1
|
Feng Y, Xue Y, Feng X, Li Z, Ren L, Guo W, Hou Y, Shu T, Zhang W, Yang Y, Zhou Y, Song K, Xiong J, Liu B, Wang J, Zhao H. Phosphodiesterase 5 inactivation in vascular smooth muscle cells aggravates aortic aneurysm and dissection. J Pathol 2025; 266:144-159. [PMID: 40145405 DOI: 10.1002/path.6411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 12/16/2024] [Accepted: 01/30/2025] [Indexed: 03/28/2025]
Abstract
Aortic aneurysm and dissection (AAD) are vascular disorders with high mortality. Previous evidence has suggested an elevated risk of AAD associated with the use of phosphodiesterase 5A (PDE5A) inhibitors. PDE5A, a cGMP-hydrolyzing enzyme, is enriched in vascular smooth muscle cells (SMCs), but the role of SMC-specific PDE5A in the pathogenesis of AAD is still unclear. In this study, PDE5A expression in human and mouse aortic tissues was analyzed by single-cell RNA sequencing (scRNA-seq), western blotting, immunofluorescence, and immunohistochemistry staining. SMC-specific PDE5A knockout (PDE5ASMC-/-) and PDE5A-overexpressing (PDE5ASMC-OE) mice were constructed and utilized, along with an AAD mouse model induced by a high-fat diet and angiotensin II (Ang II) infusion. In vivo imaging and histological analyses were performed to assess aortic pathologies. PDE5A expression was reduced in human and mouse AAD aortic tissues, primarily in SMCs. Pharmacological inhibition or genetic knockout of PDE5A in SMCs exacerbated aortic wall dilatation and elastin fiber degradation, increasing AAD incidence. In contrast, the AAD phenotype was rescued in challenged PDE5ASMC-OE mice. Mechanistically, PDE5A expression influenced myosin light chain (MLC) phosphorylation, a key regulator of SMC contractility. In AAD tissues from PDE5ASMC-/- mice, increased cGMP-dependent protein kinase (PKG) activation and decreased MLC phosphorylation indicate enhanced aortic relaxation. In conclusion, our findings suggest that PDE5A downregulation or inhibition plays a causative role in exacerbating AAD likely by potentiating cGMP/PKG-mediated aortic SMC relaxation. Our findings highlight the need for caution in the clinical use of PDE5 inhibitors in patients at risk of aortic diseases. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Animals
- Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 5/genetics
- Humans
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/drug effects
- Aortic Dissection/pathology
- Aortic Dissection/enzymology
- Aortic Dissection/genetics
- Aortic Dissection/chemically induced
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Aortic Aneurysm/pathology
- Aortic Aneurysm/enzymology
- Aortic Aneurysm/genetics
- Mice, Knockout
- Mice
- Disease Models, Animal
- Male
- Phosphodiesterase 5 Inhibitors/pharmacology
- Mice, Inbred C57BL
- Angiotensin II
- Aorta/pathology
- Aorta/enzymology
- Female
Collapse
Affiliation(s)
- Yuyao Feng
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, PR China
| | - Yunfei Xue
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Xiaohang Feng
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Zhiwei Li
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Luxia Ren
- Department of Vascular and Endovascular Surgery, Chinese PLA General Hospital, Beijing, PR China
| | - Wenjun Guo
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Yangfeng Hou
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Ting Shu
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
| | - Wensi Zhang
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Yang Yang
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Yitian Zhou
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Kai Song
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Jiang Xiong
- Department of Vascular and Endovascular Surgery, Chinese PLA General Hospital, Beijing, PR China
| | - Bao Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, PR China
| | - Jing Wang
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, PR China
| | - Hongmei Zhao
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Beijing, PR China
| |
Collapse
|
2
|
Yan J, Wang X, Cao P, Li Q, Wu H. Downregulation of miR-214 promotes dilated Cardiomyopathy Progression through PDE5A-Mediated cGMP regulation. Sci Rep 2024; 14:28070. [PMID: 39543318 PMCID: PMC11564883 DOI: 10.1038/s41598-024-78983-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a myocardial disorder resulting in a substantial decline in cardiac function and potentially leading to heart failure. This research combines bioinformatics analysis with empirical validation to explore the roles and mechanisms of miR-214 in DCM. Using the DEseq2 R package, a total of 125 differentially expressed circulating miRNAs (DE c-miRNAs) and 784 DE genes (DEGs) were identified. Cross-analysis between target genes of DE c-miRNAs and DEGs identified 124 common genes, and protein-protein interaction analysis of common genes identified 11 hub genes. Twelve DE c-miRNAs were further verified by quantifying their levels in the serum of DCM patients and healthy individuals. miR-214 levels were significantly decreased in serum from DCM patients, positively correlated with left ventricular ejection fraction and left ventricular fractional shortening. Further analysis showed that miR-214 directly targets and negatively regulates phosphodiesterase 5 A (PDE5A). Elevated PDE5A expression reduced cGMP levels; however, using sildenafil, a PDE5A inhibitor, reversed this effect, substantiating the regulatory mechanism of miR-214 on cGMP via PDE5A. These results provide new potential targets for the diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Jingjing Yan
- Graduate School, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xinzhou Wang
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, 450002, China
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002, Henan, China
| | - Panxia Cao
- Graduate School, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Qiaozhi Li
- Heart Center, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Hong Wu
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, 450002, China.
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002, Henan, China.
- Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, 450002, China.
| |
Collapse
|
3
|
Li S, Xin Q, Yan Y, Wang X, Ai H, Que B, Gong W, Nie S. Pde5 Inhibition Reduced Blood Pressure and Alleviated Target Organ Damage in Chronic Intermittent Hypoxia. J Cardiovasc Pharmacol 2024; 84:81-91. [PMID: 38030140 PMCID: PMC11230658 DOI: 10.1097/fjc.0000000000001519] [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] [Received: 05/28/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023]
Abstract
ABSTRACT The role of phosphodiesterase 5 (Pde5) in obstructive sleep apnea-induced damage remains unclear. Our study aimed to investigate the role of Pde5 in the chronic intermittent hypoxia (CIH) model. C57BL/6J wild-type (WT) mice (n = 48) and Pde5 knockout (Pde5 -/- ) mice (n = 24) were randomly assigned to CIH group and room air group. After 6 weeks, some WT mice (n = 24) in CIH group were given sildenafil or saline gavage for another 4 weeks. Blood pressure was regularly measured during the experiment. Echocardiography was used to estimate cardiac function. We collected organs from each group of mice and measured their physical indicators. Histochemical staining was used to explore the size of cardiomyocyte and fibrosis area of various organs. Cyclic guanosine monophosphate and malondialdehyde concentrations in serum were measured by ELISA assay. Compared with the RA-treated group, the 6-week CIH resulted in a significant increase in blood pressure, altered heart structure, and reduced serum cyclic guanosine monophosphate in WT mice. Pde5 -/- mice and sildenafil intragastric administration significantly reduced systolic blood pressure in CIH condition and attenuated the damage of target organs. In CIH model, we found that the cardiomyocyte size and fibrosis area of heart and kidney significantly reduced in Pde5 -/- groups. Besides, endogenous and exogenous inhibition of Pde5 reduced malondialdehyde level and inflammatory and oxidative stress markers expression in CIH condition. In this study, we found that Pde5 inhibition could reduce blood pressure and alleviate target organ damage in the CIH model, which may be mediated through the oxidative stress pathway.
Collapse
Affiliation(s)
- Siyi Li
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Qingjie Xin
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Yan Yan
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Xiao Wang
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Hui Ai
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Bin Que
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Wei Gong
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Shaoping Nie
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
4
|
Zhang W, Peng Q, Huang X, Huang Q, Zhang Z, Li F, Zheng N, Shi B, Fan Z, Maj T, Chen R. Commensal microbiome dysbiosis elicits interleukin-8 signaling to drive fibrotic skin disease. PNAS NEXUS 2024; 3:pgae273. [PMID: 39081787 PMCID: PMC11287872 DOI: 10.1093/pnasnexus/pgae273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/22/2024] [Indexed: 08/02/2024]
Abstract
Wound healing is an intensely studied topic involved in many relevant pathophysiological processes, including fibrosis. Despite the large interest in fibrosis, the network that is related to commensal microbiota and skin fibrosis remains mysterious. Here, we pay attention to keloid, a classical yet intractable skin fibrotic disease to establish the association between commensal microbiota to scaring tissue. Our histological data reveal the presence of microbiota in the keloids. 16S rRNA sequencing characterizes microbial composition and divergence between the pathological and normal skin tissues. Moreover, the data show elevation of interleukin-8 (IL-8) in both the circulation and keloid tissue, which elicited the collagen accumulation and migratory program of dermal fibroblasts via CXCR1/2 receptor. Our research provides insights into the pathology of human fibrotic diseases, advocating commensal bacteria and IL-8 signaling as useful targets in future interventions of recurrent keloid disease.
Collapse
Affiliation(s)
- Wenyu Zhang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| | - Qili Peng
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| | - Xian Huang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| | - Qing Huang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| | - Zhiliang Zhang
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Renji Ningbo Hangzhou Bay Hospital, School of Medicine, Shanghai Jiao Tong University, Binhai Second Road 1155, Ningbo 315600, China
| | - Fuli Li
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| | - Naisheng Zheng
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| | - Binsheng Shi
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| | - Zhihong Fan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Renji Ningbo Hangzhou Bay Hospital, School of Medicine, Shanghai Jiao Tong University, Binhai Second Road 1155, Ningbo 315600, China
| | - Tomasz Maj
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| | - Rui Chen
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Shanghai 200240, China
| |
Collapse
|
5
|
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
|
6
|
Kartchner D, McCoy K, Dubey J, Zhang D, Zheng K, Umrani R, Kim JJ, Mitchell CS. Literature-Based Discovery to Elucidate the Biological Links between Resistant Hypertension and COVID-19. BIOLOGY 2023; 12:1269. [PMID: 37759668 PMCID: PMC10526006 DOI: 10.3390/biology12091269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Multiple studies have reported new or exacerbated persistent or resistant hypertension in patients previously infected with COVID-19. We used literature-based discovery to identify and prioritize multi-scalar explanatory biology that relates resistant hypertension to COVID-19. Cross-domain text mining of 33+ million PubMed articles within a comprehensive knowledge graph was performed using SemNet 2.0. Unsupervised rank aggregation determined which concepts were most relevant utilizing the normalized HeteSim score. A series of simulations identified concepts directly related to COVID-19 and resistant hypertension or connected via one of three renin-angiotensin-aldosterone system hub nodes (mineralocorticoid receptor, epithelial sodium channel, angiotensin I receptor). The top-ranking concepts relating COVID-19 to resistant hypertension included: cGMP-dependent protein kinase II, MAP3K1, haspin, ral guanine nucleotide exchange factor, N-(3-Oxododecanoyl)-L-homoserine lactone, aspartic endopeptidases, metabotropic glutamate receptors, choline-phosphate cytidylyltransferase, protein tyrosine phosphatase, tat genes, MAP3K10, uridine kinase, dicer enzyme, CMD1B, USP17L2, FLNA, exportin 5, somatotropin releasing hormone, beta-melanocyte stimulating hormone, pegylated leptin, beta-lipoprotein, corticotropin, growth hormone-releasing peptide 2, pro-opiomelanocortin, alpha-melanocyte stimulating hormone, prolactin, thyroid hormone, poly-beta-hydroxybutyrate depolymerase, CR 1392, BCR-ABL fusion gene, high density lipoprotein sphingomyelin, pregnancy-associated murine protein 1, recQ4 helicase, immunoglobulin heavy chain variable domain, aglycotransferrin, host cell factor C1, ATP6V0D1, imipramine demethylase, TRIM40, H3C2 gene, COL1A1+COL1A2 gene, QARS gene, VPS54, TPM2, MPST, EXOSC2, ribosomal protein S10, TAP-144, gonadotropins, human gonadotropin releasing hormone 1, beta-lipotropin, octreotide, salmon calcitonin, des-n-octanoyl ghrelin, liraglutide, gastrins. Concepts were mapped to six physiological themes: altered endocrine function, 23.1%; inflammation or cytokine storm, 21.3%; lipid metabolism and atherosclerosis, 17.6%; sympathetic input to blood pressure regulation, 16.7%; altered entry of COVID-19 virus, 14.8%; and unknown, 6.5%.
Collapse
Affiliation(s)
- David Kartchner
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Kevin McCoy
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Janhvi Dubey
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Dongyu Zhang
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Kevin Zheng
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Rushda Umrani
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- College of Computing, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - James J. Kim
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Cassie S. Mitchell
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Center for Machine Learning at Georgia Tech, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
7
|
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
|
8
|
Samidurai A, Xi L, Das A, Kukreja RC. Beyond Erectile Dysfunction: cGMP-Specific Phosphodiesterase 5 Inhibitors for Other Clinical Disorders. Annu Rev Pharmacol Toxicol 2023; 63:585-615. [PMID: 36206989 DOI: 10.1146/annurev-pharmtox-040122-034745] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cyclic guanosine monophosphate (cGMP), an important intracellular second messenger, mediates cellular functional responses in all vital organs. Phosphodiesterase 5 (PDE5) is one of the 11 members of the cyclic nucleotide phosphodiesterase (PDE) family that specifically targets cGMP generated by nitric oxide-driven activation of the soluble guanylyl cyclase. PDE5 inhibitors, including sildenafil and tadalafil, are widely used for the treatment of erectile dysfunction, pulmonary arterial hypertension, and certain urological disorders. Preclinical studies have shown promising effects of PDE5 inhibitors in the treatment of myocardial infarction, cardiac hypertrophy, heart failure, cancer and anticancer-drug-associated cardiotoxicity, diabetes, Duchenne muscular dystrophy, Alzheimer's disease, and other aging-related conditions. Many clinical trials with PDE5 inhibitors have focused on the potential cardiovascular, anticancer, and neurological benefits. In this review, we provide an overview of the current state of knowledge on PDE5 inhibitors and their potential therapeutic indications for various clinical disorders beyond erectile dysfunction.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Lei Xi
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| |
Collapse
|
9
|
Pofi R, Giannetta E, Feola T, Galea N, Barbagallo F, Campolo F, Badagliacca R, Barbano B, Ciolina F, Defeudis G, Filardi T, Sesti F, Minnetti M, Vizza CD, Pasqualetti P, Caboni P, Carbone I, Francone M, Catalano C, Pozzilli P, Lenzi A, Venneri MA, Gianfrilli D, Isidori AM. Sex-specific effects of daily tadalafil on diabetic heart kinetics in RECOGITO, a randomized, double-blind, placebo-controlled trial. Sci Transl Med 2022; 14:eabl8503. [PMID: 35704597 DOI: 10.1126/scitranslmed.abl8503] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cyclic GMP-phosphodiesterase type 5 (PDE5) inhibition has been shown to counteract maladaptive cardiac changes triggered by diabetes in some but not all studies. We performed a single-center, 20-week, double-blind, randomized, placebo-controlled trial (NCT01803828) to assess sex differences in cardiac remodeling after PDE5 inhibition in patients with diabetic cardiomyopathy. A total of 122 men and women (45 to 80 years) with long-duration (>3 years) and well-controlled type 2 diabetes mellitus (T2DM; HbA1c < 86 mmol/mol) were selected according to echocardiographic signs of cardiac remodeling. Patients were randomly assigned (1:1) to placebo or oral tadalafil (20 mg, once daily). The primary outcome was to evaluate sex differences in cardiac torsion change. Secondary outcomes were changes in cardiovascular, metabolic, immune, and renal function. At 20 weeks, the treatment-by-sex interaction documented an improvement in cardiac torsion (-3.40°, -5.96; -0.84, P = 0.011) and fiber shortening (-1.19%, -2.24; -0.14, P = 0.027) in men but not women. The primary outcome could not be explained by differences in cGMP concentrations or tadalafil pharmacodynamics. In both sexes, tadalafil improved hsa-miR-199-5p expression, biomarkers of cardiovascular remodeling, albuminuria, renal artery resistive index, and circulating Klotho concentrations. Immune cell profiling revealed an improvement in low-grade chronic inflammation: Classic CD14++CD16- monocytes reduced, and Tie2+ monocytes increased. Nine patients (14.5%) had minor adverse reactions after tadalafil administration. Continuous PDE5 inhibition could offer a strategy to target cardiorenal complications of T2DM, with sex- and tissue-specific responses. Further studies are needed to confirm Klotho and hsa-miR-199-5p as markers for T2DM complications.
Collapse
Affiliation(s)
- Riccardo Pofi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Tiziana Feola
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy.,Neuroendocrinology, Neuromed Institute, IRCCS, 86077 Pozzilli (IS), Italy
| | - Nicola Galea
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Barbagallo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Roberto Badagliacca
- Department of Cardiovascular and Respiratory Diseases, Sapienza University of Rome, 00161 Rome, Italy
| | - Biagio Barbano
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Ciolina
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Giuseppe Defeudis
- Unit of Endocrinology and Diabetes, Department of Medicine, University Campus Bio-Medico di Roma, 00161 Rome, Italy
| | - Tiziana Filardi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Franz Sesti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marianna Minnetti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Carmine D Vizza
- Department of Cardiovascular and Respiratory Diseases, Sapienza University of Rome, 00161 Rome, Italy
| | - Patrizio Pasqualetti
- Medical Statistics and Information Technology, AFaR, Fatebenefratelli Hospital, 00161 Rome, Italy
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Iacopo Carbone
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Francone
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Carlo Catalano
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Paolo Pozzilli
- Unit of Endocrinology and Diabetes, Department of Medicine, University Campus Bio-Medico di Roma, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
10
|
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
|
11
|
Numata G, Takimoto E. Cyclic GMP and PKG Signaling in Heart Failure. Front Pharmacol 2022; 13:792798. [PMID: 35479330 PMCID: PMC9036358 DOI: 10.3389/fphar.2022.792798] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP), produced by guanylate cyclase (GC), activates protein kinase G (PKG) and regulates cardiac remodeling. cGMP/PKG signal is activated by two intrinsic pathways: nitric oxide (NO)-soluble GC and natriuretic peptide (NP)-particulate GC (pGC) pathways. Activation of these pathways has emerged as a potent therapeutic strategy to treat patients with heart failure, given cGMP-PKG signaling is impaired in heart failure with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF). Large scale clinical trials in patients with HFrEF have shown positive results with agents that activate cGMP-PKG pathways. In patients with HFpEF, however, benefits were observed only in a subgroup of patients. Further investigation for cGMP-PKG pathway is needed to develop better targeting strategies for HFpEF. This review outlines cGMP-PKG pathway and its modulation in heart failure.
Collapse
Affiliation(s)
- Genri Numata
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Department of Advanced Translational Research and Medicine in Management of Pulmonary Hypertension, The University of Tokyo Hospital, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, United States
| |
Collapse
|
12
|
Sun W, Zhou Y, Xue H, Hou H, He G, Yang Q. Endoplasmic reticulum stress mediates homocysteine-induced hypertrophy of cardiac cells through activation of cyclic nucleotide phosphodiesterase 1C. Acta Biochim Biophys Sin (Shanghai) 2022; 54:388-399. [PMID: 35538034 PMCID: PMC9828163 DOI: 10.3724/abbs.2022009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/09/2021] [Indexed: 11/25/2022] Open
Abstract
Although the association of elevated homocysteine level with cardiac hypertrophy has been reported, the molecular mechanisms by which homocysteine induces cardiac hypertrophy remain inadequately understood. In this study we aim to uncover the roles of cyclic nucleotide phosphodiesterase 1 (PDE1) and endoplasmic reticulum (ER) stress and their relationship to advance the mechanistic understanding of homocysteine-induced cardiac cell hypertrophy. H9c2 cells and primary neonatal rat cardiomyocytes are exposed to homocysteine with or without ER stress inhibitor TUDCA or PDE1-specific inhibitor Lu AF58027, or transfected with siRNAs targeting PDE1 isoforms prior to homocysteine-exposure. Cell surface area is measured and ultrastructure is examined by transmission electron microscopy. Hypertrophic markers, PDE1 isoforms, and ER stress molecules are detected by q-PCR and western blot analysis. Intracellular cGMP and cAMP are measured by ELISA. The results show that homocysteine causes the enlargement of H9c2 cells, increases the expressions of hypertrophic markers β-MHC and ANP, upregulates PDE1A and PDE1C, promotes the expressions of ER stress molecules, and causes ER dilatation and degranulation. TUDCA and Lu AF58027 downregulate β-MHC and ANP, and alleviate cell enlargement. TUDCA decreases PDE1A and PDE1C levels. Silencing of PDE1C inhibits homocysteine-induced hypertrophy, whereas PDE1A knockdown has minor effect. Both cAMP and cGMP are decreased after homocysteine-exposure, while only cAMP is restored by Lu AF58027 and TUDCA. TUDCA and Lu AF58027 also inhibit cell enlargement, downregulate ANP, β-MHC and PDE1C, and enhance cAMP level in homocysteine-exposed primary cardiomyocytes. ER stress mediates homocysteine-induced hypertrophy of cardiac cells via upregulating PDE1C expression Cyclic nucleotide, especially cAMP, is the downstream mediator of the ER stress-PDE1C signaling axis in homocysteine-induced cell hypertrophy.
Collapse
Affiliation(s)
- Wentao Sun
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300457, China
- The Institute of Cardiovascular Diseases, Tianjin University, Tianjin 300457, China
| | - Yang Zhou
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300457, China
- The Institute of Cardiovascular Diseases, Tianjin University, Tianjin 300457, China
| | - Hongmei Xue
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300457, China
- The Institute of Cardiovascular Diseases, Tianjin University, Tianjin 300457, China
- Department of Physiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Haitao Hou
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300457, China
- The Institute of Cardiovascular Diseases, Tianjin University, Tianjin 300457, China
| | - Guowei He
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300457, China
- The Institute of Cardiovascular Diseases, Tianjin University, Tianjin 300457, China
- Drug Research and Development Center, Wannan Medical College, Wuhu 241002, China
- Department of Surgery, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Qin Yang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300457, China
- The Institute of Cardiovascular Diseases, Tianjin University, Tianjin 300457, China
| |
Collapse
|
13
|
Lukowski R, Feil R. Recent developments in cGMP research: From mechanisms to medicines and back. Br J Pharmacol 2022; 179:2321-2327. [PMID: 35332531 DOI: 10.1111/bph.15824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie (IFIB), University of Tübingen, Tübingen, Germany
| |
Collapse
|
14
|
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
|
15
|
Zhang C, Mohan A, Shi H, Yan C. Sildenafil (Viagra) Aggravates the Development of Experimental Abdominal Aortic Aneurysm. J Am Heart Assoc 2022; 11:e023053. [PMID: 34984916 PMCID: PMC9238513 DOI: 10.1161/jaha.121.023053] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background cGMP‐hydrolyzing phosphodiesterase type 5 (PDE5) regulates vascular smooth muscle cell (SMC) contraction by antagonizing cGMP‐dependent protein kinase I (PKGI)–dependent SMC relaxation. SMC contractile dysfunction is implicated in the pathogenesis of aortic aneurysm. PDE5 inhibitors have been used for treating erectile dysfunction, such as drug Viagra (sildenafil). However, a few clinical cases have reported the association of Viagra usage with aortic dissection, and reduced PDE5A expression was found in human aortic aneurysm tissues. Therefore, we aimed to investigate the effect of sildenafil on experimental abdominal aortic aneurysm (AAA), the most common form of aortic aneurysm in elderly men. Methods and Results AAA was induced in C57BL/6J male mice by periaortic elastase in combination with blocking elastin/collagen formation via 3‐aminopropionitrile fumarate salt for 35 days. PDE5A protein levels detected by immunostaining were significantly reduced in mouse AAA. Sildenafil application in drinking water significantly aggravated aortic wall dilation and elastin degradation with pre‐existing moderate AAA. The phosphorylation level of myosin light chain 2 at Ser19, a biochemical marker of SMC contraction, was significantly reduced by sildenafil in AAA. Proximity ligation assay further revealed that the interaction between cGMP and PKGI was significantly increased by sildenafil in AAA, suggesting an elevation of PKGI activation in AAA. Conclusions Sildenafil treatment aggravated the degradation of elastin fibers and progression of experimental AAA by dysregulating cGMP and contractile signaling in SMCs. Our findings may raise the caution of clinical usage of Viagra in aneurysmal patients.
Collapse
Affiliation(s)
- Chongyang Zhang
- School of Medicine and Dentistry Aab Cardiovascular Research InstituteUniversity of Rochester Rochester NY.,Department of Pharmacology and Physiology School of Medicine and Dentistry University of Rochester Rochester NY
| | - Amy Mohan
- School of Medicine and Dentistry Aab Cardiovascular Research InstituteUniversity of Rochester Rochester NY
| | - Hangchuan Shi
- Department of Clinical & Translational Research University of Rochester Medical Center Rochester NY.,Department of Public Health Sciences University of Rochester Medical Center Rochester NY
| | - Chen Yan
- School of Medicine and Dentistry Aab Cardiovascular Research InstituteUniversity of Rochester Rochester NY
| |
Collapse
|
16
|
Wagner BM, Robinson JW, Healy CL, Gauthier M, Dickey DM, Yee SP, Osborn JW, O’Connell TD, Potter LR. Guanylyl cyclase-A phosphorylation decreases cardiac hypertrophy and improves systolic function in male, but not female, mice. FASEB J 2022; 36:e22069. [PMID: 34859913 PMCID: PMC8826535 DOI: 10.1096/fj.202100600rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 01/03/2023]
Abstract
Atrial natriuretic peptide (NP) and BNP increase cGMP, which reduces blood pressure and cardiac hypertrophy by activating guanylyl cyclase (GC)-A, also known as NPR-A or Npr1. Although GC-A is highly phosphorylated, and dephosphorylation inactivates the enzyme, the significance of GC-A phosphorylation to heart structure and function remains unknown. To identify in vivo processes that are regulated by GC-A phosphorylation, we substituted glutamates for known phosphorylation sites to make GC-A8E/8E mice that express an enzyme that cannot be inactivated by dephosphorylation. GC-A activity, but not protein, was increased in heart and kidney membranes from GC-A8E/8E mice. Activities were threefold higher in female compared to male cardiac ventricles. Plasma cGMP and testosterone were elevated in male and female GC-A8E/8E mice, but aldosterone was only increased in mutant male mice. Plasma and urinary creatinine concentrations were decreased and increased, respectively, but blood pressure and heart rate were unchanged in male GC-A8E/8E mice. Heart weight to body weight ratios for GC-A8E/8E male, but not female, mice were 12% lower with a 14% reduction in cardiomyocyte cross-sectional area. Subcutaneous injection of fsANP, a long-lived ANP analog, increased plasma cGMP and decreased aldosterone in male GC-AWT/WT and GC-A8E/8E mice at 15 min, but only GC-A8E/8E mice had elevated levels of plasma cGMP and aldosterone at 60 min. fsANP reduced ventricular ERK1/2 phosphorylation to a greater extent and for a longer time in the male mutant compared to WT mice. Finally, ejection fractions were increased in male but not female hearts from GC-A8E/8E mice. We conclude that increased phosphorylation-dependent GC-A activity decreases cardiac ERK activity, which results in smaller male hearts with improved systolic function.
Collapse
Affiliation(s)
- Brandon M. Wagner
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Jerid W. Robinson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Chastity L. Healy
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Madeline Gauthier
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Deborah M. Dickey
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Siu-Pok Yee
- Department of Cell Biology at the University of Connecticut Health Center, Farmington, CT 06030 USA
| | - John W. Osborn
- Department of Surgery at the University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA,,Corresponding authors: Timothy D O’Connell , Lincoln R Potter
| | - Lincoln R. Potter
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA,,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Medical School, Minneapolis, MN 55455 USA,,Corresponding authors: Timothy D O’Connell , Lincoln R Potter
| |
Collapse
|
17
|
Längst N, Adler J, Schweigert O, Kleusberg F, Cruz Santos M, Knauer A, Sausbier M, Zeller T, Ruth P, Lukowski R. Cyclic GMP-Dependent Regulation of Vascular Tone and Blood Pressure Involves Cysteine-Rich LIM-Only Protein 4 (CRP4). Int J Mol Sci 2021; 22:9925. [PMID: 34576086 PMCID: PMC8466836 DOI: 10.3390/ijms22189925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/09/2021] [Accepted: 08/25/2021] [Indexed: 01/14/2023] Open
Abstract
The cysteine-rich LIM-only protein 4 (CRP4), a LIM-domain and zinc finger containing adapter protein, has been implicated as a downstream effector of the second messenger 3',5'-cyclic guanosine monophosphate (cGMP) pathway in multiple cell types, including vascular smooth muscle cells (VSMCs). VSMCs and nitric oxide (NO)-induced cGMP signaling through cGMP-dependent protein kinase type I (cGKI) play fundamental roles in the physiological regulation of vascular tone and arterial blood pressure (BP). However, it remains unclear whether the vasorelaxant actions attributed to the NO/cGMP axis require CRP4. This study uses mice with a targeted deletion of the CRP4 gene (CRP4 KO) to elucidate whether cGMP-elevating agents, which are well known for their vasorelaxant properties, affect vessel tone, and thus, BP through CRP4. Cinaciguat, a NO- and heme-independent activator of the NO-sensitive (soluble) guanylyl cyclase (NO-GC) and NO-releasing agents, relaxed both CRP4-proficient and -deficient aortic ring segments pre-contracted with prostaglandin F2α. However, the magnitude of relaxation was slightly, but significantly, increased in vessels lacking CRP4. Accordingly, CRP4 KO mice presented with hypotonia at baseline, as well as a greater drop in systolic BP in response to the acute administration of cinaciguat, sodium nitroprusside, and carbachol. Mechanistically, loss of CRP4 in VSMCs reduced the Ca2+-sensitivity of the contractile apparatus, possibly involving regulatory proteins, such as myosin phosphatase targeting subunit 1 (MYPT1) and the regulatory light chain of myosin (RLC). In conclusion, the present findings confirm that the adapter protein CRP4 interacts with the NO-GC/cGMP/cGKI pathway in the vasculature. CRP4 seems to be part of a negative feedback loop that eventually fine-tunes the NO-GC/cGMP axis in VSMCs to increase myofilament Ca2+ desensitization and thereby the maximal vasorelaxant effects attained by (selected) cGMP-elevating agents.
Collapse
Affiliation(s)
- Natalie Längst
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Julia Adler
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Olga Schweigert
- Cardiovascular Systems Medicine and Molecular Translation, University Center of Cardiovascular Science, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (O.S.); (T.Z.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Felicia Kleusberg
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Melanie Cruz Santos
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Amelie Knauer
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Matthias Sausbier
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Tanja Zeller
- Cardiovascular Systems Medicine and Molecular Translation, University Center of Cardiovascular Science, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (O.S.); (T.Z.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| |
Collapse
|
18
|
Hofmann F. The cGMP system: components and function. Biol Chem 2021; 401:447-469. [PMID: 31747372 DOI: 10.1515/hsz-2019-0386] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022]
Abstract
The cyclic guanosine monophosphate (cGMP) signaling system is one of the most prominent regulators of a variety of physiological and pathophysiological processes in many mammalian and non-mammalian tissues. Targeting this pathway by increasing cGMP levels has been a very successful approach in pharmacology as shown for nitrates, phosphodiesterase (PDE) inhibitors and stimulators of nitric oxide-guanylyl cyclase (NO-GC) and particulate GC (pGC). This is an introductory review to the cGMP signaling system intended to introduce those readers to this system, who do not work in this area. This article does not intend an in-depth review of this system. Signal transduction by cGMP is controlled by the generating enzymes GCs, the degrading enzymes PDEs and the cGMP-regulated enzymes cyclic nucleotide-gated ion channels, cGMP-dependent protein kinases and cGMP-regulated PDEs. Part A gives a very concise introduction to the components. Part B gives a very concise introduction to the functions modulated by cGMP. The article cites many recent reviews for those who want a deeper insight.
Collapse
Affiliation(s)
- Franz Hofmann
- Pharmakologisches Institut, Technische Universität München, Biedersteiner Str. 29, D-80802 München, Germany
| |
Collapse
|
19
|
DiNicolantonio JJ, McCarty MF, Barroso-Aranda J, Assanga S, Lujan LML, O'Keefe JH. A nutraceutical strategy for downregulating TGFβ signalling: prospects for prevention of fibrotic disorders, including post-COVID-19 pulmonary fibrosis. Open Heart 2021; 8:openhrt-2021-001663. [PMID: 33879509 PMCID: PMC8061562 DOI: 10.1136/openhrt-2021-001663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- James J DiNicolantonio
- Preventive Cardiology, Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | | | | | - Simon Assanga
- Department of Research and Postgraduate Studies in Food, University of Sonora, Sonora, Mexico
| | | | - James H O'Keefe
- University of Missouri-Kansas City, Saint Lukes Mid America Heart Institute, Kansas City, Missouri, USA
| |
Collapse
|
20
|
Escudero DS, Pérez NG, Díaz RG. Myocardial Impact of NHE1 Regulation by Sildenafil. Front Cardiovasc Med 2021; 8:617519. [PMID: 33693035 PMCID: PMC7937606 DOI: 10.3389/fcvm.2021.617519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/04/2021] [Indexed: 11/13/2022] Open
Abstract
The cardiac Na+/H+ exchanger (NHE1) is a membrane glycoprotein fundamental for proper cell functioning due its multiple housekeeping tasks, including regulation of intracellular pH, Na+ concentration, and cell volume. In the heart, hyperactivation of NHE1 has been linked to the development of different pathologies. Several studies in animal models that reproduce the deleterious effects of ischemia/reperfusion injury or cardiac hypertrophy have conclusively demonstrated that NHE1 inhibition provides cardioprotection. Unfortunately, NHE1 inhibitors failed to reproduce these effects in the clinical arena. The reasons for those discrepancies are not apparent yet. However, a reasonable clue to consider would be that drugs that completely abolish the exchanger activity, including that its essential housekeeping function may not be the best therapeutic approach. Therefore, interventions tending to specifically reduce its hyperactive state without affecting its basal activity emerge as a novel potential gold standard. In this regard, a promising goal seems to be the modulation of the phosphorylation state of the cytosolic tail of the exchanger. Recent own experiments demonstrated that Sildenafil, a phosphodiesterase 5A inhibitor drug that has been widely used for the treatment of erectile dysfunction is able to decrease NHE1 phosphorylation, and hence reduce its hyperactivity. In connection, growing evidence demonstrates cardioprotective properties of Sildenafil against different cardiac pathologies, with the distinctive characteristic of directly affecting cardiac tissue without altering blood pressure. This mini-review was aimed to focus on the regulation of NHE1 activity by Sildenafil. For this purpose, experimental data reporting Sildenafil effects in different animal models of heart disease will be discussed.
Collapse
Affiliation(s)
- Daiana S Escudero
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Néstor G Pérez
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Romina G Díaz
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Abstract
The 3',5'-cyclic guanosine monophosphate (cGMP)-dependent protein kinase type I (cGKI aka PKGI) is a major cardiac effector acting downstream of nitric oxide (NO)-sensitive soluble guanylyl cyclase and natriuretic peptides (NPs), which signal through transmembrane guanylyl cyclases. Consistent with the wide distribution of the cGMP-generating guanylyl cyclases, cGKI, which usually elicits its cellular effects by direct phosphorylation of its targets, is present in multiple cardiac cell types including cardiomyocytes (CMs). Although numerous targets of cGMP/cGKI in heart were identified in the past, neither their exact patho-/physiological functions nor cell-type specific roles are clear. Herein, we inform about the current knowledge on the signal transduction downstream of CM cGKI. We believe that better insights into the specific actions of cGMP and cGKI in these cells will help to guide future studies in the search for predictive biomarkers for the response to pharmacological cGMP pathway modulation. In addition, targets downstream of cGMP/cGKI may be exploited for refined and optimized diagnostic and therapeutic strategies in different types of heart disease and their causes. Importantly, key functions of these proteins and particularly sites of regulatory phosphorylation by cGKI should, at least in principle, remain intact, although upstream signaling through the second messenger cGMP is impaired or dysregulated in a stressed or diseased heart state.
Collapse
|
24
|
Angiotensin II-Induced Cardiovascular Fibrosis Is Attenuated by NO-Sensitive Guanylyl Cyclase1. Cells 2020; 9:cells9112436. [PMID: 33171621 PMCID: PMC7695185 DOI: 10.3390/cells9112436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/24/2022] Open
Abstract
In the NO/cGMP signaling cascade, relevant in the cardiovascular system, two NO-sensitive guanylyl cyclase (NO-GC) isoforms are responsible for NO-dependent cGMP generation. Here, the impact of the major NO-GC isoform, NO-GC1, on fibrosis development in the cardiovascular system was studied in NO-GC1-deficient mice treated with AngiotensinII (AngII), known to induce vascular and cardiac remodeling. Morphometric analysis of NO-GC1 KO’s aortae demonstrated an enhanced increase of perivascular area after AngII treatment accompanied by a higher aortic collagen1 mRNA content. Increased perivascular fibrosis also occurred in cardiac vessels of AngII-treated NO-GC1 KO mice. In line, AngII-induced interstitial fibrosis was 32% more pronounced in NO-GC1 KO than in WT myocardia associated with a higher cardiac Col1 and other fibrotic marker protein content. In sum, increased perivascular and cardiac interstitial fibrosis together with the enhanced collagen1 mRNA content in AngII-treated NO-GC1-deficient mice represent an exciting manifestation of antifibrotic properties of cGMP formed by NO-GC1, a finding with great pharmaco-therapeutic implications.
Collapse
|
25
|
Abstract
Cyclic GMP (cGMP) represents a classic intracellular second messenger molecule. Over the past 2 decades, important discoveries have identified that cGMP signaling becomes deranged in heart failure (HF) and that cGMP and its main kinase effector, protein kinase G, generally oppose the biological abnormalities contributing to HF, in experimental studies. These findings have influenced the design of clinical trials of cGMP-augmenting drugs in HF patients. At present, the trial results of cGMP-augmenting therapies in HF remain mixed. As detailed in this review, strong evidence now exists that protein kinase G opposes pathologic cardiac remodeling through regulation of diverse biological processes and myocardial substrates. Potential reasons for the failures of cGMP-augmenting drugs in HF may be related to biological mechanisms opposing cGMP or because of certain features of clinical trials, all of which are discussed.
Collapse
|
26
|
Abstract
Heart failure (HF) is a common consequence of several cardiovascular diseases and is understood as a vicious cycle of cardiac and hemodynamic decline. The current inventory of treatments either alleviates the pathophysiological features (eg, cardiac dysfunction, neurohumoral activation, and ventricular remodeling) and/or targets any underlying pathologies (eg, hypertension and myocardial infarction). Yet, since these do not provide a cure, the morbidity and mortality associated with HF remains high. Therefore, the disease constitutes an unmet medical need, and novel therapies are desperately needed. Cyclic guanosine-3',5'-monophosphate (cGMP), synthesized by nitric oxide (NO)- and natriuretic peptide (NP)-responsive guanylyl cyclase (GC) enzymes, exerts numerous protective effects on cardiac contractility, hypertrophy, fibrosis, and apoptosis. Impaired cGMP signaling, which can occur after GC deactivation and the upregulation of cyclic nucleotide-hydrolyzing phosphodiesterases (PDEs), promotes cardiac dysfunction. In this study, we review the role that NO/cGMP and NP/cGMP signaling plays in HF. After considering disease etiology, the physiological effects of cGMP in the heart are discussed. We then assess the evidence from preclinical models and patients that compromised cGMP signaling contributes to the HF phenotype. Finally, the potential of pharmacologically harnessing cardioprotective cGMP to rectify the present paucity of effective HF treatments is examined.
Collapse
|
27
|
Yang D, Liu HQ, Liu FY, Tang N, Guo Z, Ma SQ, An P, Wang MY, Wu HM, Yang Z, Fan D, Tang QZ. Critical roles of macrophages in pressure overload-induced cardiac remodeling. J Mol Med (Berl) 2020; 99:33-46. [PMID: 33130927 DOI: 10.1007/s00109-020-02002-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/07/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022]
Abstract
Macrophages are integral components of the mammalian heart that show extensive expansion in response to various internal or external stimuli. After the onset of sustained pressure overload (PO), the accumulation of cardiac macrophages through local macrophage proliferation and monocyte migration has profound effects on the transition to cardiac hypertrophy and remodeling. In this review, we describe the heterogeneity and diversity of cardiac macrophages and summarize the current understanding of the important roles of macrophages in PO-induced cardiac remodeling. In addition, the possible mechanisms involved in macrophage modulation are also described. Finally, considering the significant effects of cardiac macrophages, we highlight their emerging role as therapeutic targets for alleviating pathological cardiac remodeling after PO.
Collapse
Affiliation(s)
- Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Han-Qing Liu
- Department of Thyroid and Breast, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.
- Cardiovascular Research Institute, Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Contrasting Effects of Inhibition of Phosphodiesterase 3 and 5 on Cardiac Function and Interstitial Fibrosis in Rats With Isoproterenol-Induced Cardiac Dysfunction. J Cardiovasc Pharmacol 2020; 73:195-205. [PMID: 30839513 DOI: 10.1097/fjc.0000000000000652] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myocardial relaxation and stiffness are influenced by fibrillar collagen content. Cyclic nucleotide signaling regulators have been investigated targeting more effective modulation of collagen deposition during myocardial healing process. To assess the effects of phosphodiesterase type 3 and phosphodiesterase type 5 inhibitors on cardiac function and left ventricular myocardial fibrosis in catecholamine-induced myocardial injury, sildenafil and pimobendan were administered to male Wistar rats 24 hours after isoproterenol injection. Echocardiography and electrocardiogram were performed to assess kinetic and rhythm changes during 45 days of drug administration. At the end of study, type I and type III collagen were measured through immunohistochemistry analysis, and left ventricular pressure was assessed through invasive method. Echocardiography assessment showed increased relative wall thickness at 45 days in pimobendan group with significant diastolic dysfunction and increased collagen I deposition compared with nontreated positive group (3.03 ± 0.31 vs. 2.73 ± 0.28%, P < 0.05). Diastolic pressure correlated positively with type I collagen (r = 0.54, P < 0.05). Type III collagen analysis did not demonstrate difference among the groups. Sildenafil administration attenuated type I collagen deposition (2.15 ± 0.51 vs. positive group, P < 0.05) and suggested to be related to arrhythmic events. Arrhythmic events were not related to the quantity of fibrillar collagen deposition. Although negative modulation of collagen synthesis through cyclic nucleotides signaling have shown promising results, in this study, pimobendan postconditioning resulted in increased collagen type I formation and severe diastolic dysfunction while sildenafil postconditioning reduced collagen type I deposition and attenuated diastolic dysfunction.
Collapse
|
31
|
The Role of Cyclic AMP Signaling in Cardiac Fibrosis. Cells 2019; 9:cells9010069. [PMID: 31888098 PMCID: PMC7016856 DOI: 10.3390/cells9010069] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/18/2022] Open
Abstract
Myocardial stress and injury invariably promote remodeling of the cardiac tissue, which is associated with cardiomyocyte death and development of fibrosis. The fibrotic process is initially triggered by the differentiation of resident cardiac fibroblasts into myofibroblasts. These activated fibroblasts display increased proliferative capacity and secrete large amounts of extracellular matrix. Uncontrolled myofibroblast activation can thus promote heart stiffness, cardiac dysfunction, arrhythmias, and progression to heart failure. Despite the well-established role of myofibroblasts in mediating cardiac disease, our current knowledge on how signaling pathways promoting fibrosis are regulated and coordinated in this cell type is largely incomplete. In this respect, cyclic adenosine monophosphate (cAMP) signaling acts as a major modulator of fibrotic responses activated in fibroblasts of injured or stressed hearts. In particular, accumulating evidence now suggests that upstream cAMP modulators including G protein-coupled receptors, adenylyl cyclases (ACs), and phosphodiesterases (PDEs); downstream cAMP effectors such as protein kinase A (PKA) and the guanine nucleotide exchange factor Epac; and cAMP signaling organizers such as A-kinase anchoring proteins (AKAPs) modulate a variety of fundamental cellular processes involved in myocardial fibrosis including myofibroblast differentiation, proliferation, collagen secretion, and invasiveness. The current review will discuss recent advances highlighting the role of cAMP and AKAP-mediated signaling in regulating pathophysiological responses controlling cardiac fibrosis.
Collapse
|
32
|
Li Y, Ruan X, Xu X, Li C, Qiang T, Zhou H, Gao J, Wang X. Shengmai Injection Suppresses Angiotensin II-Induced Cardiomyocyte Hypertrophy and Apoptosis via Activation of the AMPK Signaling Pathway Through Energy-Dependent Mechanisms. Front Pharmacol 2019; 10:1095. [PMID: 31616303 PMCID: PMC6764192 DOI: 10.3389/fphar.2019.01095] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/26/2019] [Indexed: 01/05/2023] Open
Abstract
Shengmai injection (SMI), a traditional Chinese herbal medicine extracted from Panax ginseng C.A. Mey., Ophiopogon japonicus (Thunb.) Ker Gawl., and Schisandra chinensis (Turcz.) Baill., has been used to treat acute and chronic heart failure. This study aimed to further clarify the effects of SMI on energy metabolism. SMI could improve cell-survival rate and also reduce myocardial cell hypertrophy and apoptosis. Mitochondria are important sites of cellular energy metabolism, and SMI protects mitochondrial function which was evaluated by mitochondrial ultrastructure, mitochondrial respiratory control ratio (RCR), and mitochondrial membrane potential (ΔΨm) in this study. The expression levels of adenosine triphosphate (ATP), adenosine diphosphate (ADP), and phosphocreatine (PCr) increased. The expression levels of free fatty acid oxidation [carnitine palmitoyltransferase-1 (CPT-1)], glucose oxidation [glucose transporter-4 (GLUT-4)], and mitochondrial biogenesis-related genes (peroxisome proliferator-activated receptor-γ coactivator-1α [PGC-1α]) were upregulated after SMI treatment. AMP-activated protein kinase (AMPK) is an important signaling pathway regulating energy metabolism and also can regulate the above-mentioned indicators. In the present study, SMI was found to promote phosphorylation of AMPK. However, the effects of SMI on fatty acid, glucose oxidation, mitochondrial biogenesis, as well as inhibiting apoptosis of hypertrophic cardiomyocytes were partly blocked by AMPK inhibitor–compound C. Moreover, decreased myocardial hypertrophy and apoptosis treated by SMI were inhibited by AMPK knockdown with shAMPK to a certain degree and AMPK knockdown almost abolished the SMI-induced increase in the expression of GLUT-4, CPT-1, and PGC-1α. These data suggest that SMI suppressed Ang II–induced cardiomyocyte hypertrophy and apoptosis via activation of the AMPK signaling pathway through energy-dependent mechanisms.
Collapse
Affiliation(s)
- Yiping Li
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaofen Ruan
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaowen Xu
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cha Li
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tingting Qiang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junjie Gao
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaolong Wang
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
33
|
MicroRNA-19a/b-3p protect the heart from hypertension-induced pathological cardiac hypertrophy through PDE5A. J Hypertens 2019; 36:1847-1857. [PMID: 29664809 PMCID: PMC6080882 DOI: 10.1097/hjh.0000000000001769] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AIM PDE5A is a leading factor contributing to cGMP signaling and cardiac hypertrophy. However, microRNA-mediated posttranscriptional regulation of PDE5A has not been reported. The aim of this study is to screen the microRNAs that are able to regulate PDE5A and explore the function of the microRNAs in cardiac hypertrophy and remodeling. METHODS AND RESULTS Although miR-19a/b-3p (microRNA-19a-3p and microRNA-19b-3p) have been reported to be differentially expressed during cardiac hypertrophy, the direct targets and the functions of this microRNA family for regulation of cardiac hypertrophy have not yet been investigated. The present study identified some direct targets and the underlying functions of miR-19a/b-3p by using bioinformatics tools and gene manipulations within mouse neonatal cardiomyocytes. Transfection of miR-19a/b-3p down-regulated endogenous expressions of PDE5A at both mRNA and protein levels with real-time PCR and western blot. Luciferase reporter assays showed that PDE5A was a direct target of miR-19a/b-3p. In mouse models of cardiac hypertrophy, we found that miR-19a/b-3p was expressed in cardiomyocytes and that its expression was reduced in pressure overload-induced hypertrophic hearts. miR-19a/b-3p transgenic mice prevented the progress of cardiac hypertrophy and cardiac remodeling in response to angiotensin II infusion with echocardiographic assessment and pressure-volume relation analysis. CONCLUSION Our study elucidates that PDE5A is a novel direct target of miR-19a/b-3p, and demonstrates that antihypertrophic roles of the miR-19a/b-3p family in Ang II-induced hypertrophy and cardiac remodeling, suggests that endogenous miR-19a/b-3p might have clinical potential to suppress cardiac hypertrophy and heart failure.
Collapse
|
34
|
Li EA, Xi W, Han YS, Brozovich FV. Phosphodiesterase expression in the normal and failing heart. Arch Biochem Biophys 2018; 662:160-168. [PMID: 30550727 DOI: 10.1016/j.abb.2018.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 01/09/2023]
Abstract
The number of patients with heart failure with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF) is increasing, and for HFpEF, no therapies have clinical benefit. It has been hypothesized that PKG attenuates pathological remodelling, and increasing cGMP would be beneficial for patients with HF. However, neither the RELAX nor NEAT-HFpEF trial showed benefit. But there is still enthusiasm for increasing cGMP in patients with HF, which highlight the need to determine the expression of PDEs in cardiac muscle. This study used immunoblotting to examine the expression of the PDEs that have been suggested to be targets for therapy of HF in both canines (normal and HFpEF) and humans (normal and HFrEF). Our results demonstrate PDE1C and PDE3A are expressed in cardiac muscle, but we could not detect the expression of PDE2A, PDE5A, PDE7A and PDE9A in cardiac tissue lysates from either normal or failing hearts. Thus, one should not expect a clinical benefit for a therapy targeting these PDEs in heart failure, which highlights the importance of rigorous demonstration of the target of therapy prior to undertaking a clinical trial.
Collapse
Affiliation(s)
- Edwin A Li
- Department of Cardiovascular Disease, Mayo Medical School, Rochester, MN, 55905, USA
| | - Wang Xi
- Biomedical Engineering and Physiology, Mayo Medical School, Rochester, MN, 55905, USA
| | - Young Soo Han
- Biomedical Engineering and Physiology, Mayo Medical School, Rochester, MN, 55905, USA
| | - Frank V Brozovich
- Department of Cardiovascular Disease, Mayo Medical School, Rochester, MN, 55905, USA.
| |
Collapse
|
35
|
Angiotensin II requires an intact cardiac thyrotropin-releasing hormone (TRH) system to induce cardiac hypertrophy in mouse. J Mol Cell Cardiol 2018; 124:1-11. [DOI: 10.1016/j.yjmcc.2018.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/13/2018] [Accepted: 09/25/2018] [Indexed: 11/18/2022]
|
36
|
Cuello F, Eaton P. Cysteine-Based Redox Sensing and Its Role in Signaling by Cyclic Nucleotide-Dependent Kinases in the Cardiovascular System. Annu Rev Physiol 2018; 81:63-87. [PMID: 30216743 DOI: 10.1146/annurev-physiol-020518-114417] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Oxidant molecules are produced in biological systems and historically have been considered causal mediators of damage and disease. While oxidants may contribute to the pathogenesis of disease, evidence continues to emerge that shows these species also play important regulatory roles in health. A major mechanism of oxidant sensing and signaling involves their reaction with reactive cysteine thiols within proteins, inducing oxidative posttranslational modifications that can couple to altered function to enable homeostatic regulation. Protein kinase A and protein kinase G are regulated by oxidants in this way, and this review focuses on our molecular-level understanding of these events and their role in regulating cardiovascular physiology during health and disease.
Collapse
Affiliation(s)
- Friederike Cuello
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Philip Eaton
- King's College London, School of Cardiovascular Medicine and Sciences, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom;
| |
Collapse
|
37
|
Lyle MA, Brozovich FV. HFpEF, a Disease of the Vasculature: A Closer Look at the Other Half. Mayo Clin Proc 2018; 93:1305-1314. [PMID: 30064827 DOI: 10.1016/j.mayocp.2018.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/12/2018] [Accepted: 05/04/2018] [Indexed: 12/31/2022]
Abstract
Patients with heart failure are commonly divided into those with reduced ejection fraction (EF<40%) and those with preserved ejection fraction (HFpEF; EF>50%). For heart failure with reduced EF, a number of therapies have been found to improve patient morbidity and mortality, and treatment is guideline based. However for patients with HFpEF, no treatment has been found to have clinical benefit. To objectively assess treatments for HFpEF, a comprehensive PubMed literature search was performed using the terms HFpEF, heart failure, smooth muscle, myosin, myosin phosphatase, and PKG (up to December 31, 2017), with an unbiased focus on pathophysiology, cell signaling, and therapy. This review provides evidence that could explain the lack of clinical benefit in treating patients with HFpEF with sildenafil and long-acting nitrates. Furthermore, the review highlights the vascular abnormalities present in patients with HFpEF, and these abnormalities of the vasculature could potentially contribute to the pathophysiology of HFpEF. Thus, focusing on HFpEF as a vascular disease could result in the development of novel and effective treatment paradigms.
Collapse
Affiliation(s)
- Melissa A Lyle
- Department of Cadiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Frank V Brozovich
- Department of Cadiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN.
| |
Collapse
|
38
|
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
|
39
|
Hofmann F. A concise discussion of the regulatory role of cGMP kinase I in cardiac physiology and pathology. Basic Res Cardiol 2018; 113:31. [PMID: 29934662 DOI: 10.1007/s00395-018-0690-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/18/2018] [Accepted: 06/13/2018] [Indexed: 12/25/2022]
Abstract
The underlying cause of cardiac hypertrophy, fibrosis, and heart failure has been investigated in great detail using different mouse models. These studies indicated that cGMP and cGMP-dependent protein kinase type I (cGKI) may ameliorate these negative phenotypes in the adult heart. Recently, evidence has been published that cardiac mitochondrial BKCa channels are a target for cGKI and that activation of mitoBKCa channels may cause some of the positive effects of conditioning in ischemia/reperfusion injury. It will be pointed out that most studies could not present convincing evidence that it is the cGMP level and the activity cGKI in specific cardiac cells that reduces hypertrophy or heart failure. However, anti-fibrotic compounds stimulating nitric oxide-sensitive guanylyl cyclase may be an upcoming therapy for abnormal cardiac remodeling.
Collapse
Affiliation(s)
- Franz Hofmann
- Institut für Pharmakologie und Toxikologie, TU München, Biedersteiner Str. 29, 80802, Munich, Germany.
| |
Collapse
|
40
|
Roles of PDE1 in Pathological Cardiac Remodeling and Dysfunction. J Cardiovasc Dev Dis 2018; 5:jcdd5020022. [PMID: 29690591 PMCID: PMC6023290 DOI: 10.3390/jcdd5020022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/05/2018] [Accepted: 04/20/2018] [Indexed: 12/16/2022] Open
Abstract
Pathological cardiac hypertrophy and dysfunction is a response to various stress stimuli and can result in reduced cardiac output and heart failure. Cyclic nucleotide signaling regulates several cardiac functions including contractility, remodeling, and fibrosis. Cyclic nucleotide phosphodiesterases (PDEs), by catalyzing the hydrolysis of cyclic nucleotides, are critical in the homeostasis of intracellular cyclic nucleotide signaling and hold great therapeutic potential as drug targets. Recent studies have revealed that the inhibition of the PDE family member PDE1 plays a protective role in pathological cardiac remodeling and dysfunction by the modulation of distinct cyclic nucleotide signaling pathways. This review summarizes recent key findings regarding the roles of PDE1 in the cardiac system that can lead to a better understanding of its therapeutic potential.
Collapse
|
41
|
Flores-Costa R, Alcaraz-Quiles J, Titos E, López-Vicario C, Casulleras M, Duran-Güell M, Rius B, Diaz A, Hall K, Shea C, Sarno R, Currie M, Masferrer JL, Clària J. The soluble guanylate cyclase stimulator IW-1973 prevents inflammation and fibrosis in experimental non-alcoholic steatohepatitis. Br J Pharmacol 2018; 175:953-967. [PMID: 29281143 PMCID: PMC5825296 DOI: 10.1111/bph.14137] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Non-alcoholic steatohepatitis (NASH) is the hepatic manifestation of metabolic syndrome and is characterized by steatosis, inflammation and fibrosis. Soluble guanylate cyclase (sGC) stimulation reduces inflammation and fibrosis in experimental models of lung, kidney and heart disease. Here, we tested whether sGC stimulation is also effective in experimental NASH. EXPERIMENTAL APPROACH NASH was induced in mice by feeding a choline-deficient, l-amino acid-defined, high-fat diet. These mice received either placebo or the sGC stimulator IW-1973 at two different doses (1 and 3 mg·kg-1 ·day-1 ) for 9 weeks. IW-1973 was also tested in high-fat diet (HFD)-induced obese mice. Steatosis, inflammation and fibrosis were assessed by Oil Red O, haematoxylin-eosin, Masson's trichrome, Sirius Red, F4/80 and α-smooth muscle actin staining. mRNA expression was assessed by quantitative PCR. Levels of IW-1973, cytokines and cGMP were determined by LC-MS/MS, Luminex and enzyme immunoassay respectively. KEY RESULTS Mice with NASH showed reduced cGMP levels and sGC expression, increased steatosis, inflammation, fibrosis, TNF-α and MCP-1 levels and up-regulated collagen types I α1 and α2, MMP2, TGF-β1 and tissue metallopeptidase inhibitor 1 expression. IW-1973 restored hepatic cGMP levels and sGC expression resulting in a dose-dependent reduction of hepatic inflammation and fibrosis. IW-1973 levels were ≈40-fold higher in liver tissue than in plasma. IW-1973 also reduced hepatic steatosis and adipocyte hypertrophy secondary to enhanced autophagy in HFD-induced obese mice. CONCLUSIONS AND IMPLICATIONS Our data indicate that sGC stimulation prevents hepatic steatosis, inflammation and fibrosis in experimental NASH. These findings warrant further evaluation of IW-1973 in the clinical setting.
Collapse
Affiliation(s)
- Roger Flores-Costa
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - José Alcaraz-Quiles
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Esther Titos
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona, Spain.,CIBERehd, Barcelona, Spain
| | - Cristina López-Vicario
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona, Spain.,CIBERehd, Barcelona, Spain
| | - Mireia Casulleras
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Marta Duran-Güell
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Bibiana Rius
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Alba Diaz
- Department of Pathology, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | | | | | - Renee Sarno
- Ironwood Pharmaceuticals Inc., Cambridge, MA, USA
| | - Mark Currie
- Ironwood Pharmaceuticals Inc., Cambridge, MA, USA
| | | | - Joan Clària
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, Barcelona, Spain.,CIBERehd, Barcelona, Spain.,Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain.,European Foundation for the Study of Chronic Liver Failure (EF-CLIF), Barcelona, Spain
| |
Collapse
|
42
|
Martín‐Sánchez P, Luengo A, Griera M, Orea MJ, López‐Olañeta M, Chiloeches A, Lara‐Pezzi E, Frutos S, Rodríguez–Puyol M, Calleros L, Rodríguez–Puyol D. H‐
ras
deletion protects against angiotensin II–induced arterial hypertension and cardiac remodeling through protein kinase G‐Iβ pathway activation. FASEB J 2018; 32:920-934. [DOI: 10.1096/fj.201700134rrrr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Paloma Martín‐Sánchez
- Department of Systems BiologyUniversidad de AlcaláMadridSpain
- Instituto Reina Sofía de Investigación en Neurológica (IRSIN)MadridSpain
- Red de Investigación Renal (REDinREN)Instituto de Salud Carlos IIIMadridSpain
| | - Alicia Luengo
- Department of Systems BiologyUniversidad de AlcaláMadridSpain
- Instituto Reina Sofía de Investigación en Neurológica (IRSIN)MadridSpain
- Red de Investigación Renal (REDinREN)Instituto de Salud Carlos IIIMadridSpain
| | - Mercedes Griera
- Department of Systems BiologyUniversidad de AlcaláMadridSpain
- Instituto Reina Sofía de Investigación en Neurológica (IRSIN)MadridSpain
- Red de Investigación Renal (REDinREN)Instituto de Salud Carlos IIIMadridSpain
| | | | - Marina López‐Olañeta
- Myocardial Pathophysiology AreaCentro Nacional de Investigaciones CardiovascularesMadridSpain
| | | | - Enrique Lara‐Pezzi
- Myocardial Pathophysiology AreaCentro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Sergio Frutos
- Department of Systems BiologyUniversidad de AlcaláMadridSpain
- Instituto Reina Sofía de Investigación en Neurológica (IRSIN)MadridSpain
- Red de Investigación Renal (REDinREN)Instituto de Salud Carlos IIIMadridSpain
| | - Manuel Rodríguez–Puyol
- Department of Systems BiologyUniversidad de AlcaláMadridSpain
- Instituto Reina Sofía de Investigación en Neurológica (IRSIN)MadridSpain
- Red de Investigación Renal (REDinREN)Instituto de Salud Carlos IIIMadridSpain
| | - Laura Calleros
- Department of Systems BiologyUniversidad de AlcaláMadridSpain
- Instituto Reina Sofía de Investigación en Neurológica (IRSIN)MadridSpain
- Red de Investigación Renal (REDinREN)Instituto de Salud Carlos IIIMadridSpain
| | - Diego Rodríguez–Puyol
- Department of MedicineUniversidad de AlcaláMadridSpain
- Instituto Reina Sofía de Investigación en Neurológica (IRSIN)MadridSpain
- Red de Investigación Renal (REDinREN)Instituto de Salud Carlos IIIMadridSpain
- Nephrology SectionResearch Unit FoundationHospital Universitario Príncipe de AsturiasAlcalá de HenaresMadridSpain
| |
Collapse
|
43
|
Crassous PA, Shu P, Huang C, Gordan R, Brouckaert P, Lampe PD, Xie LH, Beuve A. Newly Identified NO-Sensor Guanylyl Cyclase/Connexin 43 Association Is Involved in Cardiac Electrical Function. J Am Heart Assoc 2017; 6:e006397. [PMID: 29269353 PMCID: PMC5778997 DOI: 10.1161/jaha.117.006397] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Guanylyl cyclase, a heme-containing α1β1 heterodimer (GC1), produces cGMP in response to Nitric oxide (NO) stimulation. The NO-GC1-cGMP pathway negatively regulates cardiomyocyte contractility and protects against cardiac hypertrophy-related remodeling. We recently reported that the β1 subunit of GC1 is detected at the intercalated disc with connexin 43 (Cx43). Cx43 forms gap junctions (GJs) at the intercalated disc that are responsible for electrical propagation. We sought to determine whether there is a functional association between GC1 and Cx43 and its role in cardiac homeostasis. METHODS AND RESULTS GC1 and Cx43 immunostaining at the intercalated disc and coimmunoprecipitation from membrane fraction indicate that GC1 and Cx43 are associated. Mice lacking the α subunit of GC1 (GCα1 knockout mice) displayed a significant decrease in GJ function (dye-spread assay) and Cx43 membrane lateralization. In a cardiac-hypertrophic model, angiotensin II treatment disrupted the GC1-Cx43 association and induced significant Cx43 membrane lateralization, which was exacerbated in GCα1 knockout mice. Cx43 lateralization correlated with decreased Cx43-containing GJs at the intercalated disc, predictors of electrical dysfunction. Accordingly, an ECG revealed that angiotensin II-treated GCα1 knockout mice had impaired ventricular electrical propagation. The phosphorylation level of Cx43 at serine 365, a protein-kinase A upregulated site involved in trafficking/assembly of GJs, was decreased in these models. CONCLUSIONS GC1 modulates ventricular Cx43 location, hence GJ function, and partially protects from electrical dysfunction in an angiotensin II hypertrophy model. Disruption of the NO-cGMP pathway is associated with cardiac electrical disturbance and abnormal Cx43 phosphorylation. This previously unknown NO/Cx43 signaling could be a protective mechanism against stress-induced arrhythmia.
Collapse
Affiliation(s)
- Pierre-Antoine Crassous
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, NJ
| | - Ping Shu
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, NJ
| | - Can Huang
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, NJ
| | - Richard Gordan
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School-Rutgers, Newark, NJ
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Paul D Lampe
- Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School-Rutgers, Newark, NJ
| | - Annie Beuve
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, NJ
| |
Collapse
|
44
|
Wang PX, Li ZM, Cai SD, Li JY, He P, Huang Y, Feng GS, Luo HB, Chen SR, Liu PQ. C33(S), a novel PDE9A inhibitor, protects against rat cardiac hypertrophy through upregulating cGMP signaling. Acta Pharmacol Sin 2017. [PMID: 28649129 DOI: 10.1038/aps.2017.38] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphodiesterase-9A (PDE9A) expression is upregulated during cardiac hypertrophy and heart failure. Accumulating evidence suggests that PDE9A might be a promising therapeutic target for heart diseases. The present study sought to investigate the effects and underlying mechanisms of C33(S), a novel selective PDE9A inhibitor, on cardiac hypertrophy in vitro and in vivo. Treatment of neonatal rat cardiomyocytes (NRCMs) with PE (100 μmol/L) or ISO (1 μmol/L) induced cardiac hypertrophy characterized by significantly increased cell surface areas and increased expression of fetal genes (ANF and BNP). Furthermore, PE or ISO significantly increased the expression of PDE9A in the cells; whereas knockdown of PDE9A significantly alleviated PE-induced hypertrophic responses. Moreover, pretreatment with PDE9A inhibitor C33(S) (50 and 500 nmol/L) or PF-7943 (2 μmol/L) also alleviated the cardiac hypertrophic responses in PE-treated NRCMs. Abdominal aortic constriction (AAC)-induced cardiac hypertrophy and ISO-induced heart failure were established in SD rats. In ISO-treated rats, oral administration of C33(S) (9, 3, and 1 mg·kg-1·d-1, for 3 consecutive weeks) significantly increased fractional shortening (43.55%±3.98%, 54.79%±1.95%, 43.98%±7.96% vs 32.18%±6.28%), ejection fraction (72.97%±4.64%, 84.29%±1.56%, 73.41%±9.37% vs 49.17%±4.20%) and cardiac output (60.01±9.11, 69.40±11.63, 58.08±8.47 mL/min vs 48.97±2.11 mL/min) but decreased the left ventricular internal diameter, suggesting that the transition to heart failure was postponed by C33(S). We further revealed that C33(S) significantly elevated intracellular cGMP levels, phosphorylation of phospholamban (PLB) and expression of SERCA2a in PE-treated NRCMs in vitro and in ISO-induced heart failure model in vivo. Our results demonstrate that C33(S) effectively protects against cardiac hypertrophy and postpones the transition to heart failure, suggesting that it is a promising agent in the treatment of cardiac diseases.
Collapse
|
45
|
Poomvanicha M, Matthes J, Domes K, Patrucco E, Angermeier E, Laugwitz KL, Schneider T, Hofmann F. Beta-adrenergic regulation of the heart expressing the Ser1700A/Thr1704A mutated Cav1.2 channel. J Mol Cell Cardiol 2017; 111:10-16. [PMID: 28778765 DOI: 10.1016/j.yjmcc.2017.07.119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/25/2017] [Accepted: 07/28/2017] [Indexed: 01/07/2023]
Abstract
Beta-adrenergic stimulation of the heart increases ICa. PKA dependent phosphorylation of several amino acids (among them Ser 1700 and Thr 1704 in the carboxy-terminus of the Cav1.2 α1c subunit) has been implicated as decisive for the β-adrenergic up-regulation of cardiac ICa. Mutation of Ser 1700 and Thr 1704 to alanine results in the Cav1.2PKA_P2-/- mice. Cav1.2PKA_P2-/- mice display reduced cardiac L-type current. Fractional shortening and ejection fraction in the intact animal and ICa in isolated cardiomyocytes (CM) are stimulated by isoproterenol. Cardiac specific expression of the mutated Cav1.2PKA_P2-/- gene reduces Cav1.2 α1c protein concentration, ICa, and the β-adrenergic stimulation of L-type ICa in CMs. Single channels were not detected on the CM surface of the cCav1.2PKA_P2-/- hearts. This outcome supports the notion that S1700/1704 is essential for expression of the Cav1.2 channel and that isoproterenol stimulates ICa in Cav1.2PKA_P2-/- CMs.
Collapse
Affiliation(s)
- Montatip Poomvanicha
- Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany
| | - Jan Matthes
- Institut für Pharmakologie und Toxikologie, University Cologne, Germany
| | - Katrin Domes
- Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany
| | - Enrico Patrucco
- Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany
| | - Elisabeth Angermeier
- Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany
| | - Karl-Ludwig Laugwitz
- I. Medizinische Klinik und Poliklinik (Kardiologie, Angiologie & Pneumologie), Klinikum rechts der Isar-Technische Universität München, Ismaninger Straße 22, 81675 München, Germany
| | - Toni Schneider
- Institut für Neurophysiologie, University Cologne, Germany
| | - Franz Hofmann
- Institut für Pharmakologie und Toxikologie, Technische Universität München, Germany.
| |
Collapse
|
46
|
Straubinger J, Boldt K, Kuret A, Deng L, Krattenmacher D, Bork N, Desch M, Feil R, Feil S, Nemer M, Ueffing M, Ruth P, Just S, Lukowski R. Amplified pathogenic actions of angiotensin II in cysteine-rich LIM-only protein 4-negative mouse hearts. FASEB J 2017; 31:1620-1638. [PMID: 28138039 DOI: 10.1096/fj.201601186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022]
Abstract
LIM domain proteins have been identified as essential modulators of cardiac biology and pathology; however, it is unclear which role the cysteine-rich LIM-only protein (CRP)4 plays in these processes. In studying CRP4 mutant mice, we found that their hearts developed normally, but lack of CRP4 exaggerated multiple parameters of the cardiac stress response to the neurohormone angiotensin II (Ang II). Aiming to dissect the molecular details, we found a link between CRP4 and the cardioprotective cGMP pathway, as well as a multiprotein complex comprising well-known hypertrophy-associated factors. Significant enrichment of the cysteine-rich intestinal protein (CRIP)1 in murine hearts lacking CRP4, as well as severe cardiac defects and premature death of CRIP1 and CRP4 morphant zebrafish embryos, further support the notion that depleting CRP4 is incompatible with a proper cardiac development and function. Together, amplified Ang II signaling identified CRP4 as a novel antiremodeling factor regulated, at least to some extent, by cardiac cGMP.-Straubinger, J., Boldt, K., Kuret, A., Deng, L., Krattenmacher, D., Bork, N., Desch, M., Feil, R., Feil, S., Nemer, M., Ueffing, M., Ruth, P., Just, S., Lukowski, R. Amplified pathogenic actions of angiotensin II in cysteine-rich LIM-only protein 4 negative mouse hearts.
Collapse
Affiliation(s)
- Julia Straubinger
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Karsten Boldt
- Institute for Ophthalmic Research, Molecular Biology of Retinal Degenerations and Medical Proteome Center, University of Tübingen, Tübingen, Germany
| | - Anna Kuret
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lisa Deng
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Diana Krattenmacher
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Nadja Bork
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Matthias Desch
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany; and
| | - Susanne Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany; and
| | - Mona Nemer
- Laboratory of Cardiac Development and Differentiation, Department of Biochemistry, Immunology, and Microbiology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marius Ueffing
- Institute for Ophthalmic Research, Molecular Biology of Retinal Degenerations and Medical Proteome Center, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany;
| |
Collapse
|
47
|
PDE1C deficiency antagonizes pathological cardiac remodeling and dysfunction. Proc Natl Acad Sci U S A 2016; 113:E7116-E7125. [PMID: 27791092 DOI: 10.1073/pnas.1607728113] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cyclic nucleotide phosphodiesterase 1C (PDE1C) represents a major phosphodiesterase activity in human myocardium, but its function in the heart remains unknown. Using genetic and pharmacological approaches, we studied the expression, regulation, function, and underlying mechanisms of PDE1C in the pathogenesis of cardiac remodeling and dysfunction. PDE1C expression is up-regulated in mouse and human failing hearts and is highly expressed in cardiac myocytes but not in fibroblasts. In adult mouse cardiac myocytes, PDE1C deficiency or inhibition attenuated myocyte death and apoptosis, which was largely dependent on cyclic AMP/PKA and PI3K/AKT signaling. PDE1C deficiency also attenuated cardiac myocyte hypertrophy in a PKA-dependent manner. Conditioned medium taken from PDE1C-deficient cardiac myocytes attenuated TGF-β-stimulated cardiac fibroblast activation through a mechanism involving the crosstalk between cardiac myocytes and fibroblasts. In vivo, cardiac remodeling and dysfunction induced by transverse aortic constriction, including myocardial hypertrophy, apoptosis, cardiac fibrosis, and loss of contractile function, were significantly attenuated in PDE1C-knockout mice relative to wild-type mice. These results indicate that PDE1C activation plays a causative role in pathological cardiac remodeling and dysfunction. Given the continued development of highly specific PDE1 inhibitors and the high expression level of PDE1C in the human heart, our findings could have considerable therapeutic significance.
Collapse
|
48
|
Liu X, Chen Y, McCoy CW, Zhao T, Quarles DL, Pi M, Bhattacharya SK, King G, Sun Y. Differential Regulatory Role of Soluble Klothos on Cardiac Fibrogenesis in Hypertension. Am J Hypertens 2016; 29:1140-7. [PMID: 27543985 DOI: 10.1093/ajh/hpw062] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/23/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Soluble Klotho functions as an endocrine factor that plays important roles in a variety of pathophysiological processes. Soluble Klotho contains 130 KDa and 65 KDa isoforms. However, their distinct individual functional heterogeneity remains uncertain. Herein, we investigated the regulatory role of two soluble Klothos on cardiac fibrogenic responses. METHODS AND RESULTS The effect of soluble Klothos on myofibroblast differentiation, proliferation, and collagen synthesis/degradation were examined in cultured mouse cardiac myofibroblasts. The role of 130 KDa Klotho on fibrosis in hypertensive heart disease were examined in wild type (WT) and Klotho transgenic (Tg/+) mice receiving chronic angiotensin (Ang)II infusion. Our in vitro studies revealed that addition of 130 KDa soluble Klotho isoform increased collagen synthesis in a dose dependent manner. Furthermore, 130 KDa Klotho significantly stimulated myofibroblast differentiation, proliferation, and ERK phosphorylation, which were abolished by fibroblast growth factor (FGF) receptor antagonist (SU5402). In contrast, 65 KDa soluble Klotho treatment significantly suppressed myofibroblast proliferation and collagen synthesis. In vivo study further demonstrated that chronic AngII infusion lead to cardiac fibrosis in both WT and Tg/+ mice. However, cardiac collagen, TGF-β1, TIMP-2, and α-smooth muscle actin (SMA) levels were markedly upregulated in Tg/+ mice compared to WT cohort. CONCLUSION Taken together, these findings implicate that 130 KDa soluble Klotho plays a stimulatory role in cardiac myofibroblast growth and activity through FGF pathway, whereas 65 KDa soluble Klotho exerts an anti-fibrotic effect in cardiac myofibroblasts. Thus, two distinct isoforms of soluble Klotho appear to play the counter-regulatory roles in cardiac fibrogenic responses.
Collapse
Affiliation(s)
- Xue Liu
- Department of Pharmacology, Harbin Medical University, Harbin, China
| | - Yuanjian Chen
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Cody W McCoy
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Tieqiang Zhao
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Darryl L Quarles
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Min Pi
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Syamal K Bhattacharya
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Gwendalyn King
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL
| | - Yao Sun
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN;
| |
Collapse
|
49
|
Lother A, Hein L. Pharmacology of heart failure: From basic science to novel therapies. Pharmacol Ther 2016; 166:136-49. [PMID: 27456554 DOI: 10.1016/j.pharmthera.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/08/2016] [Indexed: 01/10/2023]
Abstract
Chronic heart failure is one of the leading causes for hospitalization in the United States and Europe, and is accompanied by high mortality. Current pharmacological therapy of chronic heart failure with reduced ejection fraction is largely based on compounds that inhibit the detrimental action of the adrenergic and the renin-angiotensin-aldosterone systems on the heart. More than one decade after spironolactone, two novel therapeutic principles have been added to the very recently released guidelines on heart failure therapy: the HCN-channel inhibitor ivabradine and the combined angiotensin and neprilysin inhibitor valsartan/sacubitril. New compounds that are in phase II or III clinical evaluation include novel non-steroidal mineralocorticoid receptor antagonists, guanylate cyclase activators or myosine activators. A variety of novel candidate targets have been identified and the availability of gene transfer has just begun to accelerate translation from basic science to clinical application. This review provides an overview of current pharmacology and pharmacotherapy in chronic heart failure at three stages: the updated clinical guidelines of the American Heart Association and the European Society of Cardiology, new drugs which are in clinical development, and finally innovative drug targets and their mechanisms in heart failure which are emerging from preclinical studies will be discussed.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
50
|
Abstract
cGMP controls many cellular functions ranging from growth, viability, and differentiation to contractility, secretion, and ion transport. The mammalian genome encodes seven transmembrane guanylyl cyclases (GCs), GC-A to GC-G, which mainly modulate submembrane cGMP microdomains. These GCs share a unique topology comprising an extracellular domain, a short transmembrane region, and an intracellular COOH-terminal catalytic (cGMP synthesizing) region. GC-A mediates the endocrine effects of atrial and B-type natriuretic peptides regulating arterial blood pressure/volume and energy balance. GC-B is activated by C-type natriuretic peptide, stimulating endochondral ossification in autocrine way. GC-C mediates the paracrine effects of guanylins on intestinal ion transport and epithelial turnover. GC-E and GC-F are expressed in photoreceptor cells of the retina, and their activation by intracellular Ca(2+)-regulated proteins is essential for vision. Finally, in the rodent system two olfactorial GCs, GC-D and GC-G, are activated by low concentrations of CO2and by peptidergic (guanylins) and nonpeptidergic odorants as well as by coolness, which has implications for social behaviors. In the past years advances in human and mouse genetics as well as the development of sensitive biosensors monitoring the spatiotemporal dynamics of cGMP in living cells have provided novel relevant information about this receptor family. This increased our understanding of the mechanisms of signal transduction, regulation, and (dys)function of the membrane GCs, clarified their relevance for genetic and acquired diseases and, importantly, has revealed novel targets for therapies. The present review aims to illustrate these different features of membrane GCs and the main open questions in this field.
Collapse
Affiliation(s)
- Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|