epac activation was similar in both groups. Finally, PDE2 inhibition facilitated arrhythmic events in ex vivo perfused WT hearts after reperfusion injury. Conclusion: Higher PDE2 abundance protects against ISO-induced cardiac arrhythmia by preventing the Epac- and CaMKII-mediated increases of cellular triggers. Thus, activating myocardial PDE2 may represent a novel intracellular anti-arrhythmic therapeutic strategy in HF.
Collapse
|
26
|
Colombe AS, Pidoux G. Cardiac cAMP-PKA Signaling Compartmentalization in Myocardial Infarction. Cells 2021; 10:cells10040922. [PMID: 33923648 PMCID: PMC8073060 DOI: 10.3390/cells10040922] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Under physiological conditions, cAMP signaling plays a key role in the regulation of cardiac function. Activation of this intracellular signaling pathway mirrors cardiomyocyte adaptation to various extracellular stimuli. Extracellular ligand binding to seven-transmembrane receptors (also known as GPCRs) with G proteins and adenylyl cyclases (ACs) modulate the intracellular cAMP content. Subsequently, this second messenger triggers activation of specific intracellular downstream effectors that ensure a proper cellular response. Therefore, it is essential for the cell to keep the cAMP signaling highly regulated in space and time. The temporal regulation depends on the activity of ACs and phosphodiesterases. By scaffolding key components of the cAMP signaling machinery, A-kinase anchoring proteins (AKAPs) coordinate both the spatial and temporal regulation. Myocardial infarction is one of the major causes of death in industrialized countries and is characterized by a prolonged cardiac ischemia. This leads to irreversible cardiomyocyte death and impairs cardiac function. Regardless of its causes, a chronic activation of cardiac cAMP signaling is established to compensate this loss. While this adaptation is primarily beneficial for contractile function, it turns out, in the long run, to be deleterious. This review compiles current knowledge about cardiac cAMP compartmentalization under physiological conditions and post-myocardial infarction when it appears to be profoundly impaired.
Collapse
|
27
|
Three Cases of Karoshi Without the Typical Pathomorphological Features of Cardiovascular/Cerebrovascular Disease. Am J Forensic Med Pathol 2021; 41:305-308. [PMID: 32769407 DOI: 10.1097/paf.0000000000000600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Karoshi is a term used to describe unexplained sudden death associated with overwork and has become a serious public health issue in China. Cases have occurred in physicians, university professors, engineers in high-tech companies, and blue-collar workers. The mechanisms associated with death by overwork are very complex. According to most researchers, karoshi is considered to be caused by an excessive workload that induces deterioration of underlying hypertension or atherosclerosis. These conditions inevitably lead to death from cardiovascular or cerebrovascular diseases. However, in our own experience, we have found that in some cases, the victims of karoshi were in a chronic state of overwork but without a history of cardiovascular or cerebrovascular diseases. In support of this, we have found that even autopsies have revealed few positive findings except for cardiac hypertrophy. In this article, we report 3 typical cases of karoshi but without the typical pathomorphological features of cardiovascular or cerebrovascular disease.
Collapse
|
28
|
Wang YW, Gao QW, Xiao YJ, Zhu XJ, Gao L, Zhang WH, Wang RR, Chen KS, Liu FM, Huang HL, Chen L. Bay 60-7550, a PDE2 inhibitor, exerts positive inotropic effect of rat heart by increasing PKA-mediated phosphorylation of phospholamban. Eur J Pharmacol 2021; 901:174077. [PMID: 33798601 DOI: 10.1016/j.ejphar.2021.174077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 10/21/2022]
Abstract
This study investigated the hemodynamic effect of Bay 60-7550, a phosphodiesterase type 2 (PDE2) inhibitor, in healthy rat hearts both in vivo and ex vivo and its underlying mechanisms. In vivo rat left ventricular pressure-volume loop, Langendorff isolated rat heart, Ca2+ transient of left ventricular myocyte and Western blot experiments were used in this study. The results demonstrated that Bay 60-7550 (1.5 mg/kg, i. p.) increased the in vivo rat heart contractility by enhancing stroke work, cardiac output, stroke volume, end-diastolic volume, heart rate, and ejection fraction. The simultaneous aortic pressure recording indicated that the systolic blood pressure was increased and diastolic blood pressure was decreased by Bay 60-7550. Also, the arterial elastance which is proportional to the peripheral vessel resistance was significantly decreased. Bay 60-7550 (0.001, 0.01, 0.1, 1 μmol/l) also enhanced the left ventricular development pressure in non-paced and paced modes with a decrease of heart rate in non-paced model. Bay 60-7550 (1 μmol/l) increased SERCA2a activity and SR Ca2+ content and reduced SR Ca2+ leak rate. Furthermore, Bay 60-7550 (0.1 μmol/l) increased the phosphorylation of phospholamban at 16-serine without significantly changing the phosphorylation levels of phospholamban at 17-threonine and RyR2. Bay 60-7550 increased the rat heart contractility and reduced peripheral arterial resistance may be mediated by increasing the phosphorylation of phospholamban and dilating peripheral vessels. PDE2 inhibitors which result in a positive inotropic effect and a decrease in peripheral resistance might serve as a target for developing agents for the treatment of heart failure in clinical settings.
Collapse
Affiliation(s)
- Yu-Wei Wang
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qian-Wen Gao
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu-Jie Xiao
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao-Jia Zhu
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Gao
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wen-Hui Zhang
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rong-Rong Wang
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ke-Su Chen
- School of Medicine, Nanjing University, Nanjing 210093, China
| | - Fu-Ming Liu
- First Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Hui-Li Huang
- Department of Clinical Pharmacy, No. 900 Hospital of the Chinese PLA Joint Support Forces, Fuzhou 350000, China
| | - Long Chen
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Institute of Chinese Medicine of Taizhou China Medical City, Double Tower, China Medical City, Taizhou 225300, China.
| |
Collapse
|
29
|
Chen S, Yan C. An update of cyclic nucleotide phosphodiesterase as a target for cardiac diseases. Expert Opin Drug Discov 2021; 16:183-196. [PMID: 32957823 PMCID: PMC7854486 DOI: 10.1080/17460441.2020.1821643] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Cyclic nucleotides, cAMP, and cGMP, are important second messengers of intracellular signaling and play crucial roles in cardiovascular biology and diseases. Cyclic nucleotide phosphodiesterases (PDEs) control the duration, magnitude, and compartmentalization of cyclic nucleotide signaling by catalyzing the hydrolysis of cyclic nucleotides. Individual PDEs modulate distinct signaling pathways and biological functions in the cell, making it a potential therapeutic target for the treatment of different cardiovascular disorders. The clinical success of several PDE inhibitors has ignited continued interest in PDE inhibitors and in PDE-target therapeutic strategies. AREAS COVERED This review concentrates on recent research advances of different PDE isoforms with regard to their expression patterns and biological functions in the heart. The limitations of current research and future directions are then discussed. The current and future development of PDE inhibitors is also covered. EXPERT OPINION Despite the therapeutic success of several marketed PDE inhibitors, the use of PDE inhibitors can be limited by their side effects, lack of efficacy, and lack of isoform selectivity. Advances in our understanding of the mechanisms by which cellular functions are changed through PDEs may enable the development of new approaches to achieve effective and specific PDE inhibition for various cardiac therapies.
Collapse
Affiliation(s)
- Si Chen
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
30
|
Liu Y, Chen J, Fontes SK, Bautista EN, Cheng Z. Physiological And Pathological Roles Of Protein Kinase A In The Heart. Cardiovasc Res 2021; 118:386-398. [PMID: 33483740 DOI: 10.1093/cvr/cvab008] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Protein kinase A (PKA) is a central regulator of cardiac performance and morphology. Myocardial PKA activation is induced by a variety of hormones, neurotransmitters and stress signals, most notably catecholamines secreted by the sympathetic nervous system. Catecholamines bind β-adrenergic receptors to stimulate cAMP-dependent PKA activation in cardiomyocytes. Elevated PKA activity enhances Ca2+ cycling and increases cardiac muscle contractility. Dynamic control of PKA is essential for cardiac homeostasis, as dysregulation of PKA signaling is associated with a broad range of heart diseases. Specifically, abnormal PKA activation or inactivation contributes to the pathogenesis of myocardial ischemia, hypertrophy, heart failure, as well as diabetic, takotsubo, or anthracycline cardiomyopathies. PKA may also determine sex-dependent differences in contractile function and heart disease predisposition. Here, we describe the recent advances regarding the roles of PKA in cardiac physiology and pathology, highlighting previous study limitations and future research directions. Moreover, we discuss the therapeutic strategies and molecular mechanisms associated with cardiac PKA biology. In summary, PKA could serve as a promising drug target for cardioprotection. Depending on disease types and mechanisms, therapeutic intervention may require either inhibition or activation of PKA. Therefore, specific PKA inhibitors or activators may represent valuable drug candidates for the treatment of heart diseases.
Collapse
Affiliation(s)
- Yuening Liu
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Jingrui Chen
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Shayne K Fontes
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Erika N Bautista
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| |
Collapse
|
31
|
Liang D, Xue J, Geng L, Zhou L, Lv B, Zeng Q, Xiong K, Zhou H, Xie D, Zhang F, Liu J, Liu Y, Li L, Yang J, Xue Z, Chen YH. Cellular and molecular landscape of mammalian sinoatrial node revealed by single-cell RNA sequencing. Nat Commun 2021; 12:287. [PMID: 33436583 PMCID: PMC7804277 DOI: 10.1038/s41467-020-20448-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Bioelectrical impulses intrinsically generated within the sinoatrial node (SAN) trigger the contraction of the heart in mammals. Though discovered over a century ago, the molecular and cellular features of the SAN that underpin its critical function in the heart are uncharted territory. Here, we identify four distinct transcriptional clusters by single-cell RNA sequencing in the mouse SAN. Functional analysis of differentially expressed genes identifies a core cell cluster enriched in the electrogenic genes. The similar cellular features are also observed in the SAN from both rabbit and cynomolgus monkey. Notably, Vsnl1, a core cell cluster marker in mouse, is abundantly expressed in SAN, but is barely detectable in atrium or ventricle, suggesting that Vsnl1 is a potential SAN marker. Importantly, deficiency of Vsnl1 not only reduces the beating rate of human induced pluripotent stem cell - derived cardiomyocytes (hiPSC-CMs) but also the heart rate of mice. Furthermore, weighted gene co-expression network analysis (WGCNA) unveiled the core gene regulation network governing the function of the SAN in mice. Overall, these findings reveal the whole transcriptome profiling of the SAN at single-cell resolution, representing an advance toward understanding of both the biology and the pathology of SAN. The spontaneous bioelectrical activity of pacemaker cells in sinoatrial node (SAN) triggers the heartbeats. Here, the authors perform single-cell RNA sequencing in the mouse SAN and identify molecular and cellular features of the SAN conserved in rabbit and cynomolgus monkey, identifying a new potential SAN marker.
Collapse
Affiliation(s)
- Dandan Liang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Jinfeng Xue
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200092, China
| | - Li Geng
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Liping Zhou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Bo Lv
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qiao Zeng
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ke Xiong
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Huixing Zhou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Duanyang Xie
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Fulei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Jie Liu
- Translational Center of Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yi Liu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Li Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jian Yang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Zhigang Xue
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200092, China. .,Reproductive Medicine Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Yi-Han Chen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China. .,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China. .,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China. .,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
32
|
Abstract
Cyclic nucleotide phosphodiesterases comprise an 11-member superfamily yielding near 100 isoform variants that hydrolyze cAMP or cGMP to their respective 5'-monophosphate form. Each plays a role in compartmentalized cyclic nucleotide signaling, with varying selectivity for each substrate, and conveying cell and intracellular-specific localized control. This review focuses on the 5 phosphodiesterases (PDEs) expressed in the cardiac myocyte capable of hydrolyzing cGMP and that have been shown to play a role in cardiac physiological and pathological processes. PDE1, PDE2, and PDE3 catabolize cAMP as well, whereas PDE5 and PDE9 are cGMP selective. PDE3 and PDE5 are already in clinical use, the former for heart failure, and PDE1, PDE9, and PDE5 are all being actively studied for this indication in patients. Research in just the past few years has revealed many novel cardiac influences of each isoform, expanding the therapeutic potential from their selective pharmacological blockade or in some instances, activation. PDE1C inhibition was found to confer cell survival protection and enhance cardiac contractility, whereas PDE2 inhibition or activation induces beneficial effects in hypertrophied or failing hearts, respectively. PDE3 inhibition is already clinically used to treat acute decompensated heart failure, although toxicity has precluded its long-term use. However, newer approaches including isoform-specific allosteric modulation may change this. Finally, inhibition of PDE5A and PDE9A counter pathological remodeling of the heart and are both being pursued in clinical trials. Here, we discuss recent research advances in each of these PDEs, their impact on the myocardium, and cardiac therapeutic potential.
Collapse
|
33
|
Abstract
3',5'-Cyclic guanosine monophosphate (cGMP) is a ubiquitous second messenger, which critically regulates cardiac pump function and protects from the development of cardiac hypertrophy by acting in various subcellular microdomains. Although clinical studies testing the potential of cGMP elevating drugs in patients suffering from cardiac disease showed promising results, deeper insight into the local actions of these drugs at the subcellular level are indispensable to inspire novel therapeutic strategies. Detailed information on the spatio-temporal dynamics of cGMP production and degradation can be provided by the use of fluorescent biosensors that are capable of monitoring this second messenger at different locations inside the cell with high temporal and spatial resolution. In this review, we will summarize how these emerging new tools have improved our understanding of cardiac cGMP signaling in health and disease, and attempt to anticipate future challenges in the field.
Collapse
|
34
|
Sadek MS, Cachorro E, El-Armouche A, Kämmerer S. Therapeutic Implications for PDE2 and cGMP/cAMP Mediated Crosstalk in Cardiovascular Diseases. Int J Mol Sci 2020; 21:E7462. [PMID: 33050419 PMCID: PMC7590001 DOI: 10.3390/ijms21207462] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Phosphodiesterases (PDEs) are the principal superfamily of enzymes responsible for degrading the secondary messengers 3',5'-cyclic nucleotides cAMP and cGMP. Their refined subcellular localization and substrate specificity contribute to finely regulate cAMP/cGMP gradients in various cellular microdomains. Redistribution of multiple signal compartmentalization components is often perceived under pathological conditions. Thereby PDEs have long been pursued as therapeutic targets in diverse disease conditions including neurological, metabolic, cancer and autoimmune disorders in addition to numerous cardiovascular diseases (CVDs). PDE2 is a unique member of the broad family of PDEs. In addition to its capability to hydrolyze both cAMP and cGMP, PDE2 is the sole isoform that may be allosterically activated by cGMP increasing its cAMP hydrolyzing activity. Within the cardiovascular system, PDE2 serves as an integral regulator for the crosstalk between cAMP/cGMP pathways and thereby may couple chronically adverse augmented cAMP signaling with cardioprotective cGMP signaling. This review provides a comprehensive overview of PDE2 regulatory functions in multiple cellular components within the cardiovascular system and also within various subcellular microdomains. Implications for PDE2- mediated crosstalk mechanisms in diverse cardiovascular pathologies are discussed highlighting the prospective use of PDE2 as a potential therapeutic target in cardiovascular disorders.
Collapse
Affiliation(s)
| | | | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| | - Susanne Kämmerer
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| |
Collapse
|
35
|
Abstract
The cyclic nucleotides cyclic adenosine-3′,5′-monophosphate (cAMP) and cyclic guanosine-3′,5′-monophosphate (cGMP) maintain physiological cardiac contractility and integrity. Cyclic nucleotide–hydrolysing phosphodiesterases (PDEs) are the prime regulators of cAMP and cGMP signalling in the heart. During heart failure (HF), the expression and activity of multiple PDEs are altered, which disrupt cyclic nucleotide levels and promote cardiac dysfunction. Given that the morbidity and mortality associated with HF are extremely high, novel therapies are urgently needed. Herein, the role of PDEs in HF pathophysiology and their therapeutic potential is reviewed. Attention is given to PDEs 1–5, and other PDEs are briefly considered. After assessing the role of each PDE in cardiac physiology, the evidence from pre-clinical models and patients that altered PDE signalling contributes to the HF phenotype is examined. The potential of pharmacologically harnessing PDEs for therapeutic gain is considered.
Collapse
|
36
|
Mollenhauer M, Mehrkens D, Klinke A, Lange M, Remane L, Friedrichs K, Braumann S, Geißen S, Simsekyilmaz S, Nettersheim FS, Lee S, Peinkofer G, Geisler AC, Geis B, Schwoerer AP, Carrier L, Freeman BA, Dewenter M, Luo X, El-Armouche A, Wagner M, Adam M, Baldus S, Rudolph V. Nitro-fatty acids suppress ischemic ventricular arrhythmias by preserving calcium homeostasis. Sci Rep 2020; 10:15319. [PMID: 32948795 PMCID: PMC7501300 DOI: 10.1038/s41598-020-71870-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/22/2020] [Indexed: 12/01/2022] Open
Abstract
Nitro-fatty acids are electrophilic anti-inflammatory mediators which are generated during myocardial ischemic injury. Whether these species exert anti-arrhythmic effects in the acute phase of myocardial ischemia has not been investigated so far. Herein, we demonstrate that pretreatment of mice with 9- and 10-nitro-octadec-9-enoic acid (nitro-oleic acid, NO2-OA) significantly reduced the susceptibility to develop acute ventricular tachycardia (VT). Accordingly, epicardial mapping revealed a markedly enhanced homogeneity in ventricular conduction. NO2-OA treatment of isolated cardiomyocytes lowered the number of spontaneous contractions upon adrenergic isoproterenol stimulation and nearly abolished ryanodine receptor type 2 (RyR2)-dependent sarcoplasmic Ca2+ leak. NO2-OA also significantly reduced RyR2-phosphorylation by inhibition of increased CaMKII activity. Thus, NO2-OA might be a novel pharmacological option for the prevention of VT development.
Collapse
Affiliation(s)
- Martin Mollenhauer
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany.
| | - Dennis Mehrkens
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Anna Klinke
- Clinic for General and Interventional Cardiology/ Angiology, Herz- Und Diabeteszentrum NRW, Ruhr-Universitaet Bochum, Bad Oeynhausen, Germany
| | - Max Lange
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Lisa Remane
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Kai Friedrichs
- Clinic for General and Interventional Cardiology/ Angiology, Herz- Und Diabeteszentrum NRW, Ruhr-Universitaet Bochum, Bad Oeynhausen, Germany
| | - Simon Braumann
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Simon Geißen
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Sakine Simsekyilmaz
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Felix S Nettersheim
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Samuel Lee
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Gabriel Peinkofer
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Anne C Geisler
- General and Interventional Cardiology University Heart Center Hamburg, University Hospital Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Bianca Geis
- General and Interventional Cardiology University Heart Center Hamburg, University Hospital Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Alexander P Schwoerer
- Department of Cellular and Integrative Physiology, University Medical Center Hamburg Eppendorf, DZHK (German Centre of Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lucie Carrier
- Experimental Pharmacology and Toxicology, University Hospital Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthias Dewenter
- Institute of Experimental Cardiology, University of Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Xiaojing Luo
- Department of Pharmacology and Toxicology, Technische Universitaet Dresden, Dresden, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Technische Universitaet Dresden, Dresden, Germany
| | - Michael Wagner
- Department of Pharmacology and Toxicology, Technische Universitaet Dresden, Dresden, Germany
- Clinic for Internal Medicine and Cardiology, Heart Center Dresden, Dresden, Germany
| | - Matti Adam
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Stephan Baldus
- Clinic III for Internal Medicine, Department of Cardiology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Volker Rudolph
- Clinic for General and Interventional Cardiology/ Angiology, Herz- Und Diabeteszentrum NRW, Ruhr-Universitaet Bochum, Bad Oeynhausen, Germany
| |
Collapse
|
37
|
Mohieldin AM, Pala R, Sherpa RT, Alanazi M, Alanazi A, Shamloo K, Ahsan A, AbouAlaiwi WA, Moresco JJ, Yates JR, Nauli SM. Proteomic Identification Reveals the Role of Ciliary Extracellular-Like Vesicle in Cardiovascular Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903140. [PMID: 32832346 PMCID: PMC7435257 DOI: 10.1002/advs.201903140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/07/2020] [Indexed: 06/11/2023]
Abstract
Primary cilia are shown to have membrane swelling, also known as ciliary bulbs. However, the role of these structures and their physiological relevance remains unknown. Here, it is reported that a ciliary bulb has extracellular vesicle (EV)-like characteristics. The ciliary extracellular-like vesicle (cELV) has a unique dynamic movement and can be released by mechanical fluid force. To better identify the cELV, differential multidimensional proteomic analyses are performed on the cELV. A database of 172 cELV proteins is generated, and all that examined are confirmed to be in the cELV. Repressing the expression of these proteins in vitro and in vivo inhibits cELV formation. In addition to the randomized heart looping, hydrocephalus, and cystic kidney in fish, compensated heart contractility is observed in both fish and mouse models. Specifically, low circulation of cELV results in hypotension with compensated heart function, left ventricular hypertrophy, cardiac fibrosis, and arrhythmogenic characteristics, which result in a high mortality rate in mice. Furthermore, the overall ejection fraction, stroke volume, and cardiac output are significantly decreased in mice lacking cELV. It is thus proposed that the cELV as a nanocompartment within a primary cilium plays an important role in cardiovascular functions.
Collapse
Affiliation(s)
- Ashraf M. Mohieldin
- Department of Biomedical and Pharmaceutical SciencesChapman UniversityIrvineCA92618USA
| | - Rajasekharreddy Pala
- Department of Biomedical and Pharmaceutical SciencesChapman UniversityIrvineCA92618USA
| | - Rinzhin T. Sherpa
- Department of Biomedical and Pharmaceutical SciencesChapman UniversityIrvineCA92618USA
| | - Madhawi Alanazi
- Department of Biomedical and Pharmaceutical SciencesChapman UniversityIrvineCA92618USA
| | - Ashwaq Alanazi
- Department of Biomedical and Pharmaceutical SciencesChapman UniversityIrvineCA92618USA
| | - Kiumars Shamloo
- Department of Biomedical and Pharmaceutical SciencesChapman UniversityIrvineCA92618USA
| | - Amir Ahsan
- Department of Physics, Computer Science and EngineeringChapman UniversityOrangeCA92866USA
| | - Wissam A. AbouAlaiwi
- Department of Pharmacology and Experimental TherapeuticsUniversity of ToledoToledoOH43614USA
| | - James J. Moresco
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
| | - John R. Yates
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCA92037USA
| | - Surya M. Nauli
- Department of Biomedical and Pharmaceutical SciencesChapman UniversityIrvineCA92618USA
- Department of MedicineUniversity of California IrvineIrvineCA92868USA
| |
Collapse
|
38
|
Bardsley EN, Paterson DJ. Neurocardiac regulation: from cardiac mechanisms to novel therapeutic approaches. J Physiol 2020; 598:2957-2976. [PMID: 30307615 PMCID: PMC7496613 DOI: 10.1113/jp276962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022] Open
Abstract
Cardiac sympathetic overactivity is a well-established contributor to the progression of neurogenic hypertension and heart failure, yet the underlying pathophysiology remains unclear. Recent studies have highlighted the importance of acutely regulated cyclic nucleotides and their effectors in the control of intracellular calcium and exocytosis. Emerging evidence now suggests that a significant component of sympathetic overactivity and enhanced transmission may arise from impaired cyclic nucleotide signalling, resulting from compromised phosphodiesterase activity, as well as alterations in receptor-coupled G-protein activation. In this review, we address some of the key cellular and molecular pathways that contribute to sympathetic overactivity in hypertension and discuss their potential for therapeutic targeting.
Collapse
Affiliation(s)
- E. N. Bardsley
- Wellcome Trust OXION Initiative in Ion Channels and DiseaseOxfordUK
- Burdon Sanderson Cardiac Science Centre, Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordOX1 3PTUK
| | - D. J. Paterson
- Wellcome Trust OXION Initiative in Ion Channels and DiseaseOxfordUK
- Burdon Sanderson Cardiac Science Centre, Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordOX1 3PTUK
| |
Collapse
|
39
|
Tam K, Richards DA, Aronovitz MJ, Martin GL, Pande S, Jaffe IZ, Blanton RM. Sacubitril/Valsartan Improves Left Ventricular Function in Chronic Pressure Overload Independent of Intact Cyclic Guanosine Monophosphate-dependent Protein Kinase I Alpha Signaling. J Card Fail 2020; 26:769-775. [PMID: 32464187 DOI: 10.1016/j.cardfail.2020.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 03/18/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Combined angiotensin receptor/neprilysin inhibition with sacubitril/valsartan (Sac/Val) has emerged as a therapy for heart failure. The presumed mechanism of benefit is through prevention of natriuretic peptide degradation, leading to increased cyclic guanosine monophosphate (cGMP)-dependent protein kinase (PKG) signaling. However, the specific requirement of PKG for Sac/Val effects remains untested. METHODS AND RESULTS We examined Sac/Val treatment in mice with mutation of the cGMP-dependent protein kinase I (PKGI)α leucine zipper domain, which is required for cGMP-PKGIα antiremodeling actions in vivo. Wild-type (WT) or PKG leucine zipper mutant (LZM) mice were exposed to 56-day left ventricular (LV) pressure overload by moderate (26G) transaortic constriction (TAC). At day 14 after TAC, mice were randomized to vehicle or Sac/Val by oral gavage. TAC induced the same degree of LV pressure overload in WT and LZM mice, which was not affected by Sac/Val. Although LZM mice, but not WT, developed LV dilation after TAC, Sac/Val improved cardiac hypertrophy and LV fractional shortening to the same degree in both the WT and LZM TAC mice. CONCLUSION These findings indicate the beneficial effects of Sac/Val on LV structure and function in moderate pressure overload. The unexpected finding that PKGIα mutation does not abolish the Sac/Val effects on cardiac hypertrophy and on LV function suggests that signaling other than natriuretic peptide- cGMP-PKG mediates the therapeutic benefits of neprilysin inhibition in heart failure.
Collapse
Affiliation(s)
- Kelly Tam
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Daniel A Richards
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Mark J Aronovitz
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Gregory L Martin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Suchita Pande
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Robert M Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts.
| |
Collapse
|
40
|
Karam S, Margaria JP, Bourcier A, Mika D, Varin A, Bedioune I, Lindner M, Bouadjel K, Dessillons M, Gaudin F, Lefebvre F, Mateo P, Lechène P, Gomez S, Domergue V, Robert P, Coquard C, Algalarrondo V, Samuel JL, Michel JB, Charpentier F, Ghigo A, Hirsch E, Fischmeister R, Leroy J, Vandecasteele G. Cardiac Overexpression of PDE4B Blunts β-Adrenergic Response and Maladaptive Remodeling in Heart Failure. Circulation 2020; 142:161-174. [PMID: 32264695 DOI: 10.1161/circulationaha.119.042573] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The cyclic AMP (adenosine monophosphate; cAMP)-hydrolyzing protein PDE4B (phosphodiesterase 4B) is a key negative regulator of cardiac β-adrenergic receptor stimulation. PDE4B deficiency leads to abnormal Ca2+ handling and PDE4B is decreased in pressure overload hypertrophy, suggesting that increasing PDE4B in the heart is beneficial in heart failure. METHODS We measured PDE4B expression in human cardiac tissues and developed 2 transgenic mouse lines with cardiomyocyte-specific overexpression of PDE4B and an adeno-associated virus serotype 9 encoding PDE4B. Myocardial structure and function were evaluated by echocardiography, ECG, and in Langendorff-perfused hearts. Also, cAMP and PKA (cAMP dependent protein kinase) activity were monitored by Förster resonance energy transfer, L-type Ca2+ current by whole-cell patch-clamp, and cardiomyocyte shortening and Ca2+ transients with an Ionoptix system. Heart failure was induced by 2 weeks infusion of isoproterenol or transverse aortic constriction. Cardiac remodeling was evaluated by serial echocardiography, morphometric analysis, and histology. RESULTS PDE4B protein was decreased in human failing hearts. The first PDE4B-transgenic mouse line (TG15) had a ≈15-fold increase in cardiac cAMP-PDE activity and a ≈30% decrease in cAMP content and fractional shortening associated with a mild cardiac hypertrophy that resorbed with age. Basal ex vivo myocardial function was unchanged, but β-adrenergic receptor stimulation of cardiac inotropy, cAMP, PKA, L-type Ca2+ current, Ca2+ transients, and cell contraction were blunted. Endurance capacity and life expectancy were normal. Moreover, these mice were protected from systolic dysfunction, hypertrophy, lung congestion, and fibrosis induced by chronic isoproterenol treatment. In the second PDE4B-transgenic mouse line (TG50), markedly higher PDE4B overexpression, resulting in a ≈50-fold increase in cardiac cAMP-PDE activity caused a ≈50% decrease in fractional shortening, hypertrophy, dilatation, and premature death. In contrast, mice injected with adeno-associated virus serotype 9 encoding PDE4B (1012 viral particles/mouse) had a ≈50% increase in cardiac cAMP-PDE activity, which did not modify basal cardiac function but efficiently prevented systolic dysfunction, apoptosis, and fibrosis, while attenuating hypertrophy induced by chronic isoproterenol infusion. Similarly, adeno-associated virus serotype 9 encoding PDE4B slowed contractile deterioration, attenuated hypertrophy and lung congestion, and prevented apoptosis and fibrotic remodeling in transverse aortic constriction. CONCLUSIONS Our results indicate that a moderate increase in PDE4B is cardioprotective and suggest that cardiac gene therapy with PDE4B might constitute a new promising approach to treat heart failure.
Collapse
Affiliation(s)
- Sarah Karam
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | | | - Aurélia Bourcier
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Delphine Mika
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Audrey Varin
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Ibrahim Bedioune
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Marta Lindner
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Kaouter Bouadjel
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Matthieu Dessillons
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Françoise Gaudin
- Université Paris-Saclay, Inserm, UMS-IPSIT, 92296 Châtenay-Malabry, France (F.G., V.D., P.R.)
| | - Florence Lefebvre
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Philippe Mateo
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Patrick Lechène
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Susana Gomez
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Valérie Domergue
- Université Paris-Saclay, Inserm, UMS-IPSIT, 92296 Châtenay-Malabry, France (F.G., V.D., P.R.)
| | - Pauline Robert
- Université Paris-Saclay, Inserm, UMS-IPSIT, 92296 Châtenay-Malabry, France (F.G., V.D., P.R.)
| | - Charlène Coquard
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Vincent Algalarrondo
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Jane-Lise Samuel
- UMR-S 942, Inserm, Paris University, 75010 Paris, France (J.-L.S.)
| | - Jean-Baptiste Michel
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University di Torino, 10126 Torino, Italy (J.P.M., A.G., E.H.).,UMR-S 1148, INSERM, Paris University, X. Bichat hospital, 75018 Paris, France (J.-B.M.)
| | - Flavien Charpentier
- Institut du thorax, Inserm, CNRS, Univ. Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France (F.C.)
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University di Torino, 10126 Torino, Italy (J.P.M., A.G., E.H.)
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University di Torino, 10126 Torino, Italy (J.P.M., A.G., E.H.)
| | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Jérôme Leroy
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| | - Grégoire Vandecasteele
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, 92296 Châtenay-Malabry, France (S.K., A.R., D.M., A.V., I.B., M.L., K.B., M.D., F.L., P.M., P.L., S.G., C.C., V.A., R.F., J.L., G.V.)
| |
Collapse
|
41
|
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
|
42
|
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
|
43
|
Phosphodiesterase 2 inhibition preferentially promotes NO/guanylyl cyclase/cGMP signaling to reverse the development of heart failure. Proc Natl Acad Sci U S A 2018; 115:E7428-E7437. [PMID: 30012589 PMCID: PMC6077693 DOI: 10.1073/pnas.1800996115] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a shared manifestation of several cardiovascular pathologies, including hypertension and myocardial infarction, and a limited repertoire of treatment modalities entails that the associated morbidity and mortality remain high. Impaired nitric oxide (NO)/guanylyl cyclase (GC)/cyclic guanosine-3',5'-monophosphate (cGMP) signaling, underpinned, in part, by up-regulation of cyclic nucleotide-hydrolyzing phosphodiesterase (PDE) isozymes, contributes to the pathogenesis of HF, and interventions targeted to enhancing cGMP have proven effective in preclinical models and patients. Numerous PDE isozymes coordinate the regulation of cardiac cGMP in the context of HF; PDE2 expression and activity are up-regulated in experimental and human HF, but a well-defined role for this isoform in pathogenesis has yet to be established, certainly in terms of cGMP signaling. Herein, using a selective pharmacological inhibitor of PDE2, BAY 60-7550, and transgenic mice lacking either NO-sensitive GC-1α (GC-1α-/-) or natriuretic peptide-responsive GC-A (GC-A-/-), we demonstrate that the blockade of PDE2 promotes cGMP signaling to offset the pathogenesis of experimental HF (induced by pressure overload or sympathetic hyperactivation), reversing the development of left ventricular hypertrophy, compromised contractility, and cardiac fibrosis. Moreover, we show that this beneficial pharmacodynamic profile is maintained in GC-A-/- mice but is absent in animals null for GC-1α or treated with a NO synthase inhibitor, revealing that PDE2 inhibition preferentially enhances NO/GC/cGMP signaling in the setting of HF to exert wide-ranging protection to preserve cardiac structure and function. These data substantiate the targeting of PDE2 in HF as a tangible approach to maximize myocardial cGMP signaling and enhancing therapy.
Collapse
|
44
|
Subramanian H, Froese A, Jönsson P, Schmidt H, Gorelik J, Nikolaev VO. Distinct submembrane localisation compartmentalises cardiac NPR1 and NPR2 signalling to cGMP. Nat Commun 2018; 9:2446. [PMID: 29934640 PMCID: PMC6014982 DOI: 10.1038/s41467-018-04891-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/29/2018] [Indexed: 12/11/2022] Open
Abstract
Natriuretic peptides (NPs) are important hormones that regulate multiple cellular functions including cardiovascular physiology. In the heart, two natriuretic peptide receptors NPR1 and NPR2 act as membrane guanylyl cyclases to produce 3′,5′-cyclic guanosine monophosphate (cGMP). Although both receptors protect from cardiac hypertrophy, their effects on contractility are markedly different, from little effect (NPR1) to pronounced negative inotropic and positive lusitropic responses (NPR2) with unclear underlying mechanisms. Here we use a scanning ion conductance microscopy (SICM) approach combined with Förster resonance energy transfer (FRET)-based cGMP biosensors to show that whereas NPR2 is uniformly localised on the cardiomyocyte membrane, functional NPR1 receptors are found exclusively in membrane invaginations called transverse (T)-tubules. This leads to far-reaching CNP/NPR2/cGMP signals, whereas ANP/NPR1/cGMP signals are highly confined to T-tubular microdomains by local pools of phosphodiesterase 2. This provides a previously unrecognised molecular basis for clearly distinct functional effects engaged by different cGMP producing membrane receptors. Natriuretic peptides (NPs) are important hormones that regulate cardiovascular physiology by increasing cGMP levels in cardiomyocytes. Here the authors use scanning ion conductance microscopy and a cGMP FRET sensor to identify a differential localisation pattern for the natriuretic peptide receptors within the heart.
Collapse
Affiliation(s)
- Hariharan Subramanian
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martnistr. 52, D-20246, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martnistr. 52, D-20246, Hamburg, Germany
| | - Alexander Froese
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martnistr. 52, D-20246, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martnistr. 52, D-20246, Hamburg, Germany.,Clinic of Cardiology and Pulmonology, University Medical Center Göttingen, Robert-Koch-Str. 40, D-37075, Göttingen, Germany
| | - Peter Jönsson
- Department of Chemistry, Lund University, Naturvetarvägen 14, SE-221 00, Lund, Sweden
| | - Hannes Schmidt
- Interfaculty Institute of Biochemistry, University of Tübingen, Hoppe-Seyler-Straße 4, D-72076, Tübingen, Germany
| | - Julia Gorelik
- Myocardial Function, National Heart and Lung Institute, ICTEM, Hammersmith Hospital, Imperial College London, Du Cane Road, W12 0NN, London, UK.
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martnistr. 52, D-20246, Hamburg, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martnistr. 52, D-20246, Hamburg, Germany.
| |
Collapse
|
45
|
Wiedmann F, Schulte JS, Gomes B, Zafeiriou MP, Ratte A, Rathjens F, Fehrmann E, Scholz B, Voigt N, Müller FU, Thomas D, Katus HA, Schmidt C. Atrial fibrillation and heart failure-associated remodeling of two-pore-domain potassium (K2P) channels in murine disease models: focus on TASK-1. Basic Res Cardiol 2018; 113:27. [DOI: 10.1007/s00395-018-0687-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/04/2018] [Indexed: 12/27/2022]
|
46
|
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
|
47
|
Galindo-Tovar A, Vargas ML, Kaumann AJ. Phosphodiesterase PDE2 activity, increased by isoprenaline, does not reduce β-adrenoceptor-mediated chronotropic and inotropic effects in rat heart. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:571-585. [PMID: 29556684 DOI: 10.1007/s00210-018-1480-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 02/13/2018] [Indexed: 12/12/2022]
Abstract
Myocardial PDE2 activity increases in terminal human heart failure and after isoprenaline infusion in rat heart. PDE2 inhibitors do not potentiate the murine sinoatrial tachycardia produced by noradrenaline. We investigated whether isoprenaline infusion induces PDE2 to decrease the chronotropic and inotropic effects of catecholamines in rat heart. Sprague-Dawley rats were infused with isoprenaline (2.4 mg kg-1 day-1) for 3 days. We used spontaneously beating right atria, paced right ventricular strips and left ventricular papillary muscles. The effects of the PDE2 inhibitors EHNA (10 μM) and Bay 60-7550 (0.1-1 μM) were investigated on the cardiostimulation produced by noradrenaline (ICI118551 50 nM present to block β2-adrenoceptors) and adrenaline (CGP20712A 300 nM present to block β1-adrenoceptors). Hydrolysis of cAMP by PDE2 was measured by radioenzyme assay. Bay 60-7550 but not EHNA increased sinoatrial beating. A stable tachycardia elicited by noradrenaline (10 nM) or adrenaline (1 μM) was not increased by the PDE2 inhibitors. Isoprenaline infusion increased the hydrolytic PDE2 activity threefold in left ventricle, reduced the chronotropic and inotropic effects and potency of noradrenaline and abolished the effects of adrenaline. The potency of the catecholamines was not increased by the PDE2 inhibitors. Neither EHNA nor Bay 60-7550 potentiated the effects of the catecholamines. Rat PDE2 decreased basal sinoatrial beating but did not reduce the sinoatrial tachycardia or increases of ventricular force mediated through β1- and β2-adrenoceptors. The β-adrenoceptor desensitization induced by the isoprenaline infusion was not reversed by the PDE2 inhibitors despite the increased hydrolysis of cAMP by PDE2.
Collapse
Affiliation(s)
- Alejandro Galindo-Tovar
- Grado en Farmacia, Facultad Ciencias de la Salud, Universidad Católica San Antonio de Murcia, 30107, Murcia, Spain
| | - María Luisa Vargas
- Departamento de Farmacología, Facultad de Medicina, Universidad de Murcia, Campus de Espinardo, 30100, Murcia, Spain
| | - Alberto J Kaumann
- Departamento de Farmacología, Facultad de Medicina, Universidad de Murcia, Campus de Espinardo, 30100, Murcia, Spain.
| |
Collapse
|
48
|
Ercu M, Klussmann E. Roles of A-Kinase Anchoring Proteins and Phosphodiesterases in the Cardiovascular System. J Cardiovasc Dev Dis 2018; 5:jcdd5010014. [PMID: 29461511 PMCID: PMC5872362 DOI: 10.3390/jcdd5010014] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/16/2018] [Accepted: 02/18/2018] [Indexed: 12/13/2022] Open
Abstract
A-kinase anchoring proteins (AKAPs) and cyclic nucleotide phosphodiesterases (PDEs) are essential enzymes in the cyclic adenosine 3′-5′ monophosphate (cAMP) signaling cascade. They establish local cAMP pools by controlling the intensity, duration and compartmentalization of cyclic nucleotide-dependent signaling. Various members of the AKAP and PDE families are expressed in the cardiovascular system and direct important processes maintaining homeostatic functioning of the heart and vasculature, e.g., the endothelial barrier function and excitation-contraction coupling. Dysregulation of AKAP and PDE function is associated with pathophysiological conditions in the cardiovascular system including heart failure, hypertension and atherosclerosis. A number of diseases, including autosomal dominant hypertension with brachydactyly (HTNB) and type I long-QT syndrome (LQT1), result from mutations in genes encoding for distinct members of the two classes of enzymes. This review provides an overview over the AKAPs and PDEs relevant for cAMP compartmentalization in the heart and vasculature and discusses their pathophysiological role as well as highlights the potential benefits of targeting these proteins and their protein-protein interactions for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Maria Ercu
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin 13125, Germany.
| | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin 13125, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin 13347, Germany.
| |
Collapse
|
49
|
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]
|
50
|
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
|